1
|
Warren WG, Osborn M, Duffy P, Yates A, O'Sullivan SE. Potential safety implications of fatty acid-binding protein inhibition. Toxicol Appl Pharmacol 2024; 491:117079. [PMID: 39218163 DOI: 10.1016/j.taap.2024.117079] [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: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Fatty acid-binding proteins (FABPs) are small intracellular proteins that regulate fatty acid metabolism, transport, and signalling. There are ten known human isoforms, many of which are upregulated and involved in clinical pathologies. As such, FABP inhibition may be beneficial in disease states such as cancer, and those involving the cardiovascular system, metabolism, immunity, and cognition. Recently, a potent, selective FABP5 inhibitor (ART26.12), with 90-fold selectivity to FABP3 and 20-fold selectivity to FABP7, was found to be remarkably benign, with a no-observed-adverse-effect level of 1000 mg/kg in rats and dogs, showing no genotoxicity, cardiovascular, central, or respiratory toxicity. To understand the potential implication of FABP inhibition more fully, this review systematically assessed literature investigating genetic knockout, knockdown, and pharmacological inhibition of FABP3, FABP4, FABP5, or FABP7. Analysis of the literature revealed that animals bred not to express FABPs showed the most biological effects, suggesting key roles of these proteins during development. FABP ablation sometimes exacerbated symptoms of disease models, particularly those linked to metabolism, inflammatory and immune responses, cardiac contractility, neurogenesis, and cognition. However, FABP inhibition (genetic silencing or pharmacological) had a positive effect in many more disease conditions. Several polymorphisms of each FABP gene have also been linked to pathological conditions, but it was unclear how several polymorphisms affected protein function. Overall, analysis of the literature to date suggests that pharmacological inhibition of FABPs in adults is of low risk.
Collapse
Affiliation(s)
- William G Warren
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom.
| | - Myles Osborn
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Paul Duffy
- Apconix Ltd., Alderley Park, Cheshire SK10 4TG, United Kingdom
| | - Andrew Yates
- Artelo Biosciences Limited, Alderley Park, Cheshire SK10 4TG, United Kingdom
| | | |
Collapse
|
2
|
Chen S, Pan Z, Liu M, Guo L, Jiang X, He G. Recent Advances on Small-Molecule Inhibitors of Lipocalin-like Proteins. J Med Chem 2024; 67:5144-5167. [PMID: 38525852 DOI: 10.1021/acs.jmedchem.4c00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Lipid transfer proteins (LTPs) are crucial players in nonvesicular lipid trafficking. LTPs sharing a lipocalin lipid transfer domain (lipocalin-like proteins) have a wide range of biological functions, such as regulating immune responses and cell proliferation, differentiation, and death as well as participating in the pathogenesis of inflammatory, metabolic, and neurological disorders and cancer. Therefore, the development of small-molecule inhibitors targeting these LTPs is important and has potential clinical applications. Herein, we summarize the structure and function of lipocalin-like proteins, mainly including retinol-binding proteins, lipocalins, and fatty acid-binding proteins and discuss the recent advances on small-molecule inhibitors for these protein families and their applications in disease treatment. The findings of our Perspective can provide guidance for the development of inhibitors of these LTPs and highlight the challenges that might be faced during the procedures.
Collapse
Affiliation(s)
- Siliang Chen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhaoping Pan
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingxia Liu
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linghong Guo
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Wang L, Qu F, Yu X, Yang S, Zhao B, Chen Y, Li P, Zhang Z, Zhang J, Han X, Wei D. Cortical lipid metabolic pathway alteration of early Alzheimer's disease and candidate drugs screen. Eur J Med Res 2024; 29:199. [PMID: 38528586 DOI: 10.1186/s40001-024-01730-w] [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: 03/10/2022] [Accepted: 02/12/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Lipid metabolism changes occur in early Alzheimer's disease (AD) patients. Yet little is known about metabolic gene changes in early AD cortex. METHODS The lipid metabolic genes selected from two datasets (GSE39420 and GSE118553) were analyzed with enrichment analysis. Protein-protein interaction network construction and correlation analyses were used to screen core genes. Literature analysis and molecular docking were applied to explore potential therapeutic drugs. RESULTS 60 lipid metabolic genes differentially expressed in early AD patients' cortex were screened. Bioinformatics analyses revealed that up-regulated genes were mainly focused on mitochondrial fatty acid oxidation and mediating the activation of long-chain fatty acids, phosphoproteins, and cholesterol metabolism. Down-regulated genes were mainly focused on lipid transport, carboxylic acid metabolic process, and neuron apoptotic process. Literature reviews and molecular docking results indicated that ACSL1, ACSBG2, ACAA2, FABP3, ALDH5A1, and FFAR4 were core targets for lipid metabolism disorder and had a high binding affinity with compounds including adenosine phosphate, oxidized Photinus luciferin, BMS-488043, and candidate therapeutic drugs especially bisphenol A, benzo(a)pyrene, ethinyl estradiol. CONCLUSIONS AD cortical lipid metabolism disorder was associated with the dysregulation of the PPAR signaling pathway, glycerophospholipid metabolism, adipocytokine signaling pathway, fatty acid biosynthesis, fatty acid degradation, ferroptosis, biosynthesis of unsaturated fatty acids, and fatty acid elongation. Candidate drugs including bisphenol A, benzo(a)pyrene, ethinyl estradiol, and active compounds including adenosine phosphate, oxidized Photinus luciferin, and BMS-488043 have potential therapeutic effects on cortical lipid metabolism disorder of early AD.
Collapse
Affiliation(s)
- Linshuang Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fengxue Qu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xueyun Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Sixia Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Binbin Zhao
- Institute of Gerontology, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yaojing Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Pengbo Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
- BABRI Centre, Beijing Normal University, Beijing, 100875, China
| | - Junying Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China.
- BABRI Centre, Beijing Normal University, Beijing, 100875, China.
| | - Xuejie Han
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Dongfeng Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
4
|
Cheng A, Jia W, Finkelstein DI, Stefanova N, Wang H, Sasaki T, Kawahata I, Fukunaga K. Pharmacological inhibition of FABP7 by MF 6 counteracts cerebellum dysfunction in an experimental multiple system atrophy mouse model. Acta Pharmacol Sin 2024; 45:66-75. [PMID: 37605049 PMCID: PMC10770047 DOI: 10.1038/s41401-023-01138-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/09/2023] [Indexed: 08/23/2023] Open
Abstract
Multiple system atrophy (MSA) is a rare, fatal neurodegenerative disease characterized by the accumulation of misfolded α-synuclein (αSyn) in glial cells, leading to the formation of glial cytoplasmic inclusions (GCI). We previous found that glial fatty acid-binding protein 7 (FABP7) played a crucial role in alpha-synuclein (αSyn) aggregation and toxicity in oligodendrocytes, inhibition of FABP7 by a specific inhibitor MF 6 reduced αSyn aggregation and enhanced cell viability in cultured cell lines and mouse oligodendrocyte progenitor cells. In this study we investigated whether MF 6 ameliorated αSyn-associated pathological processes in PLP-hαSyn transgenic mice (PLP-αSyn mice), a wildly used MSA mouse model with overexpressing αSyn in oligodendroglia under the proteolipid protein (PLP) promoter. PLP-αSyn mice were orally administered MF6 (0.1, 1 mg ·kg-1 ·d-1) for 32 days starting from the age of 6 months. We showed that oral administration of MF 6 significantly improved motor function assessed in a pole test, and reduced αSyn aggregation levels in both cerebellum and basal ganglia of PLP-αSyn mice. Moreover, MF 6 administration decreased oxidative stress and inflammation levels, and improved myelin levels and Purkinje neuron morphology in the cerebellum. By using mouse brain tissue slices and αSyn aggregates-treated KG-1C cells, we demonstrated that MF 6 reduced αSyn propagation to Purkinje neurons and oligodendrocytes through regulating endocytosis. Overall, these results suggest that MF 6 improves cerebellar functions in MSA by inhibiting αSyn aggregation and propagation. We conclude that MF 6 is a promising compound that warrants further development for the treatment of MSA.
Collapse
Affiliation(s)
- An Cheng
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, CA, USA.
| | - Wenbin Jia
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - David I Finkelstein
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Nadia Stefanova
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Haoyang Wang
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
5
|
Santos-Rebouças CB, Cordovil Cotrin J, Dos Santos Junior GC. Exploring the interplay between metabolomics and genetics in Parkinson's disease: Insights from ongoing research and future avenues. Mech Ageing Dev 2023; 216:111875. [PMID: 37748695 DOI: 10.1016/j.mad.2023.111875] [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: 08/22/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Parkinson's disease (PD) is a widespread neurodegenerative disorder, whose complex aetiology remains under construction. While rare variants have been associated with the monogenic PD form, most PD cases are influenced by multiple genetic and environmental aspects. Nonetheless, the pathophysiological pathways and molecular networks involved in monogenic/idiopathic PD overlap, and genetic variants are decisive in elucidating the convergent underlying mechanisms of PD. In this scenario, metabolomics has furnished a dynamic and systematic picture of the synergy between the genetic background and environmental influences that impact PD, making it a valuable tool for investigating PD-related metabolic dysfunctions. In this review, we performed a brief overview of metabolomics current research in PD, focusing on significant metabolic alterations observed in idiopathic PD from different biofluids and strata and exploring how they relate to genetic factors associated with monogenic PD. Dysregulated amino acid metabolism, lipid metabolism, and oxidative stress are the critical metabolic pathways implicated in both genetic and idiopathic PD. By merging metabolomics and genetics data, it is possible to distinguish metabolic signatures of specific genetic backgrounds and to pinpoint subgroups of PD patients who could derive personalized therapeutic benefits. This approach holds great promise for advancing PD research and developing innovative, cost-effective treatments.
