eISSN: 1509-572x
ISSN: 1641-4640
Folia Neuropathologica
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2023
vol. 61
 
Share:
Share:
Original paper

The role of synthetic ligand of PPARα in regulation of transcription of genes related to mitochondria biogenesis and dynamic in an animal model of Alzheimer’s disease

Sylwia Żulińska
1
,
Anna K. Strosznajder
2
,
Joanna B. Strosznajder
1

1.
Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
2.
University Teaching Hospital in Białystok, Poland
Folia Neuropathol 2023; 61 (2): 138-143
Online publish date: 2023/07/14
Article file
- The role (1).pdf  [0.11 MB]
Get citation
 
PlumX metrics:
 

Introduction

Alzheimer’s disease (AD) is a neurodegenerative disorder that causes the most severe dementia. During the last 30 years, the amyloid β (Aβ) hypothesis dominated in the research of AD. Despite many years of studies, the aetiology and pathomechanism of the sporadic form of AD are not elucidated and the therapeutic strategy is unsuccessful. It is widely accepted that sporadic, late-onset AD (LOAD) which attacks more than 90% of AD patients is a multifactorial disease in which environmental and genetic factors play an important role. For the past decade, extensive research has been dedicated to studying the genetics of AD [1,13]. The familial form of early-onset AD (EOAD) is primarily associated with mutations in genes encoding amyloid precursor protein (APP) or secretases responsible for APP metabolism. Recently genome-wide association studies (GWAS) have discovered many genetic loci connected with a risk of LOAD. Among them is the gene encoding peroxisome proliferator-activated receptor α (PPARα) involved in fatty acids and cholesterol metabolism [4,26]. PPARα belongs to the nuclear, non-steroid PPARs family together with receptor PPARβ/δ and receptor PPARγ, which are encoded by the corresponding genes: NR1C1, NR1C2, NR1C3 [8,32,37]. During the last decades, much data demonstrated the role of PPARs in mood alterations, depression and in other neuropsychiatric disorders and suggested their engagement in AD pathology [9,29,37]. PPARα and other members of this receptor family dimerize with retinoid X receptor (RXR). These dimers bind to PPAR-responsive regulatory elements (PPRE) and are engaged in the regulation of transcription of genes. However, a coactivator of PPARs, PGC-1α, is necessary for the activation of RXR:PPAR but at first the transcriptional corepressor must be removed as described previously [11,20].
The role of PPARα in the physiology and pathology of central nervous system (CNS) and the association between fatty acids and other lipids metabolism and AD was recently widely demonstrated and suggested [14,31,37]. It was reported that regulation of PPARα, which is involved in lipid, glucose, energy metabolism and inflammation, improves also synaptic plasticity in the AD mouse model [3,28,29]. During the last decade, the significant role of mitochondria alterations in AD pathogenesis was postulated [5,18,22,33,34,36]. The study of Zolezzi et al. [40] underlined that PPARα and PPARγ agonists (ciglitazone and WY14.643) through modulation of mitochondria fission-fusion may exert a neuroprotective effect against oxidative stress and could efficiently slow the progression of AD. Previous studies have also indicated that ligands targeting PPARα, as well as PPARγ or PPARβ/δ receptors simultaneously, could potentially offer enhanced therapeutic efficacy in the treatment of type 2 diabetes with obesity and neurodegenerative/neuropsychiatric disorders [6,10,21,38]. Additionally, it has been demonstrated that PPARγ agonists and PGC-1α activators can improve cognitive deficits, alleviate oxidative stress, and reduce inflammation in animal models of Parkinson’s disease [2].
Our previous study has shown significant changes in the transcription of genes related to mitochondria and anti-oxidative defence in an experimental model of AD [7,37]. Furthermore, last data of Qu et al. [27] indicate a neuroprotective effect of a PPAR ligand, GW7647 in an ADTg mouse model. These data suggest that activation of PPAR ameliorated iron homeostasis alteration in the brain of APP/PS1 mice and suppressed inflammation processes and lipid oxidation by activation of transcription of glutathione peroxidase 4 [27].
In our study, we investigated the impact of GW7647, a synthetic ligand of PPARα, on the mRNA expression of genes encoding proteins involved in mitochondria biogenesis and dynamics. The study was conducted in the brain cortex of 12-month-old ADTg mice (APP+) and we compared the results to age-matched control mice without the transgene (APP).

