1
|
Malebary SJ, Alromema N. iDLB-Pred: identification of disordered lipid binding residues in protein sequences using convolutional neural network. Sci Rep 2024; 14:24724. [PMID: 39433833 PMCID: PMC11494137 DOI: 10.1038/s41598-024-75700-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Proteins, nucleic acids, and lipids all interact with intrinsically disordered protein areas. Lipid-binding regions are involved in a variety of biological processes as well as a number of human illnesses. The expanding body of experimental evidence for these interactions and the dearth of techniques to anticipate them from the protein sequence serve as driving forces. Although large-scale laboratory techniques are considered to be essential for equipment for studying binding residues, they are time consuming and costly, making it challenging for researchers to predict lipid binding residues. As a result, computational techniques are being looked at as a different strategy to overcome this difficulty. To predict disordered lipid-binding residues (DLBRs), we proposed iDLB-Pred predictor utilizing benchmark dataset to compute feature through extraction techniques to identify relevant patterns and information. Various classification techniques, including deep learning methods such as Convolutional Neural Networks (CNNs), Deep Neural Networks (DNNs), Multilayer Perceptrons (MLPs), Recurrent Neural Networks (RNNs), Long Short-Term Memory (LSTM) networks, and Gated Recurrent Units (GRUs), were employed for model training. The proposed model, iDLB-Pred, was rigorously validated using metrics such as accuracy, sensitivity, specificity, and Matthew's correlation coefficient. The results demonstrate the predictor's exceptional performance, achieving accuracy rates of 81% on an independent dataset and 86% in 10-fold cross-validation.
Collapse
Affiliation(s)
- Sharaf J Malebary
- Department of Information Technology, Faculty of Computing and Information Technology-Rabigh, King Abdulaziz University, P.O. Box 344, 21911, Rabigh, Saudi Arabia.
| | - Nashwan Alromema
- Department of Computer Science, Faculty of Computing and Information Technology-Rabigh, King Abdulaziz University, P.O. Box 344, 21911, Rabigh, Saudi Arabia
| |
Collapse
|
2
|
Sanluca C, Spagnolo P, Mancinelli R, De Bartolo MI, Fava M, Maccarrone M, Carotti S, Gaudio E, Leuti A, Vivacqua G. Interaction between α-Synuclein and Bioactive Lipids: Neurodegeneration, Disease Biomarkers and Emerging Therapies. Metabolites 2024; 14:352. [PMID: 39057675 PMCID: PMC11278689 DOI: 10.3390/metabo14070352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
The present review provides a comprehensive examination of the intricate dynamics between α-synuclein, a protein crucially involved in the pathogenesis of several neurodegenerative diseases, including Parkinson's disease and multiple system atrophy, and endogenously-produced bioactive lipids, which play a pivotal role in neuroinflammation and neurodegeneration. The interaction of α-synuclein with bioactive lipids is emerging as a critical factor in the development and progression of neurodegenerative and neuroinflammatory diseases, offering new insights into disease mechanisms and novel perspectives in the identification of potential biomarkers and therapeutic targets. We delve into the molecular pathways through which α-synuclein interacts with biological membranes and bioactive lipids, influencing the aggregation of α-synuclein and triggering neuroinflammatory responses, highlighting the potential of bioactive lipids as biomarkers for early disease detection and progression monitoring. Moreover, we explore innovative therapeutic strategies aimed at modulating the interaction between α-synuclein and bioactive lipids, including the development of small molecules and nutritional interventions. Finally, the review addresses the significance of the gut-to-brain axis in mediating the effects of bioactive lipids on α-synuclein pathology and discusses the role of altered gut lipid metabolism and microbiota composition in neuroinflammation and neurodegeneration. The present review aims to underscore the potential of targeting α-synuclein-lipid interactions as a multifaceted approach for the detection and treatment of neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Chiara Sanluca
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Paolo Spagnolo
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Romina Mancinelli
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | | | - Marina Fava
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Mauro Maccarrone
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Simone Carotti
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| | - Eugenio Gaudio
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | - Alessandro Leuti
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Giorgio Vivacqua
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| |
Collapse
|
3
|
Tong B, Ba Y, Li Z, Yang C, Su K, Qi H, Zhang D, Liu X, Wu Y, Chen Y, Ling J, Zhang J, Yin X, Yu P. Targeting dysregulated lipid metabolism for the treatment of Alzheimer's disease and Parkinson's disease: Current advancements and future prospects. Neurobiol Dis 2024; 196:106505. [PMID: 38642715 DOI: 10.1016/j.nbd.2024.106505] [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: 01/28/2024] [Revised: 03/02/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024] Open
Abstract
Alzheimer's and Parkinson's diseases are two of the most frequent neurological diseases. The clinical features of AD are memory decline and cognitive dysfunction, while PD mainly manifests as motor dysfunction such as limb tremors, muscle rigidity abnormalities, and slow gait. Abnormalities in cholesterol, sphingolipid, and glycerophospholipid metabolism have been demonstrated to directly exacerbate the progression of AD by stimulating Aβ deposition and tau protein tangles. Indirectly, abnormal lipids can increase the burden on brain vasculature, induce insulin resistance, and affect the structure of neuronal cell membranes. Abnormal lipid metabolism leads to PD through inducing accumulation of α-syn, dysfunction of mitochondria and endoplasmic reticulum, and ferroptosis. Great progress has been made in targeting lipid metabolism abnormalities for the treatment of AD and PD in recent years, like metformin, insulin, peroxisome proliferator-activated receptors (PPARs) agonists, and monoclonal antibodies targeting apolipoprotein E (ApoE). This review comprehensively summarizes the involvement of dysregulated lipid metabolism in the pathogenesis of AD and PD, the application of Lipid Monitoring, and emerging lipid regulatory drug targets. A better understanding of the lipidological bases of AD and PD may pave the way for developing effective prevention and treatment methods for neurodegenerative disorders.
Collapse
Affiliation(s)
- Bin Tong
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China; School of Ophthalmology and Optometry of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Yaoqi Ba
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China; School of Ophthalmology and Optometry of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Zhengyang Li
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Caidi Yang
- The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Kangtai Su
- The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Haodong Qi
- The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Deju Zhang
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Center for Clinical Precision Medicine, Jiujiang University, Jiujiang, China; Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiao Liu
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuting Wu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Jitao Ling
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Jing Zhang
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China.
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Center for Clinical Precision Medicine, Jiujiang University, Jiujiang, China.
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China.
| |
Collapse
|
4
|
Zhai W, Zhao A, Wei C, Xu Y, Cui X, Zhang Y, Meng L, Sun L. Undetected Association Between Fatty Acids and Dementia with Lewy Bodies: A Bidirectional Two-Sample Mendelian Randomization Study. J Alzheimers Dis 2024; 100:1083-1097. [PMID: 38995791 DOI: 10.3233/jad-240267] [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] [Indexed: 07/14/2024]
Abstract
Background Although observational studies indicated connections between fatty acids (FAs) and Alzheimer's disease and dementia, uncertainty persists regarding how these relationships extend to dementia with Lewy bodies (DLB). Objective To explore the potential causal relationships between FAs and the development of DLB, thus clarifying these associations using genetic instruments to infer causality. Methods We applied a two-sample Mendelian randomization (MR) and multivariable Mendelian randomization (MVMR) approach. Genetic data were obtained from a DLB cohort, comprising 2,591 cases and 4,027 controls of European descent. Eight FAs, including linoleic acid, docosahexaenoic acid, monounsaturated fatty acid, omega-3 fatty acid, omega-6 fatty acid, polyunsaturated fatty acid, saturated fatty acid, and total fatty acid, were procured from a comprehensive GWAS of metabolic biomarkers of UK Biobank, conducted by Nightingale Health in 2020 (met-d), involving 114,999 individuals. Our analysis included inverse-variance weighted, MR-Egger, weighted-median, simple mode, and weighted-mode MR estimates. Cochran's Q-statistics, MR-PRESSO, and MR-Egger intercept test were used to quantify the heterogeneity and horizontal pleiotropy of instrumental variables. Results Only linoleic acid showed a significant genetic association with the risk of developing DLB in the univariate MR. The odds ratio for linoleic acid was 1.337 with a 95% confidence interval of 1.019-1.756 (pIVW = 0.036). Results from the MVMR showed that no FAs were associated with the incidence of DLB. Conclusions The results did not support the hypothesis that FAs could reduce the risk of developing DLB. However, elucidating the relationship between FAs and DLB risk holds potential implications for informing dietary recommendations and therapeutic approaches in DLB.
Collapse
Affiliation(s)
- Weijie Zhai
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Anguo Zhao
- Department of Urology, The Fourth Affiliated Hospital of Soochow University Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yanjiao Xu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xinran Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yan Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Lingjie Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
5
|
Tyoe O, Aryal C, Diao J. Docosahexaenoic acid promotes vesicle clustering mediated by alpha-Synuclein via electrostatic interaction. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:96. [PMID: 37823961 PMCID: PMC10611297 DOI: 10.1140/epje/s10189-023-00353-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
α-Synuclein (α-Syn) is an intrinsically disordered protein whose aggregation is associated with Parkinson's disease, dementia, and other neurodegenerative diseases known as synucleinopathies. However, the functional role of α-Syn is still unclear, although it has been shown to be involved in the regulation of neurotransmitter release via the interaction with synaptic vesicles (SVs), vesicle clustering, and SNARE complex assembly. Fatty acids have significant occupancy in synaptic vesicles; and recent studies suggest the interaction of fatty acids with α-Syn affect the formation of (pathological) aggregates, but it is less clear how fatty acids affects the functional role of α-Syn including α-Syn-membrane interactions, in particular with (SV-like) vesicles. Here, we report the concentration dependent effect of docosahexaenoic acid (DHA) in synaptic-like vesicle clustering via α-Syn interaction. Through molecular dynamics simulation, we revealed that DHA promoted vesicle clustering is due to the electrostatic interaction between DHA in the membrane and the N-terminal region of α-Syn. Moreover, this increased electrostatic interaction arises from a change in the macroscopic properties of the protein-membrane interface induced by (preferential solvation of) DHA. Our results provide insight as to how DHA regulates vesicle clustering mediated by α-Syn and may further be useful to understand its physiological as well as pathological role. Description: In physiological environments, α-Synuclein (α-Syn) localizes at nerve termini and synaptic vesicles and interacts with anionic phospholipid membranes to promote vesicle clustering. Docosahexaenoic acid (DHA) increases binding affinity between α-Syn and lipid membranes by increasing electrostatic interaction energy through modulating the local and global membrane environment and conformational properties of α-Syn.
