201
|
Hein LK, Rozaklis T, Adams MK, Hopwood JJ, Karageorgos L. Lipid composition of microdomains is altered in neuronopathic Gaucher disease sheep brain and spleen. Mol Genet Metab 2017; 121:259-270. [PMID: 28532689 DOI: 10.1016/j.ymgme.2017.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 01/16/2023]
Abstract
Gaucher disease is a lysosomal storage disorder caused by a deficiency in glucocerebrosidase activity that leads to accumulation of glucosylceramide and glucosylsphingosine. Membrane raft microdomains are discrete, highly organized microdomains with a unique lipid composition that provide the necessary environment for specific protein-lipid and protein-protein interactions to take place. In this study we purified detergent resistant membranes (DRM; membrane rafts) from the occipital cortex and spleen from sheep affected with acute neuronopathic Gaucher disease and wild-type controls. We observed significant increases in the concentrations of glucosylceramide, hexosylsphingosine, BMP and gangliosides and decreases in the percentage of cholesterol and phosphatidylcholine leading to an altered DRM composition. Altered sphingolipid/cholesterol homeostasis would dramatically disrupt DRM architecture making them less ordered and more fluid. In addition, significant changes in the length and degree of lipid saturation within the DRM microdomains in the Gaucher brain were also observed. As these DRM microdomains are involved in many cellular events, an imbalance or disruption of the cell membrane homeostasis may impair normal cell function. This disruption of membrane raft microdomains and imbalance within the environment of cellular membranes of neuronal cells may be a key factor in initiating a cascade process leading to neurodegeneration.
Collapse
Affiliation(s)
- Leanne K Hein
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia, 5001, Australia
| | - Tina Rozaklis
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia, 5001, Australia
| | - Melissa K Adams
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia, 5001, Australia
| | - John J Hopwood
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia, 5001, Australia
| | - Litsa Karageorgos
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia, 5001, Australia.
| |
Collapse
|
202
|
Caretti A, Vasso M, Bonezzi FT, Gallina A, Trinchera M, Rossi A, Adami R, Casas J, Falleni M, Tosi D, Bragonzi A, Ghidoni R, Gelfi C, Signorelli P. Myriocin treatment of CF lung infection and inflammation: complex analyses for enigmatic lipids. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:775-790. [PMID: 28439630 DOI: 10.1007/s00210-017-1373-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/04/2017] [Indexed: 12/30/2022]
Abstract
Our aim was to use quantitative and qualitative analyses to gain further insight into the role of ceramide in cystic fibrosis (CF). Sphingolipid ceramide is a known inflammatory mediator, and its accumulation in inflamed lung has been reported in different types of emphysema, chronic obstructive pulmonary disease and CF. CF is caused by a mutation of the chloride channel and associated with hyperinflammation of the respiratory airways and high susceptibility to ongoing infections. We have previously demonstrated that de novo ceramide synthesis is enhanced in lung inflammation and sustains Pseudomonas aeruginosa pulmonary infection in a CF murine model. We used liquid chromatography and matrix-assisted laser desorption/ionization (MALDI) imaging coupled with mass spectrometry, confocal laser scan microscopy and histology analyses to reveal otherwise undecipherable information. We demonstrated that (i) upregulated ceramide synthesis in the alveoli is strictly related to alveolar infection and inflammation, (ii) alveolar ceramide (C16) can be specifically targeted by nanocarrier delivery of the ceramide synthesis inhibitor myriocin (Myr) and (iii) Myr is able to downmodulate pro-inflammatory lyso-PC, favouring an increase in anti-inflammatory PCs. We concluded that Myr modulates alveolar lipids milieu, reducing hyperinflammation and favouring anti-microbial effective response in CF mouse model.
Collapse
Affiliation(s)
- Anna Caretti
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Michele Vasso
- Lita Institute, Segrate, University of Milan, Milan, Italy
| | - Fabiola Tecla Bonezzi
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Andrea Gallina
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Marco Trinchera
- Department of Medicine Clinical and Experimental, University of Insubria Medical School, Varese, Italy
| | - Alice Rossi
- Infections and Cystic Fibrosis Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Adami
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Josefina Casas
- Research Unit on Bioactive Molecules, Department of Biomedicinal Chemistry, Catalan Institute of Advanced Chemistry (IQAC/CSIC), Barcelona, Spain
| | - Monica Falleni
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Delfina Tosi
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Alessandra Bragonzi
- Infections and Cystic Fibrosis Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy
| | - Cecilia Gelfi
- Lita Institute, Segrate, University of Milan, Milan, Italy
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Department of Health Sciences, University of Milan, Via A.di Rudinì 8, 20142, Milan, Italy.
| |
Collapse
|
203
|
Santpere G, Garcia-Esparcia P, Andres-Benito P, Lorente-Galdos B, Navarro A, Ferrer I. Transcriptional network analysis in frontal cortex in Lewy body diseases with focus on dementia with Lewy bodies. Brain Pathol 2017; 28:315-333. [PMID: 28321951 DOI: 10.1111/bpa.12511] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/15/2017] [Indexed: 12/13/2022] Open
Abstract
The present study investigates global transcriptional changes in frontal cortex area 8 in incidental Lewy Body disease (iLBD), Parkinson disease (PD) and Dementia with Lewy bodies (DLB). We identified different coexpressed gene sets associated with disease stages, and gene ontology categories enriched in gene modules and differentially expressed genes including modules or gene clusters correlated to iLBD comprising upregulated dynein genes and taste receptors, and downregulated innate inflammation. Focusing on DLB, we found modules with genes significantly enriched in functions related to RNA and DNA production, mitochondria and energy metabolism, purine metabolism, chaperone and protein folding system and synapses and neurotransmission (particularly the GABAergic system). The expression of more than fifty selected genes was assessed with real time quantitative polymerase chain reaction. Our findings provide, for the first time, evidence of molecular cortical alterations in iLBD and involvement of several key metabolic pathways and gene hubs in DLB which may underlie cognitive impairment and dementia.
Collapse
Affiliation(s)
- Gabriel Santpere
- Department of Neurobiology, Yale School of Medicine, New Haven, CT.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Paula Garcia-Esparcia
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Pol Andres-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Belen Lorente-Galdos
- Department of Neurobiology, Yale School of Medicine, New Haven, CT.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Arcadi Navarro
- Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain.,Institute of Science and Technology, Centre for Genomic Regulation (CRG), Barcelona, Spain.,National Institute for Bioinformatics (INB), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat, Spain.,Institute of Neuropathology, Service of Pathologic Anatomy, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain.,Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat, Spain.,CIBERNED, Network Centre for Biomedical Research of Neurodegenerative Diseases, Institute Carlos III, Spain
| |
Collapse
|
204
|
Cipolla CM, Lodhi IJ. Peroxisomal Dysfunction in Age-Related Diseases. Trends Endocrinol Metab 2017; 28:297-308. [PMID: 28063767 PMCID: PMC5366081 DOI: 10.1016/j.tem.2016.12.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/04/2016] [Accepted: 12/07/2016] [Indexed: 12/21/2022]
Abstract
Peroxisomes carry out many key functions related to lipid and reactive oxygen species (ROS) metabolism. The fundamental importance of peroxisomes for health in humans is underscored by the existence of devastating genetic disorders caused by impaired peroxisomal function or lack of peroxisomes. Emerging studies suggest that peroxisomal function may also be altered with aging and contribute to the pathogenesis of a variety of diseases, including diabetes and its related complications, neurodegenerative disorders, and cancer. With increasing evidence connecting peroxisomal dysfunction to the pathogenesis of these acquired diseases, the possibility of targeting peroxisomal function in disease prevention or treatment becomes intriguing. Here, we review recent developments in understanding the pathophysiological implications of peroxisomal dysfunctions outside the context of inherited peroxisomal disorders.
Collapse
Affiliation(s)
- Cynthia M Cipolla
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
205
|
Taghizadeh M, Tamtaji OR, Dadgostar E, Daneshvar Kakhaki R, Bahmani F, Abolhassani J, Aarabi MH, Kouchaki E, Memarzadeh MR, Asemi Z. The effects of omega-3 fatty acids and vitamin E co-supplementation on clinical and metabolic status in patients with Parkinson's disease: A randomized, double-blind, placebo-controlled trial. Neurochem Int 2017; 108:183-189. [PMID: 28342967 DOI: 10.1016/j.neuint.2017.03.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/14/2017] [Accepted: 03/21/2017] [Indexed: 12/23/2022]
Abstract
The current research was performed to evaluate the effects of omega-3 fatty acids and vitamin E co-supplementation on clinical signs and metabolic status in people with Parkinson's disease (PD). This randomized double-blind placebo-controlled clinical trial was conducted in 60 patients with PD. Participants were randomly assigned into two groups to receive either 1000 mg omega-3 fatty acids from flaxseed oil plus 400 IU vitamin E supplements (n = 30) or placebo (n = 30) for 12 weeks. Unified Parkinson's disease rating stage (UPDRS) were recorded at baseline and the after 3-month intervention. After 12 weeks' intervention, compared with the placebo, omega-3 fatty acids and vitamin E co-supplementation led to a significant improve in UPDRS (-3.3 ± 10.0 vs. +4.4 ± 14.9, P = 0.02). Furthermore, co-supplementation decreased high-sensitivity C-reactive protein (hs-CRP) (-0.3 ± 0.6 vs. +0.3 ± 0.3 μg/mL, P < 0.001), and increased total antioxidant capacity (TAC) (+65.2 ± 68.7 vs. +16 ± 52.4 μmol/L, P = 0.003) and glutathione (GSH) concentrations (+41.4 ± 80.6 vs. -19.6 ± 55.9 μmol/L, P = 0.001) compared with the placebo. Additionally, co-supplementation meaningfully decreased insulin (-2.1 ± 4.9 vs. +1.4 ± 6.2 μIU/mL, P = 0.01), homeostasis model of assessment-estimated insulin resistance (-0.7 ± 1.8 vs.+0.3 ± 1.6, P = 0.02) and Beta cell function (-5.9 ± 13.9 vs. +5.7 ± 25.5, P = 0.03), and increased quantitative insulin sensitivity check index (+0.009 ± 0.02 vs. -0.006 ± 0.03, P = 0.03) compared with the placebo. Overall, our study demonstrated that omega-3 fatty acids and vitamin E co-supplementation in people with PD had favorable effects on UPDRS, hs-CRP, TAC, GSH and markers of insulin metabolism.
