A Mechanistic approach of Peroxisome Proliferator-Activated Receptors and its subtypes on Clinical and preclinical model of Neurodegenerative disorders
Jinu Avarachan, Anitta Augustine, Pallavi Mahadev Shinde, Venkatesh Gunasekaran*
Department of Pharmacology, KMCH College of Pharmacy, Tamil Nadu Dr MGR Medical University,
Tamil Nadu, Coimbatore, India.
*Corresponding Author E-mail: gvenkatpharma@gmail.com, jinuavarachan333@gmail.com, anittaaugustine95@gmail.com, pallavishinde7200@gmail.com
ABSTRACT:
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors, belonging to the nuclear receptor family, which has high expression of three structurally homologous PPARs isotypes (PPARα, PPARβ/δ, and PPARγ) in brain. Several studies have discovered role of PPARs in oxidative stress, mitochondrial dysfunction, neuroinflammation and production of the toxic proteins in various neurodegenerative disorders such as Parkinson disease, Alzheimer’s disease, Huntington disease, Amyotrophic Lateral Sclerosis, Multiple sclerosis etc. Currently available drugs provide symptomatic relief, but disease progression cannot be stopped, because of their unclear molecular approach. The ability of PPAR to modulate the pathways involved in these conditions paved a path for future studies. Due to increasing challenges to treat central nervous system related disorders, hence PPARs have attracted much attention nowadays. In this review, we discussed various mechanisms of PPARs subtypes in neurodegenerative disorders. We congregate the molecular evidences which support PPARs as a therapeutic target to treat neurodegenerative disorders from preclinical and clinical studies and provide a basis for the potential therapeutic use of PPAR ligands in human diseases.
KEYWORDS: Peroxisome proliferator-activated receptors, Neurodegenerative disorders, Molecular pathways
INTRODUCTION:
PPARs are also expressed in the cardiovascular (endothelial cells, vascular smooth muscle cells, and monocytes/macrophages) system and its ligands have shown a role in different cardiovascular risk factor.5,6,7 Expression of PPARS in the brain cells like neurons and glia, suggested their role in in neuroprotection and neurodegenerative disorders.8,9,10
Structure and Signaling Mechanism of PPAR:
PPARs possess 4 domains. N-terminal A/B domain contains a weak ligand-independent transactivation function called activation function 1 (AF-1). The DNA binding domain (DBD) contains two zinc-fingers that is the particularity of the nuclear receptor (NR) family. Through hinge region, the DBD is connected to the ligand binding domain (LBD). LBD possess a ligand dependent transactivation function activation function 2(AF-2). The LBD comprises a fold made by 12α helices and 4β sheets which forms a large hydrophobic pocket for ligands to bind. LBD serves as site for dimerization and cofactor interaction.11 Binding of a ligand to the receptor changes the dynamic behavior of PPARs helix12,suggesting that helix 12 plays an important role in PPARs activity.12
Expression of different genes through PPARs is controlled by complex with retinoid X receptor (RXR).13 RXR also exist in three isoforms (α, β, and γ,) and are activated by 9-cis retinoic acid(9-cis RA).14,15 RXR binds to various retinoid like natural and synthetic ligands with varying affinity for three RXR isoform.16 Formation of heterodimers with PPARs are permissive and it’s activity is regulated by either PPAR ligand or RXR ligand.17,18 .Heterodimers regarded as a basic functional unit for the signaling grid rather than RXR and PPAR monomer as separate units. During dimerization both LBD and DBD are involved, but the studies on RXR/PPAR heterodimerization have mainly focused on the LBD because of their larger and stronger dimerization interface.19,20
The binding of ligands to the PPARs make changes in the expression level of PPARs target genes encoded mRNAs and is called as ‘transactivation’.21 Binding of a ligand to the LBD of the PPARs changes the confirmation of structure and this binding cause the release of corepressors lead to activation of the receptor. Ligand binding domain get stabilized by releasing of the corepressor which result in the binding of coactivators such as PGC-1𝛼.22 Coreprssors repress target gene transcription23 Coactivators also form multiprotein complex which possess histone acetyltransferase (HAT) activity or methyl transferase activities.24 Some proliferator response elements (PPRE) are present in the promoter region of target genes and are activated by the PPAR/RXR complex. This brings RNA polymerase complex and allows gene transcription. For example, the PPREs influence insulin-sensitive genes, which enhance production of mRNAs of insulin-dependent enzymes.25,26
Physiology of PPARs and Its subtypes:
In hepatocytes, enterocytes, monocytes/macrophages, endothelial cells, smooth muscle cells, lymphocytes and non neuronal cells like microglia and astrocytes, the expression of PPARα is found to be high. It represses proinflammatory responses in different cells such as microglia and astrocytes.27,28 PPARα serves as the master regulator of hepatic lipid metabolism during fasting.29 It activates the gene expression responsible for fatty acid movement and oxidation.30 PPARα is the only isoform that binds to a different types of saturated fatty acids. They are present in tissues such as liver, intestine, kidney, abdominal adipose, and skeletal muscle, tissues and are all involved in aspects of lipid metabolism. Predominantly they are present in brain.32,33 PPAR𝛽/𝛿 can bind to both saturated and unsaturated fatty acid and their binding affinity intermediate between that of PPARα and PPARγ. like Dihomo-γ linolenic acid, arachidonic acid, and EPA and eicosanoids have been shown to activate PPAR𝛽/δ.31,34.PPAR𝛽/𝛿 mRNA and protein expression is high in neuronal cells. PPARδ has been found to decrease the expression of mediators of inflammation and adhesion molecules. Some studies proved that PPARδ ligands have the capability to inhibit cardiac hypertrophy because of their inhibitory activity on NF-κB.35,36 Three isoforms of ppar γ have been detected in human Pparℽ-1, Pparℽ-2 and Pparℽ-3.Pparℽ-1 and 3 code for the same protein while Pparℽ-2 code for a different protein. Pparℽ-1 has broad tissue distribution (heart, large and small intestines, colon, kidney, pancreas, spleen, and skeletal muscle, brain etc) Pparℽ-2 present in adipose tissue only. Pparℽ-3 found only in adipose tissue, macrophages, and colon epithelium.37 PPAR-γincreases the expression of a number of genes code for proteins involved in glucose and lipid metabolism.38 PPAR- γ has the highest expression in CNS (neurons, astrocytes, and glial cells) than other two types.27
Ppar’s and Neurodegenerative Disorders:
Cognitive and Alzheimer’s like disease:
