1
|
Ju Y, Li S, Kong X, Zhao Q. Exploring fatty acid metabolism in Alzheimer's disease: the key role of CPT1A. Sci Rep 2024; 14:31483. [PMID: 39733087 DOI: 10.1038/s41598-024-82999-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disease, and the most common type of dementia, with symptoms of progressive cognitive dysfunction and behavioral impairment. Studying the pathogenesis of AD and exploring new targets for the prevention and treatment of AD is a very worthwhile challenge. Accumulating evidence has highlighted the effects of fatty acid metabolism on AD. In this study, fatty acid metabolism was used as an entry point to understand the pathogenesis of AD and identify new targets. After identifying differentially expressed genes, multiple machine learning algorithms, carnitine palmitoyltransferase 1 A (CPT1A) was identified as the key gene for fatty acid metabolism in AD. Further single nucleus RNA sequencing analysis were performed, and the GSEA results showed that the fatty acid β-oxidation pathway was enriched only in astrocytes, and the fatty acid β-oxidation pathway was down-regulated in the AD astrocytes compared to the CN astrocytes, while CPT1A was specifically downregulated in astrocytes of AD, which was confirmed in vitro experiment subsequently, and decreased expression level of CPT1A would lead to abnormal lipid metabolism, which shapes astrocyte reactivity and injury, neuroinflammatory, and thus affects AD pathogenesis. Our findings report the involvement of CPT1A in AD. We confirm that the primary role of astrocytes for fatty acid β-oxidation, and CPT1A is localized in astrocytes. Downregulated CPT1A could be a novel potential target for the prevention and treatment of AD. Our study provides strong evidence for the involvement of fatty acid metabolism in the pathogenesis of AD.
Collapse
Affiliation(s)
- Yanxiu Ju
- Department of Neurology, Union Hospital of Jilin University, Changchun, 130000, China
- Engineering Laboratory of Memory and Cognitive Impairment Disease of Jilin Province, Union Hospital of Jilin University, Changchun, 130000, China
| | - Songtao Li
- Department of Neurology, Union Hospital of Jilin University, Changchun, 130000, China
- Engineering Laboratory of Memory and Cognitive Impairment Disease of Jilin Province, Union Hospital of Jilin University, Changchun, 130000, China
| | - Xiangyi Kong
- Key Laboratory of Lymphatic Surgery of Jilin Province, Union Hospital of Jilin University, Changchun, 130000, China
| | - Qing Zhao
- Department of Neurology, Union Hospital of Jilin University, Changchun, 130000, China.
- Engineering Laboratory of Memory and Cognitive Impairment Disease of Jilin Province, Union Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
2
|
Putri AF, Utomo DH, Tunjung WAS, Putri WA. Analysis of the anti-Alzheimer potential of bioactive compounds from Citrus hystrix DC. peel, leaf, and essential oil by network pharmacology. Heliyon 2024; 10:e33496. [PMID: 39050443 PMCID: PMC11267028 DOI: 10.1016/j.heliyon.2024.e33496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/30/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) is the most known neurodegenerative disease, and its prevalence is predicted to increase significantly. Discovering novel drugs and treatments for AD is urgently needed. Drugs from natural products have been preferred lately due to their high potential and low toxicity. Citrus hystrix DC. (kaffir lime; KL) is one such herbal plant that is found abundantly in Southeast Asia with many biological activities. In this study, the potential of bioactive compounds from KL peel, leaf, and essential oil as anti-AD agents was explored using network pharmacology. First, the compounds were identified with KNApSAcK database and related literature. Subsequently, the targets of each corresponding compound were determined with SEA Search Server and Swiss Target Prediction, while the proteins associated with AD were identified using OMIM, GenCLiP3, and DisGeNET. Furthermore, a protein-protein interaction network and a compound-target interaction network were constructed to identify the most crucial proteins and compounds in the network by employing Cytoscape v3.9.1. The study continued with pathway enrichment analysis using STRING v1.7.1, molecular docking with PyRx and SwissDock, and molecular dynamics simulation with YASARA for further confirmation. Our results showed that almost all the secondary metabolites of KL targeted AD-associated genes, with oxypeucedanin and citrusoside A showing the highest anti-AD potential and targeting essential genes, EGFR and MAPK14, respectively. These targets were associated with inflammatory and oxidative stress pathways, indicating the potential mechanism of KL in attenuating AD clinical manifestation.