Collapse
Affiliation(s)
- Owen Tyoe
- Department of Physics, University of Cincinnati College of Arts and Sciences, Cincinnati, OH, 45221, USA
| | - Chinta Aryal
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jiajie Diao
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
6
|
Battis K, Xiang W, Winkler J. The Bidirectional Interplay of α-Synuclein with Lipids in the Central Nervous System and Its Implications for the Pathogenesis of Parkinson's Disease. Int J Mol Sci 2023; 24:13270. [PMID: 37686080 PMCID: PMC10487772 DOI: 10.3390/ijms241713270] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
The alteration and aggregation of alpha-synuclein (α-syn) play a crucial role in neurodegenerative diseases collectively termed as synucleinopathies, including Parkinson's disease (PD). The bidirectional interaction of α-syn with lipids and biomembranes impacts not only α-syn aggregation but also lipid homeostasis. Indeed, lipid composition and metabolism are severely perturbed in PD. One explanation for lipid-associated alterations may involve structural changes in α-syn, caused, for example, by missense mutations in the lipid-binding region of α-syn as well as post-translational modifications such as phosphorylation, acetylation, nitration, ubiquitination, truncation, glycosylation, and glycation. Notably, different strategies targeting the α-syn-lipid interaction have been identified and are able to reduce α-syn pathology. These approaches include the modulation of post-translational modifications aiming to reduce the aggregation of α-syn and modify its binding properties to lipid membranes. Furthermore, targeting enzymes involved in various steps of lipid metabolism and exploring the neuroprotective potential of lipids themselves have emerged as novel therapeutic approaches. Taken together, this review focuses on the bidirectional crosstalk of α-syn and lipids and how alterations of this interaction affect PD and thereby open a window for therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (K.B.); (W.X.)
| |
Collapse
|
7
|
Hnath B, Chen J, Reynolds J, Choi E, Wang J, Zhang D, Sha CM, Dokholyan NV. Big versus small: The impact of aggregate size in disease. Protein Sci 2023; 32:e4686. [PMID: 37243896 PMCID: PMC10273386 DOI: 10.1002/pro.4686] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Protein aggregation results in an array of different size soluble oligomers and larger insoluble fibrils. Insoluble fibrils were originally thought to cause neuronal cell deaths in neurodegenerative diseases due to their prevalence in tissue samples and disease models. Despite recent studies demonstrating the toxicity associated with soluble oligomers, many therapeutic strategies still focus on fibrils or consider all types of aggregates as one group. Oligomers and fibrils require different modeling and therapeutic strategies, targeting the toxic species is crucial for successful study and therapeutic development. Here, we review the role of different-size aggregates in disease, and how factors contributing to aggregation (mutations, metals, post-translational modifications, and lipid interactions) may promote oligomers opposed to fibrils. We review two different computational modeling strategies (molecular dynamics and kinetic modeling) and how they are used to model both oligomers and fibrils. Finally, we outline the current therapeutic strategies targeting aggregating proteins and their strengths and weaknesses for targeting oligomers versus fibrils. Altogether, we aim to highlight the importance of distinguishing the difference between oligomers and fibrils and determining which species is toxic when modeling and creating therapeutics for protein aggregation in disease.
Collapse
Affiliation(s)
- Brianna Hnath
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jiaxing Chen
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Joshua Reynolds
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Esther Choi
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jian Wang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Dongyan Zhang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Congzhou M. Sha
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
| | - Nikolay V. Dokholyan
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of Biochemistry & Molecular BiologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of ChemistryPenn State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
8
|
Guo Q, Kawahata I, Jia W, Wang H, Cheng A, Yabuki Y, Shioda N, Fukunaga K. α-Synuclein decoy peptide protects mice against α-synuclein-induced memory loss. CNS Neurosci Ther 2023; 29:1547-1560. [PMID: 36786129 PMCID: PMC10173724 DOI: 10.1111/cns.14120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
AIMS We previously found that a decoy peptide derived from the C-terminal sequence of α-Synuclein (αSyn) prevents cytotoxic αSyn aggregation caused by fatty acid-binding protein 3 (FABP3) in vitro. In this study, we continued to utilize αSyn-derived peptides to further validate their effects on αSyn neurotoxicity and behavioral impairments in αSyn preformed fibrils (PFFs)-injected mouse model of Parkinson's disease (PD). METHODS Mice were injected with αSyn PFFs in the bilateral olfactory bulb (OB) and then were subjected to behavioral analysis at 2-week intervals post-injection. Peptides nasal administration was initiated one week after injection. Changes in phosphorylation of αSyn and neuronal damage in the OB were measured using immunostaining at week 4. The effect of peptides on the interaction between αSyn and FABP3 was examined using co-immunoprecipitation. RESULTS αSyn PFF-injected mice showed significant memory loss but no motor function impairment. Long-term nasal treatment with peptides effectively prevented memory impairment. In peptide-treated αSyn PFF-injected mice, the peptides entered the OB smoothly through the nasal cavity and were mainly concentrated in neurons in the mitral cell layer, significantly suppressing the excessive phosphorylation of αSyn and reducing the formation of αSyn-FABP3 oligomers, thereby preventing neuronal death. The addition of peptides also blocked the interaction of αSyn and FABP3 at the recombinant protein level, and its effect was strongest at molar concentrations comparable to those of αSyn and FABP3. CONCLUSIONS Our findings suggest that the αSyn decoy peptide represents a novel therapeutic approach for reducing the accumulation of toxic αSyn-FABP3 oligomers in the brain, thereby preventing the progression of synucleinopathies.
Collapse
Affiliation(s)
- Qingyun Guo
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wenbin Jia
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haoyang Wang
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - An Cheng
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasushi Yabuki
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Norifumi Shioda
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.,BRI Pharma Incorporated, Sendai, Japan
| |
Collapse
|
9
|
Hatton SL, Pandey MK. Fat and Protein Combat Triggers Immunological Weapons of Innate and Adaptive Immune Systems to Launch Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2022; 23:1089. [PMID: 35163013 PMCID: PMC8835271 DOI: 10.3390/ijms23031089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
Parkinson's disease (PD) is the second-most common neurodegenerative disease in the world, affecting up to 10 million people. This disease mainly happens due to the loss of dopaminergic neurons accountable for memory and motor function. Partial glucocerebrosidase enzyme deficiency and the resultant excess accumulation of glycosphingolipids and alpha-synuclein (α-syn) aggregation have been linked to predominant risk factors that lead to neurodegeneration and memory and motor defects in PD, with known and unknown causes. An increasing body of evidence uncovers the role of several other lipids and their association with α-syn aggregation, which activates the innate and adaptive immune system and sparks brain inflammation in PD. Here, we review the emerging role of a number of lipids, i.e., triglyceride (TG), diglycerides (DG), glycerophosphoethanolamines (GPE), polyunsaturated fatty acids (PUFA), sphingolipids, gangliosides, glycerophospholipids (GPL), and cholesterols, and their connection with α-syn aggregation as well as the induction of innate and adaptive immune reactions that trigger neuroinflammation in PD.
Collapse
Affiliation(s)
- Shelby Loraine Hatton
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
- Department of Pediatrics, Division of Human Genetics, College of Medicine, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
10
|
Saraiva MA, Florêncio MH. Buffering capacity is determinant for restoring early α-synuclein aggregation. Biophys Chem 2022; 282:106760. [DOI: 10.1016/j.bpc.2022.106760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/28/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
|
11
|
Katuwawala A, Zhao B, Kurgan L. DisoLipPred: accurate prediction of disordered lipid-binding residues in protein sequences with deep recurrent networks and transfer learning. Bioinformatics 2021; 38:115-124. [PMID: 34487138 DOI: 10.1093/bioinformatics/btab640] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/05/2021] [Accepted: 09/02/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Intrinsically disordered protein regions interact with proteins, nucleic acids and lipids. Regions that bind lipids are implicated in a wide spectrum of cellular functions and several human diseases. Motivated by the growing amount of experimental data for these interactions and lack of tools that can predict them from the protein sequence, we develop DisoLipPred, the first predictor of the disordered lipid-binding residues (DLBRs). RESULTS DisoLipPred relies on a deep bidirectional recurrent network that implements three innovative features: transfer learning, bypass module that sidesteps predictions for putative structured residues, and expanded inputs that cover physiochemical properties associated with the protein-lipid interactions. Ablation analysis shows that these features drive predictive quality of DisoLipPred. Tests on an independent test dataset and the yeast proteome reveal that DisoLipPred generates accurate results and that none of the related existing tools can be used to indirectly identify DLBR. We also show that DisoLipPred's predictions complement the results generated by predictors of the transmembrane regions. Altogether, we conclude that DisoLipPred provides high-quality predictions of DLBRs that complement the currently available methods. AVAILABILITY AND IMPLEMENTATION DisoLipPred's webserver is available at http://biomine.cs.vcu.edu/servers/DisoLipPred/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Akila Katuwawala
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Bi Zhao
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
12
|
Simon C, Soga T, Okano HJ, Parhar I. α-Synuclein-mediated neurodegeneration in Dementia with Lewy bodies: the pathobiology of a paradox. Cell Biosci 2021; 11:196. [PMID: 34798911 PMCID: PMC8605528 DOI: 10.1186/s13578-021-00709-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is epitomized by the pathognomonic manifestation of α-synuclein-laden Lewy bodies within selectively vulnerable neurons in the brain. By virtue of prion-like inheritance, the α-synuclein protein inexorably undergoes extensive conformational metamorphoses and culminate in the form of fibrillar polymorphs, instigating calamitous damage to the brain's neuropsychological networks. This epiphenomenon is nebulous, however, by lingering uncertainty over the quasi "pathogenic" behavior of α-synuclein conformers in DLB pathobiology. Despite numerous attempts, a monolithic "α-synuclein" paradigm that is able to untangle the enigma enshrouding the clinicopathological spectrum of DLB has failed to emanate. In this article, we review conceptual frameworks of α-synuclein dependent cell-autonomous and non-autonomous mechanisms that are likely to facilitate the transneuronal spread of degeneration through the neuraxis. In particular, we describe how the progressive demise of susceptible neurons may evolve from cellular derangements perpetrated by α-synuclein misfolding and aggregation. Where pertinent, we show how these bona fide mechanisms may mutually accentuate α-synuclein-mediated neurodegeneration in the DLB brain.
Collapse
Affiliation(s)
- Christopher Simon
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Tomoko Soga
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
13
|
Shan FY, Fung KM, Zieneldien T, Kim J, Cao C, Huang JH. Examining the Toxicity of α-Synuclein in Neurodegenerative Disorders. Life (Basel) 2021; 11:life11111126. [PMID: 34833002 PMCID: PMC8621244 DOI: 10.3390/life11111126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Neurodegenerative disorders are complex disorders that display a variety of clinical manifestations. The second-most common neurodegenerative disorder is Parkinson’s disease, and the leading pathological protein of the disorder is considered to be α-synuclein. Nonetheless, α-synuclein accumulation also seems to result in multiple system atrophy and dementia with Lewy bodies. In order to obtain a more proficient understanding in the pathological progression of these synucleinopathies, it is crucial to observe the post-translational modifications of α-synuclein and the conformations of α-synuclein, as well as its role in the dysfunction of cellular pathways. Abstract α-synuclein is considered the main pathological protein in a variety of neurodegenerative disorders, such as Parkinson’s disease, multiple system atrophy, and dementia with Lewy bodies. As of now, numerous studies have been aimed at examining the post-translational modifications of α-synuclein to determine their effects on α-synuclein aggregation, propagation, and oligomerization, as well as the potential cellular pathway dysfunctions caused by α-synuclein, to determine the role of the protein in disease progression. Furthermore, α-synuclein also appears to contribute to the fibrilization of tau and amyloid beta, which are crucial proteins in Alzheimer’s disease, advocating for α-synuclein’s preeminent role in neurodegeneration. Due to this, investigating the mechanisms of toxicity of α-synuclein in neurodegeneration may lead to a more proficient understanding of the timeline progression in neurodegenerative synucleinopathies and could thereby lead to the development of potent targeted therapies.