AD is chronic degenerative disease marked by memory loss and inability to function daily life.39 Amyloid cascade hypothesis considered as established paradigm for oxidative stress, neuroinflammation and energy dysmetabolism.40 Among the three isotypes (α, β/δ, γ), the role of PPARγ in AD is the most extensively studied, while evidences on α and β/δ are very less.41 PPARα gene expression was found to be significantly reduced in AD brains. Inhibitors of β-hydroxy β-methylglutaryl-CoA(HMG-CoA) reductase act as ligands of PPARα.statins.42 Binding with Leu331 and Tyr 334 residues of PPARα statins resulted in the upregulation of neurotrophins by means of PPARα mediated transcriptional activation of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) independent of the mevalonate pathway.43 Administration of fenofibrate into APP/PS1 transgenic mice enhanced the expression of PPARα, and decreased beta-site amyloid precursor protein cleaving enzyme 1 (BACE-1) mRNA and protein levels, soluble amyloid precursor protein beta (sAPPβ) and amyloid-β42 (Aβ42) releases and recommended that fenofibrate may reduce the amyloidogenic processing of amyloid precursor protein (APP) in APP/PS1 transgenic mice via PPARα/phosphoinositide 3-kinase(PI3-K) pathway.44 GW7647,another PPARα agonist also reduced the expression of sAPPβ, activity of BACE-1 and Aβ42 discharge in human neuroblastoma SH-SY5Y cells transfected with APPs.This may also involved PI3-K pathway.45 The amyloid plaque diminishing action of PPARα activators (cinnamic acid and gemfibrozil) in the hippocampus in animal model of AD (5XFAD) was demonstrated by Chandra and Pahan. Moreover, they decreased neuroinflammation and thus microglia and astrocytes activation and improved spatial learning and cognition function.46,47 PPARα agonist WY14643 can inhibit the Aβ-stimulated expression of TNFα and Interleukin-6 (IL-6) reporter genes in a dose-dependent manner.49 PPARα knockdown, but not PPARβ or PPARγ, decreased the expression of ADAM10.Gemfibrozil activation of PPARα stimulated ADAM10-mediated proteolysis of APP.50 PPARα activation by Palmitoylethanolamide (PEA) prevents reactive gliosis and neuronal damage in models of Aβ neurotoxicity.51 Wy14643 also prevented DNA damage and neuronal cell apoptosis induced by Aβ toxicity. PPARα activation decreased the expression and translocation of pro apoptotic proteins apoptosis-inducing factor (AIF) and endonuclease G (EndoG) in the NT2N (Post-mitotic neuronal cells) cell model. Brune S reported that PPARα polymorphism may be a risk factor for AD.52
PPAR β/δ has a promoting effect on learning and memory in AD rat model.53 Konttinen H showed for the first time an impairment in fatty acid oxidation in human astrocytes obtained from induced pluripotent stem cells of AD patients. PPARβ/δ agonist GW0742 corrected this impairment by means of regulating an array of genes involved in cellular metabolism, increased hippocampal neurogenesis and enhanced neuronal differentiation of neuronal progenitor cells, prevented Aβ-induced impairment of long-term potentiation in hippocampal slices.54 Chronic and uncontrolled inflammatory activation of microglial cells and the secretion of nuerotoxic chemicals around the plaques serve as major reason for neuronal death in AD. Treatment of GW0742 for a short period in 5XFAD mice demonstrated, it has a prominent effect on inflammation. PPARδ activation resulted in recruitment of microglia to amyloid accumulations and increased the clearance of the plaques.55 In a mouse model of T2DM, it has been exhibited an effect on signal transmission, synaptic plasticity, and spatial memory. It improved hippocampal long-term potentiation and also mediated enhancements in synaptic plasticity and behavior were linked with a significant recovery in hippocampal synaptic transmission.56 Intra-hippocampal infusion of GW0742 in mice reversed Aβ1–42-induced hippocampal PPARδ down-regulation, noticeably improved Aβ1– 42-induced memory deficits. neuroinflammation by decreasing nuclear NF-κB p65, TNF-α, IL-1β and apoptotic responses indicated by decrease in cleaved caspase-3 and increased ratio of B-cell lymphoma 2(Bcl-2)/Baxin in the hippocampus.57Barroso E reported PPARβ/δ deficiency results in cognitive impairment.58 Oral delivery of PPARβ/δ agonist significantly reduced amyloid plaque burden in the subiculum region (primary site for the amyloid plaque deposition) of 5XFAD mice. The changes in plaque burden were accompanied by increased expression of the amyloid degrading enzymes neprilysin and insulin degrading enzyme (IDE).59 Activated PPAR𝛽/𝛿 bind to PPRE found in the promoter region of suppressor of cytokine signaling1 (SOCS1) and it up-regulated the expression of SOCS1.The upregulated SOCS1 inhibits Janus Kinase 2 (JAK2)/STAT1 signalling pathway which will regulates the BACE1 expression.60 The Involvement of PPAR𝛽/𝛿 in neuronal maturation has been reported. This effect was demonstrated on rat cortical neuronal cultures by early increase in expression of microtubule-associated protein-2 (MAP-2), and growth-associated phosphoprotein-43(GAP-43).61
NSAIDs can bind with PPAR-γ thus justifying the role of NSAIDs in the treatment of AD. Heneka established that acute treatment with PPARγ agonist pioglitazone and ibuprofen reduces glial inflammation and Aβ levels in APPV717I transgenic mouse.62 Pioglitazone and a novel selective PPARα/γ modulator, DSP-8658, enhanced the microglial uptake of Aβ in a PPARγ dependent manner.63 Pioglitazone treatment also resulted in the phenotypic polarization of microglial cells from a proinflammatory microglia M1 state, into an anti-inflammatory M2 state, which was associated with enhanced phagocytosis of deposited Aβ. Induction of PPARγ increased microglial phagocytic capacity and reduced Aβ load and inflammation in high fat fed 5XFAD model.64 Honokiol activated PPARγ and improved spatial learning and memory impairment in APPswe/PS1dE9 transgenic mouse model of AD. PPARγ activation down regulated the expression of BACE1 which in turn reduced the production of Aβ.65 Stress due to Aβ decreases heat shock protein 90 β (HSP90β) and afterward diminishes the PPARγ level results in the down-regulation of genes related to Aβ clearance in in BV2 cells and primary microglia. The treatment with a triterpene saponin Jujuboside A (JuA) maintained the stability of PPARγ in an an Axl/ERK-dependent manner, by increasing HSP90β expression in microglia and enhanced Aβ clearance.66 MicroRNAs (miRNAs), a large family of small (20-22 nucleotides) endogenous transcripts can function as key regulators in gene silencing and translational repression.67 PPARγ is a target of miR-128.Yanqiu Liu reported that miR-128 knockout or PPARγ upregulation inhibits AD-like performances, Aβ generation, APP amyloidogenic processing and inflammatory responses.68 The angiotensin II receptor blocker telmisartan has also been reported its protective role in AD partly because of PPARγ activation.69 Neuroprotectin D1’s (NPD1) anti-amyloidogenic effects are mediated in part through activation of the PPARγ receptor.70 A study of dual PPAR β/γ agonist in mild to moderate AD patients reported some limited indications on improving cognition and higher executive function and the data supported next step clinical study of T3D-959 in mild-to-moderate AD.71
Amyotrophic lateral sclerosis (ALS):
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease, is characterized by the degeneration of both upper and lower motor neurons, which leads to muscle weakness and eventual paralysis. Valeria proposed that during ALS progression accumulation of lipid peroxidation products at critical concentrations lead to the activation of PPARγ in motor neurons as result self-protective mechanisms get triggered by the up-regulation of lipid detoxification enzymes, such as lipoprotein lipase and glutathione S-transferase α-2. PPARγ transcriptional activity limit the damage induced by lipid peroxidation derivatives and may attenuate neurodegeneration.72 The PPARγ agonist pioglitazone treatment in G93A SOD1 transgenic mouse model of ALS improved motor performance, delayed weight loss, attenuated motor neuron loss, and extended survival. It also reduced iNOS, NF-κB, and 3-nitrotyrosine immunoreactivity in the spinal cords of G93A transgenic mice.73 Pioglitazone also protected motor neurons against p38-mediated neuronal death and NF-κB-mediated glial inflammation in ALS transgenic mouse model.74 However, in humans, a placebo-controlled, randomized trial of pioglitazone as adjunct therapy for ALS showed no benefit in increasing survival.75