Collapse
Affiliation(s)
- Cíntia Barros Santos-Rebouças
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Juliana Cordovil Cotrin
- Human Genetics Service, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos Junior
- LabMet, Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Zhang Y, Liu Y, Sun J, Zhang W, Guo Z, Ma Q. Arachidonic acid metabolism in health and disease. MedComm (Beijing) 2023; 4:e363. [PMID: 37746665 PMCID: PMC10511835 DOI: 10.1002/mco2.363] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Arachidonic acid (AA), an n-6 essential fatty acid, is a major component of mammalian cells and can be released by phospholipase A2. Accumulating evidence indicates that AA plays essential biochemical roles, as it is the direct precursor of bioactive lipid metabolites of eicosanoids such as prostaglandins, leukotrienes, and epoxyeicosatrienoic acid obtained from three distinct enzymatic metabolic pathways: the cyclooxygenase pathway, lipoxygenase pathway, and cytochrome P450 pathway. AA metabolism is involved not only in cell differentiation, tissue development, and organ function but also in the progression of diseases, such as hepatic fibrosis, neurodegeneration, obesity, diabetes, and cancers. These eicosanoids are generally considered proinflammatory molecules, as they can trigger oxidative stress and stimulate the immune response. Therefore, interventions in AA metabolic pathways are effective ways to manage inflammatory-related diseases in the clinic. Currently, inhibitors targeting enzymes related to AA metabolic pathways are an important area of drug discovery. Moreover, many advances have also been made in clinical studies of AA metabolic inhibitors in combination with chemotherapy and immunotherapy. Herein, we review the discovery of AA and focus on AA metabolism in relation to health and diseases. Furthermore, inhibitors targeting AA metabolism are summarized, and potential clinical applications are discussed.
Collapse
Affiliation(s)
- Yiran Zhang
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Yingxiang Liu
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Jin Sun
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Wei Zhang
- Department of PathologyThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Zheng Guo
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Qiong Ma
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
- Department of PathologyThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| |
Collapse
|
7
|
Widyarti S, Wibowo S, Sabarudin A, Abhirama I, Sumitro SB. Dysfunctional energy and future perspective of low dose H 2O 2 as protective agent in neurodegenerative disease. Heliyon 2023; 9:e18123. [PMID: 37519743 PMCID: PMC10372669 DOI: 10.1016/j.heliyon.2023.e18123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 05/21/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
The number of people with neurodegenerative disease continues to increase every year. A new perspective is needed to overcome this disease. In this review, researchers collected information about dysfunctional energy in neurodegenerative diseases driven by mitochondria. Mitochondrial dysregulation can cause damage to the neuron system. The increase in the amount and interaction of α-synuclein with SAMM50 and GABARAPL1 in the mitochondria is one of the factors causing neurodegenerative disease. As an energy provider in the body, the existence of harmonization in the regulation of mitochondria, specifically the mitochondrial outer membrane, is important. Low-dose hydrogen peroxide (H2O2) has neuroprotective abilities to overcome the impairment function of mitochondria in neurodegenerative patients. Based on computational simulation of this case, it can be used as a basic concept for the development of the role of H2O2 in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sri Widyarti
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
| | - Syahputra Wibowo
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
- Postdoctoral Fellow, Faculty of Biology, Gadjah Mada University, Teknika Selatan Sekip Utara, 55281 Yogyakarta, Indonesia
| | - Akhmad Sabarudin
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
| | - Intan Abhirama
- Department of Neurology, Bogor Senior Hospital, Jl.Raya Tajur 16137, West Java, Indonesia
| | - Sutiman Bambang Sumitro
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
| |
Collapse
|
8
|
Jia W, Kawahata I, Cheng A, Sasaki T, Sasaoka T, Fukunaga K. Amelioration of Nicotine-Induced Conditioned Place Preference Behaviors in Mice by an FABP3 Inhibitor. Int J Mol Sci 2023; 24:ijms24076644. [PMID: 37047614 PMCID: PMC10095245 DOI: 10.3390/ijms24076644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
We previously demonstrated that fatty acid-binding protein 3 null (FABP3−/−) mice exhibit resistance to nicotine-induced conditioned place preference (CPP). Here, we confirm that the FABP3 inhibitor, MF1 ((4-(2-(1-(2-chlorophenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy) butanoic acid), successfully reduces nicotine-induced CPP scores in mice. MF1 (0.3 or 1.0 mg/kg) was orally administered 30 min before nicotine, and CPP scores were assessed in the conditioning, withdrawal, and relapse phases. MF1 treatment decreased CPP scores in a dose-dependent manner. Failure of CPP induction by MF1 (1.0 mg/kg, p.o.) was associated with the inhibition of both CaMKII and ERK activation in the nucleus accumbens (NAc) and hippocampal CA1 regions. MF1 treatment reduced nicotine-induced increases in phosphorylated CaMKII and cAMP-response element-binding protein (CREB)-positive cells. Importantly, the increase in dopamine D2 receptor (D2R) levels following chronic nicotine exposure was inhibited by MF1 treatment. Moreover, the quinpirole (QNP)-induced increase in the level of CaMKII and ERK phosphorylation was significantly inhibited by MF1 treatment of cultured NAc slices from wild type (WT) mice; however, QNP treatment had no effect on CaMKII and ERK phosphorylation levels in the NAc of D2R null mice. Taken together, these results show that MF1 treatment suppressed D2R/FABP3 signaling, thereby preventing nicotine-induced CPP induction. Hence, MF1 can be used as a novel drug to block addiction to nicotine and other drugs by inhibiting the dopaminergic system.
Collapse
Affiliation(s)
- Wenbin Jia
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - An Cheng
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Toshikuni Sasaoka
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
- BRI Pharma Inc., Sendai 982-0804, Japan
| |
Collapse
|
9
|
Weng H, Song W, Fu K, Guan Y, Cai G, Huang E, Chen X, Zou H, Ye Q. Proteomic profiling reveals the potential mechanisms and regulatory targets of sirtuin 4 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's mouse model. Front Neurosci 2023; 16:1035444. [PMID: 36760798 PMCID: PMC9905825 DOI: 10.3389/fnins.2022.1035444] [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: 09/02/2022] [Accepted: 12/06/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Parkinson's disease (PD), as a common neurodegenerative disease, currently has no effective therapeutic approaches to delay or stop its progression. There is an urgent need to further define its pathogenesis and develop new therapeutic targets. An increasing number of studies have shown that members of the sirtuin (SIRT) family are differentially involved in neurodegenerative diseases, indicating their potential to serve as targets in therapeutic strategies. Mitochondrial SIRT4 possesses multiple enzymatic activities, such as deacetylase, ADP ribosyltransferase, lipoamidase, and deacylase activities, and exhibits different enzymatic activities and target substrates in different tissues and cells; thus, mitochondrial SIRT4 plays an integral role in regulating metabolism. However, the role and mechanism of SIRT4 in PD are not fully understood. This study aimed to investigate the potential mechanism and possible regulatory targets of SIRT4 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice. Methods The expression of the SIRT4 protein in the MPTP-induced PD mouse mice or key familial Parkinson disease protein 7 knockout (DJ-1 KO) rat was compared against the control group by western blot assay. Afterwards, quantitative proteomics and bioinformatics analyses were performed to identify altered proteins in the vitro model and reveal the possible functional role of SIRT4. The most promising molecular target of SIRT4 were screened and validated by viral transfection, western blot assay and reverse transcription quantitative PCR (RT-qPCR) assays. Results The expression of the SIRT4 protein was found to be altered both in the MPTP-induced PD mouse mice and DJ-1KO rats. Following the viral transfection of SIRT4, a quantitative proteomics analysis identified 5,094 altered proteins in the vitro model, including 213 significantly upregulated proteins and 222 significantly downregulated proteins. The results from bioinformatics analyses indicated that SIRT4 mainly affected the ribosomal pathway, propionate metabolism pathway, peroxisome proliferator-activated receptor (PPAR) signaling pathway and peroxisome pathway in cells, and we screened 25 potential molecular targets. Finally, only fatty acid binding protein 4 (FABP4) in the PPAR signaling pathway was regulated by SIRT4 among the 25 molecules. Importantly, the alterations in FABP4 and PPARγ were verified in the MPTP-induced PD mouse model. Discussion Our results indicated that FABP4 in the PPAR signaling pathway is the most promising molecular target of SIRT4 in an MPTP-induced mouse model and revealed the possible functional role of SIRT4. This study provides a reference for future drug development and mechanism research with SIRT4 as a target or biomarker.