Collapse
Affiliation(s)
- Adhisa Fathirisari Putri
- Faculty of Biology, Universitas Gadjah Mada, Jl. Teknika Selatan, Sekip Utara, Yogyakarta, 55281, Indonesia
- Bioinformatics Research Center, INBIO-Indonesia, Malang, 65162, Indonesia
| | - Didik Huswo Utomo
- Bioinformatics Research Center, INBIO-Indonesia, Malang, 65162, Indonesia
- Biosystem Education Center, Brawijaya University, Malang, 65145, Indonesia
| | - Woro Anindito Sri Tunjung
- Faculty of Biology, Universitas Gadjah Mada, Jl. Teknika Selatan, Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Wahyu Aristyaning Putri
- Faculty of Biology, Universitas Gadjah Mada, Jl. Teknika Selatan, Sekip Utara, Yogyakarta, 55281, Indonesia
| |
Collapse
|
3
|
Guida F, Iannotta M, Lauritano A, Infantino R, Salviati E, Verde R, Luongo L, Sommella EM, Iannotti FA, Campiglia P, Maione S, Di Marzo V, Piscitelli F. Early biomarkers in the presymptomatic phase of cognitive impairment: changes in the endocannabinoidome and serotonergic pathways in Alzheimer's-prone mice after mTBI. Acta Neuropathol Commun 2024; 12:113. [PMID: 38992700 PMCID: PMC11241935 DOI: 10.1186/s40478-024-01820-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Despite extensive studies on the neurobiological correlates of traumatic brain injury (TBI), little is known about its molecular determinants on long-term consequences, such as dementia and Alzheimer's disease (AD). METHODS Here, we carried out behavioural studies and an extensive biomolecular analysis, including inflammatory cytokines, gene expression and the combination of LC-HRMS and MALDI-MS Imaging to elucidate the targeted metabolomics and lipidomics spatiotemporal alterations of brains from wild-type and APP-SWE mice, a genetic model of AD, at the presymptomatic stage, subjected to mild TBI. RESULTS We found that brain injury does not affect cognitive performance in APP-SWE mice. However, we detected an increase of key hallmarks of AD, including Aβ1-42 levels and BACE1 expression, in the cortices of traumatized transgenic mice. Moreover, significant changes in the expanded endocannabinoid (eCB) system, or endocannabinoidome (eCBome), occurred, including increased levels of the endocannabinoid 2-AG in APP-SWE mice in both the cortex and hippocampus, and N-acylserotonins, detected for the first time in the brain. The gene expression of enzymes for the biosynthesis and inactivation of eCBs and eCB-like mediators, and some of their main molecular targets, also underwent significant changes. We also identified the formation of heteromers between cannabinoid 1 (CB1) and serotonergic 2A (5HT2A) receptors, whose levels increased in the cortex of APP-SWE mTBI mice, possibly contributing to the exacerbated pathophysiology of AD induced by the trauma. CONCLUSIONS Mild TBI induces biochemical changes in AD genetically predisposed mice and the eCBome may play a role in the pathogenetic link between brain injury and neurodegenerative disorders also by interacting with the serotonergic system.
Collapse
Affiliation(s)
- Francesca Guida
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Monica Iannotta
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Anna Lauritano
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy
| | - Rosmara Infantino
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Emanuela Salviati
- Dipartimento di Farmacia, Università Degli Studi di Salerno, Fisciano, (SA), Italy
| | - Roberta Verde
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy
| | - Livio Luongo
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | | | - Fabio Arturo Iannotti
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy
| | - Pietro Campiglia
- Dipartimento di Farmacia, Università Degli Studi di Salerno, Fisciano, (SA), Italy
| | - Sabatino Maione
- Pharmacology Division, Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy.
- Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut sur la Nutrition et les Aliments Fonctionnels, Centre NUTRISS, Université Laval, Quebec City, Canada.