Collapse
Affiliation(s)
- Frank Y. Shan
- Department of Anatomic Pathology, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA
- Correspondence: (F.Y.S.); (T.Z.)
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Medical Center, University of Oklahoma, Norman, OK 73019, USA;
| | - Tarek Zieneldien
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
- Correspondence: (F.Y.S.); (T.Z.)
| | - Janice Kim
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA;
| |
Collapse
|
14
|
Zoey FL, Palanivel M, Padmanabhan P, Gulyás B. Parkinson's Disease: A Nanotheranostic Approach Targeting Alpha-Synuclein Aggregation. Front Cell Dev Biol 2021; 9:707441. [PMID: 34490255 PMCID: PMC8418352 DOI: 10.3389/fcell.2021.707441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders that is implicated in aging populations. As numerous developed nations are experiencing progressively aging populations today, there is a heightened propensity for the occurrence of PD cases. Alpha-synuclein (α-syn) aggregation has been considered to be the pivotal mechanism leading to PD pathogenesis. Thus, early diagnostic and therapeutic strategies targeting the misfolded α-syn protein can potentially improve the prognosis of PD. With rapid advancements in nanotechnology in the last decade, effective solutions to various neurodegenerative and oncological diseases have been suggested. This review will explore the current innovations in nanotechnology that target the α-syn aggregation pathway, and reinstate the promise they hold as effective early diagnostic and therapeutic solutions to PD.
Collapse
Affiliation(s)
- Fong LaiGuan Zoey
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imaging Probe Development Platform, Nanyang Technological University, Singapore, Singapore
| | - Mathangi Palanivel
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imaging Probe Development Platform, Nanyang Technological University, Singapore, Singapore
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imaging Probe Development Platform, Nanyang Technological University, Singapore, Singapore
- Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, Singapore
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imaging Probe Development Platform, Nanyang Technological University, Singapore, Singapore
- Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
15
|
Manzanza NDO, Sedlackova L, Kalaria RN. Alpha-Synuclein Post-translational Modifications: Implications for Pathogenesis of Lewy Body Disorders. Front Aging Neurosci 2021; 13:690293. [PMID: 34248606 PMCID: PMC8267936 DOI: 10.3389/fnagi.2021.690293] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Lewy Body Disorders (LBDs) lie within the spectrum of age-related neurodegenerative diseases now frequently categorized as the synucleinopathies. LBDs are considered to be among the second most common form of neurodegenerative dementias after Alzheimer's disease. They are progressive conditions with variable clinical symptoms embodied within specific cognitive and behavioral disorders. There are currently no effective treatments for LBDs. LBDs are histopathologically characterized by the presence of abnormal neuronal inclusions commonly known as Lewy Bodies (LBs) and extracellular Lewy Neurites (LNs). The inclusions predominantly comprise aggregates of alpha-synuclein (aSyn). It has been proposed that post-translational modifications (PTMs) such as aSyn phosphorylation, ubiquitination SUMOylation, Nitration, o-GlcNacylation, and Truncation play important roles in the formation of toxic forms of the protein, which consequently facilitates the formation of these inclusions. This review focuses on the role of different PTMs in aSyn in the pathogenesis of LBDs. We highlight how these PTMs interact with aSyn to promote misfolding and aggregation and interplay with cell membranes leading to the potential functional and pathogenic consequences detected so far, and their involvement in the development of LBDs.
Collapse
Affiliation(s)
- Nelson de Oliveira Manzanza
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lucia Sedlackova
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Raj N. Kalaria
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
16
|
Dietary intake and plasma levels of polyunsaturated fatty acids in early-stage Parkinson's disease. Sci Rep 2021; 11:12489. [PMID: 34127758 PMCID: PMC8203700 DOI: 10.1038/s41598-021-92029-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/26/2021] [Indexed: 11/09/2022] Open
Abstract
Polyunsaturated fatty acids (PUFA) are important for neuronal function and may contribute to the development of neurodegenerative diseases. Here, we investigated the correlation between dietary intake and plasma concentrations of PUFA and their associations with clinical severity in early-stage Parkinson’s disease (PD). In a case–control study with 38 patients with PD and 33 controls, we assessed dietary intake using food frequency questionnaires and simultaneously measured the plasma levels of five PUFA. No differences were observed in dietary total energy and lipid intake, including PUFA, between patients with PD and controls. However, α-linolenic acid (ALA), linoleic acid (LA), and arachidonic acid (AA) plasma levels were lower in patients with PD. The association between dietary intake and plasma PUFA concentrations was not significant in patients with PD. ALA and LA plasma levels were inversely correlated with motor severity in patients with PD, while docosahexaenoic acid and AA plasma levels were positively correlated with non-motor symptoms after controlling for age and sex.
Collapse
|
17
|
Abstract
Life expectancy, and longevity have been increasing in recent years. However, this is, in most cases, accompanied by age-related diseases. Thus, it became essential to better understand the mechanisms inherent to aging, and to establish biomarkers that characterize this physiological process. Among all biomolecules, lipids appear to be a good target for the study of these biomarkers. In fact, some lipids have already been associated with age-related diseases. With the development of analytical techniques such as Mass Spectrometry, and Nuclear Magnetic Resonance, Lipidomics has been increasingly used to study pathological, and physiological states of an organism. Thus, the study of serum, and plasma lipidome in centenarians, and elderly individuals without age-related diseases can be a useful tool for the identification of aging biomarkers, and to understand physiological aging, and longevity. This review focus on the importance of lipids as biomarkers of aging, and summarize the changes in the lipidome that have been associated with aging, and longevity.
Collapse
|
18
|
Koçancı FG. Role of Fatty Acid Chemical Structures on Underlying Mechanisms of Neurodegenerative Diseases and Gut Microbiota. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Fatma Gonca Koçancı
- Vocational High School of Health Services Department of Medical Laboratory Techniques Alanya Alaaddin Keykubat University Alanya/Antalya 07425 Turkey
| |
Collapse
|
19
|
Manna M, Murarka RK. Polyunsaturated Fatty Acid Modulates Membrane-Bound Monomeric α-Synuclein by Modulating Membrane Microenvironment through Preferential Interactions. ACS Chem Neurosci 2021; 12:675-688. [PMID: 33538574 DOI: 10.1021/acschemneuro.0c00694] [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] [Indexed: 11/29/2022] Open
Abstract
There is ample evidence that both native functions and pathogenic aggregation of α-synuclein are intimately dependent on lipid interactions and fatty acid type; the regulatory mechanism however remains unclear. In the present work, using extensive atomistic molecular dynamics simulations and enhanced-sampling, we have focused on exploring the mechanism of fatty acid dependent regulation of monomeric α-Syn100 in a native synaptic vesicle-like membrane. Our results show that α-Syn100 spontaneously binds to the membrane through its N-terminal region (residues 1-34), where the depth of membrane insertion, the structure, and orientation of the membrane-bound α-Syn100 and its impact on membrane structure are modulated by docosahexaenoic acid (DHA). DHA is a polyunsaturated fatty acid abundantly found in the brain and known to promote the oligomerization of α-synuclein. We found that DHA exhibits marked propensity to interact with monomeric α-Syn100 and modulates the microenvironment of the protein by preferentially sorting DHA-containing phospholipids, depleting other phospholipids and cholesterol as well as increasing the proportion of anionic to neutral lipids in the immediate vicinity of the protein. Owing to the unique conformational flexibility, DHA chains form more lipid-packing defects in the membrane and efficiently coat the membrane-embedded surface of the protein, compared to the saturated and monounsaturated fatty acids. DHA thus makes the bilayer more amiable to protein adsorption and less prone to α-synuclein-induced perturbation associated with cytotoxicity. Indeed, in the absence of DHA, we observed significant thinning of the local bilayer membrane induced by α-Syn100. Though α-Syn100 is predominantly α-helical in membranes studied here, in the presence of DHA we observe formation of β-sheet/β-strands in the C-terminal region (residues 35-100) of α-Syn100, which is extended out from the membrane surface. Notably, DHA induces β structure in the NAC domain of α-Syn100 and promotes extended conformations as well as large solvent exposure of this hydrophobic domain, properties that are known to facilitate self-assembly of α-synuclein. To the best of our knowledge, this study for the first time provides the atomistic insights into DHA-induced regulatory mechanism of monomeric α-synuclein, having implications in protein structure and its physiological/pathological functions.
Collapse
Affiliation(s)
- Moutusi Manna
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh 462 066, India
- Applied Phycology and Biotechnology Division, CSIR−Central Salt & Marine Chemicals Research Institute (CSIR−CSMCRI), Gijubhai Badheka Marg, Bhavnagar, Gujarat 364002, India
| | - Rajesh K. Murarka
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh 462 066, India
| |
Collapse
|
20
|
Musteikytė G, Jayaram AK, Xu CK, Vendruscolo M, Krainer G, Knowles TPJ. Interactions of α-synuclein oligomers with lipid membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183536. [PMID: 33373595 DOI: 10.1016/j.bbamem.2020.183536] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/24/2022]
Abstract
Parkinson's disease is an increasingly prevalent and currently incurable neurodegenerative disorder. At the molecular level, this disease is characterized by the formation of aberrant intracellular protein deposits known as Lewy bodies. Oligomeric forms of the protein α-synuclein (αS), which are believed to be both intermediates and by-products of Lewy body formation, are considered to be the main pathogenic species. Interactions of such oligomers with lipid membranes are increasingly emerging as a major molecular pathway underpinning their toxicity. Here we review recent progress in our understanding of the interactions of αS oligomers with lipid membranes. We highlight key structural and biophysical features of αS oligomers, the effects of these features on αS oligomer membrane binding properties, and resultant implications for understanding the etiology of Parkinson's disease. We discuss mechanistic modes of αS oligomer-lipid membrane interactions and the effects of environmental factors to such modes. Finally, we provide an overview of the current understanding of the main molecular determinants of αS oligomer toxicity in vivo.