Huntigton’s disease (HD):
HD is caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene, which encodes a polyglutamine tract in the HTT protein. Progression of HD is associated with mitochondrial dysfunction and may account for the cell-type specificity. Mutated HTT cause transcriptional dysregulation, which may lead to neuronal cell death in the brain.76,77 PGC-1α, (transcriptional dysregulation of PPARγ coactivator–1α) a co-activator that helps to regulate mitochondrial biogenesis and energy production by controlling a chain of transcriptional activities. PGC-1α was found to be upregulated several mouse models of HD and it could recover the motor phenotypes, decrease accumulation of misfolded HTT protein in the CNS and ameliorate neurodegeneration. Mijung Lee found that Beta-Lapachone can activate PGC-1α by increasing the level of Sirt-1(deacetylates PGC-1α) and CREB phosphorylation (predominant regulator of PGC-1α) and alleviated HD associated symptoms.78,79 Down regulation of PPARγ seems to play a vital role in the energy homeostasis dysregulation found in HD. Therefore PPARγ can be considered as a potential therapeutic target for this disease. Treatment with thiazolidinediones (TZD) in mouse model (R6/2) of HD expressesd improvements in progressive weight loss, motor deterioration, formation of mutant HTT aggregates, reduced expression of neuroprotective proteins such as brain-derived neurotrophic factor (BDNF) and Bcl-2. Chronic TZD treatment elevated the availability of the PPARγ protein and normalized the expression of two of its downstream genes (the glucose transporter type 4 and PPARγ coactivator-1 alpha genes).80 Mitochondrial dysfunction and oxidative stress in striatal cells that express wild-type (STHdhQ7/Q7) or mutant (STHdhQ111/Q111) huntingtin protein at physiological levels was prevented by rosiglitazone activation of PPARγ.81 The chronic administration of rosiglitazone rescued BDNF deficiency in the cerebral cortex, prevented PGC-1alpha reduction and increased Sirtuin-6 (Sirt6) protein levels in N171-82Q HD mice brain.82
Pan-PPAR agonist bezafibrate exerted neuroprotective effect in a transgenic mouse model of Huntington’s disease. It increased the levels of PGC-1α, PPARs and downstream genes, attenuated astrogliosis and neuronal atrophy in the striatum and increased the numbers of mitochondria. Bezafibrate also rescued lipid accumulation of brown adipose tissue in the HD mice.83 Activation of PPAR𝛿 by its agonistKD3010 improved motor function, reduced neurodegeneration and increased survival in mouse model of HD.PPAR𝛿 activation also decreased HTT-induced neurotoxicity in vitro and in medium of spiny-like neurons generated from stem cells derived from individuals with HD, suggesting that PPAR𝛿 activation may be a new area of treatment in HD and related disorders.84 Dickey also conducted a study with RXR agonist bexarotene in various cellular models and N171-82Q HD mouse model, found that it activates PPAR𝛿 and improve motor function, HTT protein aggregation, striatal neurodegeneartion and mouse survival.85
Multiple sclerosis (MS)
MS is a chronic inflammatory disease. Inflammatory response in CNS leads to demyelination and neuronal injury. Self-reactive T cells are believed to initiate the disease. It is categorized under the auto-immune disease. 86,87 The IL-12 family of cytokines such as IL-12, IL-23, and IL-27 play critical roles in T cell differentiation and are important modulators of MS. Jihong Xu suggested that PPARα agonist fenofibrate modulated the development of experimental autoimmune encephalomyelitis (EAE), at least in part, by suppressing the production of IL-12 family cytokines and MyD88 dependent signaling in MS model.88 Activated microglia and astrocytes are believed to contribute to disease pathology. A combination of 9-cis RA and the PPARα agonist fenofibrate or gemfibrozil inhibited, TNF-α, IL-1β, IL-6 and MCP-1 production in LPS-stimulated microglia and astrocytes.[89] Another PPARα agonist gemfibrozil can suppress relapsing remitting multiple sclerosis(RR-MS) increasing the expression of GATA-3 and decreasing the expression of T-bet through the inhibition of NO production.90,91 PPARδ(-/-) mice develop prolonged EAE in with augmented Th1/Th17 responses, suggesting an important physiological role for PPARδ in the remission and recovery of EAE.92 PPAR β/δ agonists can exert protective actions in autoimmune model of demyelinating disease. Selective PPARβ/δ agonist GW0742 reduced clinical symptoms in C57BL/6 mice. GW0742 reduced astroglial and microglial inflammatory activation and IL-1β levels in EAE brain and increased expression of some myelin genes.93 Inhibition of 12/15-lipoxygenase by baicalein induced PPARβ/δ in microglia, and alleviated EAE in mice. Induction of PARβ/δ in the CNS, more specifically in microglia, augmentted CNS inflammation of EAE.94 PPARδ agonists GW501516 and L165041 blocked interferon (IFN)-c and IL-17 production by Th1 and Th17 cells and ameliorated MOGp35-55-induced EAE in C57BL/ 6 mice. It also decreased IL-12 and IL-23 level and an increased IL-4 and IL-10 expression in the CNS and lymphoid organs and hence suggested their use in the treatment of MS.95
Use of RXR specific ligands may be highly effective when combined with PPARγ agonists in MS treatment.96 15d-PGJ2 and ciglitazone decreased IL-12 expression and differentiation of Th1 cells.97 They also showed that heterozygous PPARγ deficient mice demonstrated more severe EAE than wild type mice and more severe EAE developed following treatment with PPARγ antagonists in another studies.98 Himanshu P Raikwar also reported that PPARγ agonists15d-PGJ2 and ciglitazone, inhibited EAE through blocking IL-12 signaling leading to Th1 differentiation and the PPARγ deficient heterozygous mice (PPARγ +/-) or those treated with PPARγ antagonists developed an exacerbated EAE in with an augmented Th1 response.99 15d-PGJ2 also inhibited the proliferation of neural antigen-specific T cells from the spleen of myelin basic protein Ac1–11 TCR-transgenic mice, which spontaneously develop EAE. It also suppressed IFN-γ, IL-10, and IL-4 production by activated lymphocytes.100 Heneka investigated the effects of TZDs- pioglitazone and ciglitazone and the non-TZD PPARγ agonist GW347845 on the function of peripheral blood mononuclear cells (PBMCs) from MS patients and healthy donors, all of these PPARγ agonists decreased phytohemagglutinin (PHA) induced T cell proliferation and production of the cytokines TNF-α and IFN-γ by PBMCs through decreased bcl2 expression and induced apoptosis of activated T.101
Parkinson’s disease (PD):