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, (NA), Italy.
| |
Collapse
|
4
|
Liu H, Zhou L, Wang X, Lin Y, Yi P, Xiong Y, Zhan F, Zhou L, Dong Y, Ying J, Wu L, Xu G, Hua F. PIEZO1 as a new target for hyperglycemic stress-induced neuropathic injury: The potential therapeutic role of bezafibrate. Biomed Pharmacother 2024; 176:116837. [PMID: 38815290 DOI: 10.1016/j.biopha.2024.116837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024] Open
Abstract
Hyperglycemic stress can directly lead to neuronal damage. The mechanosensitive ion channel PIEZO1 can be activated in response to hyperglycemia, but its role in hyperglycemic neurotoxicity is unclear. The role of PIEZO1 in hyperglycemic neurotoxicity was explored by constructing a hyperglycemic mouse model and a high-glucose HT22 cell model. The results showed that PIEZO1 was significantly upregulated in response to high glucose stress. In vitro experiments have shown that high glucose stress induces changes in neuronal cell morphology and membrane tension, a key mechanism for PIEZO1 activation. In addition, high glucose stress upregulates serum/glucocorticoid-regulated kinase-1 (SGK1) and activates PIEZO1 through the Ca2+ pool and store-operated calcium entry (SOCE). PIEZO1-mediated Ca2+ influx further enhances SGK1 and SOCE, inducing intracellular Ca2+ peaks in neurons. PIEZO1 mediated intracellular Ca2+ elevation leads to calcium/calmodulin-dependent protein kinase 2α (CaMK2α) overactivation, which promotes oxidative stress and apoptosis signalling through p-CaMK2α/ERK/CREB and ox-CaMK2α/MAPK p38/NFκB p65 pathways, subsequently inducing synaptic damage and cognitive impairment in mice. The intron miR-107 of pantothenic kinase 1 (PANK1) is highly expressed in the brain and has been found to target PIEZO1 and SGK1. The PANK1 receptor is activated by peroxisome proliferator-activated receptor α (PPARα), an activator known to upregulate miR-107 levels in the brain. The clinically used lipid-lowering drug bezafibrate, a known PPARα activator, may upregulate miR-107 through the PPARɑ/PANK1 pathway, thereby inhibiting PIEZO1 and improving hyperglycemia-induced neuronal cell damage. This study provides a new idea for the pathogenesis and drug treatment of hyperglycemic neurotoxicity and diabetes-related cognitive dysfunction.
Collapse
Affiliation(s)
- Hailin Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lian Zhou
- Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Anesthesiology, Ganjiang New Area Hospital of the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xifeng Wang
- Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lanqian Zhou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yao Dong
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lidong Wu
- Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
5
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
6
|
Kwon S, Park KS, Yoon KH. Regulator of Lipid Metabolism NHR-49 Mediates Pathogen Avoidance through Precise Control of Neuronal Activity. Cells 2024; 13:978. [PMID: 38891110 PMCID: PMC11172349 DOI: 10.3390/cells13110978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Precise control of neuronal activity is crucial for the proper functioning of neurons. How lipid homeostasis contributes to neuronal activity and how much of it is regulated by cells autonomously is unclear. In this study, we discovered that absence of the lipid regulator nhr-49, a functional ortholog of the peroxisome proliferator-activated receptor (PPAR) in Caenorhabditis elegans, resulted in defective pathogen avoidance behavior against Pseudomonas aeruginosa (PA14). Functional NHR-49 was required in the neurons, and more specifically, in a set of oxygen-sensing body cavity neurons, URX, AQR, and PQR. We found that lowering the neuronal activity of the body cavity neurons improved avoidance in nhr-49 mutants. Calcium imaging in URX neurons showed that nhr-49 mutants displayed longer-lasting calcium transients in response to an O2 upshift, suggesting that excess neuronal activity leads to avoidance defects. Cell-specific rescue of NHR-49 in the body cavity neurons was sufficient to improve pathogen avoidance, as well as URX neuron calcium kinetics. Supplementation with oleic acid also improved avoidance behavior and URX calcium kinetics, suggesting that the defective calcium response in the neuron is due to lipid dysfunction. These findings highlight the role of cell-autonomous lipid regulation in neuronal physiology and immune behavior.