Material and methods

Animal model of Alzheimer’s disease
In our study, the female FVB-Tg (Thy1; APP LD2/B6) 12-month-old mice were used. These mice overexpressed human AβPP with the “London” V717I mutation under the control of a fragment of Thy1 promoter with specificity towards brain and spinal cord neurons, described as AD Tg or APP+. Mice without the transgene (APP) were used as controls. The mice in this study were extensively characterized to encompass a wide range of electrophysiological, behavioural, and biochemical features associated with AD [19,35]. The behavioural abnormalities begin at 8 weeks of age [19]. Then these mice reservedly developed agitation and cognitive impairment. These all described modifications occurred simultaneously with alterations of neurotransmitters reactivity (3-4 months of age) and then electrophysiological alterations (between 5 and 7 months) [19]. Mice used in our experiments were bred under specific pathogen-free (SPF) conditions in controlled temperature and humidity conditions and a 12-h light/dark cycle in the Animal House of the Mossakowski Medical Research Institute PAS, Warsaw, Poland. Female mice, 12 months old, were administered subcutaneously with a dose of 5 mg/kg body weight with the PPAR agonist GW7647, a cell permeable compound (2-(4-(2-(1-Cyclohexanebutyl)-3-cyclohexylureido)ethyl)phenylthio)-2-methyl­propionic acid). The GW7647 was dissolved in DMSO (3%). APP and APP + GW7647 mice treated with DMSO were used as appropriate controls. The treatment with GW7647 was conducted for a duration of 14 days, after which the mice were euthanized by decapitation. Soon after mice were decapitated, cerebral cortices were quickly isolated on ice and frozen in liquid nitrogen. The protocol was approved by the Warsaw Local Ethics Committee for Animal Experimentation and performed in accordance with the guidelines of the Polish National Ethics Committee and EU Directive 2010/63/EU. All applicable international, national and/or institutional guidelines for the care and use of animals were followed. Stringent measures were taken to minimize any potential suffering and to limit the number of animals utilized in the experiments. The research was conducted in compliance with protocols adhering to good laboratory practice and quality assurance standards.
Analysis of gene expression
RNA was isolated using the TRI reagent (Sigma-Aldrich/Merck) as described in the manufacturer’s protocols and purified by using DNase I according to the manufacturer’s protocols (Sigma-Aldrich/Merck). The concentration and purity of obtained RNA were determined spectrophotometrically (A260/A280). Reverse transcription was performed by using the High-capacity cDNA Reverse Transcription Kit according to the manufacturer’s instructions (Applied Biosystems). The level of mRNA for studied genes was analysed by using TaqMan Gene Expression Assays (Applied Biosystems) as described also by us previously [7].
Quantitative polymerase chain reaction (PCR) was performed on an Applied Biosystems 7500 Real-Time PCR System using TaqMan Gene Expression Master Mix according to the manufacturer’s instructions. The results were normalized against Gapdh and β-actin – Actb and demonstrated as a percentage of the corresponding control. The relative levels of mRNA were calculated using the ΔΔCt Method.
Statistical analysis
The results were expressed as a mean value ± SEM from 3-5 animals in each group. Differences between the means were analysed using Student t-test. Statistical analyses were performed using Graph Pad Prism version 8.0 (Graph Pad Software, San Diego, CA, USA).

Results

In this study we analysed the effect of the novel synthetic ligand of PPARα receptor, GW7647 on the transcription of genes related to mitochondria in the brain cortex of AD Tg mice.
Our findings revealed that GW7647, a novel ligand of the PPARα receptor, had a positive impact on the mRNA levels of genes involved in mitochondrial biogenesis. However, it did not show any significant influence on the expression of genes associated with mitochondrial dynamics. Analysis of our data showed that the mRNA level of Ppargc1 is significantly lower in ADTg mice vs. APP (Fig. 1A). The synthetic ligand enhanced the mRNA level of the gene coding PGC1α (the crucial protein of mitochondrial biogenesis) in APP+ as compared to APP mice (Fig. 1A). However, it is demonstrated that the mRNA level of the Ppara in APP+ is similar as in APP brain cortex of mice treated with GW7647 (Fig. 1B). Moreover, GW7647 remained without effect on the mRNA level of Nrf1 (Fig. 1C) although it significantly activates transcription of Nrf2 (Fig. 1D) and Tfam (Fig. 1E) in the brain cortex of ADTg mice (APP+) comparing to APP treated with GW7647. The following experiments focused on the effect of GW7647 on the transcription of genes coding proteins involved in mitochondrial fusion and fission. In these cases, the effect of GW7647 was compared also to the corresponding control. It was found that GW7647 did not affect the expression of genes coding proteins engaged in fusion such as MFN1, MNF2 and OPA1 (Fig. 2A-C). Moreover, in the case of gene expression coding FIS1 and DRP1 the mRNA levels of these genes were similar in investigated groups (Fig. 2D, E).