Collapse
Affiliation(s)
- Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Omid Reza Tamtaji
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Ehsan Dadgostar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Reza Daneshvar Kakhaki
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Fereshteh Bahmani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Javad Abolhassani
- Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mohammad Hossein Aarabi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Ebrahim Kouchaki
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran; Department of Neurology, School of Medicine, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
206
|
Characterization of lipid rafts in human platelets using nuclear magnetic resonance: A pilot study. Biochem Biophys Rep 2017; 10:132-136. [PMID: 28955740 PMCID: PMC5614646 DOI: 10.1016/j.bbrep.2017.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/28/2017] [Accepted: 03/15/2017] [Indexed: 11/23/2022] Open
Abstract
Lipid microdomains (‘lipid rafts’) are plasma membrane subregions, enriched in cholesterol and glycosphingolipids, which participate dynamically in cell signaling and molecular trafficking operations. One strategy for the study of the physicochemical properties of lipid rafts in model membrane systems has been the use of nuclear magnetic resonance (NMR), but until now this spectroscopic method has not been considered a clinically relevant tool. We performed a proof-of-concept study to test the feasibility of using NMR to study lipid rafts in human tissues. Platelets were selected as a cost-effective and minimally invasive model system in which lipid rafts have previously been studied using other approaches. Platelets were isolated from plasma of medication-free adult research participants (n=13) and lysed with homogenization and sonication. Lipid-enriched fractions were obtained using a discontinuous sucrose gradient. Association of lipid fractions with GM1 ganglioside was tested using HRP-conjugated cholera toxin B subunit dot blot assays. 1H high resolution magic-angle spinning nuclear magnetic resonance (HRMAS NMR) spectra obtained with single-pulse Bloch decay experiments yielded spectral linewidths and intensities as a function of temperature. Rates of lipid lateral diffusion that reported on raft size were measured with a two-dimensional stimulated echo longitudinal encode-decode NMR experiment. We found that lipid fractions at 10–35% sucrose density associated with GM1 ganglioside, a marker for lipid rafts. NMR spectra of the membrane phospholipids featured a prominent ‘centerband’ peak associated with the hydrocarbon chain methylene resonance at 1.3 ppm; the linewidth (full width at half-maximum intensity) of this ‘centerband’ peak, together with the ratio of intensities between the centerband and ‘spinning sideband’ peaks, agreed well with values reported previously for lipid rafts in model membranes. Decreasing temperature produced decreases in the 1.3 ppm peak intensity and a discontinuity at ~18 °C, for which the simplest explanation is a phase transition from Ld to Lo phases indicative of raft formation. Rates of lateral diffusion of the acyl chain lipid signal at 1.3 ppm, a quantitative measure of microdomain size, were consistent with lipid molecules organized in rafts. These results show that HRMAS NMR can characterize lipid microdomains in human platelets, a methodological advance that could be extended to other tissues in which membrane biochemistry may have physiological and pathophysiological relevance. Lipid raft properties have been studied mainly in model membranes or cell cultures. We report a novel 1H NMR approach to lipid raft characterization in human platelets. We find spectroscopy, diffusion, and phase transitions consistent with lipid rafts. NMR plus bioassays may be used to study raft-mediated cell function in human tissues.
Collapse
|
207
|
Membrane cholesterol access into a G-protein-coupled receptor. Nat Commun 2017; 8:14505. [PMID: 28220900 PMCID: PMC5321766 DOI: 10.1038/ncomms14505] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 01/05/2017] [Indexed: 12/25/2022] Open
Abstract
Cholesterol is a key component of cell membranes with a proven modulatory role on the function and ligand-binding properties of G-protein-coupled receptors (GPCRs). Crystal structures of prototypical GPCRs such as the adenosine A2A receptor (A2AR) have confirmed that cholesterol finds stable binding sites at the receptor surface suggesting an allosteric role of this lipid. Here we combine experimental and computational approaches to show that cholesterol can spontaneously enter the A2AR-binding pocket from the membrane milieu using the same portal gate previously suggested for opsin ligands. We confirm the presence of cholesterol inside the receptor by chemical modification of the A2AR interior in a biotinylation assay. Overall, we show that cholesterol's impact on A2AR-binding affinity goes beyond pure allosteric modulation and unveils a new interaction mode between cholesterol and the A2AR that could potentially apply to other GPCRs. G-protein-coupled receptors trigger several signalling pathways and their activity was proposed to be allosteric modulated by cholesterol. Here the authors use molecular dynamics simulations and ligand binding assays to show that membrane cholesterol can bind to adenosine A2A receptor orthosteric site.
Collapse
|
208
|
Conte C, Roscini L, Sardella R, Mariucci G, Scorzoni S, Beccari T, Corte L. Toll Like Receptor 4 Affects the Cerebral Biochemical Changes Induced by MPTP Treatment. Neurochem Res 2017; 42:493-500. [PMID: 28108849 DOI: 10.1007/s11064-016-2095-6] [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: 08/05/2016] [Revised: 10/17/2016] [Accepted: 10/27/2016] [Indexed: 02/06/2023]
Abstract
The etiology and pathogenesis of Parkinson's disease (PD) are still unclear. However, multiple lines of evidence suggest a critical role of the toll like receptor 4 (TLR4) in inflammatory response and neuronal death. Neuroinflammation may be associated with the misfolding and aggregation of proteins accompanied by a change in their secondary structure. Recent findings also suggest that biochemical perturbations in cerebral lipid content could contribute to the pathogenesis of central nervous system (CNS) disorders, including PD. Thus, it is of great importance to determine the biochemical changes that occur in PD. In this respect, Fourier Transform Infrared (FTIR) spectroscopy represents a useful tool to detect molecular alterations in biological systems in response to stress stimuli. By relying upon FTIR approach, this study was designed to elucidate the potential role of TLR4 in biochemical changes induced by methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxin in a mouse model of PD. The analysis of the FTIR spectra was performed in different brain regions of both wild type (WT) and toll like receptor 4-deficient (TLR4-/-) mice. It revealed that each brain region exhibited a characteristic molecular fingerprint at baseline, with no significant differences between genotypes. Conversely, WT and TLR4-/- mice showed differential biochemical response to MPTP toxicity, principally related to lipid and protein composition. These differences appeared to be characteristic for each brain area. Furthermore, the present study showed that WT mice resulted more vulnerable than TLR4-/- animals to striatal dopamine (DA) depletion following MPTP treatment. These results support the hypothesis of a possible involvement of TLR4 in biochemical changes occurring in neurodegeneration.
Collapse
Affiliation(s)
- Carmela Conte
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy.
| | - Luca Roscini
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy
| | - Roccaldo Sardella
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy
| | - Giuseppina Mariucci
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy
| | - Stefania Scorzoni
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy
| | - Laura Corte
- Department of Pharmaceutical Sciences, Section of Biochemical and Health Sciences, University of Perugia, Via del Giochetto, 06123, Perugia, Italy
| |
Collapse
|
209
|
Eriksson I, Nath S, Bornefall P, Giraldo AMV, Öllinger K. Impact of high cholesterol in a Parkinson's disease model: Prevention of lysosomal leakage versus stimulation of α-synuclein aggregation. Eur J Cell Biol 2017; 96:99-109. [PMID: 28109635 DOI: 10.1016/j.ejcb.2017.01.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 01/04/2017] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease is characterized by accumulation of intraneuronal cytoplasmic inclusions, Lewy bodies, which mainly consist of aggregated α-synuclein. Controversies exist as to whether high blood cholesterol is a risk factor for the development of the disease and whether statin treatment could have a protective effect. Using a model system of BE(2)-M17 neuroblastoma cells treated with the neurotoxin 1-methyl-4-phenylpyridinium (MPP+), we found that MPP+-induced cell death was accompanied by cholesterol accumulation in a lysosomal-like pattern in pre-apoptotic cells. To study the effects of lysosomal cholesterol accumulation, we increased lysosomal cholesterol through pre-treatment with U18666A and found delayed leakage of lysosomal contents into the cytosol, which reduced cell death. This suggests that increased lysosomal cholesterol is a stress response mechanism to protect lysosomal membrane integrity in response to early apoptotic stress. However, high cholesterol also stimulated the accumulation of α-synuclein. Treatment with the cholesterol-lowering drug lovastatin reduced MPP+-induced cell death by inhibiting the production of reactive oxygen species, but did not prevent lysosomal cholesterol increase nor affect α-synuclein accumulation. Our study indicates a dual role of high cholesterol in Parkinson's disease, in which it acts both as a protector against lysosomal membrane permeabilization and as a stimulator of α-synuclein accumulation.
Collapse
Affiliation(s)
- Ida Eriksson
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | - Sangeeta Nath
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | - Per Bornefall
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | - Ana Maria Villamil Giraldo
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | - Karin Öllinger
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
210
|
Petrov AM, Kasimov MR, Zefirov AL. Cholesterol in the Pathogenesis of Alzheimer's, Parkinson's Diseases and Autism: Link to Synaptic Dysfunction. Acta Naturae 2017; 9:26-37. [PMID: 28461971 PMCID: PMC5406657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Indexed: 11/30/2022] Open
Abstract
In our previous review, we described brain cholesterol metabolism in control conditions and in the case of some rare neurological pathologies linked to defects in the genes which are directly involved in the synthesis and/or traffic of cholesterol. Here, we have analyzed disruptions in cholesterol homeostasis in widespread neurodegenerative diseases (Alzheimer's and Parkinson's diseases) and autism spectrum disorders. We particularly focused on the synaptic dysfunctions that could arise from changes in both membrane cholesterol availability and oxysterol production. Notably, alterations in the brain cholesterol metabolism and neurotransmission occur in the early stages of these pathologies and the polymorphism of the genes associated with cholesterol homeostasis and synaptic communication affects the risk of onset and severity of these diseases. In addition, pharmacological and genetic manipulations of brain cholesterol homeostasis in animal models frequently have marked effects on the progression of neurodegenerative diseases. Thus, the development of Alzheimer's, Parkinson's and autism spectrum disorders may be partially associated with an imbalance of cholesterol homeostasis that leads to changes in the membrane cholesterol and oxysterol levels that, in turn, modulates key steps in the synaptic transmission.
Collapse
Affiliation(s)
- A. M. Petrov
- Kazan State Medical University, Normal Physiology department, Butlerova str. 49, Kazan, 420012, Russia
| | - M. R. Kasimov
- Kazan State Medical University, Normal Physiology department, Butlerova str. 49, Kazan, 420012, Russia
| | - A. L. Zefirov
- Kazan State Medical University, Normal Physiology department, Butlerova str. 49, Kazan, 420012, Russia
| |
Collapse
|
211
|
Pintado-Sierra M, García-Álvarez I, Bribián A, Medina-Rodríguez EM, Lebrón-Aguilar R, Garrido L, de Castro F, Fernández-Mayoralas A, Quintanilla-López JE. A comprehensive profiling of sulfatides in myelin from mouse brain using liquid chromatography coupled to high-resolution accurate tandem mass spectrometry. Anal Chim Acta 2016; 951:89-98. [PMID: 27998489 DOI: 10.1016/j.aca.2016.11.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 11/17/2022]
Abstract
Sulfatides are sulfoglycolipids found in the myelin sheath. The composition ratio of sulfatide molecular species changes with age, and it has also been associated with the pathogenesis of various human central nervous system diseases. However, profiling sulfatides in biological samples is difficult, due to the great variety of molecular species. In this work, a new, easy and reliable liquid chromatography-electrospray tandem mass spectrometry (LC-ESI(+)-MS/MS) method has been developed to profile sulfatide content in biological samples of myelin. The 'wrong-way-round' ionization effect has been described for this type of molecules for the first time, making it possible to correctly identify as many as 37 different sulfatides in mouse brain myelin samples, including molecules with different fatty acid chain lengths and varying degrees of unsaturation and hydroxylation. A chemometric analysis of their relative abundances showed that the main difference among individuals of different ages was the content of sulfatides with odd-numbered fatty acid chains, in addition to hydroxylated species.
Collapse
Affiliation(s)
- M Pintado-Sierra
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | - I García-Álvarez
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain; Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda", 45071, Toledo, Spain
| | - A Bribián
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda", 45071, Toledo, Spain; Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| | - E M Medina-Rodríguez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda", 45071, Toledo, Spain
| | - R Lebrón-Aguilar
- Instituto de Química-Física 'Rocasolano' (CSIC), Serrano 119, 28006, Madrid, Spain
| | - L Garrido
- Instituto de Ciencia y Tecnología de Polímeros (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | - F de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda", 45071, Toledo, Spain; Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| | - A Fernández-Mayoralas
- Instituto de Química Orgánica General (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | | |
Collapse
|
212
|
Periole X. Interplay of G Protein-Coupled Receptors with the Membrane: Insights from Supra-Atomic Coarse Grain Molecular Dynamics Simulations. Chem Rev 2016; 117:156-185. [PMID: 28073248 DOI: 10.1021/acs.chemrev.6b00344] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
G protein-coupled receptors (GPCRs) are central to many fundamental cellular signaling pathways. They transduce signals from the outside to the inside of cells in physiological processes ranging from vision to immune response. It is extremely challenging to look at them individually using conventional experimental techniques. Recently, a pseudo atomistic molecular model has emerged as a valuable tool to access information on GPCRs, more specifically on their interactions with their environment in their native cell membrane and the consequences on their supramolecular organization. This approach uses the Martini coarse grain (CG) model to describe the receptors, lipids, and solvent in molecular dynamics (MD) simulations and in enough detail to allow conserving the chemical specificity of the different molecules. The elimination of unnecessary degrees of freedom has opened up large-scale simulations of the lipid-mediated supramolecular organization of GPCRs. Here, after introducing the Martini CGMD method, we review these studies carried out on various members of the GPCR family, including rhodopsin (visual receptor), opioid receptors, adrenergic receptors, adenosine receptors, dopamine receptor, and sphingosine 1-phosphate receptor. These studies have brought to light an interesting set of novel biophysical principles. The insights range from revealing localized and heterogeneous deformations of the membrane bilayer at the surface of the protein, specific interactions of lipid molecules with individual GPCRs, to the effect of the membrane matrix on global GPCR self-assembly. The review ends with an overview of the lessons learned from the use of the CGMD method, the biophysical-chemical findings on lipid-protein interplay.