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disease with multiple motor and non-motor features that contribute to the impairment of health-related quality of life (QOL), the death of dopaminergic neurons from substantia nigra pars compacta (SNPC) of basal ganglia is the central cause of PD.102,103 The first cause of dopamine decrease is the death of dopaminergic neurons in the substantia nigra. It is possible to control progression of neuronal damage in PD by controlling oxidative stress, neuroinflammation and mitochondrial dysfunction induced by ROS generation.104 In brain regions such as striatum, substantia nigra, cortex and hippocampus the expression of PPARγ was found to be high.105 Pioglitazone protected the dopaminergic neurons in the substantia nigra of rats by reducing microglial proliferation and NFκB activation in 6-hydroxydopamine in (6-OHDA) model of PD. Pioglitazone treatment also improved motor impairment and exerted neuroprotective effects by enhancing hippocampal neurogenesis in 6-OHDA-lesioned rats.106,107 Pioglitazone inhibited the enzyme monoamine oxidase-B(MAO-B) activity which prevent conversion of 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) to MPP+ and protected from MPTP-induced neurotoxicity.108 PPARγ activation by pioglitazone also protected neuronal cells by decreasing IκBα induction, blocking NFκB activation, iNOS induction and NO-mediated toxicity in MPTP model of PD.109 Pioglitazone showed some improvement in behavioral deficit on rotenone induced PD in rats and also reversed dopamine deficit in striatum.110 Several studies using rosigliatzone also reported for its beneficial effects on PD. Rosiglitazone prevented enhanced phosphorylation of α-Syn which is a potential drug target and GSK3β at Tyr216 and also decreased αSyn oligomers in dopaminergic neurons in seipin-nKO mice.111,112 Eduardo Maria Normand showed that liposome encapsulated rosiglitazone enhanced neuroprotective effect on the retina and CNS in a rotenone-induced model of PD.113 PPARγ activation by rosiglitazone increases mitochondrial biogenesis, oxygen consumption and suppresses free radical generation and autophagy in sporadic and PTEN-induced kinase 1associated PD models.114 Another PPARγ agonist MDG548 attenuated increased microgliosis in MPTP induced PD model of mice. MDG548 has higher receptor affinity and BBB permeability than TZD.115 Pioglitazone also reduced microglial activation and the levels of inflammatory mediators. In dopaminergic neurons it reduced nitrotyrosine expression and also significantly reducd the numbers of activated astrocytes in the striatum and substantia nigra regions of the brain. Pioglitazone protected dopaminergic neurons against LPS insult by inhibiting iNOS expression and NO generation, which is potentially mediated through inhibition of p38 MAPK activity.116 Brage Brakedal conducted a retrospective clinical study and reported that the use of glitazones is associated with a decreased risk of incident PD in populations with diabetes.117
PPARδ subtype is highly expressed in hypothalamus and plays an important role maintaining homeostasis in the brain. PPARδ agonists L-165041 and GW-501516 inhibited caspase-3 activation and protected SH-SY5Y neuroblastoma cells from MPP+ mediated apoptosis and also reduced MPTP-induced dopamine depletion and its metabolites levels in the striatum region of the brain.118 GW501516 also protected dopaminergic neurons against degeneration which reduces endoplasmic reticulum stress by IRE1α-caspase-12-mediated pathway in rotenone rat model of PD.119 Another PPAR𝛽/𝛿 agonist GW0742 plays a role to reduce cognitive impairment associated with PD by restoring tyrosine hydroxylase levels in the brain.120 PPAR𝛼 agonist fenofibrate, a widely used lipid lowering agent prevents dopaminergic cell loss in the SNPC and improved the striatal loss of tyrosine hydroxylase immunoreactivity in the MPTP-induced parkinsonism model of mice.121 and reduced oxidative stress induced by MPTP and was confirmed by measuring the levels of glutathione peroxidas (Increased), superoxide dismutase(decreased) and hydroperoxide lipid (decreased).122
CONCLUSION:
Many in vitro and in vivo studies have undergone to demonstrate the role of Peroxisome proliferator-activated receptors (PPAR) as a therapeutic target to treat various disorders. Role of three types of PPARS (PPARα, PPARβ, PPARγ) in neurodegenerative disorders such as Parkinson disease, Alzheimer’s disease, Huntington disease, Amyotrophic Lateral Sclerosis, Multiple sclerosis etc. have been studied widely. Currently available drugs provide symptomatic relief, but disease progression cannot be stopped. So drugs which can inhibit the disease progression are needed. Findings obtained from the various studies suggested that PPAR agonists can regulate several molecular pathways involved in these conditions and reduce disease progression. Current progresses of knowledge regarding the specific biological activity of PPARs in these conditions provide a foundation for the potential therapeutic use of PPAR ligands in human diseases.
CONFLICT OF INTEREST:
Authors declare no conflict of research interests.
ACKNOWLEDGEMENT:
No funding support for this study. Thanks for the KMCH College of Pharmacy and Management for the free journal access facilities.
REFERENCES:
1. Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun. 2017;483(4): 1166-1177.
2. Warden A, Truitt J, Merriman M, Ponomareva O, Jameson K, Ferguson LB, Mayfield RD, Harris RA. Localization of PPAR isotypes in the adult mouse and human brain. Sci Rep. 2016;6: 27618.
3. Oliveira AC, Bertollo CM, Rocha LT, Nascimento EB Jr, Costa KA, Coelho MM. Antinociceptive and antiedematogenic activities of fenofibrate, an agonist of PPAR alpha, and pioglitazone, an agonist of PPAR gamma. Eur J Pharmacol. 2007;561(1-3): 194-201.
4. Merlin NJ, Nair CC, Dharan SS. Peroxisome Proliferator-Activated Receptors (PPARs)–A Review. Asian Journal of Research in Pharmaceutical Science. 2014;4(1): 32-7.
5. Mirza AZ, Althagafi II, 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.
6. Marion-Letellier R, Savoye G, Ghosh S. Fatty acids, eicosanoids and PPAR gamma. Eur J Pharmacol. 2016 Aug 15;785: 44-49.
7. Chigurupati S, Dhanaraj SA, Balakumar P. A step ahead of PPARγ full agonists to PPARγ partial agonists: therapeutic perspectives in the management of diabetic insulin resistance. Eur J Pharmacol. 2015; 755:50-7.
8. Jiang Q, Heneka M, Landreth GE. The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimer's disease: therapeutic implications. CNS Drugs. 2008;22(1): 1-14.
9. Escribano L, Simón AM, Pérez-Mediavilla A, Salazar-Colocho P, Del Río J, Frechilla D. Rosiglitazone reverses memory decline and hippocampal glucocorticoid receptor down-regulation in an Alzheimer's disease mouse model. Biochem Biophys Res Commun. 2009; 379(2): 406-10.
10. Bordet R, Ouk T, Petrault O, Gelé P, Gautier S, Laprais M, Deplanque D, Duriez P, Staels B, Fruchart JC, Bastide M. PPAR: a new pharmacological target for neuroprotection in stroke and neurodegenerative diseases. Biochem Soc Trans.2006;34(Pt 6): 13416.
11. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, Milburn MV. Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature. 1998; 395(6698): 137-43.
12. Kallenberger BC, Love JD, Chatterjee VK, Schwabe JW. A dynamic mechanism of nuclear receptor activation and its perturbation in a human disease. Nat Struct Biol. 2003 Feb;10(2): 136-40.
13. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev. 1999;20(5): 649-88.
14. Mangelsdorf DJ, Borgmeyer U, Heyman RA, Zhou JY, Ong ES, Oro AE, Kakizuka A, Evans RM. Characterization of three RXR genes that mediate the action of 9-cis retinoic acid. Genes Dev. 1992;6(3): 329-44.
15. Chandra V, Huang P, Hamuro Y, Raghuram S, Wang Y, Burris TP, Rastinejad F. Structure of the intact PPAR-gamma-RXR- nuclear receptor complex on DNA. Nature.2008;456(7220): 350-6.