Collapse
Affiliation(s)
- Huidan Weng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Wenjing Song
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Kangyue Fu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yunqian Guan
- Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guoen Cai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - En Huang
- The School of Basic Medical Sciences, Fujian Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Haiqiang Zou
- Department of Neurosurgery, General Hospital of Southern Theatre Command, PLA, Guangzhou, Guangdong, China,Haiqiang Zou,
| | - Qinyong Ye
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China,*Correspondence: Qinyong Ye,
| |
Collapse
|
10
|
Cheng A, Wang YF, Shinoda Y, Kawahata I, Yamamoto T, Jia WB, Yamamoto H, Mizobata T, Kawata Y, Fukunaga K. Fatty acid-binding protein 7 triggers α-synuclein oligomerization in glial cells and oligodendrocytes associated with oxidative stress. Acta Pharmacol Sin 2022; 43:552-562. [PMID: 33935286 PMCID: PMC8888578 DOI: 10.1038/s41401-021-00675-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/31/2021] [Indexed: 02/03/2023] Open
Abstract
We previously show that fatty acid-binding protein 3 (FABP3) triggers α-synuclein (Syn) accumulation and induces dopamine neuronal cell death in Parkinson disease mouse model. But the role of fatty acid-binding protein 7 (FABP7) in the brain remains unclear. In this study we investigated whether FABP7 was involved in synucleinopathies. We showed that FABP7 was co-localized and formed a complex with Syn in Syn-transfected U251 human glioblastoma cells, and treatment with arachidonic acid (100 M) significantly promoted FABP7-induced Syn aggregation, which was associated with cell death. We demonstrated that synthetic FABP7 ligand 6 displayed a high affinity against FABP7 with Kd value of 209 nM assessed in 8-anilinonaphthalene-1-sulfonic acid (ANS) assay; ligand 6 improved U251 cell survival via disrupting the FABP7-Syn interaction. We showed that activation of phospholipase A2 (PLA2) by psychosine (10 M) triggered oligomerization of endogenous Syn and FABP7, and induced cell death in both KG-1C human oligodendroglia cells and oligodendrocyte precursor cells (OPCs). FABP7 ligand 6 (1 M) significantly decreased Syn oligomerization and aggregation thereby prevented KG-1C and OPC cell death. This study demonstrates that FABP7 triggers α-synuclein oligomerization through oxidative stress, while FABP7 ligand 6 can inhibit FABP7-induced Syn oligomerization and aggregation, thereby rescuing glial cells and oligodendrocytes from cell death.
Collapse
Affiliation(s)
- An Cheng
- grid.69566.3a0000 0001 2248 6943Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yi-fei Wang
- grid.69566.3a0000 0001 2248 6943Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuharu Shinoda
- grid.69566.3a0000 0001 2248 6943Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- grid.69566.3a0000 0001 2248 6943Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tetsunori Yamamoto
- grid.69566.3a0000 0001 2248 6943Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wen-bin Jia
- grid.69566.3a0000 0001 2248 6943Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hanae Yamamoto
- grid.265107.70000 0001 0663 5064Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Tomohiro Mizobata
- grid.265107.70000 0001 0663 5064Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Yasushi Kawata
- grid.265107.70000 0001 0663 5064Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
11
|
Zhang X, Xu Y, Ma L, Yu K, Niu Y, Xu X, Shi Y, Guo S, Xue X, Wang Y, Qiu S, Cui J, Wang H, Tian X, Miao Y, Meng F, Qiao Y, Yu Y, Wang J. Essential roles of exosome and circRNA_101093 on ferroptosis desensitization in lung adenocarcinoma. Cancer Commun (Lond) 2022; 42:287-313. [PMID: 35184419 PMCID: PMC9017758 DOI: 10.1002/cac2.12275] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/20/2021] [Accepted: 02/08/2022] [Indexed: 12/16/2022] Open
Abstract
Background Resistance to ferroptosis, a regulated cell death caused by iron‐dependent excessive accumulation of lipid peroxides, has recently been linked to lung adenocarcinoma (LUAD). Intracellular antioxidant systems are required for protection against ferroptosis. The purpose of the present study was to investigate whether and how extracellular system desensitizes LUAD cells to ferroptosis. Methods Established human lung fibroblasts MRC‐5, WI38, and human LUAD H1650, PC9, H1975, H358, A549, and H1299 cell lines, tumor and matched normal adjacent tissues of LUAD, and plasma from healthy individuals and LUAD patients were used in this study. Immunohistochemistry and immunoblotting were used to analyze protein expression, and quantitative reverse transcription‐PCR was used to analyze mRNA expression. Cell viability, cell death, and the lipid reactive oxygen species generation were measured to evaluate the responses to ferroptosis. Exosomes were observed using transmission electron microscope. The localization of arachidonic acid (AA) was detected using click chemistry labeling followed by confocal microscopy. Interactions between RNAs and proteins were detected using RNA pull‐down, RNA immunoprecipitation and photoactivatable ribonucleoside‐enhanced crosslinking and immunoprecipitation methods. Proteomic analysis was used to investigate RNA‐regulated proteins, and metabolomic analysis was performed to analyze metabolites. Cell‐derived xenograft, patient‐derived xenograft, cell‐implanted intrapulmonary LUAD mouse models and plasma/tissue specimens from LUAD patients were used to validate the molecular mechanism. Results Plasma exosome from LUAD patients specifically reduced lipid peroxidation and desensitized LUAD cells to ferroptosis. A potential explanation is that exosomal circRNA_101093 (cir93) maintained an elevation in intracellular cir93 in LUAD to modulate AA, a poly‐unsaturated fatty acid critical for ferroptosis‐associated increased peroxidation in the plasma membrane. Mechanistically, cir93 interacted with and increased fatty acid‐binding protein 3 (FABP3), which transported AA and facilitated its reaction with taurine. Thus, global AA was reduced, whereas N‐arachidonoyl taurine (NAT, the product of AA and taurine) was induced. Notably, the role of NAT in suppressing AA incorporation into the plasma membrane was also revealed. In pre‐clinical in vivo models, reducing exosome improved ferroptosis‐based treatment. Conclusion Exosome and cir93 are essential for desensitizing LUAD cells to ferroptosis, and blocking exosome may be helpful for future LUAD treatment.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Laboratory Medicine Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Yunhua Xu
- Shanghai Lung Cancer Center Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Lifang Ma
- Department of Laboratory Medicine Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Keke Yu
- Department of Bio‐bank Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Yongjie Niu
- Shanghai Municipal Hospital of Traditional Chinese Medicine Shanghai University of Traditional Chinese Medicine Shanghai 200071 P. R. China
| | - Xin Xu
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Yi Shi
- Bio‐X Institutes Key Laboratory for the Genetics of Developmental and Neuropsychiatric disorder Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Susu Guo
- Department of Clinical Laboratory Medicine Shanghai Tenth People's Hospital of Tongji University Shanghai 200072 P. R. China
| | - Xiangfei Xue
- Department of Clinical Laboratory Medicine Shanghai Tenth People's Hospital of Tongji University Shanghai 200072 P. R. China
| | - Yikun Wang
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Shiyu Qiu
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Jiangtao Cui
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Hong Wang
- School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai 200025 P. R. China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Yayou Miao
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Fanyu Meng
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Yongxia Qiao
- School of Public Health Shanghai Jiao Tong University School of Medicine Shanghai 200025 P. R. China
| | - Yongchun Yu
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
| | - Jiayi Wang
- Department of Laboratory Medicine Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200030 P. R. China
- Bio‐X Institutes Key Laboratory for the Genetics of Developmental and Neuropsychiatric disorder Shanghai Jiao Tong University Shanghai 200030 P. R. China
| |
Collapse
|
12
|
Cheng A, Fukunaga K. [Role of fatty acid-binding protein 7 and novel therapeutic approach in synucleinopathies]. Nihon Yakurigaku Zasshi 2022; 157:396-400. [PMID: 36328545 DOI: 10.1254/fpj.22056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The synucleinopathies are neurodegenerative disease caused by abnormal accumulation of the 140-amino acid-containing protein α-synuclein (αSyn), including Parkinson's disease (PD), diffuse Lewy body dementia (DLBD), and multiple system atrophy (MSA). In patients with PD and DLBD, αSyn is misfolded in neurons, and its aggregation forms Lewy bodies (LB) and Lewy neurites (LN). On the other hand, in patients with MSA, αSyn accumulates primarily in oligodendrocytes (OLGs) and forms glial inclusion bodies (GCIs), a typical pathological feature of MSA. We recently demonstrated a making complex between αSyn and fatty acid-binding proteins (FABPs) in synucleinopathies and received wide attention. Fatty acid-binding protein 3 (FABP3) in dopamine nerves, and fatty acid-binding protein 7 (FABP7) in glial cells promoted αSyn accumulation and aggregation, respectively and caused cell death. Here, we introduced the current studies about the role of αSyn and FABP7 in MSA and novel therapeutic approach targeting for FABP7.
Collapse
Affiliation(s)
- An Cheng
- Department of CNS drug innovation, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Kohji Fukunaga
- Department of CNS drug innovation, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
13
|
A novel fatty acid-binding protein 5 and 7 inhibitor ameliorates oligodendrocyte injury in multiple sclerosis mouse models. EBioMedicine 2021; 72:103582. [PMID: 34624687 PMCID: PMC8502714 DOI: 10.1016/j.ebiom.2021.103582] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disease characterised by the demyelination of mature oligodendrocytes in the central nervous system. Recently, several studies have indicated the vital roles of fatty acid-binding proteins (FABPs) 5 and 7 in regulating the immune response. METHODS We assessed a novel FABP5/FABP7 inhibitor, FABP ligand 6 (MF 6), as a potential therapeutic for MS therapy. In vivo, we established MOG35-55-administered experimental autoimmune encephalomyelitis (EAE) mice as an MS mouse model, followed by prophylactic and symptomatic treatment with MF 6. The therapeutic effect of MF 6 was determined using behavioural and biochemical analyses. In vitro, MF 6 effects on astrocytes and oligodendrocytes were examined using both astrocyte primary culture and KG-1C cell lines. FINDINGS Prophylactic and symptomatic MF 6 therapy reduced myelin loss and clinical EAE symptoms. Furthermore, oxidative stress levels and GFAP-positive and ionised calcium-binding adaptor protein-1-positive cells were reduced in the spinal cord of MF 6-treated mice. In addition, MF 6 attenuated lipopolysaccharide-stimulated interleukin-1β and tumour necrosis factor-α accumulation in primary astrocyte culture. Moreover, MF 6 indicated a powerful protective function for the mitochondria in the oligodendrocytes of EAE mice via FABP5 inhibition. INTERPRETATIONS MF 6 is a potent inhibitor of FABP5 and FABP7; targeted inhibition of the two proteins may confer potential therapeutic effects in MS via immune inhibition and oligodendrocyte protection. FUNDING This work was supported by the Strategic Research Program for Brain Sciences from the Japan Agency for Medical Research and Development (JP17dm0107071, JP18dm0107071, JP19dm0107071, and JP20dm0107071).