Collapse
Affiliation(s)
- Xavier Periole
- Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen , Nijenborgh 7, 9747AG Groningen, The Netherlands
| |
Collapse
|
213
|
Díaz M, Fabelo N, Casañas-Sánchez V, Marin R, Gómez T, Quinto-Alemany D, Pérez JA. Hippocampal Lipid Homeostasis in APP/PS1 Mice is Modulated by a Complex Interplay Between Dietary DHA and Estrogens: Relevance for Alzheimer's Disease. J Alzheimers Dis 2016; 49:459-81. [PMID: 26519437 DOI: 10.3233/jad-150470] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Current evidence suggests that lipid homeostasis in the hippocampus is affected by different genetic, dietary, and hormonal factors, and that its deregulation may be associated with the onset and progression of Alzheimer's disease (AD). However, the precise levels of influence of each of these factors and their potential interactions remain largely unknown, particularly during neurodegenerative processes. In the present study, we have performed multifactorial analyses of the combined effects of diets containing different doses of docosahexaenoic acid (DHA), estrogen status (ovariectomized animals receiving vehicle or 17β-estradiol), and genotype (wild-type or transgenic APP/PS1 mice) in hippocampal lipid profiles. We have observed that the three factors affect lipid classes and fatty acid composition to different extents, and that strong interactions between these factors exist. The most aberrant lipid profiles were observed in APP/PS1 animals receiving DHA-poor diets and deprived of estrogens. Conversely, wild-type animals under a high-DHA diet and receiving estradiol exhibited a lipid profile that closely resembled that of the hippocampus of control animals. Interestingly, though the lipid signatures of APP/PS1 hippocampi markedly differed from wild-type, administration of a high-DHA diet in the presence of estrogens gave rise to a lipid profile that approached that of control animals. Paralleling changes in lipid composition, patterns of gene expression of enzymes involved in lipid biosynthesis were also altered and affected by combination of experimental factors. Overall, these results indicate that hippocampal lipid homeostasis is strongly affected by hormonal and dietary conditions, and that manipulation of these factors might be incorporated in AD therapeutics.
Collapse
Affiliation(s)
- Mario Díaz
- Department of Animal Physiology, Laboratory of Membrane Physiology and Biophysics, University of La Laguna, Tenerife, Spain
| | - Noemí Fabelo
- Department of Animal Physiology, Laboratory of Membrane Physiology and Biophysics, University of La Laguna, Tenerife, Spain
| | | | - Raquel Marin
- Department of Physiology, Laboratory of Cellular Neurobiology, University of La Laguna, Tenerife, Spain
| | - Tomás Gómez
- Department of Animal Physiology, Laboratory of Membrane Physiology and Biophysics, University of La Laguna, Tenerife, Spain
| | - David Quinto-Alemany
- Department of Animal Physiology, Laboratory of Membrane Physiology and Biophysics, University of La Laguna, Tenerife, Spain
| | - José A Pérez
- Department of Genetics, University of La Laguna, Tenerife, Spain
| |
Collapse
|
214
|
Marin R, Fabelo N, Martín V, Garcia-Esparcia P, Ferrer I, Quinto-Alemany D, Díaz M. Anomalies occurring in lipid profiles and protein distribution in frontal cortex lipid rafts in dementia with Lewy bodies disclose neurochemical traits partially shared by Alzheimer's and Parkinson's diseases. Neurobiol Aging 2016; 49:52-59. [PMID: 27768960 DOI: 10.1016/j.neurobiolaging.2016.08.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 10/21/2022]
Abstract
Lipid rafts are highly dynamic membrane microdomains intimately associated with cell signaling. Compelling evidence has demonstrated that alterations in lipid rafts are associated with neurodegenerative diseases such Alzheimer's disease, but at present, whether alterations in lipid raft microdomains occur in other types of dementia such dementia with Lewy bodies (DLB) remains unknown. Our analyses reveal that lipid rafts from DLB exhibit aberrant lipid profiles including low levels of n-3 long-chain polyunsaturated fatty acids (mainly docosahexaenoic acid), plasmalogens and cholesterol, and reduced unsaturation and peroxidability indexes. As a consequence, lipid raft resident proteins holding principal factors of the β-amyloidogenic pathway, including β-amyloid precursor protein, presenilin 1, β-secretase, and PrP, are redistributed between lipid rafts and nonraft domains in DLB frontal cortex. Meta-analysis discloses certain similarities in the altered composition of lipid rafts between DLB and Parkinson's disease which are in line with the spectrum of Lewy body diseases. In addition, redistribution of proteins linked to the β-amyloidogenic pathway in DLB can facilitate generation of β-amyloid, thus providing mechanistic clues to the intriguing convergence of Alzheimer's disease pathology, particularly β-amyloid deposition, in DLB.
Collapse
Affiliation(s)
- Raquel Marin
- Departamento de Fisiología, Universidad de La Laguna, Tenerife, Spain
| | - Noemí Fabelo
- Departamento de Biología Animal, Universidad de La Laguna, Tenerife, Spain
| | | | - Paula Garcia-Esparcia
- Instituto Neuropatología, Servicio Anatomía Patológica, Hospital Universitario de Bellvitge, Universidad de Barcelona, CIBERNED, Hospitalet de Llobregat, Spain
| | - Isidre Ferrer
- Instituto Neuropatología, Servicio Anatomía Patológica, Hospital Universitario de Bellvitge, Universidad de Barcelona, CIBERNED, Hospitalet de Llobregat, Spain
| | | | - Mario Díaz
- Departamento de Biología Animal, Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
215
|
Plasma metabolic profile delineates roles for neurodegeneration, pro-inflammatory damage and mitochondrial dysfunction in the FMR1 premutation. Biochem J 2016; 473:3871-3888. [PMID: 27555610 DOI: 10.1042/bcj20160585] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/23/2016] [Indexed: 12/19/2022]
Abstract
Carriers of premutation CGG expansions in the fragile X mental retardation 1 (FMR1) gene are at higher risk of developing a late-onset neurodegenerative disorder named Fragile X-associated tremor ataxia syndrome (FXTAS). Given that mitochondrial dysfunction has been identified in fibroblasts, PBMC and brain samples from carriers as well as in animal models of the premutation and that mitochondria are at the center of intermediary metabolism, the aim of the present study was to provide a complete view of the metabolic pattern by uncovering plasma metabolic perturbations in premutation carriers. To this end, metabolic profiles were evaluated in plasma from 23 premutation individuals and 16 age- and sex-matched controls. Among the affected pathways, mitochondrial dysfunction was associated with a Warburg-like shift with increases in lactate levels and altered Krebs' intermediates, neurotransmitters, markers of neurodegeneration and increases in oxidative stress-mediated damage to biomolecules. The number of CGG repeats correlated with a subset of plasma metabolites, which are implicated not only in mitochondrial disorders but also in other neurological diseases, such as Parkinson's, Alzheimer's and Huntington's diseases. For the first time, the identified pathways shed light on disease mechanisms contributing to morbidity of the premutation, with the potential of assessing metabolites in longitudinal studies as indicators of morbidity or disease progression, especially at the early preclinical stages.
Collapse
|
216
|
Ferrazza R, Cogo S, Melrose H, Bubacco L, Greggio E, Guella G, Civiero L, Plotegher N. LRRK2 deficiency impacts ceramide metabolism in brain. Biochem Biophys Res Commun 2016; 478:1141-6. [PMID: 27539321 DOI: 10.1016/j.bbrc.2016.08.082] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/13/2016] [Indexed: 12/30/2022]
Abstract
Mutations in LRRK2 gene cause inherited Parkinson's disease (PD) and variations around LRRK2 act as risk factor for disease. Similar to sporadic disease, LRRK2-linked cases show late onset and, typically, the presence of proteinaceous inclusions named Lewy bodies (LBs) in neurons. Recently, defects on ceramide (Cer) metabolism have been recognized in PD. In particular, heterozygous mutations in the gene encoding for glucocerebrosidase (GBA1), a lysosomal enzyme converting glucosyl-ceramides (Glc-Cer) into Cer, increase the risk of developing PD. Although several studies have linked LRRK2 with membrane-related processes and autophagic-lysosomal pathway regulation, whether this protein impinges on the Cer pathway has not been addressed. Here, using a targeted lipidomics approach, we report an altered sphingolipid composition in Lrrk2(-/-) mouse brains. In particular, we observe a significant increase of Cer levels in Lrrk2(-/-) mice and direct effects on GBA1. Collectively, our results suggest a link between LRRK2 and Cer metabolism, providing new insights into the possible role of this protein in sphingolipids metabolism, with implications for PD therapeutics.
Collapse
Affiliation(s)
- Ruggero Ferrazza
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, Via Sommarive 14, 38123, Povo, Trento, Italy
| | - Susanna Cogo
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131, Padova, Italy
| | - Heather Melrose
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, 32224, USA
| | - Luigi Bubacco
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131, Padova, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131, Padova, Italy
| | - Graziano Guella
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, Via Sommarive 14, 38123, Povo, Trento, Italy; Biophysical Institute, CNR, Via alla Cascata 56/C, 38123, Povo, Trento, Italy
| | - Laura Civiero
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131, Padova, Italy.
| | - Nicoletta Plotegher
- Department of Biology, University of Padova, Via U. Bassi 58/b, 35131, Padova, Italy.
| |
Collapse
|
217
|
Kim JM, Cha SH, Choi YR, Jou I, Joe EH, Park SM. DJ-1 deficiency impairs glutamate uptake into astrocytes via the regulation of flotillin-1 and caveolin-1 expression. Sci Rep 2016; 6:28823. [PMID: 27346864 PMCID: PMC4922019 DOI: 10.1038/srep28823] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 06/10/2016] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a common chronic and progressive neurodegenerative disorder. Although the cause of PD is still poorly understood, mutations in many genes including SNCA, parkin, PINK1, LRRK2, and DJ-1 have been identified in the familial forms of PD. It was recently proposed that alterations in lipid rafts may cause the neurodegeneration shown in PD. Here, we observe that DJ-1 deficiency decreased the expression of flotillin-1 (flot-1) and caveolin-1 (cav-1), the main protein components of lipid rafts, in primary astrocytes and MEF cells. As a mechanism, DJ-1 regulated flot-1 stability by direct interaction, however, decreased cav-1 expression may not be a direct effect of DJ-1, but rather as a result of decreased flot-1 expression. Dysregulation of flot-1 and cav-1 by DJ-1 deficiency caused an alteration in the cellular cholesterol level, membrane fluidity, and alteration in lipid rafts-dependent endocytosis. Moreover, DJ-1 deficiency impaired glutamate uptake into astrocytes, a major function of astrocytes in the maintenance of CNS homeostasis, by altering EAAT2 expression. This study will be helpful to understand the role of DJ-1 in the pathogenesis of PD, and the modulation of lipid rafts through the regulation of flot-1 or cav-1 may be a novel therapeutic target for PD.