16. Bissonnette RP, Brunner T, Lazarchik SB, Yoo NJ, Boehm MF, Green DR, Heyman RA. 9-cis retinoic acid inhibition of activation-induced apoptosis is mediated via regulation of fas ligand and requires retinoic acid receptor and retinoid X receptor activation. Mol Cell Biol. 1995;15(10): 5576-85.
17. Schulman IG, Li C, Schwabe JW, Evans RM. The phantom ligand effect: allosteric control of transcription by the retinoid X receptor. Genes Dev. 1997;11(3): 299-308.
18. Westin S, Kurokawa R, Nolte RT, Wisely GB, McInerney EM, Rose DW, Milburn MV,Rosenfeld MG, Glass CK. Interactions controlling the assembly of nuclear- receptor heterodimers and co-activators. Nature. 1998;395(6698): 199-202.
19. Vivat-Hannah V, Bourguet W, Gottardis M, Gronemeyer H. Separation of retinoid X receptor homo- and heterodimerization functions. Mol Cell Biol. 2003;23(21): 7678-88.
20. Bourguet W, Ruff M, Chambon P, Gronemeyer H, Moras D. Crystal structure of the ligand-binding domain of the human nuclear receptor RXR-alpha. Nature. 1995;375(6530): 377-82.
21. Kliewer SA, Umesono K, Noonan DJ, Heyman RA, Evans RM. Convergence of 9-cis retinoic acid and peroxisome proliferator signalling pathways through heterodimer formation of their receptors. Nature. 1992;358(6389): 771-4.
22. Mattson MP. Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab. 2012;16(6): 706-22.
23. Guan HP, Ishizuka T, Chui PC, Lehrke M, Lazar MA. Corepressors selectively control the transcriptional activity of PPARgamma in adipocytes. Genes Dev. 2005;19(4): 453-61.
24. Xu L, Glass CK, Rosenfeld MG. Coactivator and corepressor complexes in nuclear receptor function. Curr Opin Genet Dev. 1999;9(2): 140-7.
25. Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab. 2005;1(6): 361-70.
26. Krishna PN, Mohite YM. Insilico Activity Prediction of Thiazolidinediones Derivatives. Asian Journal of Pharmaceutical Analysis. 2018;8(1): 39-44.
27. Moreno S, Farioli-Vecchioli S, Cerù MP. Immunolocalization of peroxisomeproliferator-activated receptors and retinoid X receptors in the adult rat CNS.Neuroscience. 2004;123(1): 131-45.
28. Xu J, Storer PD, Chavis JA, Racke MK, Drew PD. Agonists for the peroxisomeproliferator-activated receptor-alpha and the retinoid X receptor inhibit inflammatory responses of microglia. J Neurosci Res. 2005;81(3): 403-11.
29. Kersten S, Stienstra R. The role and regulation of the peroxisome proliferator activated receptor alpha in human liver. Biochimie. 2017;136: 75-84.
30. Roy A, Jana M, Corbett GT, Ramaswamy S, Kordower JH, Gonzalez FJ, Pahan K.Regulation of cyclic AMP response element binding and hippocampal plasticity-related genes by peroxisome proliferator-activated receptor α. Cell Rep. 2013;4(4): 724-37.
31. Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, Sternbach DD, Lehmann JM, Wisely GB, Willson TM, Kliewer SA, Milburn MV. Molecular recognition of fatty acids by peroxisome proliferator-activated receptors. Mol Cell. 1999; 3(3): 397-403.
32. Auboeuf D, Rieusset J, Fajas L, Vallier P, Frering V, Riou JP, Staels B,Auwerx J, Laville M, Vidal H. Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-alpha in humans: no alteration in adipose tissue of obese and NIDDM patients. Diabetes. 1997;46(8): 1319-27.
33. Kalinin S, Richardson JC, Feinstein DL. A PPARdelta agonist reduces amyloid burden and brain inflammation in a transgenic mouse model of Alzheimer's disease. Curr Alzheimer Res. 2009; 6(5): 431-7.
34. Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc Natl Acad Sci U S A. 1997; 94(9): 4312-7.
35. Graham TL, Mookherjee C, Suckling KE, Palmer CN, Patel L. The PPARdelta agonist GW0742X reduces atherosclerosis in LDLR(-/-) mice. Atherosclerosis. 2005; 181(1): 29-37.
36. Balakumar P, Rose M, Ganti SS, Krishan P, Singh M. PPAR dual agonists: are they opening Pandora's Box? Pharmacol Res. 2007; 56(2) :91-8.
37. Fajas L, Auboeuf D, Raspé E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, Vidal-Puig A, Flier J, Briggs MR, Staels B, Vidal H, Auwerx J. The organization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem. 1997;272(30): 18779-89.
38. Balakumar P, Rose M, Ganti SS, Krishan P, Singh M. PPAR dual agonists: are they opening Pandora's Box? Pharmacol Res. 2007; 56(2): 91-8.
39. Sanmugam K. Depression is a risk factor for Alzheimer disease-review. Research Journal of Pharmacy and Technology. 2015; 8(8): 1056-8.
40. Galimberti D, Scarpini E. Pioglitazone for the treatment of Alzheimer's disease. Expert OpinInvestig Drugs. 2017; 26(1): 97-101.
41. D'Orio B, Fracassi A, Ceru MP, Moreno S. Targeting PPARalpha in Alzheimer's disease. Current Alzheimer Research. 2018; 15(4): 345-54.
42. Roy A, Jana M, Kundu M, Corbett GT, Rangaswamy SB, Mishra RK, Luan CH, Gonzalez FJ, Pahan K. HMG-CoA Reductase Inhibitors Bind to PPARα to Upregulate Neurotrophin Expression in the Brain and Improve Memory in Mice. Cell Metab.2015; 22(2): 253-65.
43. Lekshmi RS, Shanmugasundaram P. Neuroprotective Properties of Statins. Research Journal of Pharmacy and Technology. 2018;11(8): 3581-4.]
44. Zhang H, Gao Y, Qiao PF, Zhao FL, Yan Y. Fenofibrate reduces amyloidogenic processing of APP in APP/PS1 transgenic mice via PPAR-α/PI3-K pathway. Int J Dev Neurosci. 2014; 38: 223-31.
45. Zhang H, Gao Y, Qiao PF, Zhao FL, Yan Y. PPAR-α agonist regulates amyloid-β generation via inhibiting BACE-1 activity in human neuroblastoma SH-SY5Y cells transfected with APPswe gene. Mol Cell Biochem. 2015;408(1-2): 37-46.
46. Subamalani S, Sasikumar A, Vijayaragavan R, Senthilkumar S, Kumar SM, Raj LS, Kannan I. Effect of Acorus calamus Linn on histomorphometric changes in the CA1 and CA3 regions of Hippocampus in Wistar Albino rats. Research Journal of Pharmacy and Technology. 2019;12(7): 3531-6.
47. Koo JH, Kwon IS, Kang EB, Lee CK, Lee NH, Kwon MG, Cho IH, Cho JY. Neuroprotective effects of treadmill exercise on BDNF and PI3-K/Akt signaling pathway in the cortex of transgenic mice model of Alzheimer's disease. J Exerc Nutrition Biochem. 2013;17(4): 151-60.
48. Chandra S, Pahan K. Gemfibrozil, a Lipid-Lowering Drug, Lowers Amyloid Plaque Pathology and Enhances Memory in a Mouse Model of Alzheimer’s Disease via Peroxisome Proliferator-Activated Receptor α. Journal of Alzheimer's disease reports. 2019;3(1): 149-68.