Collapse
|
14
|
Zhang J, Liu L, Zhang L, Chen S, Chen Y, Cai C. Targeted fatty acid metabolomics to discover Parkinson's disease associated metabolic alteration. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4781. [PMID: 34523199 DOI: 10.1002/jms.4781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/31/2021] [Accepted: 08/07/2021] [Indexed: 06/13/2023]
Abstract
The pathogenesis of Parkinson's disease (PD) remains to be elucidated, and the metabolomics analysis has the potential to identify metabolic profiles that are involved in PD pathogenesis. Here we applied a target metabolomics approach to measure the plasma levels of 158 fatty acid metabolites in a discovery cohort including 42 PD patients and 54 health volunteers, and found two upregulated (arachidonic acid and 13-hydroxy-octadecatrienoic acid) and eleven down-regulated (docosahexaenoic acid, lyso-platelet-activating factor, 12-hydroxy-eicosatetraenoic acid, dihydroxy-eicosatrienoic acids, dihidroxy-octadecenoic acids, 17,18-dihydroxy-eicosatetraenoic acid, and hydroperoxy-octadecadienoic acids) metabolites as primary candidate marker of PD. A support vector machine algorithm with primary candidate marker was used in an independent validation cohort to identify PD. Arachidonic acid and 13-hydroxy-octadecatrienoic acid were evaluated as an effective tool in that area under the receiver operating characteristic curve reached 0.995 and 0.912 in the validation set for diagnosing PD from healthy volunteers. Besides, the sensitivity and specificity of arachidonic acid as diagnostic factor of PD in validation set were 100% and 94.10%. Similarly, the sensitivity and specificity of 13-hydroxy-octadecatrienoic acid were 100% and 82.40% for identifying PD. This target fatty acid metabolomics demonstrated a series of plasma fatty acid metabolite as PD candidate marker with high efficiency and provided insights into the understanding of PD metabolic regulation.
Collapse
Affiliation(s)
- Junjie Zhang
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Lulu Liu
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Lijiang Zhang
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Simei Chen
- Neurology Department, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Yusen Chen
- Neurology Department, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Chun Cai
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
15
|
Fatty Acid Binding Protein 3 (FABP3) and Apolipoprotein E4 (ApoE4) as Lipid Metabolism-Related Biomarkers of Alzheimer's Disease. J Clin Med 2021; 10:jcm10143009. [PMID: 34300173 PMCID: PMC8303862 DOI: 10.3390/jcm10143009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Lipid metabolism-related biomarkers gain increasing researchers interest in the field of neurodegenerative disorders. Mounting evidence have indicated the role of fatty acid-binding proteins and pathology lipid metabolism in Alzheimer’s Disease (AD). The imbalance of fatty acids (FA) and lipids may negatively affect brain functions related to neurodegenerative disorders. The ApoE4 and FABP3 proteins may reflect processes leading to neurodegeneration. This study aimed to evaluate the relationship between the CSF levels of FABP3 and ApoE4 proteins and cognitive decline as well as the diagnostic performance of these candidate biomarkers in AD and mild cognitive impairment (MCI). Methods: A total of 70 subjects, including patients with AD, MCI, and non-demented controls, were enrolled in the study. CSF concentrations of FABP3 and ApoE4 were measured using immunoassay technology. Results: Significantly higher CSF concentrations of FABP3 and ApoE4 were observed in AD patients compared to MCI subjects and individuals without cognitive impairment. Both proteins were inversely associated with Aβ42/40 ratio: ApoE4 (rho = −0.472, p < 0.001), and FABP3 (rho = −0.488, p < 0.001) in the whole study group, respectively. Additionally, FABP3 was negatively correlated with Mini-Mental State Examination score in the whole study cohort (rho = −0.585 p < 0.001). Conclusion: Presented results indicate the pivotal role of FABP3 and ApoE4 in AD pathology as lipid-related biomarkers, but studies on larger cohorts are needed.
Collapse
|
16
|
Cheng A, Jia W, Kawahata I, Fukunaga K. Impact of Fatty Acid-Binding Proteins in α-Synuclein-Induced Mitochondrial Injury in Synucleinopathy. Biomedicines 2021; 9:biomedicines9050560. [PMID: 34067791 PMCID: PMC8156290 DOI: 10.3390/biomedicines9050560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Synucleinopathies are diverse diseases with motor and cognitive dysfunction due to progressive neuronal loss or demyelination, due to oligodendrocyte loss in the brain. While the etiology of neurodegenerative disorders (NDDs) is likely multifactorial, mitochondrial injury is one of the most vital factors in neuronal loss and oligodendrocyte dysfunction, especially in Parkinson’s disease, dementia with Lewy body, multiple system atrophy, and Krabbe disease. In recent years, the abnormal accumulation of highly neurotoxic α-synuclein in the mitochondrial membrane, which leads to mitochondrial dysfunction, was well studied. Furthermore, fatty acid-binding proteins (FABPs), which are members of a superfamily and are essential in fatty acid trafficking, were reported to trigger α-synuclein oligomerization in neurons and glial cells and to target the mitochondrial outer membrane, thereby causing mitochondrial loss. Here, we provide an updated overview of recent findings on FABP and α-synuclein interactions and mitochondrial injury in NDDs.
Collapse
Affiliation(s)
- An Cheng
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
| | - Wenbin Jia
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
| | - Ichiro Kawahata
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Kohji Fukunaga
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
- Correspondence: ; Tel.: +81-(22)-795-6837
| |
Collapse
|
17
|
Guo Q, Kawahata I, Degawa T, Ikeda-Matsuo Y, Sun M, Han F, Fukunaga K. Fatty Acid-Binding Proteins Aggravate Cerebral Ischemia-Reperfusion Injury in Mice. Biomedicines 2021; 9:529. [PMID: 34068550 PMCID: PMC8150391 DOI: 10.3390/biomedicines9050529] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Fatty acid-binding proteins (FABPs) regulate the intracellular dynamics of fatty acids, mediate lipid metabolism and participate in signaling processes. However, the therapeutic efficacy of targeting FABPs as novel therapeutic targets for cerebral ischemia is not well established. Previously, we synthesized a novel FABP inhibitor, i.e., FABP ligand 6 [4-(2-(5-(2-chlorophenyl)-1-(4-isopropylphenyl)-1H-pyrazol-3-yl)-4-fluorophenoxy)butanoic acid] (referred to here as MF6). In this study, we analyzed the ability of MF6 to ameliorate transient middle cerebral artery occlusion (tMCAO) and reperfusion-induced injury in mice. A single MF6 administration (3.0 mg/kg, per os) at 0.5 h post-reperfusion effectively reduced brain infarct volumes and neurological deficits. The protein-expression levels of FABP3, FABP5 and FABP7 in the brain gradually increased after tMCAO. Importantly, MF6 significantly suppressed infarct volumes and the elevation of FABP-expression levels at 12 h post-reperfusion. MF6 also inhibited the promotor activity of FABP5 in human neuroblastoma cells (SH-SY5Y). These data suggest that FABPs elevated infarct volumes after ischemic stroke and that inhibiting FABPs ameliorated the ischemic injury. Moreover, MF6 suppressed the inflammation-associated prostaglandin E2 levels through microsomal prostaglandin E synthase-1 expression in the ischemic hemispheres. Taken together, the results imply that the FABP inhibitor MF6 can potentially serve as a neuroprotective therapeutic for ischemic stroke.
Collapse
Affiliation(s)
- Qingyun Guo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan; (Q.G.); (I.K.); (T.D.); (M.S.)