Collapse
Affiliation(s)
- Jin-Mo Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Seon-Heui Cha
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Ilo Jou
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
218
|
Exogenous Alpha-Synuclein Alters Pre- and Post-Synaptic Activity by Fragmenting Lipid Rafts. EBioMedicine 2016; 7:191-204. [PMID: 27322472 PMCID: PMC4909369 DOI: 10.1016/j.ebiom.2016.03.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/24/2016] [Accepted: 03/24/2016] [Indexed: 12/28/2022] Open
Abstract
Alpha-synuclein (αSyn) interferes with multiple steps of synaptic activity at pre-and post-synaptic terminals, however the mechanism/s by which αSyn alters neurotransmitter release and synaptic potentiation is unclear. By atomic force microscopy we show that human αSyn, when incubated with reconstituted membrane bilayer, induces lipid rafts' fragmentation. As a consequence, ion channels and receptors are displaced from lipid rafts with consequent changes in their activity. The enhanced calcium entry leads to acute mobilization of synaptic vesicles, and exhaustion of neurotransmission at later stages. At the post-synaptic terminal, an acute increase in glutamatergic transmission, with increased density of PSD-95 puncta, is followed by disruption of the interaction between N-methyl-d-aspartate receptor (NMDAR) and PSD-95 with ensuing decrease of long term potentiation. While cholesterol loading prevents the acute effect of αSyn at the presynapse; inhibition of casein kinase 2, which appears activated by reduction of cholesterol, restores the correct localization and clustering of NMDARs. Extracellular αSyn disrupts lipid raft platforms with consequent mislocalization of several pre- and post-synaptic proteins. αSyn-driven changes in raft-partitioning of proteins blunt neurotransmission and LTP. Cholesterol loading and inhibition of CK2 restore αSyn-induced alterations of the post-synaptic density assembly.
Alpha-synuclein (αSyn), a cytosolic protein that can be released from neurons, becomes pathogenic when expressed at high levels, as in Parkinson's disease, due to multiplication of αSyn gene. We show that the mechanism responsible for the defects in synaptic vesicles mobilization and post-synaptic activity induced by extracellular αSyn is the fragmentation of lipid rafts, cholesterol-rich microdomains of the plasma membrane. Moreover restoration of lipid raft platforms and raft-partitioning of surface proteins prevents the alteration of synaptic transmission caused by exposure of neurons to αSyn.
Collapse
|
219
|
Miville-Godbout E, Bourque M, Morissette M, Al-Sweidi S, Smith T, Mochizuki A, Senanayake V, Jayasinghe D, Wang L, Goodenowe D, Di Paolo T. Plasmalogen Augmentation Reverses Striatal Dopamine Loss in MPTP Mice. PLoS One 2016; 11:e0151020. [PMID: 26959819 PMCID: PMC4784967 DOI: 10.1371/journal.pone.0151020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/23/2016] [Indexed: 01/08/2023] Open
Abstract
Plasmalogens are a class of glycerophospholipids shown to play critical roles in membrane structure and function. Decreased plasmalogens are reported in the brain and blood of Parkinson’s disease (PD) patients. The present study investigated the hypothesis that augmenting plasmalogens could protect striatal dopamine neurons that degenerate in response to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment in mice, a PD model. First, in a pre-treatment experiment male mice were treated for 10 days with the docosahexaenoic acid (DHA)-plasmalogen precursor PPI-1011 (10, 50 and 200 mg/kg). On day 5 mice received MPTP and were killed on day 11. Next, in a post-treatment study, male mice were treated with MPTP and then received daily for 5 days PPI-1011 (5, 10 and 50 mg/kg). MPTP treatment reduced serum plasmalogen levels, striatal contents of dopamine (DA) and its metabolites, serotonin, DA transporter (DAT) and vesicular monoamine transporter 2 (VMAT2). Pre-treatment with PPI-1011 (10 and 50 mg/kg) prevented all MPTP-induced effects. Positive correlations were measured between striatal DA contents and serum plasmalogen levels as well as striatal DAT and VMAT2 specific binding. Post-treatment with PPI-1011 prevented all MPTP-induced effects at 50 mg/kg but not at lower doses. Positive correlations were measured between striatal DA contents and serum plasmalogen levels as well as striatal DAT and VMAT2 specific binding in the post-treatment experiment. PPI-1011 treatment (10 days at 5, 10 and 50 mg/kg) of intact mice left unchanged striatal biogenic amine contents. These data demonstrate that treatment with a plasmalogen precursor is capable of protecting striatal dopamine markers in an animal model of PD.
Collapse
Affiliation(s)
- Edith Miville-Godbout
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, Quebec City, Canada
- Faculty of Pharmacy, Laval University, Quebec City, Canada
| | - Mélanie Bourque
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, Quebec City, Canada
- Faculty of Pharmacy, Laval University, Quebec City, Canada
| | - Marc Morissette
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, Quebec City, Canada
| | - Sara Al-Sweidi
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, Quebec City, Canada
| | - Tara Smith
- Phenomenome Discoveries Inc., Saskatoon, Canada
| | - Asuka Mochizuki
- Phenomenome Discoveries Inc., Saskatoon, Canada
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | - Li Wang
- Phenomenome Discoveries Inc., Saskatoon, Canada
| | | | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre de Recherche du CHU de Québec, CHUL, Quebec City, Canada
- Faculty of Pharmacy, Laval University, Quebec City, Canada
- * E-mail:
| |
Collapse
|
220
|
Kuter K, Kratochwil M, Berghauzen-Maciejewska K, Głowacka U, Sugawa MD, Ossowska K, Dencher NA. Adaptation within mitochondrial oxidative phosphorylation supercomplexes and membrane viscosity during degeneration of dopaminergic neurons in an animal model of early Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2016; 1862:741-753. [PMID: 26844379 DOI: 10.1016/j.bbadis.2016.01.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 01/02/2023]
Abstract
In Parkinson's disease (PD) motor symptoms are not observed until loss of 70% of dopaminergic neurons in substantia nigra (SN), preventing early diagnosis. Mitochondrial dysfunction was indicated in neuropathological process already at early PD stages. Aging and oxidative stress, the main factors in PD pathogenesis, cause membrane stiffening, which could influence functioning of membrane-bound oxidative phosphorylation (OxPhos) complexes (Cxs) in mitochondria. In 6-OHDA rat model, medium-sized dopaminergic lesion was used to study mitochondrial membrane viscosity and changes at the level of OxPhos Cxs and their higher assembled states-supercomplexes (SCxs), during the early degeneration processes and after it. We observed loss of dopaminergic phenotype in SN and decreased dopamine level in striatum (STR) before actual death of neurons in SN. Behavioural deficits induced by lesion were reversed despite progressing neurodegeneration. Along with degeneration process in STR, mitochondrial Cx I performance and amount decreased in almost all forms of SCxs. Also, progressing decrease of Cx IV performance in SCxs (I1III2IV3-1, I1IV2-1) in STR was observed during degeneration. In SN, SCxs containing Cx I increased protein amount and a shifted individual Cx I1 into superassembled states. Importantly, mitochondrial membrane viscosity changed in parallel with altered SCxs performance. We show for the first time changes at the level of mitochondrial membrane viscosity influencing SCxs function after dopaminergic system degeneration. It implicates that altered mitochondrial membrane viscosity could play an important role in regulation of mitochondria functioning and pathomechanisms of PD. The data obtained are also discussed in relation to compensatory processes observed.
Collapse
Affiliation(s)
- Katarzyna Kuter
- Department of Neuropsychopharmacology, Polish Academy of Sciences, Smętna St. 12, 31-343 Kraków, Poland.
| | - Manuela Kratochwil
- Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany
| | | | - Urszula Głowacka
- Department of Neuropsychopharmacology, Polish Academy of Sciences, Smętna St. 12, 31-343 Kraków, Poland
| | - Michiru D Sugawa
- Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany; Clinical Neurobiology, Charité-Universitätsmedizin, D-12203 Berlin, Germany
| | - Krystyna Ossowska
- Department of Neuropsychopharmacology, Polish Academy of Sciences, Smętna St. 12, 31-343 Kraków, Poland
| | - Norbert A Dencher
- Physical Biochemistry, Department of Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Str. 4, D-64287 Darmstadt, Germany
| |
Collapse
|
221
|
Membrane omega-3 fatty acids modulate the oligomerisation kinetics of adenosine A2A and dopamine D2 receptors. Sci Rep 2016; 6:19839. [PMID: 26796668 PMCID: PMC4726318 DOI: 10.1038/srep19839] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/01/2015] [Indexed: 12/03/2022] Open
Abstract
Membrane levels of docosahexaenoic acid (DHA), an essential omega-3 polyunsaturated fatty acid (ω-3 PUFA), are decreased in common neuropsychiatric disorders. DHA modulates key cell membrane properties like fluidity, thereby affecting the behaviour of transmembrane proteins like G protein-coupled receptors (GPCRs). These receptors, which have special relevance for major neuropsychiatric disorders have recently been shown to form dimers or higher order oligomers, and evidence suggests that DHA levels affect GPCR function by modulating oligomerisation. In this study, we assessed the effect of membrane DHA content on the formation of a class of protein complexes with particular relevance for brain disease: adenosine A2A and dopamine D2 receptor oligomers. Using extensive multiscale computer modelling, we find a marked propensity of DHA for interaction with both A2A and D2 receptors, which leads to an increased rate of receptor oligomerisation. Bioluminescence resonance energy transfer (BRET) experiments performed on living cells suggest that this DHA effect on the oligomerisation of A2A and D2 receptors is purely kinetic. This work reveals for the first time that membrane ω-3 PUFAs play a key role in GPCR oligomerisation kinetics, which may have important implications for neuropsychiatric conditions like schizophrenia or Parkinson’s disease.
Collapse
|
222
|
Postila PA, Vattulainen I, Róg T. Selective effect of cell membrane on synaptic neurotransmission. Sci Rep 2016; 6:19345. [PMID: 26782980 PMCID: PMC4725992 DOI: 10.1038/srep19345] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022] Open
Abstract
Atomistic molecular dynamics simulations were performed with 13 non-peptidic neurotransmitters (NTs) in three different membrane environments. The results provide compelling evidence that NTs are divided into membrane-binding and membrane-nonbinding molecules. NTs adhere to the postsynaptic membrane surface whenever the ligand-binding sites of their synaptic receptors are buried in the lipid bilayer. In contrast, NTs that have extracellular ligand-binding sites do not have a similar tendency to adhere to the membrane surface. This finding is a seemingly simple yet important addition to the paradigm of neurotransmission, essentially dividing it into membrane-independent and membrane-dependent mechanisms. Moreover, the simulations also indicate that the lipid composition especially in terms of charged lipids can affect the membrane partitioning of NTs. The revised paradigm, highlighting the importance of cell membrane and specific lipids for neurotransmission, should to be of interest to neuroscientists, drug industry and the general public alike.
Collapse
Affiliation(s)
- Pekka A. Postila
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- Department of Chemistry and Biochemistry, University of California San Diego, 92093-0340 San Diego, CA, USA
| | - Ilpo Vattulainen
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
- MEMPHYS– Center for Biomembrane Physics, University of Southern Denmark, Odense, Denmark
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014, Helsinki, Finland
| | - Tomasz Róg
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| |
Collapse
|
223
|
Bienias K, Fiedorowicz A, Sadowska A, Prokopiuk S, Car H. Regulation of sphingomyelin metabolism. Pharmacol Rep 2016; 68:570-81. [PMID: 26940196 DOI: 10.1016/j.pharep.2015.12.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/24/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022]
Abstract
Sphingolipids (SFs) represent a large class of lipids playing diverse functions in a vast number of physiological and pathological processes. Sphingomyelin (SM) is the most abundant SF in the cell, with ubiquitous distribution within mammalian tissues, and particularly high levels in the Central Nervous System (CNS). SM is an essential element of plasma membrane (PM) and its levels are crucial for the cell function. SM content in a cell is strictly regulated by the enzymes of SM metabolic pathways, which activities create a balance between SM synthesis and degradation. The de novo synthesis via SM synthases (SMSs) in the last step of the multi-stage process is the most important pathway of SM formation in a cell. The SM hydrolysis by sphingomyelinases (SMases) increases the concentration of ceramide (Cer), a bioactive molecule, which is involved in cellular proliferation, growth and apoptosis. By controlling the levels of SM and Cer, SMSs and SMases maintain cellular homeostasis. Enzymes of SM cycle exhibit unique properties and diverse tissue distribution. Disturbances in their activities were observed in many CNS pathologies. This review characterizes the physiological roles of SM and enzymes controlling SM levels as well as their involvement in selected pathologies of the Central Nervous System, such as ischemia/hypoxia, Alzheimer disease (AD), Parkinson disease (PD), depression, schizophrenia and Niemann Pick disease (NPD).