Collapse
Affiliation(s)
- Saebom Kwon
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Kyoung-hye Yoon
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| |
Collapse
|
7
|
Steinke I, Singh M, Amin R. Dual PPAR delta/gamma agonists offer therapeutic potential for Alzheimer's disease. Neural Regen Res 2024; 19:1175-1176. [PMID: 37905851 PMCID: PMC11467939 DOI: 10.4103/1673-5374.386410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/15/2023] [Accepted: 08/26/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Ian Steinke
- Department of Drug Discovery and Development, Auburn University, Auburn Alabama, AL, USA
| | - Meenakshi Singh
- Department of Drug Discovery and Development, Auburn University, Auburn Alabama, AL, USA
| | - Rajesh Amin
- Department of Drug Discovery and Development, Auburn University, Auburn Alabama, AL, USA
| |
Collapse
|
8
|
de la Monte SM. Conquering Insulin Network Dysfunctions in Alzheimer's Disease: Where Are We Today? J Alzheimers Dis 2024; 101:S317-S343. [PMID: 39422949 DOI: 10.3233/jad-240069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Functional impairments in the brain's insulin and insulin-like growth factor (IGF) signal transduction networks are recognized mediators of dysregulated energy metabolism, a major driver of the Alzheimer's disease (AD) neurodegeneration cascade. AD-associated insulin-deficient and insulin-resistant states mimic those of diabetes mellitus and affect all cell types in the brain. Besides accounting for abundant amyloid-β and hyperphosphorylated tau lesions in AD, insulin/IGF pathway dysfunctions cause cortical atrophy, loss of synaptic plasticity, white matter myelin/oligodendrocyte degeneration, astrocyte and microglial neuroinflammation and oxidative stress, deficits in energy metabolism, mitochondrial dysfunction, and microvascular disease. These same neuropathological processes have been linked to cognitive impairment in type 2 diabetes mellitus, Parkinson's disease, and vascular dementia. Strategies to address metabolic mediators of cognitive impairment have been borrowed from diabetes and other insulin-resistant diseases and leveraged on preclinical AD model data. The repurposing of diabetes drugs led to clinical trials with intranasal insulin, followed by insulin sensitizers including metformin and peroxisome-proliferator-activated receptor agonists, and then incretin mimetics primarily targeting GLP-1 receptors. In addition, other glucose-lowering agents have been tested for their efficacy in preventing cognitive declines. The strengths and limitations of these approaches are discussed. The main conclusion of this review is that we have now arrived at a stage in which it is time to address long-term deficits in trophic factor availability and receptor responsiveness, signaling abnormalities that extend beyond insulin and include IGFs and interconnected pathways, and the need for multi-pronged rather than single-pronged therapeutic targeting to remediate AD and other forms of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Pathology and Laboratory Medicine, Medicine, Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
9
|
Hu P, Li K, Peng X, Kan Y, Li H, Zhu Y, Wang Z, Li Z, Liu HY, Cai D. Nuclear Receptor PPARα as a Therapeutic Target in Diseases Associated with Lipid Metabolism Disorders. Nutrients 2023; 15:4772. [PMID: 38004166 PMCID: PMC10674366 DOI: 10.3390/nu15224772] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Lipid metabolic diseases have substantial morbidity and mortality rates, posing a significant threat to human health. PPARα, a member of the peroxisome proliferator-activated receptors (PPARs), plays a crucial role in lipid metabolism and immune regulation. Recent studies have increasingly recognized the pivotal involvement of PPARα in diverse pathological conditions. This comprehensive review aims to elucidate the multifaceted role of PPARα in metabolic diseases including liver diseases, diabetes-related diseases, age-related diseases, and cancers, shedding light on the underlying molecular mechanisms and some regulatory effects of natural/synthetic ligands of PPARα. By summarizing the latest research findings on PPARα, we aim to provide a foundation for the possible therapeutic exploitation of PPARα in lipid metabolic diseases.
Collapse
Affiliation(s)
- Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Kaiqi Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Xiaoxu Peng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Yufei Kan
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Hao Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Yanli Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Ziyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Zhaojian Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (P.H.); (K.L.); (X.P.); (Y.K.); (H.L.); (Y.Z.); (Z.W.); (Z.L.)