Collapse
Affiliation(s)
- Greta Musteikytė
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Akhila K Jayaram
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Catherine K Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Georg Krainer
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom; Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, United Kingdom.
| |
Collapse
|
21
|
Nuber S, Nam AY, Rajsombath MM, Cirka H, Hronowski X, Wang J, Hodgetts K, Kalinichenko LS, Müller CP, Lambrecht V, Winkler J, Weihofen A, Imberdis T, Dettmer U, Fanning S, Selkoe DJ. A Stearoyl-Coenzyme A Desaturase Inhibitor Prevents Multiple Parkinson Disease Phenotypes in α-Synuclein Mice. Ann Neurol 2020; 89:74-90. [PMID: 32996158 PMCID: PMC7756464 DOI: 10.1002/ana.25920] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/20/2022]
Abstract
Objective Parkinson disease (PD) has useful symptomatic treatments that do not slow the neurodegenerative process, and no significant disease‐modifying treatments are approved. A key therapeutic target in PD is α‐synuclein (αS), which is both genetically implicated and accumulates in Lewy bodies rich in vesicles and other lipid membranes. Reestablishing αS homeostasis is a central goal in PD. Based on previous lipidomic analyses, we conducted a mouse trial of a stearoyl–coenzyme A desaturase (SCD) inhibitor (“5b”) that prevented αS‐positive vesicular inclusions and cytotoxicity in cultured human neurons. Methods Oral dosing and brain activity of 5b were established in nontransgenic mice. 5b in drinking water was given to mice expressing wild‐type human αS (WT) or an amplified familial PD αS mutation (E35K + E46K + E61K ["3K"]) beginning near the onset of nigral and cortical neurodegeneration and the robust PD‐like motor syndrome in 3K. Motor phenotypes, brain cytopathology, and SCD‐related lipid changes were quantified in 5b‐ versus placebo‐treated mice. Outcomes were compared to effects of crossing 3K to SCD1−/− mice. Results 5b treatment reduced αS hyperphosphorylation in E46K‐expressing human neurons, in 3K neural cultures, and in both WT and 3K αS mice. 5b prevented subtle gait deficits in WT αS mice and the PD‐like resting tremor and progressive motor decline of 3K αS mice. 5b also increased αS tetramers and reduced proteinase K‐resistant lipid‐rich aggregates. Similar benefits accrued from genetically deleting 1 SCD allele, providing target validation. Interpretation Prolonged reduction of brain SCD activity prevented PD‐like neuropathology in multiple PD models. Thus, an orally available SCD inhibitor potently ameliorates PD phenotypes, positioning this approach to treat human α‐synucleinopathies. ANN NEUROL 2021;89:74–90
Collapse
Affiliation(s)
- Silke Nuber
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alice Y Nam
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Molly M Rajsombath
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Haley Cirka
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Junmin Wang
- Chemical Biology & Proteomics, Biogen, Cambridge, MA, USA
| | - Kevin Hodgetts
- Laboratory for Drug Discovery in Neurodegeneration, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Vera Lambrecht
- Division of Molecular Neurology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Jürgen Winkler
- Division of Molecular Neurology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Weihofen
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | - Thibaut Imberdis
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Xylaki M, Boumpoureka I, Kokotou MG, Marras T, Papadimitriou G, Kloukina I, Magrioti V, Kokotos G, Vekrellis K, Emmanouilidou E. Changes in the cellular fatty acid profile drive the proteasomal degradation of α-synuclein and enhance neuronal survival. FASEB J 2020; 34:15123-15145. [PMID: 32931072 DOI: 10.1096/fj.202001344r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 01/04/2023]
Abstract
Parkinson's disease is biochemically characterized by the deposition of aberrant aggregated α-synuclein in the affected neurons. The aggregation properties of α-synuclein greatly depend on its affinity to bind cellular membranes via a dynamic interaction with specific lipid moieties. In particular, α-synuclein can interact with arachidonic acid (AA), a polyunsaturated fatty acid, in a manner that promotes the formation of α-helix enriched assemblies. In a cellular context, AA is released from membrane phospholipids by phospholipase A2 (PLA2 ). To investigate the impact of PLA2 activity on α-synuclein aggregation, we have applied selective PLA2 inhibitors to a SH-SY5Y cellular model where the expression of human wild-type α-synuclein is correlated with a gradual accumulation of soluble oligomers and subsequent cell death. We have found that pharmacological and genetic inhibition of GIVA cPLA2 resulted in a dramatic decrease of intracellular oligomeric and monomeric α-synuclein significantly promoting cell survival. Our data suggest that alterations in the levels of free fatty acids, and especially AA and adrenic acid, promote the formation of α-synuclein conformers which are more susceptible to proteasomal degradation. This mechanism is active only in living cells and is generic since it does not depend on the absolute quantity of α-synuclein, the presence of disease-linked point mutations, the expression system or the type of cells. Our findings indicate that the α-synuclein-fatty acid interaction can be a critical determinant of the conformation and fate of α-synuclein in the cell interior and, as such, cPLA2 inhibitors could serve to alleviate the intracellular, potentially pathological, α-synuclein burden.
Collapse
Affiliation(s)
- Mary Xylaki
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioanna Boumpoureka
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maroula G Kokotou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Marras
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Papadimitriou
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Ismini Kloukina
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Victoria Magrioti
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Vekrellis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelia Emmanouilidou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
23
|
Sengupta U, Puangmalai N, Bhatt N, Garcia S, Zhao Y, Kayed R. Polymorphic α-Synuclein Strains Modified by Dopamine and Docosahexaenoic Acid Interact Differentially with Tau Protein. Mol Neurobiol 2020; 57:2741-2765. [PMID: 32350746 PMCID: PMC7253398 DOI: 10.1007/s12035-020-01913-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/30/2020] [Indexed: 12/22/2022]
Abstract
The pathological hallmark of synucleinopathies, including Parkinson’s disease (PD), is the aggregation of α-synuclein (α-Syn) protein. Even so, tau protein pathology is abundantly found in these diseases. Both α-Syn and tau can exist as polymorphic aggregates, a phenomenon that has been widely studied, mostly in their fibrillar assemblies. We have previously discovered that in addition to α-Syn oligomers, oligomeric tau is also present in the brain tissues of patients with PD and dementia with Lewy bodies (DLB). However, the effect of interaction between polymorphic α-Syn oligomers and tau has not been scrupulously studied. Here, we have explored the structural and functional diversity of distinct α-Syn oligomers, prepared by modifying the protein with dopamine (DA) and docosahexaenoic acid (DHA). The two α-Syn oligomers differed in aggregate size, conformation, sensitivity to proteinase K digestion, tryptic digestion, and toxicity, suggesting them as distinct α-Syn oligomeric strains. We examined their internalization mechanisms in primary neurons and seeding propensity in inducing α-Syn aggregation. Using a combined approach of molecular and cellular techniques, we observed that the tau aggregates cross-seeded with the individual α-Syn oligomeric strains differed in their biochemical and biological properties, suggesting two distinct tau strains. The tau aggregate cross-seeded with the DA-modified α-Syn oligomeric strain possessed a potent intracellular tau seeding propensity. This study provides a comprehensive analysis of unique strain-specific interaction between oligomeric α-Syn and tau. Furthermore, this study allows us to speculate that distinct α-Syn-tau interactions inducing tau aggregation might be an underlying mechanism of neurodegeneration in PD.
Collapse
Affiliation(s)
- Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, USA
| | - Stephanie Garcia
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, USA
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, USA
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, USA.
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, USA.
| |
Collapse
|
24
|
Davidi D, Schechter M, Elhadi SA, Matatov A, Nathanson L, Sharon R. α-Synuclein Translocates to the Nucleus to Activate Retinoic-Acid-Dependent Gene Transcription. iScience 2020; 23:100910. [PMID: 32120069 PMCID: PMC7052517 DOI: 10.1016/j.isci.2020.100910] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/06/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
α-Synuclein (α-Syn) protein is implicated in the pathogenesis of Parkinson disease (PD). It is primarily cytosolic and interacts with cell membranes. α-Syn also occurs in the nucleus. Here we investigated the mechanisms involved in nuclear translocation of α-Syn. We analyzed alterations in gene expression following induced α-Syn expression in SH-SY5Y cells. Analysis of upstream regulators pointed at alterations in transcription activity of retinoic acid receptors (RARs) and additional nuclear receptors. We show that α-Syn binds RA and translocates to the nucleus to selectively enhance gene transcription. Nuclear translocation of α-Syn is regulated by calreticulin and is leptomycin-B independent. Importantly, nuclear translocation of α-Syn following RA treatment enhances its toxicity in cultured neurons and the expression levels of PD-associated genes, including ATPase cation transporting 13A2 (ATP13A2) and PTEN-induced kinase1 (PINK1). The results link a physiological role for α-Syn in the regulation of RA-mediated gene transcription and its toxicity in the synucleinopathies.
Collapse
Affiliation(s)
- Dana Davidi
- Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Meir Schechter
- Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Suaad Abd Elhadi
- Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Adar Matatov
- Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel
| | - Lubov Nathanson
- Institute for Neuro Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Ronit Sharon
- Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, 9112001 Jerusalem, Israel.
| |
Collapse
|
25
|
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: 28] [Impact Index Per Article: 5.6] [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
|
26
|
Ugalde CL, Lawson VA, Finkelstein DI, Hill AF. The role of lipids in α-synuclein misfolding and neurotoxicity. J Biol Chem 2019; 294:9016-9028. [PMID: 31064841 DOI: 10.1074/jbc.rev119.007500] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The misfolding and aggregation of α-synuclein (αsyn) in the central nervous system is associated with a group of neurodegenerative disorders referred to as the synucleinopathies. In addition to being a pathological hallmark of disease, it is now well-established that upon misfolding, αsyn acquires pathogenic properties, such as neurotoxicity, that can contribute to disease development. The mechanisms that produce αsyn misfolding and the molecular events underlying the neuronal damage caused by these misfolded species are not well-defined. A consistent observation that may be relevant to αsyn's pathogenicity is its ability to associate with lipids. This appears important not only to how αsyn aggregates, but also to the mechanism by which the misfolded protein causes intracellular damage. This review discusses the current literature reporting a role of lipids in αsyn misfolding and neurotoxicity in various synucleinopathy disorders and provides an overview of current methods to assess protein misfolding and pathogenicity both in vitro and in vivo.
Collapse
Affiliation(s)
- Cathryn L Ugalde
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086, Australia, .,the Departments of Microbiology and Immunology and.,the Howard Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia, and
| | | | - David I Finkelstein
- the Howard Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Andrew F Hill
- From the Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3086, Australia, .,Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia, and
| |
Collapse
|
27
|
Alza NP, Iglesias González PA, Conde MA, Uranga RM, Salvador GA. Lipids at the Crossroad of α-Synuclein Function and Dysfunction: Biological and Pathological Implications. Front Cell Neurosci 2019; 13:175. [PMID: 31118888 PMCID: PMC6504812 DOI: 10.3389/fncel.2019.00175] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/11/2019] [Indexed: 12/15/2022] Open
Abstract
Since its discovery, the study of the biological role of α-synuclein and its pathological implications has been the subject of increasing interest. The propensity to adopt different conformational states governing its aggregation and fibrillation makes this small 14-kDa cytosolic protein one of the main etiologic factors associated with degenerative disorders known as synucleinopathies. The structure, function, and toxicity of α-synuclein and the possibility of different therapeutic approaches to target the protein have been extensively investigated and reviewed. One intriguing characteristic of α-synuclein is the different ways in which it interacts with lipids. Though in-depth studies have been carried out in this field, the information they have produced is puzzling and the precise role of lipids in α-synuclein biology and pathology and vice versa is still largely unknown. Here we provide an overview and discussion of the main findings relating to α-synuclein/lipid interaction and its involvement in the modulation of lipid metabolism and signaling.