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan; (Q.G.); (I.K.); (T.D.); (M.S.)
| | - Tomohide Degawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan; (Q.G.); (I.K.); (T.D.); (M.S.)
| | - Yuri Ikeda-Matsuo
- Laboratory of Pharmacology, Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanagawa-Machi, Kanazawa 920-1181, Japan;
| | - Meiling Sun
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan; (Q.G.); (I.K.); (T.D.); (M.S.)
| | - Feng Han
- School of Pharmacy, Nanjing Medical School, Nanjing 211166, China;
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan; (Q.G.); (I.K.); (T.D.); (M.S.)
| |
Collapse
|
18
|
Fukui N, Yamamoto H, Miyabe M, Aoyama Y, Hongo K, Mizobata T, Kawahata I, Yabuki Y, Shinoda Y, Fukunaga K, Kawata Y. An α-synuclein decoy peptide prevents cytotoxic α-synuclein aggregation caused by fatty acid binding protein 3. J Biol Chem 2021; 296:100663. [PMID: 33862084 PMCID: PMC8131325 DOI: 10.1016/j.jbc.2021.100663] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022] Open
Abstract
α-synuclein (αSyn) is a protein known to form intracellular aggregates during the manifestation of Parkinson’s disease. Previously, it was shown that αSyn aggregation was strongly suppressed in the midbrain region of mice that did not possess the gene encoding the lipid transport protein fatty acid binding protein 3 (FABP3). An interaction between these two proteins was detected in vitro, suggesting that FABP3 may play a role in the aggregation and deposition of αSyn in neurons. To characterize the molecular mechanisms that underlie the interactions between FABP3 and αSyn that modulate the cellular accumulation of the latter, in this report, we used in vitro fluorescence assays combined with fluorescence microscopy, transmission electron microscopy, and quartz crystal microbalance assays to characterize in detail the process and consequences of FABP3–αSyn interaction. We demonstrated that binding of FABP3 to αSyn results in changes in the aggregation mechanism of the latter; specifically, a suppression of fibrillar forms of αSyn and also the production of aggregates with an enhanced cytotoxicity toward mice neuro2A cells. Because this interaction involved the C-terminal sequence region of αSyn, we tested a peptide derived from this region of αSyn (αSynP130-140) as a decoy to prevent the FABP3–αSyn interaction. We observed that the peptide competitively inhibited binding of αSyn to FABP3 in vitro and in cultured cells. We propose that administration of αSynP130-140 might be used to prevent the accumulation of toxic FABP3-αSyn oligomers in cells, thereby preventing the progression of Parkinson’s disease.
Collapse
Affiliation(s)
- Naoya Fukui
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Hanae Yamamoto
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Moe Miyabe
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Yuki Aoyama
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Kunihiro Hongo
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan; Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Tottori, Japan; Center for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan
| | - Tomohiro Mizobata
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan; Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Tottori, Japan; Center for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Kawata
- Department of Chemistry and Biotechnology, Faculty of Engineering/Graduate School of Engineering, Tottori University, Tottori, Japan; Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Tottori, Japan; Center for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan.
| |
Collapse
|
19
|
Sambra V, Echeverria F, Valenzuela A, Chouinard-Watkins R, Valenzuela R. Docosahexaenoic and Arachidonic Acids as Neuroprotective Nutrients throughout the Life Cycle. Nutrients 2021; 13:986. [PMID: 33803760 PMCID: PMC8003191 DOI: 10.3390/nu13030986] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/08/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
The role of docosahexaenoic acid (DHA) and arachidonic acid (AA) in neurogenesis and brain development throughout the life cycle is fundamental. DHA and AA are long-chain polyunsaturated fatty acids (LCPUFA) vital for many human physiological processes, such as signaling pathways, gene expression, structure and function of membranes, among others. DHA and AA are deposited into the lipids of cell membranes that form the gray matter representing approximately 25% of the total content of brain fatty acids. Both fatty acids have effects on neuronal growth and differentiation through the modulation of the physical properties of neuronal membranes, signal transduction associated with G proteins, and gene expression. DHA and AA have a relevant role in neuroprotection against neurodegenerative pathologies such as Alzheimer's disease and Parkinson's disease, which are associated with characteristic pathological expressions as mitochondrial dysfunction, neuroinflammation, and oxidative stress. The present review analyzes the neuroprotective role of DHA and AA in the extreme stages of life, emphasizing the importance of these LCPUFA during the first year of life and in the developing/prevention of neurodegenerative diseases associated with aging.
Collapse
Affiliation(s)
- Verónica Sambra
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (V.S.); (F.E.)
| | - Francisca Echeverria
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (V.S.); (F.E.)
| | - Alfonso Valenzuela
- Faculty of Medicine, School of Nutrition, Universidad de Los Andes, Santiago 8380000, Chile;
| | - Raphaël Chouinard-Watkins
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada;
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile; (V.S.); (F.E.)
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada;
| |
Collapse
|
20
|
Matsuo K, Kawahata I, Melki R, Bousset L, Owada Y, Fukunaga K. Suppression of α-synuclein propagation after intrastriatal injection in FABP3 null mice. Brain Res 2021; 1760:147383. [PMID: 33636166 DOI: 10.1016/j.brainres.2021.147383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 01/08/2021] [Accepted: 02/13/2021] [Indexed: 01/04/2023]
Abstract
Accumulation and aggregation of α-synuclein (αSyn) trigger neuronal loss in the substantia nigra pars compacta (SNpc), which in turn causes motor symptoms in Parkinson's disease. We previously demonstrated that fatty acid-binding protein 3 (FABP3), an intracellular fatty acid carrier protein, enhances αSyn neurotoxicity in SNpc and motor impairments after intranigral injection of αSyn fibrils. However, the temporal profile of αSyn fibril spread and their toxicity remains unclear. In the present study, we investigated the temporal profile of αSyn fibril spread and its toxicity, which induces intracellular fibril formation. Monomeric and fibrillar aSyn assemblies were labeled with ATTO550 to distinguish the exogenous form from the endogenous species and injected into bilateral striatum in Fabp3+/+ (wild type) and Fabp3-/- mice. Accumulation of both monomeric and fibrillar exogenous αSyn in the SNpc was drastically decreased in Fabp3-/- mice compared to that in the Fabp3+/+ counterparts. Deletion of Fabp3 also prevented exogenous αSyn fibril-induced seeding of the endogenous αSyn into aggregates containing phosphorylated and filamentous forms in the SNpc. Consistent with these results, loss of dopaminergic neurons and subsequent impaired motor behavior were attenuated in Fabp3-/- mice. These results highlight the crucial role of FABP3 in pathogenic αSyn accumulation and its seeding ability. Taken together, FABP3 could be a potential therapeutic target against αSyn propagation in synucleinopathies.
Collapse
Affiliation(s)
- Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| | - Ronald Melki
- CEA, Institut François Jacob (MIRcen) and CNRS, Laboratory of Neurodegenerative Diseases, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Luc Bousset
- CEA, Institut François Jacob (MIRcen) and CNRS, Laboratory of Neurodegenerative Diseases, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-0872, Japan.
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
21
|
Comprehensive metabolic profiling of Parkinson's disease by liquid chromatography-mass spectrometry. Mol Neurodegener 2021; 16:4. [PMID: 33485385 PMCID: PMC7825156 DOI: 10.1186/s13024-021-00425-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Background Parkinson’s disease (PD) is a prevalent neurological disease in the elderly with increasing morbidity and mortality. Despite enormous efforts, rapid and accurate diagnosis of PD is still compromised. Metabolomics defines the final readout of genome-environment interactions through the analysis of the entire metabolic profile in biological matrices. Recently, unbiased metabolic profiling of human sample has been initiated to identify novel PD metabolic biomarkers and dysfunctional metabolic pathways, however, it remains a challenge to define reliable biomarker(s) for clinical use. Methods We presented a comprehensive metabolic evaluation for identifying crucial metabolic disturbances in PD using liquid chromatography-high resolution mass spectrometry-based metabolomics approach. Plasma samples from 3 independent cohorts (n = 460, 223 PD, 169 healthy controls (HCs) and 68 PD-unrelated neurological disease controls) were collected for the characterization of metabolic changes resulted from PD, antiparkinsonian treatment and potential interferences of other diseases. Unbiased multivariate and univariate analyses were performed to determine the most promising metabolic signatures from all metabolomic datasets. Multiple linear regressions were applied to investigate the associations of metabolites with age, duration time and stage of PD. The combinational biomarker model established by binary logistic regression analysis was validated by 3 cohorts. Results A list of metabolites including amino acids, acylcarnitines, organic acids, steroids, amides, and lipids from human plasma of 3 cohorts were identified. Compared with HC, we observed significant reductions of fatty acids (FFAs) and caffeine metabolites, elevations of bile acids and microbiota-derived deleterious metabolites, and alterations in steroid hormones in drug-naïve PD. Additionally, we found that L-dopa treatment could affect plasma metabolome involved in phenylalanine and tyrosine metabolism and alleviate the elevations of bile acids in PD. Finally, a metabolite panel of 4 biomarker candidates, including FFA 10:0, FFA 12:0, indolelactic acid and phenylacetyl-glutamine was identified based on comprehensive discovery and validation workflow. This panel showed favorable discriminating power for PD. Conclusions This study may help improve our understanding of PD etiopathogenesis and facilitate target screening for therapeutic intervention. The metabolite panel identified in this study may provide novel approach for the clinical diagnosis of PD in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00425-8.
Collapse
|
22
|
Wang Y, Shinoda Y, Cheng A, Kawahata I, Fukunaga K. Epidermal Fatty Acid-Binding Protein 5 (FABP5) Involvement in Alpha-Synuclein-Induced Mitochondrial Injury under Oxidative Stress. Biomedicines 2021; 9:biomedicines9020110. [PMID: 33499263 PMCID: PMC7911662 DOI: 10.3390/biomedicines9020110] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
The accumulation of α-synuclein (αSyn) has been implicated as a causal factor in the pathogenesis of Parkinson’s disease (PD). There is growing evidence that supports mitochondrial dysfunction as a potential primary cause of dopaminergic neuronal death in PD. Here, we focused on reciprocal interactions between αSyn aggregation and mitochondrial injury induced by oxidative stress. We further investigated whether epidermal fatty acid-binding protein 5 (FABP5) is related to αSyn oligomerization/aggregation and subsequent disturbances in mitochondrial function in neuronal cells. In the presence of rotenone, a mitochondrial respiratory chain complex I inhibitor, co-overexpression of FABP5 with αSyn significantly decreased the viability of Neuro-2A cells compared to that of αSyn alone. Under these conditions, FABP5 co-localized with αSyn in the mitochondria, thereby reducing mitochondrial membrane potential. Furthermore, we confirmed that pharmacological inhibition of FABP5 by its ligand prevented αSyn accumulation in mitochondria, which led to cell death rescue. These results suggested that FABP5 is crucial for mitochondrial dysfunction related to αSyn oligomerization/aggregation in the mitochondria induced by oxidative stress in neurons.