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009, China
| |
Collapse
|
10
|
Sarnowski C, Huan T, Ma Y, Joehanes R, Beiser A, DeCarli CS, Heard-Costa NL, Levy D, Lin H, Liu CT, Liu C, Meigs JB, Satizabal CL, Florez JC, Hivert MF, Dupuis J, De Jager PL, Bennett DA, Seshadri S, Morrison AC. Multi-tissue epigenetic analysis identifies distinct associations underlying insulin resistance and Alzheimer's disease at CPT1A locus. Clin Epigenetics 2023; 15:173. [PMID: 37891690 PMCID: PMC10612362 DOI: 10.1186/s13148-023-01589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Insulin resistance (IR) is a major risk factor for Alzheimer's disease (AD) dementia. The mechanisms by which IR predisposes to AD are not well-understood. Epigenetic studies may help identify molecular signatures of IR associated with AD, thus improving our understanding of the biological and regulatory mechanisms linking IR and AD. METHODS We conducted an epigenome-wide association study of IR, quantified using the homeostatic model assessment of IR (HOMA-IR) and adjusted for body mass index, in 3,167 participants from the Framingham Heart Study (FHS) without type 2 diabetes at the time of blood draw used for methylation measurement. We identified DNA methylation markers associated with IR at the genome-wide level accounting for multiple testing (P < 1.1 × 10-7) and evaluated their association with neurological traits in participants from the FHS (N = 3040) and the Religious Orders Study/Memory and Aging Project (ROSMAP, N = 707). DNA methylation profiles were measured in blood (FHS) or dorsolateral prefrontal cortex (ROSMAP) using the Illumina HumanMethylation450 BeadChip. Linear regressions (ROSMAP) or mixed-effects models accounting for familial relatedness (FHS) adjusted for age, sex, cohort, self-reported race, batch, and cell type proportions were used to assess associations between DNA methylation and neurological traits accounting for multiple testing. RESULTS We confirmed the strong association of blood DNA methylation with IR at three loci (cg17901584-DHCR24, cg17058475-CPT1A, cg00574958-CPT1A, and cg06500161-ABCG1). In FHS, higher levels of blood DNA methylation at cg00574958 and cg17058475 were both associated with lower IR (P = 2.4 × 10-11 and P = 9.0 × 10-8), larger total brain volumes (P = 0.03 and P = 9.7 × 10-4), and smaller log lateral ventricular volumes (P = 0.07 and P = 0.03). In ROSMAP, higher levels of brain DNA methylation at the same two CPT1A markers were associated with greater risk of cognitive impairment (P = 0.005 and P = 0.02) and higher AD-related indices (CERAD score: P = 5 × 10-4 and 0.001; Braak stage: P = 0.004 and P = 0.01). CONCLUSIONS Our results suggest potentially distinct epigenetic regulatory mechanisms between peripheral blood and dorsolateral prefrontal cortex tissues underlying IR and AD at CPT1A locus.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Tianxiao Huan
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
| | - Yiyi Ma
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Alexa Beiser
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | | | - Nancy L Heard-Costa
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Daniel Levy
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Chunyu Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claudia L Satizabal
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Harvard University, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Josée Dupuis
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Epidemiology, Biostatistics and Occupational Health, School of Population and Global Health, McGill University, Montreal, Canada
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sudha Seshadri
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
11
|
Comerota MM, Gedam M, Xiong W, Jin F, Deng L, Wang MC, Wang J, Zheng H. Oleoylethanolamide facilitates PPARα and TFEB signaling and attenuates Aβ pathology in a mouse model of Alzheimer's disease. Mol Neurodegener 2023; 18:56. [PMID: 37580742 PMCID: PMC10426131 DOI: 10.1186/s13024-023-00648-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Age is the strongest risk factor for the development of Alzheimer's disease (AD). Besides the pathological hallmarks of β-amyloid (Aβ) plaques and neurofibrillary tangles, emerging evidence demonstrates a critical role of microglia and neuroinflammation in AD pathogenesis. Oleoylethanolamide (OEA) is an endogenous lipid amide that has been shown to promote lifespan and healthspan in C. elegans through regulation of lysosome-to-nucleus signaling and cellular metabolism. The goal of our study was to determine the role of OEA in the mediation of microglial activity and AD pathology using its stable analog, KDS-5104. METHODS We used primary microglial cultures and genetic and pharmacological approaches to examine the signaling mechanisms and functional roles of OEA in mediating Aβ phagocytosis and clearance, lipid metabolism and inflammasome formation. Further, we tested the effect of OEA in vivo in acute LPS-induced neuroinflammation and by chronic treatment of 5xFAD mice. RESULTS We found that OEA activates PPARα signaling and its downstream cell-surface receptor CD36 activity. In addition, OEA promotes TFEB lysosomal function in a PPARα-dependent but mTORC1-independent manner, the combination of which leads to enhanced microglial Aβ uptake and clearance. These are associated with the suppression of LPS-induced lipid droplet accumulation and inflammasome activation. Chronic treatment of 5xFAD mice with KDS-5104 restored dysregulated lipid profiles, reduced reactive gliosis and Aβ pathology and rescued cognitive impairments. CONCLUSION Together, our study provides support that augmenting OEA-mediated lipid signaling may offer therapeutic benefit against aging and AD through modulating lipid metabolism and microglia phagocytosis and clearance.