Collapse
Affiliation(s)
- Natalia P Alza
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional del Sur, Bahía Blanca, Argentina.,Departamento de Química, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Pablo A Iglesias González
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Melisa A Conde
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional del Sur, Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Romina M Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional del Sur, Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Gabriela A Salvador
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional del Sur, Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| |
Collapse
|
28
|
Alecu I, Bennett SAL. Dysregulated Lipid Metabolism and Its Role in α-Synucleinopathy in Parkinson's Disease. Front Neurosci 2019; 13:328. [PMID: 31031582 PMCID: PMC6470291 DOI: 10.3389/fnins.2019.00328] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 12/23/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, the main pathological hallmark of which is the accumulation of α-synuclein (α-syn) and the formation of filamentous aggregates called Lewy bodies in the brainstem, limbic system, and cortical areas. Lipidomics is a newly emerging field which can provide fresh insights and new answers that will enhance our capacity for early diagnosis, tracking disease progression, predicting critical endpoints, and identifying risk in pre-symptomatic persons. In recent years, lipids have been implicated in many aspects of PD pathology. Biophysical and lipidomic studies have demonstrated that α-syn binds preferentially not only to specific lipid families but also to specific molecular species and that these lipid-protein complexes enhance its interaction with synaptic membranes, influence its oligomerization and aggregation, and interfere with the catalytic activity of cytoplasmic lipid enzymes and lysosomal lipases, thereby affecting lipid metabolism. The genetic link between aberrant lipid metabolism and PD is even more direct, with mutations in GBA and SMPD1 enhancing PD risk in humans and loss of GALC function increasing α-syn aggregation and accumulation in experimental murine models. Moreover, a number of lipidomic studies have reported PD-specific lipid alterations in both patient brains and plasma, including alterations in the lipid composition of lipid rafts in the frontal cortex. A further aspect of lipid dysregulation promoting PD pathogenesis is oxidative stress and inflammation, with proinflammatory lipid mediators such as platelet activating factors (PAFs) playing key roles in arbitrating the progressive neurodegeneration seen in PD linked to α-syn intracellular trafficking. Lastly, there are a number of genetic risk factors of PD which are involved in normal lipid metabolism and function. Genes such as PLA2G6 and SCARB2, which are involved in glycerophospholipid and sphingolipid metabolism either directly or indirectly are associated with risk of PD. This review seeks to describe these facets of metabolic lipid dysregulation as they relate to PD pathology and potential pathomechanisms involved in disease progression, while highlighting incongruous findings and gaps in knowledge that necessitate further research.
Collapse
Affiliation(s)
- Irina Alecu
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, Centre for Catalysis and Research Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Steffany A. L. Bennett
- Neural Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, Centre for Catalysis and Research Innovation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
29
|
Deryusheva E, Nemashkalova E, Galloux M, Richard C, Eléouët J, Kovacs D, Belle K, Tompa P, Uversky V, Permyakov S. Does Intrinsic Disorder in Proteins Favor Their Interaction with Lipids? Proteomics 2019; 19:e1800098. [DOI: 10.1002/pmic.201800098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 12/09/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Evgenia Deryusheva
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
| | - Ekaterina Nemashkalova
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
| | - Marie Galloux
- VIM, INRAUniversité Paris‐Saclay Jouy‐en‐Josas 78350 France
| | | | | | - Denis Kovacs
- VIB, Structural Biology Research CenterVrije Universiteit Brussel Brussels 1050 Belgium
| | - Karo Belle
- VIB, Structural Biology Research CenterVrije Universiteit Brussel Brussels 1050 Belgium
| | - Peter Tompa
- VIB, Structural Biology Research CenterVrije Universiteit Brussel Brussels 1050 Belgium
- Institute of EnzymologyResearch Centre for Natural Sciences of the Hungarian Academy of Sciences Budapest 1117 Hungary
| | - Vladimir Uversky
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research InstituteMorsani College of MedicineUniversity of South Florida Tampa FL 33612 USA
| | - Sergei Permyakov
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
| |
Collapse
|
30
|
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by a progressive loss of dopaminergic neurons from the nigrostriatal pathway, formation of Lewy bodies, and microgliosis. During the past decades multiple cellular pathways have been associated with PD pathology (i.e., oxidative stress, endosomal-lysosomal dysfunction, endoplasmic reticulum stress, and immune response), yet disease-modifying treatments are not available. We have recently used genetic data from familial and sporadic cases in an unbiased approach to build a molecular landscape for PD, revealing lipids as central players in this disease. Here we extensively review the current knowledge concerning the involvement of various subclasses of fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterols, and lipoproteins in PD pathogenesis. Our review corroborates a central role for most lipid classes, but the available information is fragmented, not always reproducible, and sometimes differs by sex, age or PD etiology of the patients. This hinders drawing firm conclusions about causal or associative effects of dietary lipids or defects in specific steps of lipid metabolism in PD. Future technological advances in lipidomics and additional systematic studies on lipid species from PD patient material may improve this situation and lead to a better appreciation of the significance of lipids for this devastating disease.
Collapse
|
31
|
Abstract
Peroxisomes are key metabolic organelles, which contribute to cellular lipid metabolism, e.g. the β-oxidation of fatty acids and the synthesis of myelin sheath lipids, as well as cellular redox balance. Peroxisomal dysfunction has been linked to severe metabolic disorders in man, but peroxisomes are now also recognized as protective organelles with a wider significance in human health and potential impact on a large number of globally important human diseases such as neurodegeneration, obesity, cancer, and age-related disorders. Therefore, the interest in peroxisomes and their physiological functions has significantly increased in recent years. In this review, we intend to highlight recent discoveries, advancements and trends in peroxisome research, and present an update as well as a continuation of two former review articles addressing the unsolved mysteries of this astonishing organelle. We summarize novel findings on the biological functions of peroxisomes, their biogenesis, formation, membrane dynamics and division, as well as on peroxisome-organelle contacts and cooperation. Furthermore, novel peroxisomal proteins and machineries at the peroxisomal membrane are discussed. Finally, we address recent findings on the role of peroxisomes in the brain, in neurological disorders, and in the development of cancer.
Collapse
Affiliation(s)
- Markus Islinger
- Institute of Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, Medical Faculty Manheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Alfred Voelkl
- Institute for Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | - H Dariush Fahimi
- Institute for Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | | |
Collapse
|
32
|
Schommer J, Marwarha G, Nagamoto-Combs K, Ghribi O. Palmitic Acid-Enriched Diet Increases α-Synuclein and Tyrosine Hydroxylase Expression Levels in the Mouse Brain. Front Neurosci 2018; 12:552. [PMID: 30127714 PMCID: PMC6087752 DOI: 10.3389/fnins.2018.00552] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/20/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Accumulation of the α-synuclein (α-syn) protein and depletion of dopaminergic neurons in the substantia nigra are hallmarks of Parkinson's disease (PD). Currently, α-syn is under scrutiny as a potential pathogenic factor that may contribute to dopaminergic neuronal death in PD. However, there is a significant gap in our knowledge on what causes α-syn to accumulate and dopaminergic neurons to die. It is now strongly suggested that the nature of our dietary intake influences both epigenetic changes and disease-related genes and may thus potentially increase or reduce our risk of developing PD. Objective: In this study, we determined the extent to which a 3 month diet enriched in the saturated free fatty acid palmitate (PA) influences levels of α-syn and tyrosine hydroxylase, the rate limiting enzyme in dopamine synthesis in mice brains. Methods: We fed the m-Thy1-αSyn (m-Thy1) mouse model for PD and its matched control, the B6D2F1/J (B6D2) mouse a PA-enriched diet or a normal diet for 3 months. Levels of α-syn, tyrosine hydroxylase, and the biogenic amines dopamine and dopamine metabolites, serotonin and noradrenaline were determined. Results: We found that the PA-enriched diet induces an increase in α-syn and TH protein and mRNA expression levels in m-Thy1 transgenic mice. We also show that, while it didn't affect levels of biogenic amine content in the B6D2 mice, the PA-enriched diet significantly reduces dopamine metabolites and increases the level of serotonin in m-Thy1 mice. Conclusion: Altogether, our results demonstrate that a diet rich in the saturated fatty acid palmitate can modulate levels of α-syn, TH, dopamine, and serotonin which all are proteins and neurochemicals that play key roles in increasing or reducing the risk for many neurodegenerative diseases including PD.
Collapse
Affiliation(s)
- Jared Schommer
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Kumi Nagamoto-Combs
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
33
|
Fecchio C, Palazzi L, de Laureto PP. α-Synuclein and Polyunsaturated Fatty Acids: Molecular Basis of the Interaction and Implication in Neurodegeneration. Molecules 2018; 23:molecules23071531. [PMID: 29941855 PMCID: PMC6099649 DOI: 10.3390/molecules23071531] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/19/2018] [Accepted: 06/23/2018] [Indexed: 12/31/2022] Open
Abstract
α-Synuclein (α-syn) is a 140-amino acid protein, the physiological function of which has yet to be clarified. It is involved in several neurodegenerative disorders, and the interaction of the protein with brain lipids plays an important role in the pathogenesis of Parkinson’s disease (PD). Polyunsaturated fatty acids (PUFA) are highly abundant in the brain where they play critical roles in neuronal membrane fluidity and permeability, serve as energy reserves and function as second messengers in cell signaling. PUFA concentration and composition in the brain are altered with age when also an increase of lipid peroxidation is observed. Considering that PD is clearly correlated with oxidative stress, PUFA abundance and composition became of great interest in neurodegeneration studies because of PUFA’s high propensity to oxidize. The high levels of the PUFA docosahexaenoic acid (DHA) in brain areas containing α-syn inclusions in patients with PD further support the hypothesis of possible interactions between α-syn and DHA. Additionally, a possible functional role of α-syn in sequestering the early peroxidation products of fatty acids was recently proposed. Here, we provide an overview of the current knowledge regarding the molecular interactions between α-syn and fatty acids and the effect exerted by the protein on their oxidative state. We highlight recent findings supporting a neuroprotective role of the protein, linking α-syn, altered lipid composition in neurodegenerative disorders and PD development.