49. Combs CK, Bates P, Karlo JC, Landreth GE. Regulation of beta-amyloid stimulated proinflammatory responses by peroxisome proliferator-activated receptor alpha. Neurochem Int. 2001; 39(5-6):449-57.
50. Corbett GT, Gonzalez FJ, Pahan K. Activation of peroxisome proliferator-activated receptor α stimulates ADAM10-mediated proteolysis of APP. Proc NatlAcad Sci U S A. 2015;112(27):8445-50.
51. Scuderi C, Valenza M, Stecca C, Esposito G, Carratù MR, Steardo L. Palmitoylethanolamide exerts neuroprotective effects in mixed neuroglialcultures and organotypic hippocampal slices via peroxisome proliferator-activated receptor-α. J Neuroinflammation. 2012;9:49.
52. Brune S, Kölsch H, Ptok U, Majores M, Schulz A, Schlosser R, Rao ML, Maier W, Heun R. Polymorphism in the peroxisome proliferator-activated receptor alphagene influences the risk for Alzheimer's disease. J Neural Transm (Vienna). 2003Sep;110(9):1041-50.
53. Khorasani A, Abbasnejad M, Esmaeili-Mahani S. Phytohormone abscisic acid ameliorates cognitive impairments in streptozotocin-induced rat model of Alzheimer's disease through PPARβ/δ and PKA signaling. Int J Neurosci. 2019;129(11):1053-1065.
54. Konttinen H, Gureviciene I, Oksanen M, Grubman A, Loppi S, Huuskonen MT, Korhonen P, Lampinen R, Keuters M, Belaya I, Tanila H, Kanninen KM, Goldsteins G, Landreth G, Koistinaho J, Malm T. PPARβ/δ-agonist GW0742 ameliorates dysfunction in fatty acid oxidation in PSEN1ΔE9 astrocytes. Glia. 2019;67(1):146-159.
55. Malm T, Mariani M, Donovan LJ, Neilson L, Landreth GE. Activation of thenuclear receptor PPARδ is neuroprotective in a transgenic mouse model of Alzheimer's disease through inhibition of inflammation. J Neuroinflammation. 2015;12:7.
56. Abdel-Rahman EA, Bhattacharya S, Buabeid M, Majrashi M, Bloemer J, Tao YX, Dhanasekaran M, Escobar M, Amin R, Suppiramaniam V. PPAR-δ Activation Ameliorates Diabetes-Induced Cognitive Dysfunction by Modulating Integrin-linked Kinase and AMPA Receptor Function. J Am Coll Nutr. 2019;38(8):693-702.
57. An YQ, Zhang CT, Du Y, Zhang M, Tang SS, Hu M, Long Y, Sun HB, Hong H. PPARδ agonist GW0742 ameliorates Aβ1-42-induced hippocampal neurotoxicity in mice. Metab Brain Dis. 2016;31(3):663-71.
58. Barroso E, del Valle J, Porquet D, Vieira Santos AM, Salvadó L, Rodríguez- Rodríguez R, Gutiérrez P, Anglada-Huguet M, Alberch J, Camins A, Palomer X, Pallàs M, Michalik L, Wahli W, Vázquez-Carrera M. Tau hyperphosphorylation and increased BACE1 and RAGE levels in the cortex of PPARβ/δ-null mice. Biochim Biophys Acta. 2013;1832(8):1241-8.
59. Kalinin S, Richardson JC, Feinstein DL. A PPARdelta agonist reduces amyloidburden and brain inflammation in a transgenic mouse model of Alzheimer's disease. Curr Alzheimer Res. 2009;6(5):431-7.
60. Lee WJ, Ham SA, Lee GH, Choi MJ, Yoo H, Paek KS, Lim DS, Hong K, Hwang JS,Seo HG. Activation of peroxisome proliferator-activated receptor delta suppresses BACE1 expression by up-regulating SOCS1 in a JAK2/STAT1-dependent manner. J Neurochem. 2019;151(3):370-385..
61. Benedetti E, Di Loreto S, D'Angelo B, Cristiano L, d'Angelo M, Antonosante A, Fidoamore A, Golini R, Cinque B, Cifone MG, Ippoliti R, Giordano A, Cimini A. The PPARβ/δ Agonist GW0742 Induces Early Neuronal Maturation of Cortical Post- Mitotic Neurons: Role of PPARβ/δ in Neuronal Maturation. J Cell Physiol. 2016;231(3):597-606.
62. Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C,O'Banion K, Klockgether T, Van Leuven F, Landreth GE. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain. 2005;128(Pt 6):1442-53.
63. Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT. PPARγ/RXRα-induced and CD36-mediated microglial amyloid-β phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci. 2012;32(48):17321-31.
64. Medrano-Jiménez E, Carrillo IJ, Pedraza-Escalona M, Ramírez-Serrano CE, Álvarez-Arellano L, Cortés-Mendoza J, Herrera-Ruiz M, Jiménez-Ferrer E, Zamilpa A, Tortoriello J, Pedraza-Alva G. Malvaparviflora extract ameliorates the deleterious effects of a high fat diet on the cognitive deficit in a mouse model of Alzheimer’s disease by restoring microglial function via a PPAR-γ-dependent mechanism. Journal of neuroinflammation. 2019;16(1):143.
65. Wang D, Dong X, Wang C. Honokiol ameliorates amyloidosis and neuroinflammation and improves cognitive impairment in Alzheimer’s disease transgenic mice. Journal of Pharmacology and Experimental Therapeutics. 2018;366(3):470-8.
66. Zhang M, Qian C, Zheng ZG, Qian F, Wang Y, Thu PM, Zhang X, Zhou Y, Tu L, LiuQ, Li HJ, Yang H, Li P, Xu X. Jujuboside A promotes Aβ clearance and ameliorates cognitive deficiency in Alzheimer's disease through activating Axl/HSP90/PPARγpathway. Theranostics. 2018;8(15):4262-4278.
67. Vishnoi A, Rani S. MiRNA Biogenesis and Regulation of Diseases: An Overview.MethodsMol Biol. 2017;1509:1-10,Ullah S, John P, Bhatti A. MicroRNAs with a role in gene regulation and in human diseases. MolBiol Rep. 2014;41(1):225-32.
68. Liu Y, Zhang Y, Liu P, Bai H, Li X, Xiao J, Yuan Q, Geng S, Yin H, Zhang H, Wang Z, Li J, Wang S, Wang Y. MicroRNA-128 knockout inhibits the development of Alzheimer's disease by targeting PPARγ in mouse models. Eur J Pharmacol. 2019;843:134-144.
69. Tsukuda K, Mogi M, Iwanami J, Min LJ, Sakata A, Jing F, Iwai M, Horiuchi M. Cognitive deficit in amyloid-beta-injected mice was improved by pretreatment with a low dose of telmisartan partly because of peroxisome proliferator- activated receptor-gamma activation. Hypertension. 2009;54(4):782-7.
70. Zhao Y, Calon F, Julien C, Winkler JW, Petasis NA, Lukiw WJ, Bazan NG. Docosahexaenoic acid-derived neuroprotectin D1 induces neuronal survival via secretase- and PPARγ-mediated mechanisms in Alzheimer's disease models. PLoSOne. 2011;6(1):e15816.
71. 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(3):1085-1103.