Collapse
Affiliation(s)
- Michele M Comerota
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Manasee Gedam
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Feng Jin
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Lisheng Deng
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- HHMI Janelia Research Campus, Ashburn, VA, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA.
- Department of Molecular and Human Genetics, Houston, TX, USA.
| |
Collapse
|
12
|
Semikasev E, Ahlemeyer B, Acker T, Schänzer A, Baumgart-Vogt E. Rise and fall of peroxisomes during Alzheimer´s disease: a pilot study in human brains. Acta Neuropathol Commun 2023; 11:80. [PMID: 37170361 PMCID: PMC10176950 DOI: 10.1186/s40478-023-01567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Peroxisomes are eukaryotic organelles that rapidly change in number depending on the metabolic requirement of distinct cell types and tissues. In the brain, these organelles are essential for neuronal migration and myelination during development and their dysfunction is associated with age-related neurodegenerative diseases. Except for one study analysing ABCD3-positive peroxisomes in neurons of the frontal neocortex of Alzheimer disease (AD) patients, no data on other brain regions or peroxisomal proteins are available. In the present morphometric study, we quantified peroxisomes labelled with PEX14, a metabolism-independent peroxisome marker, in 13 different brain areas of 8 patients each either with low, intermediate or high AD neuropathological changes compared to 10 control patients. Classification of patient samples was based on the official ABC score. During AD-stage progression, the peroxisome density decreased in the area entorhinalis, parietal/occipital neocortex and cerebellum, it increased and in later AD-stage patients decreased in the subiculum and hippocampal CA3 region, frontal neocortex and pontine gray and it remained unchanged in the gyrus dentatus, temporal neocortex, striatum and inferior olive. Moreover, we investigated the density of catalase-positive peroxisomes in a subset of patients (> 80 years), focussing on regions with significant alterations of PEX14-positive peroxisomes. In hippocampal neurons, only one third of all peroxisomes contained detectable levels of catalase exhibiting constant density at all AD stages. Whereas the density of all peroxisomes in neocortical neurons was only half of the one of the hippocampus, two thirds of them were catalase-positive exhibiting increased levels at higher ABC scores. In conclusion, we observed spatiotemporal differences in the response of peroxisomes to different stages of AD-associated pathologies.
Collapse
Affiliation(s)
- Eugen Semikasev
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, 35385, Giessen, Germany
- Department of Neurosurgery, University Hospital of Giessen, Klinikstr. 33, 35392, Giessen, Germany
| | - Barbara Ahlemeyer
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, 35385, Giessen, Germany.
| | - Till Acker
- Institute of Neuropathology, Justus-Liebig University, Arndtstr. 16, 35392, Giessen, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig University, Arndtstr. 16, 35392, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, 35385, Giessen, Germany.