Collapse
Affiliation(s)
- Kamil Bienias
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland
| | - Anna Fiedorowicz
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland; Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland
| | - Sławomir Prokopiuk
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland.
| |
Collapse
|
224
|
Houston JE, Kraft M, Scherf U, Evans RC. Sequential detection of multiple phase transitions in model biological membranes using a red-emitting conjugated polyelectrolyte. Phys Chem Chem Phys 2016; 18:12423-7. [DOI: 10.1039/c6cp01553k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Charge-mediated assembly of an anionic poly(thiophene) leads to a highly sensitive probe of membrane order.
Collapse
Affiliation(s)
- Judith E. Houston
- School of Chemistry
- University of Dublin
- Trinity College
- Dublin 2
- Ireland
| | - Mario Kraft
- Macromolecular Chemistry Group (buwmacro) and Institute for Polymer Technology
- Bergische Universität Wuppertal
- Wuppertal
- Germany
| | - Ullrich Scherf
- Macromolecular Chemistry Group (buwmacro) and Institute for Polymer Technology
- Bergische Universität Wuppertal
- Wuppertal
- Germany
| | - Rachel C. Evans
- School of Chemistry
- University of Dublin
- Trinity College
- Dublin 2
- Ireland
| |
Collapse
|
225
|
Lemire S, Jeromin A, Boisselier É. Membrane binding of Neuronal Calcium Sensor-1 (NCS1). Colloids Surf B Biointerfaces 2015; 139:138-47. [PMID: 26705828 DOI: 10.1016/j.colsurfb.2015.11.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/29/2015] [Accepted: 11/22/2015] [Indexed: 01/10/2023]
Abstract
Neuronal Calcium Sensor-1 (NCS1) belongs to the family of Neuronal Calcium Sensor (NCS) proteins. NCS1 is composed of four EF-hand motifs and an N-terminal myristoylation. However, the presence of a calcium-myristoyl switch in NCS1 and its role in the membrane binding are controversial. The model of Langmuir lipid monolayers is thus used to mimic the cell membrane in order to characterize the membrane interactions of NCS1. Two binding parameters are calculated from monolayer measurements: the maximum insertion pressure, up to which protein binding is energetically favorable, and the synergy, reporting attractive or repulsive interactions with the lipid monolayers. Binding membrane measurements performed in the presence of myristoylated NCS1 reveal better binding interactions for phospholipids composed of phosphoethanolamine polar head groups and unsaturated fatty acyl chains. In the absence of calcium, the membrane binding measurements are drastically modified and suggest that the protein is more strongly bound to the membrane. Indeed, the binding of calcium by three EF-hand motifs of NCS1 leads to a conformation change. NCS1 arrangement at the membrane could thus be reshuffled for better interactions with its substrates. The N-terminal peptide of NCS1 is composed of two amphiphilic helices involved in the membrane interactions of NCS1. Moreover, the presence of the myristoyl group has a weak influence on the membrane binding of NCS1 suggesting the absence of a calcium-myristoyl switch mechanism in this protein. The myristoylation could thus have a structural role required in the folding/unfolding of NCS1 which is essential to its multiple biological functions.
Collapse
Affiliation(s)
- Samuel Lemire
- CUO-Recherche, Hôpital du Saint-Sacrement, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Québec, Québec, Canada
| | | | - Élodie Boisselier
- CUO-Recherche, Hôpital du Saint-Sacrement, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
226
|
Cha SH, Choi YR, Heo CH, Kang SJ, Joe EH, Jou I, Kim HM, Park SM. Loss of parkin promotes lipid rafts-dependent endocytosis through accumulating caveolin-1: implications for Parkinson's disease. Mol Neurodegener 2015; 10:63. [PMID: 26627850 PMCID: PMC4666086 DOI: 10.1186/s13024-015-0060-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023] Open
Abstract
Background Parkinson’s disease (PD) is characterized by progressive loss of midbrain dopaminergic neurons, resulting in motor dysfunctions. While most PD is sporadic in nature, a significant subset can be linked to either autosomal dominant or recessive mutations. PARK2, encoding the E3 ubiquitin ligase, parkin, is the most frequently mutated gene in autosomal recessive early onset PD. It has recently been reported that PD-associated gene products such as PINK1, α-synuclein, LRRK2, and DJ-1, as well as parkin associate with lipid rafts, suggesting that the dysfunction of these proteins in lipid rafts may be a causal factor of PD. Therefore here, we examined the relationship between lipid rafts-related proteins and parkin. Results We identified caveolin-1 (cav-1), which is one of the major constituents of lipid rafts at the plasma membrane, as a substrate of parkin. Loss of parkin function was found to disrupt the ubiquitination and degradation of cav-1, resulting in elevated cav-1 protein level in cells. Moreover, the total cholesterol level and membrane fluidity was altered by parkin deficiency, causing dysregulation of lipid rafts-dependent endocytosis. Further, cell-to-cell transmission of α-synuclein was facilitated by parkin deficiency. Conclusions Our results demonstrate that alterations in lipid rafts by the loss of parkin via cav-1 may be a causal factor of PD, and cav-1 may be a novel therapeutic target for PD.
Collapse
Affiliation(s)
- Seon-Heui Cha
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Korea
| | - Cheol-Ho Heo
- Department of Chemistry, Ajou University, Suwon, Korea
| | - Seo-Jun Kang
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Korea
| | - Ilo Jou
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Korea
| | | | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea. .,Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
227
|
Kubo SI. Membrane lipids as therapeutic targets for Parkinson’s disease: a possible link between Lewy pathology and membrane lipids. Expert Opin Ther Targets 2015; 20:1301-1310. [DOI: 10.1517/14728222.2016.1086340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
228
|
Shaikh SR, Boyle S, Edidin M. A high fat diet containing saturated but not unsaturated fatty acids enhances T cell receptor clustering on the nanoscale. Prostaglandins Leukot Essent Fatty Acids 2015; 100:1-4. [PMID: 26143085 PMCID: PMC4554807 DOI: 10.1016/j.plefa.2015.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 05/07/2015] [Indexed: 01/16/2023]
Abstract
Cell culture studies show that the nanoscale lateral organization of surface receptors, their clustering or dispersion, can be altered by changing the lipid composition of the membrane bilayer. However, little is known about similar changes in vivo, which can be effected by changing dietary lipids. We describe the use of a newly developed method, k-space image correlation spectroscopy, kICS, for analysis of quantum dot fluorescence to show that a high fat diet can alter the nanometer-scale clustering of the murine T cell receptor, TCR, on the surface of naive CD4(+) T cells. We found that diets enriched primarily in saturated fatty acids increased TCR nanoscale clustering to a level usually seen only on activated cells. Diets enriched in monounsaturated or n-3 polyunsaturated fatty acids had no effect on TCR clustering. Also none of the high fat diets affected TCR clustering on the micrometer scale. Furthermore, the effect of the diets was similar in young and middle aged mice. Our data establish proof-of-principle that TCR nanoscale clustering is sensitive to the composition of dietary fat.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/metabolism
- Diet, High-Fat
- Fatty Acids/metabolism
- Fatty Acids, Monounsaturated/metabolism
- Fatty Acids, Omega-3/metabolism
- Mice
- Mice, Transgenic
- Protein Multimerization
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Spectrum Analysis/methods
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry & Molecular Biology, East Carolina Diabetes & Obesity Institute, East Carolina University, 600 Moye Blvd, Greenville, NC 27834, USA.
| | - Sarah Boyle
- Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Michael Edidin
- Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
229
|
Farmer K, Smith CA, Hayley S, Smith J. Major Alterations of Phosphatidylcholine and Lysophosphotidylcholine Lipids in the Substantia Nigra Using an Early Stage Model of Parkinson's Disease. Int J Mol Sci 2015; 16:18865-77. [PMID: 26274953 PMCID: PMC4581276 DOI: 10.3390/ijms160818865] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/06/2015] [Accepted: 08/06/2015] [Indexed: 01/14/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease affecting the nigrostriatal pathway, where patients do not manifest motor symptoms until >50% of neurons are lost. Thus, it is of great importance to determine early neuronal changes that may contribute to disease progression. Recent attention has focused on lipids and their role in pro- and anti-apoptotic processes. However, information regarding the lipid alterations in animal models of PD is lacking. In this study, we utilized high performance liquid chromatography electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS) and novel HPLC solvent methodology to profile phosphatidylcholines and sphingolipids within the substantia nigra. The ipsilateral substantia nigra pars compacta was collected from rats 21 days after an infusion of 6-hydroxydopamine (6-OHDA), or vehicle into the anterior dorsal striatum. We identified 115 lipid species from their mass/charge ratio using the LMAPS Lipid MS Predict Database. Of these, 19 lipid species (from phosphatidylcholine and lysophosphotidylcholine lipid classes) were significantly altered by 6-OHDA, with most being down-regulated. The two lipid species that were up-regulated were LPC (16:0) and LPC (18:1), which are important for neuroinflammatory signalling. These findings provide a first step in the characterization of lipid changes in early stages of PD-like pathology and could provide novel targets for early interventions in PD.
Collapse
Affiliation(s)
- Kyle Farmer
- Carleton University Department of Neuroscience, 1125 Colonel By Drive, Life Sciences Research Building, Ottawa, ON K1S 5B6, Canada.
| | - Catherine A Smith
- Carleton University Department of Neuroscience, 1125 Colonel By Drive, Life Sciences Research Building, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Carleton University Department of Neuroscience, 1125 Colonel By Drive, Life Sciences Research Building, Ottawa, ON K1S 5B6, Canada.
| | - Jeffrey Smith
- Carleton University Department of Chemistry and Institute of Biochemistry, 1125 Colonel By Drive, Steacie Building, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
230
|
Gegg ME, Sweet L, Wang BH, Shihabuddin LS, Sardi SP, Schapira AHV. No evidence for substrate accumulation in Parkinson brains with GBA mutations. Mov Disord 2015; 30:1085-9. [PMID: 26096906 PMCID: PMC4529481 DOI: 10.1002/mds.26278] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/20/2015] [Accepted: 05/03/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To establish whether Parkinson's disease (PD) brains previously described to have decreased glucocerebrosidase activity exhibit accumulation of the lysosomal enzyme's substrate, glucosylceramide, or other changes in lipid composition. METHODS Lipidomic analyses and cholesterol measurements were performed on the putamen (n = 5-7) and cerebellum (n = 7-14) of controls, Parkinson's disease brains with heterozygote GBA1 mutations (PD+GBA), or sporadic PD. RESULTS Total glucosylceramide levels were unchanged in both PD+GBA and sporadic PD brains when compared with controls. No changes in glucosylsphingosine (deacetylated glucosylceramide), sphingomyelin, gangliosides (GM2, GM3), or total cholesterol were observed in either putamen or cerebellum. CONCLUSIONS This study did not demonstrate glucocerebrosidase substrate accumulation in PD brains with heterozygote GBA1 mutations in areas of the brain with low α-synuclein pathology.