Collapse
Affiliation(s)
- Chiara Fecchio
- Department of Biomedical Sciences, University of Padova; Padova 35131, Italy.
| | - Luana Palazzi
- Department of Pharmaceutical and Pharmacological Sciences, CRIBI, University of Padova; Padova 35131, Italy.
| | | |
Collapse
|
34
|
Seipin deficiency in mice causes loss of dopaminergic neurons via aggregation and phosphorylation of α-synuclein and neuroinflammation. Cell Death Dis 2018; 9:440. [PMID: 29670081 PMCID: PMC5906676 DOI: 10.1038/s41419-018-0471-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/12/2018] [Accepted: 03/09/2018] [Indexed: 12/16/2022]
Abstract
Seipin gene is originally found in type 2 congenital generalized lipodystrophy (CGL2) to involve lipid droplet formation. Recently, decrease of seipin expression is reported in substantia nigra of Parkinson’s disease patients. Dopaminergic neurons in substantia nigra pars compacta expressed the seipin protein. The objective of this study is to investigate influence of the seipin deficiency on dopaminergic neurons and motor behaviors. Neuronal seipin knockout (seipin-nKO) mice (3–12 months of age) displayed an age-related deficit in motor coordination. The number of dopaminergic neurons in seipin-nKO mice was age dependently reduced with increase in cleaved caspase-3. The levels of αSyn oligomers and oligomer phosphorylation (S129), but not αSyn monomers, were elevated in dopaminergic neurons and substantia nigra of seipin-nKO mice. The PPARγ expression in seipin-nKO mice was reduced. In seipin-nKO mice, the phosphorylation of GSK3β was increased at Tyr216 and was reduced at Ser9, which was corrected by the PPARγ agonist rosiglitazone. The increased IL-6 level in seipin-nKO mice was sensitive to rosiglitazone and GSK3β inhibitor AR-A014418. The enhanced phosphorylation of αSyn was prevented by rosiglitazone and AR-A014418, while the increase in αSyn oligomers was corrected only by rosiglitazone. The treatment of seipin-nKO mice with rosiglitazone and AR-A014418 rescued the death of dopaminergic neurons, which was accompanied by the improvement of motor coordination. Therefore, the results indicate that seipin deficiency causes an age-related loss of dopaminergic neurons and impairment of motor coordination through reducing PPARγ to enhance aggregation and phosphorylation of αSyn and neuroinflammation.
Collapse
|
35
|
Galvagnion C. The Role of Lipids Interacting with α-Synuclein in the Pathogenesis of Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2018; 7:433-450. [PMID: 28671142 DOI: 10.3233/jpd-171103] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
α-synuclein is a small protein abundantly expressed in the brain and mainly located in synaptic terminals. The conversion of α-synuclein into oligomers and fibrils is the hallmark of a range of neurodegenerative disorders including Parkinson's disease and dementia with Lewy bodies. α-synuclein is disordered in solution but can adopt an α-helical conformation upon binding to lipid membranes. This lipid-protein interaction plays an important role in its proposed biological function, i.e., synaptic plasticity, but can also entail the aggregation of the protein. Both the chemical properties of the lipids and the lipid-to-protein-ratio have been reported to modulate the aggregation propensity of α-synuclein. In this review, the influence of changes in the nature and levels of lipids on the aggregation propensity of α-synuclein in vivo and in vitro will be discussed within a common general framework. In particular, while biophysical measurements and kinetic analyses of the time courses of α-synuclein aggregation in the presence of different types of lipid vesicles allow a mechanistic dissection of the influence of the lipids on α-synuclein aggregation, biological studies of cellular and animal models of Parkinson's disease allow the determination of changes in lipid levels and properties associated with the disease.
Collapse
Affiliation(s)
- Céline Galvagnion
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Physical Biology, Heinrich Heine Universität, Düsseldorf, Germany
| |
Collapse
|
36
|
Rawat A, Langen R, Varkey J. Membranes as modulators of amyloid protein misfolding and target of toxicity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1863-1875. [PMID: 29702073 DOI: 10.1016/j.bbamem.2018.04.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/19/2018] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
Abnormal protein aggregation is a hallmark of various human diseases. α-Synuclein, a protein implicated in Parkinson's disease, is found in aggregated form within Lewy bodies that are characteristically observed in the brains of PD patients. Similarly, deposits of aggregated human islet amyloid polypeptide (IAPP) are found in the pancreatic islets in individuals with type 2 diabetes mellitus. Significant number of studies have focused on how monomeric, disaggregated proteins transition into various amyloid structures leading to identification of a vast number of aggregation promoting molecules and processes over the years. Inasmuch as these factors likely enhance the formation of toxic, misfolded species, they might act as risk factors in disease. Cellular membranes, and particularly certain lipids, are considered to be among the major players for aggregation of α-synuclein and IAPP, and membranes might also be the target of toxicity. Past studies have utilized an array of biophysical tools, both in vitro and in vivo, to expound the membrane-mediated aggregation. Here, we focus on membrane interaction of α-synuclein and IAPP, and how various kinds of membranes catalyze or modulate the aggregation of these proteins and how, in turn, these proteins disrupt membrane integrity, both in vitro and in vivo. The membrane interaction and subsequent aggregation has been briefly contrasted to aggregation of α-synuclein and IAPP in solution. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
Affiliation(s)
- Anoop Rawat
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, United States
| | - Ralf Langen
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, United States.
| | - Jobin Varkey
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
37
|
Impact of DHA intake in a mouse model of synucleinopathy. Exp Neurol 2017; 301:39-49. [PMID: 29229294 DOI: 10.1016/j.expneurol.2017.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/26/2017] [Accepted: 12/06/2017] [Indexed: 11/22/2022]
Abstract
Polyunsaturated fatty acids omega-3 (n-3 PUFA), such as docosahexaenoic acid (DHA), have been shown to prevent, and partially reverse, neurotoxin-induced nigrostriatal denervation in animal models of Parkinson's disease (PD). However, the accumulation of α-synuclein (αSyn) in cerebral tissues is equally important to the pathophysiology. To determine whether DHA intake improves various aspects related to synucleinopathy, ninety male mice overexpressing human αSyn under the Thy-1 promoter (Thy1-αSyn) were fed one of three diets (specially formulated control, low n-3 PUFA or high DHA) and compared to non-transgenic C57/BL6 littermate mice exposed to a control diet. Thy1-αSyn mice displayed impaired motor skills, lower dopaminergic neuronal counts within the substantia nigra (-13%) in parallel to decreased levels of the striatal dopamine transporter (DAT) (-24%), as well as reduced NeuN (-41%) and synaptic proteins PSD-95 (-51%), synaptophysin (-80%) and vesicular acetylcholine transporter (VChAT) (-40%) in the cerebral cortex compared to C57/BL6 mice. However, no significant difference in dopamine concentrations was observed by HPLC analysis between Thy1-αSyn and non-transgenic C57/BL6 littermates under the control diet. The most striking finding was a favorable effect of DHA on the survival/longevity of Thy1-αSyn mice (+51% survival rate at 12months of age). However, dietary DHA supplementation did not have a significant effect on other parameters examined in this study, despite increased striatal dopamine concentrations. While human αSyn monomers and oligomers were detected in the cortex of Thy1-αSyn mice, the effects of the diets were limited to a small increase of 42kDa oligomers in insoluble protein fractions upon n-3 PUFA deprivation. Overall, our data indicate that a diet rich in n-3 PUFA has a beneficial effect on the longevity of a murine model of α-synucleinopathy without a major impact on the dopamine system and motor impairments, nor αSyn levels.
Collapse
|
38
|
De Franceschi G, Fecchio C, Sharon R, Schapira AHV, Proukakis C, Bellotti V, de Laureto PP. α-Synuclein structural features inhibit harmful polyunsaturated fatty acid oxidation, suggesting roles in neuroprotection. J Biol Chem 2017; 292:6927-6937. [PMID: 28232489 PMCID: PMC5409462 DOI: 10.1074/jbc.m116.765149] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/22/2017] [Indexed: 01/23/2023] Open
Abstract
α-Synuclein (aS) is a protein abundant in presynaptic nerve terminals in Parkinson disease (PD) and is a major component of intracellular Lewy bodies, the pathological hallmark of neurodegenerative disorders such as PD. Accordingly, the relationships between aS structure, its interaction with lipids, and its involvement in neurodegeneration have attracted great interest. Previously, we reported on the interaction of aS with brain polyunsaturated fatty acids, in particular docosahexaenoic acid (DHA). aS acquires an α-helical secondary structure in the presence of DHA and, in turn, affects DHA structural and aggregative properties. Moreover, aS forms a covalent adduct with DHA. Here, we provide evidence that His-50 is the main site of this covalent modification. To better understand the role of His-50, we analyzed the effect of DHA on aS-derived species: a naturally occurring variant, H50Q; an oxidized aS in which all methionines are sulfoxides (aS4ox); a fully lysine-alkylated aS (acetyl-aS); and aS fibrils, testing their ability to be chemically modified by DHA. We show, by mass spectrometry and spectroscopic techniques, that H50Q and aS4ox are modified by DHA, whereas acetyl-aS is not. We correlated this modification with aS structural features, and we suggest a possible functional role of aS in sequestering the early peroxidation products of fatty acids, thereby reducing the level of highly reactive lipid species. Finally, we show that fibrillar aS loses almost 80% of its scavenging activity, thus lacking a potentially protective function. Our findings linking aS scavenging activity with brain lipid composition suggest a possible etiological mechanism in some neurodegenerative disorders.
Collapse
Affiliation(s)
- Giorgia De Franceschi
- From the Department of Pharmaceutical Sciences, CRIBI, Biotechnology Centre, University of Padova, 35121 Padova, Italy
| | - Chiara Fecchio
- From the Department of Pharmaceutical Sciences, CRIBI, Biotechnology Centre, University of Padova, 35121 Padova, Italy
| | - Ronit Sharon
- the Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, 9112102 Jerusalem, Israel
| | - Anthony H V Schapira
- the Department of Clinical Neuroscience, Institute of Neurology, University College London, NW32PF London, United Kingdom
| | - Christos Proukakis
- the Department of Clinical Neuroscience, Institute of Neurology, University College London, NW32PF London, United Kingdom
| | - Vittorio Bellotti
- the Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, London, United Kingdom, and.,the Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, 27100 Pavia, Italy
| | - Patrizia Polverino de Laureto
- From the Department of Pharmaceutical Sciences, CRIBI, Biotechnology Centre, University of Padova, 35121 Padova, Italy,
| |
Collapse
|
39
|
van Diggelen F, Tepper AWJW, Apetri MM, Otzen DE. α-Synuclein Oligomers: A Study in Diversity. Isr J Chem 2016. [DOI: 10.1002/ijch.201600116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Femke van Diggelen
- Crossbeta Biosciences; Padualaan 8 3584CH Utrecht The Netherlands
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| | | | | | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| |
Collapse
|
40
|
Disease-Toxicant Interactions in Parkinson's Disease Neuropathology. Neurochem Res 2016; 42:1772-1786. [PMID: 27613618 DOI: 10.1007/s11064-016-2052-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/15/2022]
Abstract
Human disease commonly manifests as a result of complex genetic and environmental interactions. In the case of neurodegenerative diseases, such as Parkinson's disease (PD), understanding how environmental exposures collude with genetic polymorphisms in the central nervous system to cause dysfunction is critical in order to develop better treatment strategies, therapies, and a more cohesive paradigm for future research. The intersection of genetics and the environment in disease etiology is particularly relevant in the context of their shared pathophysiological mechanisms. This review offers an integrated view of disease-toxicant interactions in PD. Particular attention is dedicated to how mutations in the genes SNCA, parkin, leucine-rich repeat kinase 2 (LRRK2) and DJ-1, as well as dysfunction of the ubiquitin proteasome system, may contribute to PD and how exposure to heavy metals, pesticides and illicit drugs may further the consequences of these mutations to exacerbate PD and PD-like disorders. Although the toxic effects induced by exposure to these environmental factors may not be the primary causes of PD, their mechanisms of action are critical for our current understanding of the neuropathologies driving PD. Elucidating how environment and genetics collude to cause pathogenesis of PD will facilitate the development of more effective treatments for the disease. Additionally, we discuss the neuroprotection exerted by estrogen and other compounds that may prevent PD and provide an overview of current treatment strategies and therapies.