72. Benedusi V, Martorana F, Brambilla L, Maggi A, Rossi D. The peroxisome proliferator-activated receptor γ (PPARγ) controls natural protective mechanisms against lipid peroxidation in amyotrophic lateral sclerosis. J Biol Chem. 2012;287(43):35899-911.
73. Kiaei M, Kipiani K, Chen J, Calingasan NY, Beal MF. Peroxisome proliferator- activated receptor-gamma agonist extends survival in transgenic mouse model of amyotrophic lateral sclerosis. Exp Neurol. 2005;191(2):331-6.
74. Shibata N, Kawaguchi-Niida M, Yamamoto T, Toi S, Hirano A, Kobayashi M. Effects of the PPARgamma activator pioglitazone on p38 MAP kinase and IkappaBalpha in the spinal cord of a transgenic mouse model of amyotrophic lateral sclerosis. Neuropathology. 2008;28(4):387-98.
75. Dupuis L, Dengler R, Heneka MT, Meyer T, Zierz S, Kassubek J, Fischer W, Steiner F, Lindauer E, Otto M, Dreyhaupt J. A randomized, double blind, placebo-controlled trial of pioglitazone in combination with riluzole in amyotrophic lateral sclerosis. PloS one. 2012;7(6):e37885.
76. Nance MA, US Huntington Disease Genetic Testing Group. Genetic testing of children at risk for Huntington's disease. Neurology. 1997;49(4):1048-53.).
77. Sharma S, Taliyan R. Transcriptional dysregulation in Huntington’s disease: the role of histone deacetylases. Pharmacological research. 2015;100:157-69.
78. Tsunemi T, Ashe TD, Morrison BE, Soriano KR, Au J, Roque RA, Lazarowski ER, Damian VA, Masliah E, La Spada AR. PGC-1α rescues Huntington’s disease proteotoxicity by preventing oxidative stress and promoting TFEB function. Science translational medicine. 2012;4(142):142ra97-.)
79. Lee M, Ban JJ, Chung JY, Im W, Kim M. Amelioration of Huntington's disease phenotypes by Beta-Lapachone is associated with increases in Sirt1 expression, CREB phosphorylation and PGC-1α deacetylation. PloS one. 2018;13(5).
80. Chiang MC, Chen CM, Lee MR, Chen HW, Chen HM, Wu YS, Hung CH, Kang JJ, Chang CP, Chang C, Wu YR. Modulation of energy deficiency in Huntington's disease via activation of the peroxisome proliferator-activated receptor gamma. Human molecular genetics. 2010;19(20):4043-58.
81. Quintanilla RA, Jin YN, Fuenzalida K, Bronfman M, Johnson GV. Rosiglitazone Treatment Prevents Mitochondrial Dysfunction in Mutant Huntingtin-expressing cells possible role of peroxisome proliferator-activated receptor-γ (pparγ) in the pathogenesis of huntington disease. Journal of biological chemistry. 2008;283(37):25628-37.
82. Jin J, Albertz J, Guo Z, Peng Q, Rudow G, Troncoso JC, Ross CA, Duan W. Neuroprotective effects of PPAR‐γ agonist rosiglitazone in N171‐82Q mouse model of Huntington's disease. Journal of neurochemistry. 2013;125(3):410-9.
83. Johri A, Calingasan NY, Hennessey TM, Sharma A, Yang L, Wille E, Chandra A, Beal MF. Pharmacologic activation of mitochondrial biogenesis exerts widespread beneficial effects in a transgenic mouse model of Huntington's disease. Human molecular genetics. 2012;21(5):1124-37.
84. Dickey AS, Pineda VV, Tsunemi T, Liu PP, Miranda HC, Gilmore-Hall SK, Lomas N, Sampat KR, Buttgereit A, Torres MJ, Flores AL. PPAR-δ is repressed in Huntington's disease, is required for normal neuronal function and can be targeted therapeutically. Nature medicine. 2016;22(1):37.
85. Dickey AS, Sanchez DN, Arreola M, Sampat KR, Fan W, Arbez N, Akimov S, Van Kanegan MJ, Ohnishi K, Gilmore-Hall SK, Flores AL. PPARδ activation by bexarotene promotes neuroprotection by restoring bioenergetic and quality control homeostasis. Science translational medicine. 2017 Dec 6;9(419):eaal2332.
86. Stinissen P, Medaer R, Raus J. Myelin reactive T cells in the autoimmunepathogenesis of multiple sclerosis. Mult Scler. 1998;4(3):203-11.
87. Varshney P, Saini P. An Overview of DRF in the treatment of Multiple Sclerosis. Research Journal of Pharmacy and Technology. 2020;13(6):2992-6.
88. Xu J, Racke MK, Drew PD. Peroxisome proliferator-activated receptor-alpha agonist fenofibrate regulates IL-12 family cytokine expression in the CNS:relevance to multiple sclerosis. J Neurochem. 2007;103(5):1801-10.
89. Xu J, Chavis JA, Racke MK, Drew PD. Peroxisome proliferator-activated receptor-alpha and retinoid X receptor agonists inhibit inflammatory responses of astrocytes. J Neuroimmunol. 2006;176(1-2):95-105.
90. Dasgupta S, Roy A, Jana M, Hartley DM, Pahan K. Gemfibrozil ameliorates relapsing-remitting experimental autoimmune encephalomyelitis independent of peroxisome proliferator-activated receptor-alpha. Mol Pharmacol. 2007;72(4):934-46.
91. Balakrishnan N, Panda AB, Raj NR, Shrivastava A, Prathani R. The evaluation of nitric oxide scavenging activity of Acalypha indica Linn root. Asian Journal of Research in Chemistry. 2009;2(2):148-50.
92. Kanakasabai S, Walline CC, Chakraborty S, Bright JJ. PPARδ deficient mice develop elevated Th1/Th17 responses and prolonged experimental autoimmune encephalomyelitis. Brain Res. 2011;1376:101-12.
93. Polak PE, Kalinin S, Dello Russo C, Gavrilyuk V, Sharp A, Peters JM, Richardson J, Willson TM, Weinberg G, Feinstein DL. Protective effects of a peroxisome proliferator-activated receptor-beta/delta agonist in experimental autoimmune encephalomyelitis. J Neuroimmunol. 2005;168(1-2):65-75.
94. Xu J, Zhang Y, Xiao Y, Ma S, Liu Q, Dang S, Jin M, Shi Y, Wan B, Zhang Y. Inhibition of 12/15-lipoxygenase by baicalein induces microglia PPARβ/δ: a potential therapeutic role for CNS autoimmune disease. Cell Death Dis. 2013;4(4):e569.
95. Kanakasabai S, Chearwae W, Walline CC, Iams W, Adams SM, Bright JJ. Peroxisome proliferator-activated receptor delta agonists inhibit T helper type 1 (Th1) and Th17 responses in experimental allergic encephalomyelitis. Immunology. 2010;130(4):572-88.
96. Diab A, Hussain RZ, Lovett-Racke AE, Chavis JA, Drew PD, Racke MK. Ligands for the peroxisome proliferator-activated receptor-gamma and the retinoid X receptor exert additive anti-inflammatory effects on experimental autoimmune encephalomyelitis. J Neuroimmunol. 2004;148(1-2):116-26.