Collapse
Affiliation(s)
- Matthew E Gegg
- Department of Clinical Neuroscience, UCL Institute of Neurology, London, UK
| | - Lindsay Sweet
- Genzyme, a Sanofi Company, Framingham, Massachusetts, USA
| | - Bing H Wang
- Genzyme, a Sanofi Company, Framingham, Massachusetts, USA
| | | | | | | |
Collapse
|
231
|
Khan MZ, He L. The role of polyunsaturated fatty acids and GPR40 receptor in brain. Neuropharmacology 2015; 113:639-651. [PMID: 26005184 DOI: 10.1016/j.neuropharm.2015.05.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/02/2015] [Accepted: 05/08/2015] [Indexed: 01/15/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are found in abundance in the nervous system. They perform significant functions for example boosting synaptogenesis, neurogenesis, inducing antinociception, stimulating gene expression and neuronal activity, preventing apoptosis and neuroinflammation. G-protein-coupled receptor 40 (GPR40), also called free fatty acid receptor 1 (FFA1), is ubiquitously expressed in various regions of the human brain including the olfactory bulb, midbrain, medulla oblongata, hippocampus, hypothalamus, cerebral cortex, cerebellum and in the spinal cord. GPR40, when binding with polyunsaturated fatty acids (PUFAs) has shown promising therapeutic potential. This review presents current knowledge regarding the pharmacological properties of GPR40 and addresses its functions in brain, with a focus on neurodevelopment & neurogenesis. Furthermore, the demonstration of GPR40 involvement in several neuropathological conditions such as apoptosis, inflammatory pain, Alzheimer's disease and Parkinson's disease. Although the results are encouraging, further research is needed to clarify their role in the treatment of inflammatory pain, Alzheimer's disease and Parkinson's disease. This article is part of the Special Issue entitled 'Lipid Sensing G Protein-Coupled Receptors in the CNS'.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
232
|
Dyall SC. Long-chain omega-3 fatty acids and the brain: a review of the independent and shared effects of EPA, DPA and DHA. Front Aging Neurosci 2015; 7:52. [PMID: 25954194 PMCID: PMC4404917 DOI: 10.3389/fnagi.2015.00052] [Citation(s) in RCA: 546] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/28/2015] [Indexed: 12/19/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) exhibit neuroprotective properties and represent a potential treatment for a variety of neurodegenerative and neurological disorders. However, traditionally there has been a lack of discrimination between the different omega-3 PUFAs and effects have been broadly accredited to the series as a whole. Evidence for unique effects of eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and more recently docosapentaenoic acid (DPA) is growing. For example, beneficial effects in mood disorders have more consistently been reported in clinical trials using EPA; whereas, with neurodegenerative conditions such as Alzheimer’s disease, the focus has been on DHA. DHA is quantitatively the most important omega-3 PUFA in the brain, and consequently the most studied, whereas the availability of high purity DPA preparations has been extremely limited until recently, limiting research into its effects. However, there is now a growing body of evidence indicating both independent and shared effects of EPA, DPA and DHA. The purpose of this review is to highlight how a detailed understanding of these effects is essential to improving understanding of their therapeutic potential. The review begins with an overview of omega-3 PUFA biochemistry and metabolism, with particular focus on the central nervous system (CNS), where DHA has unique and indispensable roles in neuronal membranes with levels preserved by multiple mechanisms. This is followed by a review of the different enzyme-derived anti-inflammatory mediators produced from EPA, DPA and DHA. Lastly, the relative protective effects of EPA, DPA and DHA in normal brain aging and the most common neurodegenerative disorders are discussed. With a greater understanding of the individual roles of EPA, DPA and DHA in brain health and repair it is hoped that appropriate dietary recommendations can be established and therapeutic interventions can be more targeted and refined.
Collapse
Affiliation(s)
- Simon C Dyall
- Faculty of Health and Social Sciences, Bournemouth University Bournemouth, UK
| |
Collapse
|
233
|
Plasmalogen precursor analog treatment reduces levodopa-induced dyskinesias in parkinsonian monkeys. Behav Brain Res 2015; 286:328-37. [PMID: 25771209 DOI: 10.1016/j.bbr.2015.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 01/28/2023]
Abstract
L-DOPA-induced dyskinesias (LID) remain a serious obstacle in the treatment of Parkinson's disease (PD). The objective of this study was to test a new target for treatment of dyskinesias, ethanolamine plasmalogens (PlsEtn). PlsEtn play critical roles in membrane structure mediated functions and as a storage depot of polyunsaturated fatty acids such as docosahexaenoic acid (DHA, omega-3) known to reduce dyskinesias. The motor effect of a daily treatment for 12 days of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Macaca fascicularis monkeys with DHA (100mg/kg) was compared to the DHA-PlsEtn precursor PPI-1011 (50mg/kg). PPI-1011 and DHA reduced LID while maintaining the antiparkinsonian activity of l-DOPA, however the PPI-1011 effect was observed at the first behavioral time point analyzed following drug administration (day 2) whereas the effect of DHA was not observed until after 10 days of administration. DHA treatment increased plasma DHA levels 2-3× whereas PPI-1011 had no effect. DHA and PPI-1011 increased DHA-PlsEtn levels by 1.5-2× while DHA-phosphatidylethanolamine (PtdEtn) levels remained unaffected. DHA treatment also elevated very long chain fatty acid containing PtdEtn and reduced non-DHA containing PtdEtn and PlsEtn levels. PPI-1011 had no effect on these systems. LID scores were inversely correlated with serum DHA-PlsEtn/total PlsEtn ratios levels in DHA and PPI-1011 treated monkeys. Hence, the antidyskinetic activity of DHA and PPI-1011 in MPTP monkeys appears to be associated with the increase of serum DHA-PlsEtn concentrations. This is the first study reporting an antidyskinetic response to augmentation of DHA-PlsEtn using a plasmalogen precursor thus providing a novel drug target for dyskinesias.
Collapse
|
234
|
Guardia-Laguarta C, Area-Gomez E, Schon EA, Przedborski S. Novel subcellular localization for α-synuclein: possible functional consequences. Front Neuroanat 2015; 9:17. [PMID: 25755636 PMCID: PMC4337379 DOI: 10.3389/fnana.2015.00017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/04/2015] [Indexed: 12/21/2022] Open
Abstract
α-synuclein (α-syn) is one of the genes that when mutated or overexpressed causes Parkinson’s Disease (PD). Initially, it was described as a synaptic terminal protein and later was found to be localized at mitochondria. Mitochondria-associated membranes (MAM) have emerged as a central endoplasmic reticulum (ER) subcellular compartments where key functions of the cell occur. These domains, enriched in cholesterol and anionic phospholipids, are where calcium homeostasis, lipid transfer, and cholesterol metabolism are regulated. Some proteins, related to mitochondrial dynamics and function, are also localized to this area. Several neurodegenerative diseases have shown alterations in MAM functions and resident proteins, including Charcot Marie-Tooth and Alzheimer’s disease (AD). We have recently reported that MAM function is downregulated in cell and mouse models of PD expressing pathogenic mutations of α-syn. This review focuses on the possible role of α-syn in these cellular domains and the early pathogenic features of PD that could be explained by α-syn-MAM disturbances.
Collapse
Affiliation(s)
| | - Estela Area-Gomez
- Departments of Neurology, Columbia University Medical Center New York, NY, USA
| | - Eric A Schon
- Departments of Neurology, Columbia University Medical Center New York, NY, USA ; Departments of Genetics and Development, Columbia University Medical Center New York, NY, USA
| | - Serge Przedborski
- Departments of Pathology, Columbia University Medical Center New York, NY, USA
| |
Collapse
|
235
|
Waugh MG. PIPs in neurological diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1066-82. [PMID: 25680866 DOI: 10.1016/j.bbalip.2015.02.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
Abstract
Phosphoinositide (PIP) lipids regulate many aspects of cell function in the nervous system including receptor signalling, secretion, endocytosis, migration and survival. Levels of PIPs such as PI4P, PI(4,5)P2 and PI(3,4,5)P3 are normally tightly regulated by phosphoinositide kinases and phosphatases. Deregulation of these biochemical pathways leads to lipid imbalances, usually on intracellular endosomal membranes, and these changes have been linked to a number of major neurological diseases including Alzheimer's, Parkinson's, epilepsy, stroke, cancer and a range of rarer inherited disorders including brain overgrowth syndromes, Charcot-Marie-Tooth neuropathies and neurodevelopmental conditions such as Lowe's syndrome. This article analyses recent progress in this area and explains how PIP lipids are involved, to varying degrees, in almost every class of neurological disease. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Mark G Waugh
- Lipid and Membrane Biology Group, Institute for Liver and Digestive Health, UCL, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
236
|
Sardi SP, Cheng SH, Shihabuddin LS. Gaucher-related synucleinopathies: the examination of sporadic neurodegeneration from a rare (disease) angle. Prog Neurobiol 2015; 125:47-62. [PMID: 25573151 DOI: 10.1016/j.pneurobio.2014.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/01/2014] [Accepted: 12/27/2014] [Indexed: 10/24/2022]
Abstract
Gaucher disease, the most common lysosomal storage disease, is caused by a recessively inherited deficiency in glucocerebrosidase and subsequent accumulation of toxic lipid substrates. Heterozygous mutations in the lysosomal glucocerebrosidase gene (GBA1) have recently been recognized as the highest genetic risk factor for the development of α-synuclein aggregation disorders ("synucleinopathies"), including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Despite the wealth of experimental, clinical and genetic evidence that supports the association between mutant genotypes and synucleinopathy risk, the precise mechanisms by which GBA1 mutations lead to PD and DLB remain unclear. Decreased glucocerebrosidase activity has been demonstrated to promote α-synuclein misprocessing. Furthermore, aberrant α-synuclein species have been reported to downregulate glucocerebrosidase activity, which further contributes to disease progression. In this review, we summarize the recent findings that highlight the complexity of this pathogenetic link and how several pathways that connect glucocerebrosidase insufficiency with α-synuclein misprocessing have emerged as potential therapeutic targets. From a translational perspective, we discuss how various therapeutic approaches to lysosomal dysfunction have been explored for the treatment of GBA1-related synucleinopathies, and potentially, for non-GBA1-associated neurodegenerative diseases. In summary, the link between GBA1 and synucleinopathies has become the paradigm of how the study of a rare lysosomal disease can transform the understanding of the etiopathology, and hopefully the treatment, of a more prevalent and multifactorial disorder.
Collapse
Affiliation(s)
- S Pablo Sardi
- Genzyme, a Sanofi Company, 49 New York Avenue, Framingham, MA 01701, USA.
| | - Seng H Cheng
- Genzyme, a Sanofi Company, 49 New York Avenue, Framingham, MA 01701, USA
| | | |
Collapse
|
237
|
Abbott SK, Li H, Muñoz SS, Knoch B, Batterham M, Murphy KE, Halliday GM, Garner B. Altered ceramide acyl chain length and ceramide synthase gene expression in Parkinson’s disease. Mov Disord 2014; 29:518-26. [PMID: 24822250 DOI: 10.1002/mds.25729] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Genetic studies have provided increasing evidence that ceramide homeostasis plays a role in neurodegenerative diseases including Parkinson’s disease (PD). It is known that the relative amounts of different ceramide molecular species, as defined by their fatty acyl chain length, regulate ceramide function in lipid membranes and in signaling pathways. In the present study we used a comprehensive sphingolipidomic case-control approach to determine the effects of PD on ceramide composition in postmortem brain tissue from the anterior cingulate cortex (a region with significant PD pathology) and the occipital cortex (spared in PD), also assessing mRNA expression of the major ceramide synthase genes that regulate ceramide acyl chain composition in the same tissue using quantitative PCR. In PD anterior cingulate cortex but not occipital cortex, total ceramide and sphingomyelin levels were reduced from control levels by 53% (P < 0.001) and 42% (P < 0.001), respectively. Of the 13 ceramide and 15 sphingomyelin molecular lipid species identified and quantified, there was a significant shift in the ceramide acyl chain composition toward shorter acyl chain length in the PD anterior cingulate cortex. This PD-associated change in ceramide acyl chain composition was accompanied by an upregulation of ceramide synthase-1 gene expression, which we consider may represent a response to reduced ceramide levels. These data suggest a significant shift in ceramide function in lipid membranes and signaling pathways occurs in regions with PD pathology. Identifying the regulatory mechanisms precipitating this change may provide novel targets for future therapeutics.