Collapse
|
23
|
Dopamine D2 Long Receptors Are Critical for Caveolae-Mediated α-Synuclein Uptake in Cultured Dopaminergic Neurons. Biomedicines 2021; 9:biomedicines9010049. [PMID: 33429895 PMCID: PMC7826971 DOI: 10.3390/biomedicines9010049] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
α-synuclein accumulation into dopaminergic neurons is a pathological hallmark of Parkinson's disease. We previously demonstrated that fatty acid-binding protein 3 (FABP3) is critical for α-synuclein uptake and propagation to accumulate in dopaminergic neurons. FABP3 is abundant in dopaminergic neurons and interacts with dopamine D2 receptors, specifically the long type (D2L). Here, we investigated the importance of dopamine D2L receptors in the uptake of α-synuclein monomers and their fibrils. We employed mesencephalic neurons derived from dopamine D2L
-/-, dopamine D2 receptor null (D2 null), FABP3-/-, and wild type C57BL6 mice, and analyzed the uptake ability of fluorescence-conjugated α-synuclein monomers and fibrils. We found that D2L receptors are co-localized with FABP3. Immunocytochemistry revealed that TH+ D2L-/- or D2 null neurons do not take up α-synuclein monomers. The deletion of α-synuclein C-terminus completely abolished the uptake to dopamine neurons. Likewise, dynasore, a dynamin inhibitor, and caveolin-1 knockdown also abolished the uptake. D2L and FABP3 were also critical for α-synuclein fibrils uptake. D2L and accumulated α-synuclein fibrils were well co-localized. These data indicate that dopamine D2L with a caveola structure coupled with FABP3 is critical for α-synuclein uptake by dopaminergic neurons, suggesting a novel pathogenic mechanism of synucleinopathies, including Parkinson's disease.
Collapse
|
24
|
Jia W, Wilar G, Kawahata I, Cheng A, Fukunaga K. Impaired Acquisition of Nicotine-Induced Conditioned Place Preference in Fatty Acid-Binding Protein 3 Null Mice. Mol Neurobiol 2021; 58:2030-2045. [PMID: 33411237 DOI: 10.1007/s12035-020-02228-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Nicotine causes psychological dependence through its interactions with nicotinic acetylcholine receptors in the brain. We previously demonstrated that fatty acid-binding protein 3 (FABP3) colocalizes with dopamine D2 receptors (D2Rs) in the dorsal striatum, and FABP3 deficiency leads to impaired D2R function. Moreover, D2R null mice do not exhibit increased nicotine-induced conditioned place preference (CPP) following chronic nicotine administration. To investigate the role of FABP3 in nicotine-induced CPP, FABP3 knockout (FABP3-/-) mice were evaluated using a CPP apparatus following consecutive nicotine administration (0.5 mg/kg) for 14 days. Importantly, nicotine-induced CPP was suppressed in the conditioning, withdrawal, and relapse phases in FABP3-/- mice. To resolve the mechanisms underlying impaired nicotine-induced CPP in these mice, we assessed c-Fos expression and Ca2+/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) signaling in both dopamine D1 receptor (D1R)- and D2R-positive neurons in the nucleus accumbens (NAc). Notably, 64% of dopamine receptor-positive neurons in the mouse NAc expressed both D1R and D2R. Impaired nicotine-induced CPP was correlated with lack of responsiveness of both CaMKII and ERK phosphorylation. The number of D2R-positive neurons was increased in FABP3-/- mice, while the number of D1R-positive neurons and the responsiveness of c-Fos expression to nicotine were decreased. The aberrant c-Fos expression was closely correlated with CaMKII but not ERK phosphorylation levels in the NAc of FABP3-/- mice. Taken together, these results indicate that impaired D2R signaling due to lack of FABP3 may affect D1R and c-Fos signaling and underlie nicotine-induced CPP behaviors.
Collapse
Affiliation(s)
- Wenbin Jia
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, 980-0845, Japan
| | - Gofarana Wilar
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, 980-0845, Japan.,Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, JL. Raya Bandung-Sumedang KM 20.5 Jatinangor, Sumedang, Jawa Barat, 45363, Indonesia
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, 980-0845, Japan
| | - An Cheng
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, 980-0845, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, 980-0845, Japan. .,, Sendai, Japan.
| |
Collapse
|
25
|
Cheng A, Kawahata I, Fukunaga K. Fatty Acid Binding Protein 5 Mediates Cell Death by Psychosine Exposure through Mitochondrial Macropores Formation in Oligodendrocytes. Biomedicines 2020; 8:biomedicines8120635. [PMID: 33419250 PMCID: PMC7766880 DOI: 10.3390/biomedicines8120635] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Oligodendrocytes, the myelinating cells in the central nervous system (CNS), are critical for producing myelin throughout the CNS. The loss of oligodendrocytes is associated with multiple neurodegenerative disorders mediated by psychosine. However, the involvement of psychosine in the critical biochemical pathogenetic mechanism of the loss of oligodendrocytes and myelin in krabbe disease (KD) remains unclear. Here, we addressed how oligodendrocytes are induced by psychosine treatment in both KG-1C human oligodendroglial cells and mouse oligodendrocyte precursor cells. We found that fatty acid binding protein 5 (FABP5) expressed in oligodendrocytes accelerates mitochondria-induced glial death by inducing mitochondrial macropore formation through voltage-dependent anion channels (VDAC-1) and BAX. These two proteins mediate mitochondrial outer membrane permeabilization, thereby leading to the release of mitochondrial DNA and cytochrome C into the cytosol, and the activation of apoptotic caspases. Furthermore, we confirmed that the inhibition of FABP5 functions by shRNA and FABP5-specific ligands blocking mitochondrial macropore formation, thereby rescuing psychosine-induced oligodendrocyte death. Taken together, we identified FABP5 as a critical factor in mitochondrial injury associated with psychosine-induced apoptosis in oligodendrocytes.
Collapse
|
26
|
Motornov VA, Tabolin AA, Nelyubina YV, Nenajdenko VG, Ioffe SL. Acid‐Mediated Three Component Assembly of 4‐Fluoropyrazoles from α‐Fluoronitroalkenes, Hydrazines, and Aldehydes. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000841] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Vladimir A. Motornov
- N. D. Zelinsky Institute of Organic Chemistry Russian Academy of Sciences Leninsky prosp. 47 119991 Moscow Russia
- Higher Chemical College D. I. Mendeleev University of Chemical Technology of Russia Miusskaya sq. 9 125047 Moscow Russia
| | - Andrey A. Tabolin
- N. D. Zelinsky Institute of Organic Chemistry Russian Academy of Sciences Leninsky prosp. 47 119991 Moscow Russia
| | - Yulia V. Nelyubina
- A. N. Nesmeyanov Institute of Organoelement Compounds Russian Academy of Sciencesm Vavilov str. 28 119991 Moscow Russia
| | - Valentine G. Nenajdenko
- Department of Chemistry M. V. Lomonosov Moscow State University Leninskie Gory 1 119991 Moscow Russia
| | - Sema L. Ioffe
- N. D. Zelinsky Institute of Organic Chemistry Russian Academy of Sciences Leninsky prosp. 47 119991 Moscow Russia
| |
Collapse
|
27
|
Yabuki Y, Liu J, Kawahata I, Izumi H, Shinoda Y, Koga K, Ueno S, Shioda N, Fukunaga K. Anti-Epileptic Effects of FABP3 Ligand MF1 through the Benzodiazepine Recognition Site of the GABA A Receptor. Int J Mol Sci 2020; 21:ijms21155525. [PMID: 32752296 PMCID: PMC7432285 DOI: 10.3390/ijms21155525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 11/16/2022] Open
Abstract
Recently, we developed the fatty acid-binding protein 3 (FABP3) ligand MF1 (4-(2-(1-(2-chlorophenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy) butanoic acid) as a therapeutic candidate for α-synucleinopathies. MF1 shows affinity towards γ-aminobutyric acid type-A (GABAA) receptor, but its effect on the receptor remains unclear. Here, we investigate the pharmacological properties of MF1 on the GABAA receptor overexpressed in Neuro2A cells. While MF1 (1–100 μm) alone failed to evoke GABA currents, MF1 (1 μm) promoted GABA currents during GABA exposure (1 and 10 μm). MF1-promoted GABA currents were blocked by flumazenil (10 μm) treatment, suggesting that MF1 enhances receptor function via the benzodiazepine recognition site. Acute and chronic administration of MF1 (0.1, 0.3 and 1.0 mg/kg, p.o.) significantly attenuated status epilepticus (SE) and the mortality rate in pilocarpine (PILO: 300 mg/kg, i.p.)-treated mice, similar to diazepam (DZP: 5.0 mg/kg, i.p.). The anti-epileptic effects of DZP (5.0 mg/kg, i.p.) and MF1 (0.3 mg/kg, p.o.) were completely abolished by flumazenil (25 mg/kg, i.p.) treatment. Pentylenetetrazol (PTZ: 90 mg/kg, i.p.)-induced seizures in mice were suppressed by DZP (5.0 mg/kg, i.p.), but not MF1. Collectively, this suggests that MF1 is a mild enhancer of the GABAA receptor and exercises anti-epileptic effects through the receptor’s benzodiazepine recognition site in PILO-induced SE models.