Collapse
|
41
|
Ren C, Hu X, Li X, Zhou Q. Ultra-trace graphene oxide in a water environment triggers Parkinson's disease-like symptoms and metabolic disturbance in zebrafish larvae. Biomaterials 2016; 93:83-94. [PMID: 27085073 DOI: 10.1016/j.biomaterials.2016.03.036] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/13/2016] [Accepted: 03/25/2016] [Indexed: 02/09/2023]
Abstract
Over the past decade, the safety of nanomaterials has attracted attention due to their rapid development. The relevant health threat of these materials remains largely unknown, particularly at environmentally or biologically relevant ultra-trace concentrations. To address this, we first found that graphene oxide (GO, a carbon nanomaterial that receives extensive attention across various disciplines) at concentrations of 0.01 μg/L-1 μg/L induced Parkinson's disease-like symptoms in zebrafish larvae. In this model, zebrafish showed a loss of more than 90% of dopamine neurons, a 69-522% increase in Lewy bodies (α-synuclein and ubiquitin) and significantly disturbed locomotive activity. Moreover, it was also shown that GO was able to translocate from the water environment to the brain and localize to the nucleus of the diencephalon, thereby inducing structural and morphological damage in the mitochondria. Cell apoptosis and senescence were triggered via oxidative stress, as shown by the upregulation of caspase 8 and β-galactosidase. Using metabolomics, we found that the upregulation of amino acid and some fatty acids (e.g. dodecanoic acid, hexadecanoic acid, octadecenoic acid, nonanoic acid, arachidonic acid, eicosanoic acid, propanoic acid and benzenedicarboxylic acid) metabolism and the downregulation of some other fatty acids (e.g. butanoic acid, phthalic acid and docosenoic acid) are linked to these Parkinson's disease-like symptoms. These findings broaden our understanding of nanomaterial safety at ultra-trace concentrations.
Collapse
Affiliation(s)
- Chaoxiu Ren
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Xueyan Li
- Department of Gastroenterology, General Hospital of Shenyang Military Region, Shenyang 110016, Liaoning Province, China
| | - Qixing Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China.
| |
Collapse
|
42
|
Hatano T, Saiki S, Okuzumi A, Mohney RP, Hattori N. Identification of novel biomarkers for Parkinson's disease by metabolomic technologies. J Neurol Neurosurg Psychiatry 2016; 87:295-301. [PMID: 25795009 DOI: 10.1136/jnnp-2014-309676] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/06/2015] [Indexed: 11/04/2022]
Abstract
OBJECTIVE The pathogenesis of Parkinson's disease (PD) involves complex interactions between environmental and genetic factors. Metabolomics can shed light on alterations in metabolic pathways in many diseases, including neurodegenerative diseases. In the present study, we attempted to elucidate the candidate metabolic pathway(s) associated with PD. METHODS Serum samples were collected from 35 individuals with idiopathic PD without dementia and 15 healthy age-matched control participants without PD. This analysis used a combination of three independent platforms: ultrahigh-performance liquid chromatography/tandem mass spectrometry (UPLC/MS/MS) optimised for basic species, UPLC/MS/MS optimised for acidic species and gas chromatography/MS (GC/MS). RESULTS The metabolomic profiles of PD were clearly different from normal controls. PD profiles had significantly lower levels of tryptophan, caffeine and its metabolites, bilirubin and ergothioneine, and significantly higher levels of levodopa metabolites and biliverdin than those of normal controls. Alterations in the bilirubin/biliverdin ratio and ergothioneine can indicate oxidative stress intensity and may suggest elevated oxidative stress and/or insufficient ability for scavenging free radicals, which could contribute to PD pathogenesis. Decreased serum tryptophan level is associated with psychiatric problems in PD. A decrease in serum caffeine levels is consistent with an inverse association of caffeine consumption with development of PD based on past epidemiological studies. CONCLUSIONS Metabolomic analysis detected biomarkers associated with PD pathogenesis and disease progression. Since critical metabolic biomarkers need to be identified in PD, future studies should include assay validation and replication in independent cohorts.
Collapse
Affiliation(s)
- Taku Hatano
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
43
|
Sivachenko A, Gordon HB, Kimball SS, Gavin EJ, Bonkowsky JL, Letsou A. Neurodegeneration in a Drosophila model of adrenoleukodystrophy: the roles of the Bubblegum and Double bubble acyl-CoA synthetases. Dis Model Mech 2016; 9:377-87. [PMID: 26893370 PMCID: PMC4852500 DOI: 10.1242/dmm.022244] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/17/2016] [Indexed: 12/21/2022] Open
Abstract
Debilitating neurodegenerative conditions with metabolic origins affect millions of individuals worldwide. Still, for most of these neurometabolic disorders there are neither cures nor disease-modifying therapies, and novel animal models are needed for elucidation of disease pathology and identification of potential therapeutic agents. To date, metabolic neurodegenerative disease has been modeled in animals with only limited success, in part because existing models constitute analyses of single mutants and have thus overlooked potential redundancy within metabolic gene pathways associated with disease. Here, we present the first analysis of a very-long-chain acyl-CoA synthetase (ACS) double mutant. We show that the Drosophila bubblegum(bgm) and double bubble(dbb) genes have overlapping functions, and that the consequences of double knockout of both bubblegum and double bubble in the fly brain are profound, affecting behavior and brain morphology, and providing the best paradigm to date for an animal model of adrenoleukodystrophy (ALD), a fatal childhood neurodegenerative disease associated with the accumulation of very-long-chain fatty acids. Using this more fully penetrant model of disease to interrogate brain morphology at the level of electron microscopy, we show that dysregulation of fatty acid metabolism via disruption of ACS function in vivois causal of neurodegenerative pathologies that are evident in both neuronal cells and their supporting cell populations, and leads ultimately to lytic cell death in affected areas of the brain. Finally, in an extension of our model system to the study of human disease, we describe our identification of an individual with leukodystrophy who harbors a rare mutation in SLC27a6(encoding a very-long-chain ACS), a human homolog of bgm and dbb.
Collapse
Affiliation(s)
- Anna Sivachenko
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Hannah B Gordon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Suzanne S Kimball
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Erin J Gavin
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84112, USA
| | - Anthea Letsou
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
44
|
Schulte EC, Altmaier E, Berger HS, Do KT, Kastenmüller G, Wahl S, Adamski J, Peters A, Krumsiek J, Suhre K, Haslinger B, Ceballos-Baumann A, Gieger C, Winkelmann J. Alterations in Lipid and Inositol Metabolisms in Two Dopaminergic Disorders. PLoS One 2016; 11:e0147129. [PMID: 26808974 PMCID: PMC4726488 DOI: 10.1371/journal.pone.0147129] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/28/2015] [Indexed: 12/23/2022] Open
Abstract
Background Serum metabolite profiling can be used to identify pathways involved in the pathogenesis of and potential biomarkers for a given disease. Both restless legs syndrome (RLS) and Parkinson`s disease (PD) represent movement disorders for which currently no blood-based biomarkers are available and whose pathogenesis has not been uncovered conclusively. We performed unbiased serum metabolite profiling in search of signature metabolic changes for both diseases. Methods 456 metabolites were quantified in serum samples of 1272 general population controls belonging to the KORA cohort, 82 PD cases and 95 RLS cases by liquid-phase chromatography and gas chromatography separation coupled with tandem mass spectrometry. Genetically determined metabotypes were calculated using genome-wide genotyping data for the 1272 general population controls. Results After stringent quality control, we identified decreased levels of long-chain (polyunsaturated) fatty acids of individuals with PD compared to both RLS (PD vs. RLS: p = 0.0001 to 5.80x10-9) and general population controls (PD vs. KORA: p = 6.09x10-5 to 3.45x10-32). In RLS, inositol metabolites were increased specifically (RLS vs. KORA: p = 1.35x10-6 to 3.96x10-7). The impact of dopaminergic drugs was reflected in changes in the phenylalanine/tyrosine/dopamine metabolism observed in both individuals with RLS and PD. Conclusions A first discovery approach using serum metabolite profiling in two dopamine-related movement disorders compared to a large general population sample identified significant alterations in the polyunsaturated fatty acid metabolism in PD and implicated the inositol metabolism in RLS. These results provide a starting point for further studies investigating new perspectives on factors involved in the pathogenesis of the two diseases as well as possible points of therapeutic intervention.
Collapse
Affiliation(s)
- Eva C. Schulte
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany
- Institut für Humangenetik, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Elisabeth Altmaier
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Hannah S. Berger
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany
- Institut für Humangenetik, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Kieu Trinh Do
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Jan Krumsiek
- Institute of Computational Biology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Qatar Foundation–Education City, PO Box 24144, Doha, Qatar
| | - Bernhard Haslinger
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany
| | - Andres Ceballos-Baumann
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany
- Schön Klinik München Schwabing, Munich, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Juliane Winkelmann
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany
- Institut für Humangenetik, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institut für Humangenetik, Technische Universität München, 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology and Neurosciences, Stanford University, Palo Alto, CA, 94304, United States of America
- * E-mail: ;
| |
Collapse
|
45
|
Fröhlich C, Zschiebsch K, Gröger V, Paarmann K, Steffen J, Thurm C, Schropp EM, Brüning T, Gellerich F, Radloff M, Schwabe R, Lachmann I, Krohn M, Ibrahim S, Pahnke J. Activation of Mitochondrial Complex II-Dependent Respiration Is Beneficial for α-Synucleinopathies. Mol Neurobiol 2015; 53:4728-44. [PMID: 26319560 PMCID: PMC4965489 DOI: 10.1007/s12035-015-9399-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/17/2015] [Indexed: 02/06/2023]
Abstract
Parkinson’s disease and dementia with Lewy bodies are major challenges in research and clinical medicine world-wide and contribute to the most common neurodegenerative disorders. Previously, specific mitochondrial polymorphisms have been found to enhance clearance of amyloid-β from the brain of APP-transgenic mice leading to beneficial clinical outcome. It has been discussed whether specific mitochondrial alterations contribute to disease progression or even prevent toxic peptide deposition, as seen in many neurodegenerative diseases. Here, we investigated α-synuclein-transgenic C57BL/6J mice with the A30P mutation, and a novel A30P C57BL/6J mouse model with three mitochondrial DNA polymorphisms in the ND3, COX3 and mtRNAArg genes, as found in the inbred NOD/LtJ mouse strain. We were able to detect that the new model has increased mitochondrial complex II-respiration which occurs in parallel to neuronal loss and improved motor performance, although it exhibits higher amounts of high molecular weight species of α-synuclein. High molecular weight aggregates of different peptides are controversially discussed in the light of neurodegeneration. A favourable hypothesis states that high molecular weight species are protective and of minor importance for the pathogenesis of neurodegenerative disorders as compared to the extreme neurotoxic monomers and oligomers. Summarising, our results point to a potentially protective and beneficial effect of specific mitochondrial polymorphisms which cause improved mitochondrial complex II-respiration in α-synucleinopathies, an effect that could be exploited further for pharmaceutical interventions.