97. Natarajan C, Bright JJ. Peroxisome proliferator-activated receptor-gamma agonists inhibit experimental allergic encephalomyelitis by blocking IL-12 production, IL-12 signaling and Th1 differentiation. Genes Immun. 2002;3(2):59-70.
98. Raikwar HP, Muthian G, Rajasingh J, Johnson C, Bright JJ. PPARgamma antagonists exacerbate neural antigen-specific Th1 response and experimental allergic encephalomyelitis. J Neuroimmunol. 2005;167(1-2):99-107.
99. Raikwar HP, Muthian G, Rajasingh J, Johnson CN, Bright JJ. PPARgamma antagonists reverse the inhibition of neural antigen-specific Th1 response andexperimental allergic encephalomyelitis by Ciglitazone and 15-deoxy- Delta12,14-prostaglandin J2. J Neuroimmunol. 2006;178(1-2):76-86.
100. Diab A, Deng C, Smith JD, Hussain RZ, Phanavanh B, Lovett-Racke AE, Drew PD, Racke MK. Peroxisome proliferator-activated receptor-gamma agonist 15-deoxy- Delta(12,14)-prostaglandin J(2) ameliorates experimental autoimmune encephalomyelitis. J Immunol. 2002;168(5):2508-15.
101. Schmidt S, Moric E, Schmidt M, Sastre M, Feinstein DL, Heneka MT. Anti- inflammatory and antiproliferative actions of PPAR-gamma agonists on T lymphocytes derived from MS patients. J Leukoc Biol. 2004;75(3):478-85.
102. Kahale VP, Upadhay PR, Mhaiskar AJ, Shelat PS, Mundhada DR. To Access the Efficacy of Rutin on 6-Hydroxydopamine induced Animal Model of Memory Impairment in Parkinson's Disease. Research Journal of Pharmacology and Pharmacodynamics. 2013;5(6):331-6,
103. Baul HS, Rajiniraja M. Molecular Docking Studies of Selected Flavonoids on Inducible Nitric Oxide Synthase (INOS) in Parkinson's Disease. Research Journal of Pharmacy and Technology. 2018;11(8):3685-8.
104. Agarwal S, Yadav A, Chaturvedi RK. Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem Biophys Res Commun. 2017;483(4):1166-1177.
105. Moraes LA, Piqueras L, Bishop-Bailey D. Peroxisome proliferator-activated receptors and inflammation. Pharmacology & therapeutics. 2006;110(3):371-85.
106. Machado MM, Bassani TB, Cóppola-Segovia V, Moura EL, Zanata SM, Andreatini R, Vital MA. PPAR-γ agonist pioglitazone reduces microglial proliferation and NF-κB activation in the substantia nigra in the 6-hydroxydopamine model of Parkinson’s disease. Pharmacological reports. 2019;71(4):556-64.
107. Bonato JM, Bassani TB, Milani H, Vital MA, de Oliveira RM. Pioglitazone reduces mortality, prevents depressive-like behavior, and impacts hippocampal neurogenesis in the 6-OHDA model of Parkinson's disease in rats. Experimental neurology. 2018;300:188-200.
108. Chaturvedi RK, Beal MF. PPAR: a therapeutic target in Parkinson’s disease. Journal of neurochemistry. 2008;106(2):506-18.
109. Dehmer T, Heneka MT, Sastre M, Dichgans J, Schulz JB. Protection by pioglitazone in the MPTP model of Parkinson's disease correlates with IκBα induction and block of NFκB and iNOS activation. Journal of neurochemistry. 2004;88(2):494-501.
110. Ulusoy GK, Celik T, Kayir H, Gürsoy M, Isik AT, Uzbay TI. Effects of pioglitazone and retinoic acid in a rotenone model of Parkinson's disease. Brain research bulletin. 2011;85(6):380-4.
111. Wang L, Hong J, Wu Y, Liu G, Yu W, Chen L. Seipin deficiency in mice causes loss of dopaminergic neurons via aggregation and phosphorylation of α-synuclein and neuroinflammation. Cell death & disease. 2018;9(5):1-3.
112. Baul HS, Rajiniraja M. Favorable binding of Quercetin to α-Synuclein as potential target in Parkinson disease: An Insilico approach. Research Journal of Pharmacy and Technology. 2018;11(1):203-6.
113. Normando EM, Davis BM, De Groef L, Nizari S, Turner LA, Ravindran N, Pahlitzsch M, Brenton J, Malaguarnera G, Guo L, Somavarapu S. The retina as an early biomarker of neurodegeneration in a rotenone-induced model of Parkinson’s disease: evidence for a neuroprotective effect of rosiglitazone in the eye and brain. Acta neuropathologica communications. 2016;4(1):86.
114. Corona JC, de Souza SC, Duchen MR. PPARγ activation rescues mitochondrial function from inhibition of complex I and loss of PINK1. Experimental neurology. 2014;253:16-27.
115. Nevin DK, Peters MB, Carta G, Fayne D, Lloyd DG. Integrated virtual screening for the identification of novel and selective peroxisome proliferator-activated receptor (PPAR) scaffolds. Journal of medicinal chemistry. 2012;55(11):4978-89.
116. Xing B, Xin T, Hunter RL, Bing G. Pioglitazone inhibition of lipopolysaccharide-induced nitric oxide synthase is associated with altered activity of p38 MAP kinase and PI3K/Akt. Journal of neuroinflammation. 2008;5(1):4.
117. Brakedal B, Flønes I, Reiter SF, Torkildsen Ø, Dölle C, Assmus J, Haugarvoll K, Tzoulis C. Glitazone use associated with reduced risk of Parkinson's disease. Movement Disorders. 2017;32(11):1594-9.
118. Iwashita A, Muramatsu Y, Yamazaki T, Muramoto M, Kita Y, Yamazaki S, Mihara K, Moriguchi A, Matsuoka N. Neuroprotective efficacy of the peroxisome proliferator-activated receptor δ-selective agonists in vitro and in vivo. Journal of Pharmacology and Experimental Therapeutics. 2007;320(3):1087-96.
119. Tong Q, Wu L, Gao Q, Ou Z, Zhu D, Zhang Y. PPARβ/δ agonist provides neuroprotection by suppression of IRE1α–Caspase-12-mediated endoplasmic reticulum stress pathway in the rotenone rat model of Parkinson’s disease. Molecular neurobiology. 2016;53(6):3822-31.
120. R Das N, P Gangwal R, V Damre M, T Sangamwar A, S Sharma S. A PPAR-β/δ agonist is neuroprotective and decreases cognitive impairment in a rodent model of Parkinson’s disease. Current neurovascular research. 2014;11(2):114-24.
121. Kreisler A, Gelé P, Wiart JF, Lhermitte M, Destée A, Bordet R. Lipid-lowering drugs in the MPTP mouse model of Parkinson's disease: fenofibrate has a neuroprotective effect, whereas bezafibrate and HMG-CoA reductase inhibitors do not. Brain research. 2007;1135:77-84.
122. Barbiero JK, Santiago R, Tonin FS, Boschen S, da Silva LM, de Paula Werner MF, da Cunha C, Lima MM, Vital MA. PPAR-α agonist fenofibrate protects against the damaging effects of MPTP in a rat model of Parkinson's disease. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2014;53:35-44.
Received on 24.06.2020 Modified on 18.09.2020
Accepted on 12.11.2020 © RJPT All right reserved
Research J. Pharm. and Tech. 2021; 14(7):3967-3975.
DOI: 10.52711/0974-360X.2021.00688