Collapse
Affiliation(s)
- Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (Y.Y.); (J.L.); (I.K.); (H.I.); (Y.S.)
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan;
| | - Jiaqi Liu
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (Y.Y.); (J.L.); (I.K.); (H.I.); (Y.S.)
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (Y.Y.); (J.L.); (I.K.); (H.I.); (Y.S.)
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (Y.Y.); (J.L.); (I.K.); (H.I.); (Y.S.)
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (Y.Y.); (J.L.); (I.K.); (H.I.); (Y.S.)
| | - Kohei Koga
- Department of Neurophysiology, Hyogo College of Medicine, Nishinomiya 663-8501, Japan;
- Department of Neurophysiology, Graduate School of Medicine, Hirosaki University, Hirosaki 036-8216, Japan;
| | - Shinya Ueno
- Department of Neurophysiology, Graduate School of Medicine, Hirosaki University, Hirosaki 036-8216, Japan;
| | - Norifumi Shioda
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan;
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (Y.Y.); (J.L.); (I.K.); (H.I.); (Y.S.)
- Correspondence: ; Tel.: +81-22-795-6836; Fax: 81-22-795-6835
| |
Collapse
|
28
|
Kawahata I, Fukunaga K. Degradation of Tyrosine Hydroxylase by the Ubiquitin-Proteasome System in the Pathogenesis of Parkinson's Disease and Dopa-Responsive Dystonia. Int J Mol Sci 2020; 21:ijms21113779. [PMID: 32471089 PMCID: PMC7312529 DOI: 10.3390/ijms21113779] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 12/16/2022] Open
Abstract
Nigrostriatal dopaminergic systems govern physiological functions related to locomotion, and their dysfunction leads to movement disorders, such as Parkinson’s disease and dopa-responsive dystonia (Segawa disease). Previous studies revealed that expression of the gene encoding nigrostriatal tyrosine hydroxylase (TH), a rate-limiting enzyme of dopamine biosynthesis, is reduced in Parkinson’s disease and dopa-responsive dystonia; however, the mechanism of TH depletion in these disorders remains unclear. In this article, we review the molecular mechanism underlying the neurodegeneration process in dopamine-containing neurons and focus on the novel degradation pathway of TH through the ubiquitin-proteasome system to advance our understanding of the etiology of Parkinson’s disease and dopa-responsive dystonia. We also introduce the relation of α-synuclein propagation with the loss of TH protein in Parkinson’s disease as well as anticipate therapeutic targets and early diagnosis of these diseases.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Correspondence: (I.K.); (K.F.); Tel.: +81-22-795-6838 (I.K.); +81-22-795-6836 (K.F.); Fax: +81-22-795-6835 (I.K. & K.F.)
| | - Kohji Fukunaga
- Correspondence: (I.K.); (K.F.); Tel.: +81-22-795-6838 (I.K.); +81-22-795-6836 (K.F.); Fax: +81-22-795-6835 (I.K. & K.F.)
| |
Collapse
|
29
|
Shinoda Y, Wang Y, Yamamoto T, Miyachi H, Fukunaga K. Analysis of binding affinity and docking of novel fatty acid-binding protein (FABP) ligands. J Pharmacol Sci 2020; 143:264-271. [PMID: 32499096 DOI: 10.1016/j.jphs.2020.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/18/2020] [Accepted: 05/12/2020] [Indexed: 01/22/2023] Open
Abstract
Fatty acid-binding proteins (FABPs) belong to a family of proteins that transports fatty acids in the cytosol and regulates cellular functions like membrane phospholipid synthesis, lipid metabolism, and mitochondrial β oxidation. In this study, we synthesized ten novel derivatives from BMS309403, a biphenyl azole compound specific for FABP4, and analyzed their affinity and specificity for FABP3, FABP4, and FABP5, which possess 60% of homology in amino acid sequence. Here, we used 1-anilinonaphthalene 8-sulfonic acid (ANS) displacement assay and found that Ligand 1 has highest affinity for FABP3, with comparable affinity for FABP4 and FABP5. The apparent dissociation constant of BMS309403 was identical to that of arachidonic acid and docosahexaenoic acid. Docking studies with X-ray structural data showed that these novel derivatives obtained by the substitution of phenoxyacetic acid in BMS309403 but not BMS309403 have high or moderate affinity for FABP3. We further found that substitution of a phenyl group and alkyl group caused steric hindrance between 16F, the portal loop and 115L, 117L, respectively, leading to decrease in their affinity for FABPs. In conclusion, our study provides a novel strategy for development of specific ligand for each FABP.
Collapse
Affiliation(s)
- Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Japan
| | - Yifei Wang
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Japan
| | - Tetsunori Yamamoto
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Japan
| | - Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Japan.
| |
Collapse
|
30
|
Fatty Acid Binding Protein 3 Enhances the Spreading and Toxicity of α-Synuclein in Mouse Brain. Int J Mol Sci 2020; 21:ijms21062230. [PMID: 32210174 PMCID: PMC7139546 DOI: 10.3390/ijms21062230] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/07/2020] [Accepted: 03/17/2020] [Indexed: 01/08/2023] Open
Abstract
Oligomerization and/or aggregation of α-synuclein (α-Syn) triggers α-synucleinopathies such as Parkinson’s disease and dementia with Lewy bodies. It is known that α-Syn can spread in the brain like prions; however, the mechanism remains unclear. We demonstrated that fatty acid binding protein 3 (FABP3) promotes propagation of α-Syn in mouse brain. Animals were injected with mouse or human α-Syn pre-formed fibrils (PFF) into the bilateral substantia nigra pars compacta (SNpc). Two weeks after injection of mouse α-Syn PFF, wild-type (WT) mice exhibited motor and cognitive deficits, whereas FABP3 knock-out (Fabp3−/−) mice did not. The number of phosphorylated α-Syn (Ser-129)-positive cells was significantly decreased in Fabp3−/− mouse brain compared to that in WT mice. The SNpc was unilaterally infected with AAV-GFP/FABP3 in Fabp3−/− mice to confirm the involvement of FABP3 in the development of α-Syn PFF toxicity. The number of tyrosine hydroxylase (TH)- and phosphorylated α-Syn (Ser-129)-positive cells following α-Syn PFF injection significantly decreased in Fabp3−/− mice and markedly increased by AAV-GFP/FABP3 infection. Finally, we confirmed that the novel FABP3 inhibitor MF1 significantly antagonized motor and cognitive impairments by preventing α-Syn spreading following α-Syn PFF injection. Overall, FABP3 enhances α-Syn spreading in the brain following α-Syn PFF injection, and the FABP3 ligand MF1 represents an attractive therapeutic candidate for α-synucleinopathy.
Collapse
|
31
|
Haga H, Yamada R, Izumi H, Shinoda Y, Kawahata I, Miyachi H, Fukunaga K. Novel fatty acid-binding protein 3 ligand inhibits dopaminergic neuronal death and improves motor and cognitive impairments in Parkinson's disease model mice. Pharmacol Biochem Behav 2020; 191:172891. [PMID: 32126223 DOI: 10.1016/j.pbb.2020.172891] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/22/2020] [Accepted: 02/28/2020] [Indexed: 11/19/2022]
Abstract
The main symptom of Parkinson's disease (PD) is motor dysfunction and remarkably approximately 30-40% of PD patients exhibit cognitive impairments. Recently, we have developed MF8, a heart-type fatty acid-binding protein (FABP3)-specific ligand, which can inhibit α-synuclein (α-syn) oligomerization induced by arachidonic acid in FABP3 overexpressing neuro2A cells. The present study aimed to determine whether MF8 attenuates dopaminergic neuronal death and motor and cognitive impairments in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice model. MF8 can penetrate the blood-brain barrier and its peak brain concentration (21.5 ± 2.1 nM) was achieved 6 h after the oral administration (1.0 mg/kg). We also compared its effects and pharmacological action with those of L-DOPA (3,4-dihydroxy-l-phenylalanine). PD model mice were developed by administering MPTP (25 mg/kg, i.p.) once a day for five consecutive days. Twenty-four hours after the final MPTP injection, mice were administered MF8 (0.3, 1.0 mg/kg, p.o.) or L-DOPA (25 mg/kg, i.p.) once a day for 28 consecutive days and subjected to behavioral and histochemical studies. MF8 (1.0 mg/kg, p.o.), but not L-DOPA, inhibited the dopaminergic neuronal death in the ventral tegmental area and the substantia nigra pars compacta region of the MPTP-treated mice. MF8 also improved both, motor and cognitive functions, while L-DOPA ameliorated only motor dysfunction. Taken together, our results showed that MF8 attenuated the MPTP-induced dopaminergic neuronal death associated with PD pathology. We present MF8 as a novel disease-modifying therapeutic molecule for PD, which acts via a mechanism different from that of L-DOPA.