Collapse
Affiliation(s)
- Christina Fröhlich
- Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany
| | - Katja Zschiebsch
- Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany.,University of Frankfurt, Institute of Clinical Pharmacology/ZAFES, Frankfurt, Germany
| | - Victoria Gröger
- Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany.,Fraunhofer Institute for Cell Therapy and Immunology (IZI), Halle, Germany
| | - Kristin Paarmann
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Postboks 4950 Nydalen, 0424, Oslo, Norway.,Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany.,LIED, University of Lübeck (UzL), Lübeck, Germany
| | - Johannes Steffen
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Postboks 4950 Nydalen, 0424, Oslo, Norway.,Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany.,LIED, University of Lübeck (UzL), Lübeck, Germany
| | - Christoph Thurm
- Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany
| | - Eva-Maria Schropp
- Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany
| | - Thomas Brüning
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Postboks 4950 Nydalen, 0424, Oslo, Norway.,Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany
| | - Frank Gellerich
- Department of Neurology, University of Magdeburg/Leibniz Institut for Neurobiology, Magdeburg, Germany
| | - Martin Radloff
- Institute for Mathematical Stochastics, University of Magdeburg, Magdeburg, Germany
| | - Rainer Schwabe
- Institute for Mathematical Stochastics, University of Magdeburg, Magdeburg, Germany
| | | | - Markus Krohn
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Postboks 4950 Nydalen, 0424, Oslo, Norway.,Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany
| | | | - Jens Pahnke
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Postboks 4950 Nydalen, 0424, Oslo, Norway. .,Department of Neurology, Neurodegeneration Research Lab (NRL), University of Magdeburg, Magdeburg, Germany. .,LIED, University of Lübeck (UzL), Lübeck, Germany. .,Department of Bioorganic Chemistry, Leibniz-Institute of Plant Biochemistry (IPB), Halle, Germany.
| |
Collapse
|
46
|
Yang JE, Park JS, Cho E, Jung S, Paik SR. Robust polydiacetylene-based colorimetric sensing material developed with amyloid fibrils of α-synuclein. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:1802-1810. [PMID: 25602613 DOI: 10.1021/la504645m] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Robust polydiacetylene-based colorimetric sensing material has been developed with amyloid fibrils of α-synuclein in the presence of 10,12-pentacosadiynoic acid (PCDA) by taking advantage of the specific fatty acid interaction of α-synuclein and structural regularity of the self-assembled product of amyloid fibrils. PCDA facilitated not only self-oligomerization of α-synuclein but also its fibrillation into the fibrils with increased thickness. Upon UV irradiation, the PCDA-containing amyloid fibrils (AF-PCDAs) turned blue, which then became red following heat treatment. The blue-to-red color transition was also observed with other stimuli of pH and ethanol. AF-PCDAs were demonstrated to be mechanically stable since not only the individual colors of blue and red but also their colorimetric transition were not affected by a number of sonications which readily disrupted the polydiaceylene (PDA) vesicles with the instant loss of color. Therefore, AF-PCDA can be considered to be a novel PDA-based colorimetric sensing material with high mechanical strength, which has the potential to be employed in various areas involving advanced sensing technologies.
Collapse
Affiliation(s)
- Jee Eun Yang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, College of Engineering, Seoul National University , Seoul 151-744, Korea
| | | | | | | | | |
Collapse
|
47
|
Chaudhary H, Stefanovic AND, Subramaniam V, Claessens MMAE. Membrane interactions and fibrillization of α-synuclein play an essential role in membrane disruption. FEBS Lett 2015; 588:4457-63. [PMID: 25448986 DOI: 10.1016/j.febslet.2014.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 12/22/2022]
Abstract
We studied α-synuclein (αS) aggregation in giant vesicles, and observed dramatic membrane disintegration, as well as lipid incorporation into micrometer-sized suprafibrillar aggregates. In the presence of dye-filled vesicles, dye leakage and fibrillization happen concurrently. However, growing fibrils do not impair the integrity of phospholipid vesicles that have a low affinity for αS. Seeding αS aggregation accelerates dye leakage, indicating that oligomeric species are not required to explain the observed effect. The evolving picture suggests that fibrils that appear in solution bind membranes and recruit membrane-bound monomers, resulting in lipid extraction, membrane destabilization and the formation of lipid-containing suprafibrillar aggregates.
Collapse
Affiliation(s)
- Himanshu Chaudhary
- Nanobiophysics Group, MESA+ Institute for Nanotechnology, Department of Science and Technology, University Twente, 7500 AE Enschede, The Netherlands
| | | | | | | |
Collapse
|
48
|
Dietary factors in the etiology of Parkinson's disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:672838. [PMID: 25688361 PMCID: PMC4320877 DOI: 10.1155/2015/672838] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 11/07/2014] [Accepted: 11/08/2014] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. The majority of cases do not arise from purely genetic factors, implicating an important role of environmental factors in disease pathogenesis. Well-established environmental toxins important in PD include pesticides, herbicides, and heavy metals. However, many toxicants linked to PD and used in animal models are rarely encountered. In this context, other factors such as dietary components may represent daily exposures and have gained attention as disease modifiers. Several in vitro, in vivo, and human epidemiological studies have found a variety of dietary factors that modify PD risk. Here, we critically review findings on association between dietary factors, including vitamins, flavonoids, calorie intake, caffeine, alcohol, and metals consumed via food and fatty acids and PD. We have also discussed key data on heterocyclic amines that are produced in high-temperature cooked meat, which is a new emerging field in the assessment of dietary factors in neurological diseases. While more research is clearly needed, significant evidence exists that specific dietary factors can modify PD risk.
Collapse
|
49
|
Ahola-Olli AV, Pitkänen N, Kettunen J, Oikonen MK, Mikkilä V, Lehtimäki T, Kähönen M, Pahkala K, Niinikoski H, Kangas AJ, Soininen P, Ala-Korpela M, Viikari JS, Rönnemaa T, Simell O, Raitakari OT. Interactions between genetic variants and dietary lipid composition: effects on circulating LDL cholesterol in children. Am J Clin Nutr 2014; 100:1569-77. [PMID: 25411292 DOI: 10.3945/ajcn.114.085027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Elevated serum low-density lipoprotein (LDL) cholesterol is a predictor of cardiovascular disease events, and the quality of dietary fat is known to influence serum concentrations of LDL cholesterol in children. Interindividual differences in response to diet exist, but the underlying genetic factors remain largely unknown. OBJECTIVE We aimed to identify genetic variants that modify the variation in serum lipid response to dietary fat quality. DESIGN We used data from 2 longitudinal Finnish cohorts designed to study risk factors for cardiovascular diseases. Large-scale genotyping was performed with Metabochip in a long-term randomized controlled dietary intervention trial, the Special Turku Coronary Risk Factor Intervention Project (STRIP), for discovery of genetic polymorphisms. The observational Cardiovascular Risk in Young Finns Study (YFS) with genome-wide genetic data was used as a replication sample for the initial findings. Dietary records were used to calculate the ratio of unsaturated to saturated fats. Interaction models and multiple follow-ups were used in the analysis. RESULTS In the STRIP cohort, a variant within the PARK2 locus, rs9364628, showed moderate interaction with dietary fat quality and a consistent direction of effect in both scans on serum LDL-cholesterol concentration in children aged 5 and 7 y (P < 0.0084 and P < 0.0057, respectively). In the YFS cohort, we were unable to replicate the initial discovery signal, but rs12207186 within the PARK2 locus and dietary lipid quality had a stronger interaction effect on serum LDL-cholesterol concentration (P < 9.44 × 10(-5)) than did rs9364628 in children aged 6 y. CONCLUSION This genotyping study involving 2 cohorts of healthy Finnish children indicates a possible interaction between PARK2 variants and dietary fat quality on serum LDL-cholesterol concentration. This trial was registered at clinicaltrials.gov as NCT00223600.
Collapse
Affiliation(s)
- Ari V Ahola-Olli
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Niina Pitkänen
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Johannes Kettunen
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Mervi K Oikonen
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Vera Mikkilä
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Terho Lehtimäki
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Mika Kähönen
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Katja Pahkala
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Harri Niinikoski
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Antti J Kangas
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Pasi Soininen
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Mika Ala-Korpela
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Jorma S Viikari
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Tapani Rönnemaa
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Olli Simell
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| | - Olli T Raitakari
- From the Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland (AVA-O, NP, MKO, VM, KP, and TR); the Department of Food and Environmental Sciences (VM) and the Institute for Molecular Medicine Finland (FIMM) (JK), University of Helsinki, Helsinki, Finland; the Department of Clinical Chemistry, Fimlab Laboratories, Pirkanmaa Hospital District, School of Medicine, University of Tampere, Tampere, Finland (TL); the Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland (MK); Paavo Nurmi Centre, Sports & Exercise Medicine Unit, Department of Physical Activity and Health, University of Turku, Turku, Finland (KP); the Department of Pediatrics, Turku University Hospital, Turku, Finland (HN and OS); Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (AJK, PS, and MA-K); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (PS and MA-K); Oulu University Hospital, Oulu, Finland (MA-K); Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (MA-K); the Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, Turku, Finland (JSV and TR); and the Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland (OTR)
| |
Collapse
|
50
|
Dong J, Beard JD, Umbach DM, Park Y, Huang X, Blair A, Kamel F, Chen H. Dietary fat intake and risk for Parkinson's disease. Mov Disord 2014; 29:1623-30. [PMID: 25186946 DOI: 10.1002/mds.26032] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 08/04/2014] [Accepted: 08/25/2014] [Indexed: 01/12/2023] Open
Abstract
Previous epidemiological studies have generated inconsistent results regarding the associations between dietary fat intakes and risk for Parkinson's disease (PD). We therefore prospectively examined these associations in the National Institutes of Health-American Association of Retired Persons (NIH-AARP) Diet and Health Study. A 124-item food frequency questionnaire was administered at baseline in1995 to 1996, and PD diagnosis was self-reported at the follow-up survey in 2004 to 2006. A total of 1,087 cases with a PD diagnosis between 2000 and 2006 and 299,617 controls were included in the analyses. Overall, intakes of fats and other macronutrients were not associated with PD risk. However, we found a weak positive association between n-6 polyunsaturated fatty acids (PUFA) and the risk for PD. After adjusting for potential confounders, the odds ratio (OR) and 95% confidence interval (CI) between extreme quintiles of n-6 PUFA intake was 1.23 (95% CI = 1.02-1.49, P for trend = 0.02). A similar association was observed for the intake of linoleic acid. Results were similar among men and among women. Our study suggests that fat intake in general is not related to the risk for PD. The weak positive association between intake of n-6 PUFA and PD risk needs further investigation.
Collapse
Affiliation(s)
- Jing Dong
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | | | |
Collapse
|