Discussion

Our data indicated the beneficial effect of PPARα synthetic agonist, GW7647 in ADTg mice. Previous studies have reported that the activation of PPAR, a nuclear receptor and transcription factor and potent regulator of lipids and energy metabolism, exerts a beneficial effect on synaptic plasticity in animal models of AD [29]. Furthermore, it has been demonstrated that the regulation of PPARα by APP influences the pharmacological modulation of synaptic activity [28]. Synaptic dysfunction, synaptic degeneration and synaptosis were indicated as very early events of AD pathology [24,39]. These alterations, closely associated with mitochondrial function, may occur prior to the onset of typical neuropathological changes and could potentially contribute to cognitive impairment. Moreover, subsequent studies have provided further evidence that dysregulation of lipid metabolism and impaired PPAR function may have a significant impact on the progression of AD [16].
Our data revealed the favourable impact of the PPARα synthetic agonist GW7647 on mitochondrial biogenesis. GW7647 by activating the transcription of genes related to mitochondria may exert a neuroprotective effect. Our results demonstrated elevated mRNA levels of Ppargc1a, Nrf2, and Tfam genes in GW7647-treated AD Tg mice compared to control animals without the transgene (APP + GW7647). These results imply a potentially important role of the nuclear receptor PPARα in AD, as well as the promising neuroprotective effect of its novel synthetic agonist [17,30]. Through the PPARα/PGC-1α pathway, GW7647 has the potential to influence transcription processes and multiple molecular pathways. PGC-1α has been identified as a crucial regulator of several genes involved in oxidative stress, mitochondrial function, neuroinflammation, apoptosis, autophagy, and various other processes related to brain function under both physiological and pathological conditions. PGC-1α belongs to the family of transcriptional coactivators together with PGC-1α and PGC1-related coactivator (PRC). It regulates the transcription of numerous genes, including those encoding nuclear receptors such as all members of the PPARs family and estrogen-related receptors (ERR). It also modulates other transcription factors involved in mitochondrial biogenesis, such as NRF2 and TFAM. The synthetic agonist GW7647 significantly enhances the expression of Ppargc1a and then the transcription of the gene coding NRF2 and TFAM. NRF2 (nuclear factor, erythroid2-related factor 2) is one of the most important molecular regulators engaged in antioxidant defence, calcium and iron homeostasis. NRF2 plays an important role in several signalling pathways in inflammation processes, immunity, and autophagy and in mitochondria [12,23,25]. It was discovered that in mitochondria biogenesis, NRF2 interacts with TFAM in the regulation of mtDNA. Our study indicated that the expression of the gene coding TFAM is activated by GW7647 in the AD Tg brain cortex. TFAM is a mitochondrial DNA binding protein that determines mitochondrial genome maintenance (mtDNA) and which response may have an impact on mitochondria homeostasis [15]. In summary, our findings indicate that GW7647, by activating the transcription of genes involved in mitochondria biogenesis, may have a beneficial effect in an experimental model of AD (Fig. 3). Further studies are currently underway to gain a better understanding of the mechanism underlying GW7647’s action in the brains of AD animal models.

Funding

This work is supported by National Science Centre (PL) Grant 2019/35/N/NZ4/03706.