Collapse
|
238
|
Saab S, Mazzocco J, Creuzot-Garcher CP, Bron AM, Bretillon L, Acar N. Plasmalogens in the retina: From occurrence in retinal cell membranes to potential involvement in pathophysiology of retinal diseases. Biochimie 2014; 107 Pt A:58-65. [DOI: 10.1016/j.biochi.2014.07.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/26/2014] [Indexed: 10/24/2022]
|
239
|
Shukla AK, Ratnasekhar C, Pragya P, Chaouhan HS, Patel DK, Chowdhuri DK, Mudiam MKR. Metabolomic Analysis Provides Insights on Paraquat-Induced Parkinson-Like Symptoms in Drosophila melanogaster. Mol Neurobiol 2014; 53:254-269. [PMID: 25428622 DOI: 10.1007/s12035-014-9003-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/13/2014] [Indexed: 11/29/2022]
Abstract
Paraquat (PQ) exposure causes degeneration of the dopaminergic neurons in an exposed organism while altered metabolism has a role in various neurodegenerative disorders. Therefore, the study presented here was conceived to depict the role of altered metabolism in PQ-induced Parkinson-like symptoms and to explore Drosophila as a potential model organism for such studies. Metabolic profile was generated in control and in flies that were fed PQ (5, 10, and 20 mM) in the diet for 12 and 24 h concurrent with assessment of indices of oxidative stress, dopaminergic neurodegeneration, and behavioral alteration. PQ was found to significantly alter 24 metabolites belonging to different biological pathways along with significant alterations in the above indices. In addition, PQ attenuated brain dopamine content in the exposed organism. The study demonstrates that PQ-induced alteration in the metabolites leads to oxidative stress and neurodegeneration in the exposed organism along with movement disorder, a phenotype typical of Parkinson-like symptoms. The study is relevant in the context of Drosophila and humans because similar alteration in the metabolic pathways has been observed in both PQ-exposed Drosophila and in postmortem samples of patients with Parkinsonism. Furthermore, this study provides advocacy towards the applicability of Drosophila as an alternate model organism for pre-screening of environmental chemicals for their neurodegenerative potential with altered metabolism.
Collapse
Affiliation(s)
- Arvind Kumar Shukla
- Embryotoxicology Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
| | - Ch Ratnasekhar
- Analytical Chemistry Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Campus, Lucknow, India
| | - Prakash Pragya
- Embryotoxicology Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
| | - Hitesh Singh Chaouhan
- Embryotoxicology Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Campus, Lucknow, India
| | - Devendra Kumar Patel
- Analytical Chemistry Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Campus, Lucknow, India
| | - Debapratim Kar Chowdhuri
- Embryotoxicology Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Campus, Lucknow, India.
| | - Mohana Krishna Reddy Mudiam
- Analytical Chemistry Section, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR Campus, Lucknow, India.
| |
Collapse
|
240
|
Dorninger F, Brodde A, Braverman NE, Moser AB, Just WW, Forss-Petter S, Brügger B, Berger J. Homeostasis of phospholipids - The level of phosphatidylethanolamine tightly adapts to changes in ethanolamine plasmalogens. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:117-28. [PMID: 25463479 PMCID: PMC4331674 DOI: 10.1016/j.bbalip.2014.11.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/04/2014] [Accepted: 11/10/2014] [Indexed: 01/19/2023]
Abstract
Ethanolamine plasmalogens constitute a group of ether glycerophospholipids that, due to their unique biophysical and biochemical properties, are essential components of mammalian cellular membranes. Their importance is emphasized by the consequences of defects in plasmalogen biosynthesis, which in humans cause the fatal disease rhizomelic chondrodysplasia punctata (RCDP). In the present lipidomic study, we used fibroblasts derived from RCDP patients, as well as brain tissue from plasmalogen-deficient mice, to examine the compensatory mechanisms of lipid homeostasis in response to plasmalogen deficiency. Our results show that phosphatidylethanolamine (PE), a diacyl glycerophospholipid, which like ethanolamine plasmalogens carries the head group ethanolamine, is the main player in the adaptation to plasmalogen insufficiency. PE levels were tightly adjusted to the amount of ethanolamine plasmalogens so that their combined levels were kept constant. Similarly, the total amount of polyunsaturated fatty acids (PUFAs) in ethanolamine phospholipids was maintained upon plasmalogen deficiency. However, we found an increased incorporation of arachidonic acid at the expense of docosahexaenoic acid in the PE fraction of plasmalogen-deficient tissues. These data show that under conditions of reduced plasmalogen levels, the amount of total ethanolamine phospholipids is precisely maintained by a rise in PE. At the same time, a shift in the ratio between ω-6 and ω-3 PUFAs occurs, which might have unfavorable, long-term biological consequences. Therefore, our findings are not only of interest for RCDP but may have more widespread implications also for other disease conditions, as for example Alzheimer's disease, that have been associated with a decline in plasmalogens. PE accurately compensates for the lack of plasmalogens in vitro and in vivo. PE levels decrease to adapt to excess of ethanolamine plasmalogens (PlsEtn). Plasmalogen deficiency favors incorporation of arachidonic acid into PE. Docosahexaenoic acid in ethanolamine phospholipids decreases upon PlsEtn depletion.
Collapse
Affiliation(s)
- Fabian Dorninger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| | - Alexander Brodde
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany.
| | - Nancy E Braverman
- Department of Human Genetics and Pediatrics, McGill University-Montreal Children's Hospital, 4060 Ste-Catherine West, PT-406.2, Montreal, QC H3Z 2Z3, Canada.
| | - Ann B Moser
- Peroxisomal Diseases Laboratory, The Hugo W Moser Research Institute, The Kennedy Krieger Institute, 707 N. Broadway, Baltimore, MD 21205, USA.
| | - Wilhelm W Just
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany.
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| | - Britta Brügger
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany.
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| |
Collapse
|
241
|
Shah A, Chen D, Boda AR, Foster LJ, Davis MJ, Hill MM. RaftProt: mammalian lipid raft proteome database. Nucleic Acids Res 2014; 43:D335-8. [PMID: 25392410 PMCID: PMC4383944 DOI: 10.1093/nar/gku1131] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RaftProt (http://lipid-raft-database.di.uq.edu.au/) is a database of mammalian lipid raft-associated proteins as reported in high-throughput mass spectrometry studies. Lipid rafts are specialized membrane microdomains enriched in cholesterol and sphingolipids thought to act as dynamic signalling and sorting platforms. Given their fundamental roles in cellular regulation, there is a plethora of information on the size, composition and regulation of these membrane microdomains, including a large number of proteomics studies. To facilitate the mining and analysis of published lipid raft proteomics studies, we have developed a searchable database RaftProt. In addition to browsing the studies, performing basic queries by protein and gene names, searching experiments by cell, tissue and organisms; we have implemented several advanced features to facilitate data mining. To address the issue of potential bias due to biochemical preparation procedures used, we have captured the lipid raft preparation methods and implemented advanced search option for methodology and sample treatment conditions, such as cholesterol depletion. Furthermore, we have identified a list of high confidence proteins, and enabled searching only from this list of likely bona fide lipid raft proteins. Given the apparent biological importance of lipid raft and their associated proteins, this database would constitute a key resource for the scientific community.
Collapse
Affiliation(s)
- Anup Shah
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - David Chen
- School of Information and Communication Technology, Griffith University, Brisbane, QLD, Australia
| | - Akash R Boda
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Leonard J Foster
- Centre for High-Throughput Biology, University of British Columbia, British Columbia, Canada
| | - Melissa J Davis
- Systems Biology Laboratory, Melbourne School of Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Michelle M Hill
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
242
|
Toops KA, Tan LX, Jiang Z, Radu RA, Lakkaraju A. Cholesterol-mediated activation of acid sphingomyelinase disrupts autophagy in the retinal pigment epithelium. Mol Biol Cell 2014; 26:1-14. [PMID: 25378587 PMCID: PMC4279221 DOI: 10.1091/mbc.e14-05-1028] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
How autophagy is regulated in the postmitotic retinal pigment epithelium (RPE) is unclear. Visual cycle metabolites and cholesterol that accumulate in the RPE inhibit autophagic flux by activating acid sphingomyelinase (ASMase). Increased ceramide promotes tubulin acetylation, which prevents autophagosome traffic. ASMase inhibition restores RPE autophagy. Autophagy is an essential mechanism for clearing damaged organelles and proteins within the cell. As with neurodegenerative diseases, dysfunctional autophagy could contribute to blinding diseases such as macular degeneration. However, precisely how inefficient autophagy promotes retinal damage is unclear. In this study, we investigate innate mechanisms that modulate autophagy in the retinal pigment epithelium (RPE), a key site of insult in macular degeneration. High-speed live imaging of polarized adult primary RPE cells and data from a mouse model of early-onset macular degeneration identify a mechanism by which lipofuscin bisretinoids, visual cycle metabolites that progressively accumulate in the RPE, disrupt autophagy. We demonstrate that bisretinoids trap cholesterol and bis(monoacylglycero)phosphate, an acid sphingomyelinase (ASMase) cofactor, within the RPE. ASMase activation increases cellular ceramide, which promotes tubulin acetylation on stabilized microtubules. Live-imaging data show that autophagosome traffic and autophagic flux are inhibited in RPE with acetylated microtubules. Drugs that remove excess cholesterol or inhibit ASMase reverse this cascade of events and restore autophagosome motility and autophagic flux in the RPE. Because accumulation of lipofuscin bisretinoids and abnormal cholesterol homeostasis are implicated in macular degeneration, our studies suggest that ASMase could be a potential therapeutic target to ensure the efficient autophagy that maintains RPE health.
Collapse
Affiliation(s)
- Kimberly A Toops
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, McPherson Eye Research Institute, and
| | - Li Xuan Tan
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53706
| | - Zhichun Jiang
- Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90024
| | - Roxana A Radu
- Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90024
| | - Aparna Lakkaraju
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, McPherson Eye Research Institute, and Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
243
|
Moyano AL, Li G, Lopez-Rosas A, Månsson JE, van Breemen RB, Givogri MI. Distribution of C16:0, C18:0, C24:1, and C24:0 sulfatides in central nervous system lipid rafts by quantitative ultra-high-pressure liquid chromatography tandem mass spectrometry. Anal Biochem 2014; 467:31-9. [PMID: 25205652 DOI: 10.1016/j.ab.2014.08.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/26/2014] [Indexed: 01/18/2023]
Abstract
Sulfated galactosylceramides (sulfatides) are glycosphingolipids associated with cholesterol- and sphingolipid-enriched membrane microdomains (lipid rafts) and are highly expressed in brain tissue. Although it is known that sulfatide species show heterogeneity in their fatty acid acyl group composition throughout brain development, their lipid raft distribution and biological relevance is poorly understood. We validated a fast and sensitive ultra-high-pressure liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) method to measure developmentally regulated sulfatide species (C16:0, C18:0, C24:1, and C24:0) in central nervous system (CNS) lipid rafts isolated without using detergent. Our UHPLC-MS/MS assay showed good accuracy and precision with a linear range of 5 to 1,000 nM for C18:0 and C24:1 sulfatides and 10 to 1,000 nM for C16:0 and C24:0 sulfatides. We applied this quantitative analysis to detergent-free lipid rafts isolated from wild-type mice and arylsulfatase A-deficient (ASA knockout) mice that accumulate sulfatides. All four sulfatide species were more abundant in raft membranes than in non-raft membranes, with a significant increase in lipid rafts isolated from ASA knockout mice. This is the first description of an analytical method to study these sulfatide species in raft and non-raft membranes and has the potential to be applied to preparations from other tissues.