Collapse
Affiliation(s)
- Hidaka Haga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryo Yamada
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, Tokyo, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
32
|
Xu H, Diolintzi A, Storch J. Fatty acid-binding proteins: functional understanding and diagnostic implications. Curr Opin Clin Nutr Metab Care 2019; 22:407-412. [PMID: 31503024 PMCID: PMC9940447 DOI: 10.1097/mco.0000000000000600] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Fatty acid-binding proteins (FABPs) are a family of small, abundant proteins with highly tissue-specific expression patterns whose different functions remain incompletely understood. The purpose of this review is to summarize recent findings regarding FABP functions and mechanisms of action, including their potential utilization as serum markers of tissue-specific metabolic diseases. RECENT FINDINGS FABPs are important not only in their tissues of origin but also appear to influence the metabolism and function of tissues distal to their sites of expression. This may be secondary to metabolic changes in their primary tissues, and/or a result of FABP secretion from these tissues leading to effects on distal sites. Their levels in the circulation are increasingly explored as potential biomarkers for tissue-specific disease prognosis and progression. SUMMARY The nine fatty acid-binding members of the FABP family have unique tissue-specific functions and important secondary effects on tissues in which they are not expressed. For many of the FABPs, circulating levels may be indicative of disease processes related to their primary tissues, and may influence physiological function in distal tissues.
Collapse
Affiliation(s)
- Heli Xu
- Department of Nutritional Sciences, Rutgers University, New Brunswick,
- Rutgers Center for Lipid Research, New Jersey, USA
| | - Anastasia Diolintzi
- Department of Kinesiology and Health, New Jersey, USA
- Rutgers Center for Lipid Research, New Jersey, USA
| | - Judith Storch
- Department of Nutritional Sciences, Rutgers University, New Brunswick,
- Rutgers Center for Lipid Research, New Jersey, USA
| |
Collapse
|
33
|
Kawahata I, Bousset L, Melki R, Fukunaga K. Fatty Acid-Binding Protein 3 is Critical for α-Synuclein Uptake and MPP +-Induced Mitochondrial Dysfunction in Cultured Dopaminergic Neurons. Int J Mol Sci 2019; 20:ijms20215358. [PMID: 31661838 PMCID: PMC6862506 DOI: 10.3390/ijms20215358] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022] Open
Abstract
α-Synuclein is an abundant neuronal protein that accumulates in insoluble inclusions in Parkinson′s disease and other synucleinopathies. Fatty acids partially regulate α-Synuclein accumulation, and mesencephalic dopaminergic neurons highly express fatty acid-binding protein 3 (FABP3). We previously demonstrated that FABP3 knockout mice show decreased α-Synuclein oligomerization and neuronal degeneration of tyrosine hydroxylase (TH)-positive neurons in vivo. In this study, we newly investigated the importance of FABP3 in α-Synuclein uptake, 1-methyl-4-phenylpyridinium (MPP+)-induced axodendritic retraction, and mitochondrial dysfunction. To disclose the issues, we employed cultured mesencephalic neurons derived from wild type or FABP3−/− C57BL6 mice and performed immunocytochemical analysis. We demonstrated that TH+ neurons from FABP3+/+ mice take up α-Synuclein monomers while FABP3−/− TH+ neurons do not. The formation of filamentous α-Synuclein inclusions following treatment with MPP+ was observed only in FABP3+/+, and not in FABP3−/− neurons. Notably, detailed morphological analysis revealed that FABP−/− neurons did not exhibit MPP+-induced axodendritic retraction. Moreover, FABP3 was also critical for MPP+-induced reduction of mitochondrial activity and the production of reactive oxygen species. These data indicate that FABP3 is critical for α-Synuclein uptake in dopaminergic neurons, thereby preventing synucleinopathies, including Parkinson′s disease.
Collapse
Affiliation(s)
- Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| | - Luc Bousset
- CEA, Institut François Jacob (MIRcen) and CNRS, Laboratory of Neurodegenerative Diseases, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Ronald Melki
- CEA, Institut François Jacob (MIRcen) and CNRS, Laboratory of Neurodegenerative Diseases, 18 Route du Panorama, 92265 Fontenay-aux-Roses, France.
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan.
| |
Collapse
|
34
|
Fukunaga K, Matsuo K, Cheng A, Shinoda Y. Discovery of disease-modifying drug inhibiting alpha-synuclein aggregation in Lewy body dementia. IBRO Rep 2019. [DOI: 10.1016/j.ibror.2019.07.1153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
35
|
Falomir-Lockhart LJ, Cavazzutti GF, Giménez E, Toscani AM. Fatty Acid Signaling Mechanisms in Neural Cells: Fatty Acid Receptors. Front Cell Neurosci 2019; 13:162. [PMID: 31105530 PMCID: PMC6491900 DOI: 10.3389/fncel.2019.00162] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/08/2019] [Indexed: 12/15/2022] Open
Abstract
Fatty acids (FAs) are typically associated with structural and metabolic roles, as they can be stored as triglycerides, degraded by β-oxidation or used in phospholipids’ synthesis, the main components of biological membranes. It has been shown that these lipids exhibit also regulatory functions in different cell types. FAs can serve as secondary messengers, as well as modulators of enzymatic activities and substrates for cytokines synthesis. More recently, it has been documented a direct activity of free FAs as ligands of membrane, cytosolic, and nuclear receptors, and cumulative evidence has emerged, demonstrating its participation in a wide range of physiological and pathological conditions. It has been long known that the central nervous system is enriched with poly-unsaturated FAs, such as arachidonic (C20:4ω-6) or docosohexaenoic (C22:6ω-3) acids. These lipids participate in the regulation of membrane fluidity, axonal growth, development, memory, and inflammatory response. Furthermore, a whole family of low molecular weight compounds derived from FAs has also gained special attention as the natural ligands for cannabinoid receptors or key cytokines involved in inflammation, largely expanding the role of FAs as precursors of signaling molecules. Nutritional deficiencies, and alterations in lipid metabolism and lipid signaling have been associated with developmental and cognitive problems, as well as with neurodegenerative diseases. The molecular mechanism behind these effects still remains elusive. But in the last two decades, different families of proteins have been characterized as receptors mediating FAs signaling. This review focuses on different receptors sensing and transducing free FAs signals in neural cells: (1) membrane receptors of the family of G Protein Coupled Receptors known as Free Fatty Acid Receptors (FFARs); (2) cytosolic transport Fatty Acid-Binding Proteins (FABPs); and (3) transcription factors Peroxisome Proliferator-Activated Receptors (PPARs). We discuss how these proteins modulate and mediate direct regulatory functions of free FAs in neural cells. Finally, we briefly discuss the advantages of evaluating them as potential targets for drug design in order to manipulate lipid signaling. A thorough characterization of lipid receptors of the nervous system could provide a framework for a better understanding of their roles in neurophysiology and, potentially, help for the development of novel drugs against aging and neurodegenerative processes.
Collapse
Affiliation(s)
- Lisandro Jorge Falomir-Lockhart
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Gian Franco Cavazzutti
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Ezequiel Giménez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| | - Andrés Martín Toscani
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Centro Científico Tecnológico - La Plata, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP), La Plata, Argentina
| |
Collapse
|
36
|
Matsuo K, Cheng A, Yabuki Y, Takahata I, Miyachi H, Fukunaga K. Inhibition of MPTP-induced α-synuclein oligomerization by fatty acid-binding protein 3 ligand in MPTP-treated mice. Neuropharmacology 2019; 150:164-174. [PMID: 30930168 DOI: 10.1016/j.neuropharm.2019.03.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/24/2019] [Accepted: 03/25/2019] [Indexed: 10/27/2022]
Abstract
Accumulation and aggregation of α-synuclein (αSyn) triggers dopaminergic (DAergic) neuronal loss in Parkinson's disease (PD). This pathological event is partly facilitated by the presence of long-chain polyunsaturated fatty acids (LC-PUFAs), including arachidonic acid. The intracellular transport and metabolism of LC-PUFAs are mediated by fatty acid-binding proteins (FABPs). We previously reported that heart-type FABP (FABP3) interacts with αSyn, thereby promoting αSyn oligomerization in DAergic neurons in the substantia nigra pars compacta (SNpc) following 1-methyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. This αSyn oligomerization is prevented in Fabp3 gene knock out mice. We document a novel FABP3 ligand, MF1 (4-(2-(1-(2-chlorophenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy)butanoic acid), that inhibits αSyn accumulation in DA neurons, thereby inhibiting the oligomerization of αSyn, loss of DAergic neurons, and PD-like motor deficits in MPTP-treated mice. Chronic oral administration of MF1 (0.3 or 1.0 mg/kg/day) significantly improved motor impairments and inhibited MPTP-induced accumulation and oligomerization of αSyn in the SNpc, and in turn prevented loss of tyrosine hydroxylase (TH)-positive cells in the SNpc. MF1 administration (0.1, 0.3, or 1.0 mg/kg/day) also restored MPTP-induced cognitive impairments. Although chronic administration of l-DOPA (3,4-dihydroxl-l-phenylalanine; 25 mg/kg/day, i.p.) also improved motor deficits, it failed to improve the cognitive impairments. In addition, l-DOPA failed to inhibit DAergic neuronal loss and αSyn pathologies in the SNpc. In summary, the novel FABP3 ligand MF1 rescues MPTP-induced behavioural and neuropathological features, suggesting that MF1 may be a disease-modifying drug candidate for synucleinopathies.
Collapse
Affiliation(s)
- Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - An Cheng
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ibuki Takahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, Tokyo, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|