Disclosure

The authors report no conflict of interest.
References
1. Bellenguez C, Küçükali F, Jansen IE, Kleineidam L, Moreno-Grau S, Amin N, Naj AC, Campos-Martin R, Grenier-Boley B, Andrade V, Holmans PA, Boland A. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nat Genet 2022; 54: 412-436.
2. Botta M, Audano M, Sahebkar A, Sirtori CR, Mitro N, Ruscica M. PPAR agonists and metabolic syndrome: an established role? Int J Mol Sci 2018; 19: 1197.
3. Bougarne N, Weyers B, Desmet SJ, Deckers J, Ray DW, Staels B, Bosscher KD. Molecular actions of PPAR in lipid metabolism and inflammation. Endocr Rev 2018; 39: 760-802.
4. 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 alpha gene influences the risk for Alzheimer’s disease. J Neural Transm (Vienna) 2003; 110: 1041-1050.
5. Camandola S, Mattson MP. Brain metabolism in health, aging, and neurodegeneration. EMBO J 2017; 36: 1474-1492.
6. Cheng HS, Tan WR, Low ZS, Marvalim C, Lee JYH, Tan NS. Exploration and development of PPAR modulators in health and disease: an update of clinical evidence. Int J Mol Sci 2019; 20: 5055.
7. Cieślik M, Czapski GA, Wójtowicz S, Wieczorek I, Wencel PL, Strosznajder RP, Jaber V, Lukiw WJ, Strosznajder JB. Alterations of transcription of genes coding anti-oxidative and mitochondria-related proteins in amyloid  toxicity: Relevance to Alzheimer’s disease. Mol Neurobiol 2020; 57: 1374-1388.
8. Corrales P, Vidal-Puig A, Medina-Gómez G. PPARs and metabolic disorders associated with challenged adipose tissue plasticity. Int J Mol Sci 2018; 19: 2124.
9. D’Agostino G, Cristiano C, Lyons DJ, Citraro R, Russo E, Avagliano C, Russo R, Raso GM, Meli R, De Sarro G, Heisler LK, Calignano A. Peroxisome proliferator-activated receptor alpha plays a crucial role in behavioral repetition and cognitive flexibility in mice. Mol Metab 2015; 4: 528-536.
10. Das SR, Everett BM, Birtcher KK, Brown JM, Januzzi JL, Jr., Kalyani RR, Kosiborod M, Magwire M, Morris PB, Neumiller, Sperling LS. 2020 Expert consensus decision pathway on novel therapies for cardiovascular risk reduction in patients with type 2 diabetes: A report of the American College of Cardiology solution set oversight committee. J Am Coll Cardiol 2020; 76: 1117-1145.
11. Delerive P, Wu Y, Burris TP, Chin WW, Suen CS. PGC-1 functions as a transcriptional coactivator for the retinoid X receptors. J Biol Chem 2002; 277: 3913-3917.
12. Gazaryan IG, Thomas B. The status of Nrf2-based therapeutics: current perspectives and future prospects. Neural Regen Res 2016; 11: 1708-1711.
13. Hardy J, Escott-Price V. Genes, pathways and risk prediction in Alzheimer’s disease. Hum Mol Genet 2019; 28: R235-R240.
14. Jęśko H, Wencel PL, Lukiw WJ, Strosznajder RP. Modulatory effects of fingolimod (FTY720) on the expression of sphingolipid metabolism-related genes in an animal model of Alzheimer’s disease. Mol Neurobiol 2019; 56: 174-185.
15. Kang I, Chu CT, Kaufman BA. The mitochondrial transcription factor TFAM in neurodegeneration: emerging evidence and mechanisms. FEBS Lett 2018; 592: 793-811.
16. Kao YC, Ho PC, Tu YK, Jou IM, Tsai KJ. Lipids and Alzheimer’s disease. Int J Mol Sci 2020; 21: 1505.
17. Lin Y, Wang Y, Li PF. PPAR: An emerging target of metabolic syndrome, neurodegenerative and cardiovascular diseases. Front Endocrinol 2022; 13: 1074911.
18. Monte de la SM, Wands JR. Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer’s disease. J Alzheimers Dis 2006; 9: 167-181.
19. Moechars D, Dewachter I, Lorent K, Reversé D, Baekelandt V, Naidu A, Tesseur I, Spittael K, Haute CV, Checler F, Godaux E, Cordell B, Leuven FV. Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain. J Biol Chem 1999; 274: 6483-6492.
20. Mulholland DJ, Dedhar S, Coetzee GA, Nelson CC. Interaction of nuclear receptors with the Wnt/beta-catenin/Tcf signaling axis: Wnt you like to know? Endocr Rev 2005; 26: 898-915.