Collapse
Affiliation(s)
- Ana Lis Moyano
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Guannan Li
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Aurora Lopez-Rosas
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jan-Eric Månsson
- Department of Clinical Chemistry, The Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Richard B van Breemen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Maria Irene Givogri
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
244
|
Systems-based analyses of brain regions functionally impacted in Parkinson's disease reveals underlying causal mechanisms. PLoS One 2014; 9:e102909. [PMID: 25170892 PMCID: PMC4149353 DOI: 10.1371/journal.pone.0102909] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 06/25/2014] [Indexed: 12/20/2022] Open
Abstract
Detailed analysis of disease-affected tissue provides insight into molecular mechanisms contributing to pathogenesis. Substantia nigra, striatum, and cortex are functionally connected with increasing degrees of alpha-synuclein pathology in Parkinson's disease. We undertook functional and causal pathway analysis of gene expression and proteomic alterations in these three regions, and the data revealed pathways that correlated with disease progression. In addition, microarray and RNAseq experiments revealed previously unidentified causal changes related to oligodendrocyte function and synaptic vesicle release, and these and other changes were reflected across all brain regions. Importantly, subsets of these changes were replicated in Parkinson's disease blood; suggesting peripheral tissue may provide important avenues for understanding and measuring disease status and progression. Proteomic assessment revealed alterations in mitochondria and vesicular transport proteins that preceded gene expression changes indicating defects in translation and/or protein turnover. Our combined approach of proteomics, RNAseq and microarray analyses provides a comprehensive view of the molecular changes that accompany functional loss and alpha-synuclein pathology in Parkinson's disease, and may be instrumental to understand, diagnose and follow Parkinson's disease progression.
Collapse
|
245
|
Wang A, Lin Y, Wu Y, Zhang D. Macronutrients intake and risk of Parkinson's disease: A meta-analysis. Geriatr Gerontol Int 2014; 15:606-16. [DOI: 10.1111/ggi.12321] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Aimin Wang
- Department of Nursing; Qingdao University Medical College; Qingdao China
| | - Yan Lin
- Department of Epidemiology and Health Statistics; Qingdao University Medical College; Qingdao China
| | - Yili Wu
- Department of Epidemiology and Health Statistics; Qingdao University Medical College; Qingdao China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics; Qingdao University Medical College; Qingdao China
| |
Collapse
|
246
|
Fernandez-Echevarria C, Díaz M, Ferrer I, Canerina-Amaro A, Marin R. Aβ promotes VDAC1 channel dephosphorylation in neuronal lipid rafts. Relevance to the mechanisms of neurotoxicity in Alzheimer's disease. Neuroscience 2014; 278:354-66. [PMID: 25168729 DOI: 10.1016/j.neuroscience.2014.07.079] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 11/25/2022]
Abstract
Voltage-dependent anion channel (VDAC) is a mitochondrial protein abundantly found in neuronal lipid rafts. In these membrane domains, VDAC is associated with a complex of signaling proteins that trigger neuroprotective responses. Loss of lipid raft integrity may result in disruption of multicomplex association and alteration of signaling responses that may ultimately promote VDAC activation. Some data have demonstrated that VDAC at the neuronal membrane may be involved in the mechanisms of amyloid beta (Aβ)-induced neurotoxicity, through yet unknown mechanisms. Aβ is generated from amyloid precursor protein (APP), and is released to the extracellular space where it may undergo self-aggregation. Aβ aggregate deposition in the form of senile plaques may lead to Alzheimer's disease (AD) neuropathology, although other pathological hallmarks (such as hyper-phosphorylated Tau deposition) also participate in this neurodegenerative process. The present study demonstrates that VDAC1 associates with APP and Aβ in lipid rafts of neurons. Interaction of VDAC1 with APP was observed in lipid rafts from the frontal and entorhinal cortex of human brains affected by AD at early stages (I-IV/0-B of Braak and Braak). Furthermore, Aβ exposure enhanced the dephosphorylation of VDAC1 that correlated with cell death. Both effects were reverted in the presence of tyrosine phosphatase inhibitors. VDAC1 dephosphorylation was corroborated in lipid rafts of AD brains. These results demonstrate that Aβ is involved in alterations of the phosphorylation state of VDAC in neuronal lipid rafts. Modulation of this channel may contribute to the development and progression of AD pathology.
Collapse
Affiliation(s)
- C Fernandez-Echevarria
- Laboratory of Cellular Neurobiology, Department of Physiology, University of La Laguna, School of Medicine, Sta. Cruz de Tenerife, Spain
| | - M Díaz
- Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Faculty of Biology, University of La Laguna, Sta. Cruz de Tenerife, Spain
| | - I Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, University of Barcelona, IDIBELL, CIBERNED, Hospitalet de Llobregat, Barcelona, Spain
| | - A Canerina-Amaro
- Laboratory of Cellular Neurobiology, Department of Physiology, University of La Laguna, School of Medicine, Sta. Cruz de Tenerife, Spain
| | - R Marin
- Laboratory of Cellular Neurobiology, Department of Physiology, University of La Laguna, School of Medicine, Sta. Cruz de Tenerife, Spain.
| |
Collapse
|
247
|
Casañas-Sánchez V, Pérez JA, Fabelo N, Herrera-Herrera AV, Fernández C, Marín R, González-Montelongo MC, Díaz M. Addition of docosahexaenoic acid, but not arachidonic acid, activates glutathione and thioredoxin antioxidant systems in murine hippocampal HT22 cells: potential implications in neuroprotection. J Neurochem 2014; 131:470-83. [DOI: 10.1111/jnc.12833] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 07/12/2014] [Accepted: 07/21/2014] [Indexed: 01/20/2023]
Affiliation(s)
- Verónica Casañas-Sánchez
- Department of Genetics; University Institute of Tropical Diseases and Public Health; University of La Laguna; Tenerife Spain
| | - José A. Pérez
- Department of Genetics; University Institute of Tropical Diseases and Public Health; University of La Laguna; Tenerife Spain
| | - Noemí Fabelo
- Laboratory of Membrane Physiology and Biophysics; Department of Animal Biology; University of La Laguna; Tenerife Spain
| | | | - Cecilia Fernández
- Laboratory of Cellular Neurobiology; Department of Physiology; University of La Laguna; Tenerife Spain
| | - Raquel Marín
- Laboratory of Cellular Neurobiology; Department of Physiology; University of La Laguna; Tenerife Spain
| | - María C. González-Montelongo
- Laboratory of Membrane Physiology and Biophysics; Department of Animal Biology; University of La Laguna; Tenerife Spain
| | - Mario Díaz
- Laboratory of Membrane Physiology and Biophysics; Department of Animal Biology; University of La Laguna; Tenerife Spain
| |
Collapse
|
248
|
Fabelo N, Martín V, Marín R, Moreno D, Ferrer I, Díaz M. Altered lipid composition in cortical lipid rafts occurs at early stages of sporadic Alzheimer's disease and facilitates APP/BACE1 interactions. Neurobiol Aging 2014; 35:1801-12. [DOI: 10.1016/j.neurobiolaging.2014.02.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/18/2014] [Accepted: 02/05/2014] [Indexed: 12/31/2022]
|
249
|
González-Redondo R, García-García D, Clavero P, Gasca-Salas C, García-Eulate R, Zubieta JL, Arbizu J, Obeso JA, Rodríguez-Oroz MC. Grey matter hypometabolism and atrophy in Parkinson's disease with cognitive impairment: a two-step process. ACTA ACUST UNITED AC 2014; 137:2356-67. [PMID: 24951642 DOI: 10.1093/brain/awu159] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The pathophysiological process underlying cognitive decline in Parkinson's disease is not well understood. Cerebral atrophy and hypometabolism have been described in patients with Parkinson's disease and dementia or mild cognitive impairment with respect to control subjects. However, the exact relationships between atrophy and hypometabolism are still unclear. To determine the extension and topographical distribution of hypometabolism and atrophy in the different cognitive states of Parkinson's disease, we examined 46 patients with Parkinson's disease (19 female, 27 male; 71.7 ± 5.9 years old; 14.6 ± 4.2 years of disease evolution; modified Hoehn and Yahr mean stage 3.1 ± 0.7). Cognitive status was diagnosed as normal in 14 patients, as mild cognitive impairment in 17 and as dementia in 15 patients. Nineteen normal subjects (eight female, 11 male; 68.1 ± 3.2 years old) were included as controls. (18)F-fluorodeoxyglucose positron emission tomography and magnetic resonance imaging scans were obtained, co-registered, corrected for partial volume effect and spatially normalized to the Montreal Neurological Institute space in each subject. Smoothing was applied to the positron emission tomography and magnetic resonance imaging scans to equalize their effective smoothness and resolution (10 mm and 12 mm full-width at half-maximum and Gaussian kernel, respectively). Z-score maps for atrophy and for hypometabolism were obtained by comparing individual images to the data set of control subjects. For each group of patients, a paired Student's t-test was performed to statistically compare the two Z-map modalities (P < 0.05 false discovery rate corrected) using the direct voxel-based comparison technique. In patients with mild cognitive impairment, hypometabolism exceeded atrophy in the angular gyrus, occipital, orbital and anterior frontal lobes. In patients with dementia, the hypometabolic areas observed in the group with mild cognitive impairment were replaced by areas of atrophy, which were surrounded by extensive zones of hypometabolism. Areas where atrophy was more extended than hypometabolism were found in the precentral and supplementary motor areas in both patients with mild cognitive impairment and with dementia, and in the hippocampus and temporal lobe in patients with dementia. These findings suggest that there is a gradient of severity in cortical changes associated with the development of cognitive impairment in Parkinson's disease in which hypometabolism and atrophy represent consecutive stages of the same process in most of the cortical regions affected.
Collapse
Affiliation(s)
- Rafael González-Redondo
- 1 Department of Neurology, Clinica Universidad de Navarra Medical School, Pamplona, Spain2 Neuroscience Centre, CIMA, Pamplona, Spain3 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - David García-García
- 2 Neuroscience Centre, CIMA, Pamplona, Spain3 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Pedro Clavero
- 1 Department of Neurology, Clinica Universidad de Navarra Medical School, Pamplona, Spain2 Neuroscience Centre, CIMA, Pamplona, Spain3 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Carmen Gasca-Salas
- 1 Department of Neurology, Clinica Universidad de Navarra Medical School, Pamplona, Spain2 Neuroscience Centre, CIMA, Pamplona, Spain3 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Reyes García-Eulate
- 4 Department of Radiology, Clinica Universidad de Navarra Medical School, Pamplona, Spain
| | - José L Zubieta
- 4 Department of Radiology, Clinica Universidad de Navarra Medical School, Pamplona, Spain
| | - Javier Arbizu
- 5 Department of Nuclear Medicine, Clinica Universidad de Navarra Medical School, Pamplona, Spain
| | - José A Obeso
- 1 Department of Neurology, Clinica Universidad de Navarra Medical School, Pamplona, Spain2 Neuroscience Centre, CIMA, Pamplona, Spain3 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - María C Rodríguez-Oroz
- 1 Department of Neurology, Clinica Universidad de Navarra Medical School, Pamplona, Spain2 Neuroscience Centre, CIMA, Pamplona, Spain3 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
250
|
Ariga T. Pathogenic role of ganglioside metabolism in neurodegenerative diseases. J Neurosci Res 2014; 92:1227-42. [PMID: 24903509 DOI: 10.1002/jnr.23411] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 12/13/2022]
Abstract
Ganglioside metabolism is altered in several neurodegenerative diseases, and this may participate in several events related to the pathogenesis of these diseases. Most changes occur in specific areas of the brain and their distinct membrane microdomains or lipid rafts. Antiganglioside antibodies may be involved in dysfunction of the blood-brain barrier and disease progression in these diseases. In lipid rafts, interactions of glycosphingolipids, including ganglioside, with proteins may be responsible for the misfolding events that cause the fibril and/or aggregate processing of disease-specific proteins, such as α-synuclein, in Parkinson's disease, huntingtin protein in Huntington's disease, and copper-zinc superoxide dismutase in amyotrophic lateral sclerosis. Targeting ganglioside metabolism may represent an underexploited opportunity to design novel therapeutic strategies for neurodegeneration in these diseases.
Collapse
Affiliation(s)
- Toshio Ariga
- Institute of Molecular Medicine and Genetics, Institute of Neuroscience, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| |
Collapse
|