| |
Collapse
|
13
|
Garcia G, Pinto S, Ferreira S, Lopes D, Serrador MJ, Fernandes A, Vaz AR, de Mendonça A, Edenhofer F, Malm T, Koistinaho J, Brites D. Emerging Role of miR-21-5p in Neuron-Glia Dysregulation and Exosome Transfer Using Multiple Models of Alzheimer's Disease. Cells 2022; 11:3377. [PMID: 36359774 PMCID: PMC9655962 DOI: 10.3390/cells11213377] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 08/25/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with neuron-glia dysfunction and dysregulated miRNAs. We previously reported upregulated miR-124/miR-21 in AD neurons and their exosomes. However, their glial distribution, phenotypic alterations and exosomal spread are scarcely documented. Here, we show glial cell activation and miR-21 overexpression in mouse organotypic hippocampal slices transplanted with SH-SY5Y cells expressing the human APP695 Swedish mutation. The upregulation of miR-21 only in the CSF from a small series of mild cognitive impairment (MCI) AD patients, but not in non-AD MCI individuals, supports its discriminatory potential. Microglia, neurons, and astrocytes differentiated from the same induced pluripotent stem cells from PSEN1ΔE9 AD patients all showed miR-21 elevation. In AD neurons, miR-124/miR-21 overexpression was recapitulated in their exosomes. In AD microglia, the upregulation of iNOS and miR-21/miR-146a supports their activation. AD astrocytes manifested a restrained inflammatory profile, with high miR-21 but low miR-155 and depleted exosomal miRNAs. Their immunostimulation with C1q + IL-1α + TNF-α induced morphological alterations and increased S100B, inflammatory transcripts, sAPPβ, cytokine release and exosomal miR-21. PPARα, a target of miR-21, was found to be repressed in all models, except in neurons, likely due to concomitant miR-125b elevation. The data from these AD models highlight miR-21 as a promising biomarker and a disease-modifying target to be further explored.
Collapse
Affiliation(s)
- Gonçalo Garcia
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Sara Pinto
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Sofia Ferreira
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Daniela Lopes
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Maria João Serrador
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Adelaide Fernandes
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Central Nervous System, Blood and Peripheral Inflammation Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Ana Rita Vaz
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | | | - Frank Edenhofer
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
14
|
PPARα Signaling: A Candidate Target in Psychiatric Disorder Management. Biomolecules 2022; 12:biom12050723. [PMID: 35625650 PMCID: PMC9138493 DOI: 10.3390/biom12050723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
Peroxisome proliferator-activator receptors (PPARs) regulate lipid and glucose metabolism, control inflammatory processes, and modulate several brain functions. Three PPAR isoforms have been identified, PPARα, PPARβ/δ, and PPARγ, which are expressed in different tissues and cell types. Hereinafter, we focus on PPARα involvement in the pathophysiology of neuropsychiatric and neurodegenerative disorders, which is underscored by PPARα localization in neuronal circuits involved in emotion modulation and stress response, and its role in neurodevelopment and neuroinflammation. A multiplicity of downstream pathways modulated by PPARα activation, including glutamatergic neurotransmission, upregulation of brain-derived neurotrophic factor, and neurosteroidogenic effects, encompass mechanisms underlying behavioral regulation. Modulation of dopamine neuronal firing in the ventral tegmental area likely contributes to PPARα effects in depression, anhedonia, and autism spectrum disorder (ASD). Based on robust preclinical evidence and the initial results of clinical studies, future clinical trials should assess the efficacy of PPARα agonists in the treatment of mood and neurodevelopmental disorders, such as depression, schizophrenia, and ASD.
Collapse
|
15
|
Lin Y, Wang Y, Li PF. PPARα: An emerging target of metabolic syndrome, neurodegenerative and cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1074911. [PMID: 36589809 PMCID: PMC9800994 DOI: 10.3389/fendo.2022.1074911] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a ligand-activated transcription factor that is involved in lipid metabolism of various tissues. Different metabolites of fatty acids and agonists like fibrates activate PPARα for its transactivative or repressive function. PPARα is known to affect diverse human diseases, and we focus on advanced studies of its transcriptional regulation in these diseases. In MAFLD, PPARα shows a protective function with its upregulation of lipid oxidation and mitochondrial biogenesis and transcriptional repression of inflammatory genes, which is similar in Alzheimer's disease and cardiovascular disease. Activation of PPARα also prevents the progress of diabetes complications; however, its role in diabetes and cancers remains uncertain. Some PPARα-specific agonists, such as Wy14643 and fenofibrate, have been applied in metabolic syndrome treatment, which might own potential in wider application. Future studies may further explore the functions and interventions of PPARα in cancer, diabetes, immunological diseases, and neurodegenerative disease.
Collapse
Affiliation(s)
- Yijun Lin
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| | - Pei-feng Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| |
Collapse
|