21. Nicholls SJ, Uno K. Peroxisome proliferator-activated receptor (PPAR /) agonists as a potential target to reduce cardiovascular risk in diabetes. Diab Vasc Dis Res 2012; 9: 89-94.
22. Oliver D, Reddy PH. Dynamics of dynamin-related protein 1 in Alzheimer’s disease and other neurodegenerative diseases. Cells 2019; 8: 961.
23. Pajares M, Jiménez-Moreno N, García-Yagüe Á J, Escoll M, de Ceballos ML, Van Leuven F, Rábano A, Yamamato M, Rijo AI, Cuadardo A. Transcription factor NFE2L2/NRF2 is a regulator of macroautophagy genes. Autophagy 2016; 12: 1902-1916.
24. Pelucchi S, Gardoni F, Di Luca M, Marcello E. Synaptic dysfunction in early phases of Alzheimer’s disease. Handb Clin Neurol 2022; 184: 417-438.
25. Petsouki E, Cabrera SNS, Heiss EH. AMPK and NRF2: Interactive players in the same team for cellular homeostasis? Free Radic Biol Med 2022; 190: 75-93.
26. Picard M, McEwen BS, Epel ES, Sandi C. An energetic view of stress: Focus on mitochondria. Front Neuroendocrinol 2018; 49: 72-85.
27. Qu XX, He JH, Cui ZQ, Yang T, Sun XH. PPAR- agonist GW7647 protects against oxidative stress and iron deposit via GPx4 in a transgenic mouse model of Alzheimer’s diseases. ACS Chem Neurosci 2022; 13: 207-216.
28. Sáez-Orellana F, Leroy T, Ribeiro F, Kreis A, Leroy K, Lalloyer F, Baugé E, Staels B, Duyckaerts C, Brion JP, Gailly P, Octave JN, Pierrot N. Regulation of PPAR by APP in Alzheimer disease affects the pharmacological modulation of synaptic activity. JCI Insight 2021; 6: e150099.
29. Sáez-Orellana F, Octave JN, Pierrot N. Alzheimer’s disease, a lipid story: involvement of peroxisome proliferator-activated receptor . Cells 2020; 9: 1215.
30. Sagheddu C, Melis M, Muntoni AL, Pistis M. Repurposing peroxisome proliferator-activated receptor agonists in neurological and psychiatric disorders. Pharmaceuticals (Basel) 2021; 14: 1025.
31. Snowden SG, Ebshiana AA, Hye A, An Y, Pletnikova O, O’Brien R, Troncoso J, Legido-Quigley C, Thambisetty M. Association between fatty acid metabolism in the brain and Alzheimer disease neuropathology and cognitive performance: A nontargeted metabolomic study. PLoS Med 2017; 14: e1002266.
32. Strosznajder AK, Wójtowicz S, Jeżyna MJ, Sun GY, Strosznajder JB. Recent insights on the role of PPAR-/ in neuroinflammation and neurodegeneration, and its potential target for therapy. Neuro-molecular Med 2021; 23: 86-98.
33. Swerdlow RH, Burns JM, Khan SM. The Alzheimer’s disease mitochondrial cascade hypothesis: progress and perspectives. Biochim Biophys Acta 2014; 1842: 1219-1231.
34. Swerdlow RH. Mitochondria and mitochondrial cascades in Alzheimer’s disease. J Alzheimers Dis 2018; 62: 1403-1416.
35. Van Dorpe J, Smeijers L, Dewachter I, Nuyens D, Spittaels K, Van Den Haute C, Mercken M, Moechars D, Laenen I, Kuiperi C, Bruynseels K, Tesseur I, Loos R, Vanderstichele H, Checler F, Sciot R, Van Leuven F. Prominent cerebral amyloid angiopathy in transgenic mice overexpressing the london mutant of human APP in neurons. Am J Pathol 2000; 157: 1283-1298.
36. Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Mol Neurodegener 2020; 15: 30.
37. Wójtowicz S, Strosznajder AK, Jeżyna M, Strosznajder JB. 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.
38. Yajima K, Hirose H, Fujita H, Seto Y, Fujita H, Ukeda K, Miyashita K, Kawi T, Yamamoto Y, Ogawa T, Yamada T, Saruta T. Combination therapy with PPARgamma and PPARalpha agonists increases glucose-stimulated insulin secretion in db/db mice. Am J Physiol Endocrinol Metab 2003; 284: E966-971.
39. Zhang H, Jiang X, Ma L, Wei W, Li Z, Chang S, Wen J, Sun J, Li H. Role of A in Alzheimer’s-related synaptic dysfunction. Front Cell Dev Biol 2022; 10: 964075.
40. Zolezzi JM, Inestrosa NC. Peroxisome proliferator-activated receptors and Alzheimer’s disease: hitting the blood-brain barrier. Mol Neurobiol 2013; 48: 438-451.
Copyright: © 2023 Mossakowski Medical Research Centre Polish Academy of Sciences and the Polish Association of Neuropathologists. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.