251
|
Mukherjee J, Ladwa RM, Liang C, Syed AU. Elevated Monoamine Oxidase-A in Anterior Cingulate of Post-Mortem Human Parkinson's Disease: A Potential Surrogate Biomarker for Lewy Bodies? Cells 2022; 11:cells11244000. [PMID: 36552764 PMCID: PMC9777299 DOI: 10.3390/cells11244000] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 11/27/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Lewy bodies (LB) play a neuropathological role in Parkinson's disease (PD). Our goal was to evaluate LB using anti-ubiquitin immunohistochemistry (UIHC) and find correlations with monoamine oxidase-A (MAO-A) using imaging agent, [18F]FAZIN3. Human post-mortem anterior cingulate (AC) and corpus callosum (CC) from control subjects (CN), n = 6; age 81-90 LB = 0 and PD, n = 6, age 77-89, LB = III-IV were sectioned (10 μm slices). Brain slices were immunostained with anti-ubiquitin for LB (UIHC) and analyzed using QuPath for percent anti-ubiquitin per unit area (μm2). Adjacent brain slices were incubated with [18F]FAZIN3 and cortical layers I-III, IV-VI and CC (white matter) regions were quantified for the binding of [18F]FAZIN3. UIHC was correlated with [18F]FAZIN3 binding. All PD brains were positively UIHC stained and confirmed presence of LB. Outer cortical layers (I-III) of PD AC had 21% UIHC while inner layers (IV-VI) had >75% UIHC. In the CN brains LB were absent (<1% UIHC). Increased [18F]FAZIN3 binding to MAO-A in AC was observed in all PD subjects. [18F]FAZIN3 ratio in PD was AC/CC = 3.57 while in CN subjects it was AC/CC = 2.24. Increases in UIHC μm2 correlated with [18F]FAZIN3 binding to MAO-A in DLU/mm2. Increased [18F]FAZIN3 binding to MAO-A in PD is a potential novel "hot spot" PET imaging approach.
Collapse
|
252
|
Baek JM, Jung WH, Yu ES, Ahn DJ, Ryu YS. In Vitro Membrane Platform for the Visualization of Water Impermeability across the Liquid-Ordered Phase under Hypertonic Conditions. J Am Chem Soc 2022; 144:21887-21896. [PMID: 36367984 DOI: 10.1021/jacs.2c06626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Passive water penetration across the cell membrane by osmotic diffusion is essential for the homeostasis of cell volume, in addition to the protein-assisted active transportation of water. Since membrane components can regulate water permeability, controlling compositional variation during the volume regulatory process is a prerequisite for investigating the underlying mechanisms of water permeation and related membrane dynamics. However, the lack of a viable in vitro membrane platform in hypertonic solutions impedes advanced knowledge of cell volume regulation processes, especially cholesterol-enriched lipid domains called lipid rafts. By reconstituting the liquid-ordered (Lo) domain as a likeness of lipid rafts, we verified suppressed water permeation across the Lo domains, which had yet to be confirmed with experimental demonstrations despite a simulation approach. With the help of direct transfer of the Lo domains from vesicles to supported lipid membranes, the biological roles of lipid composition in suppressed water translocation were experimentally confirmed. Additionally, the improvement in membrane stability under hypertonic conditions was demonstrated based on molecular dynamics simulations.
Collapse
Affiliation(s)
- Ji Min Baek
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Woo Hyuk Jung
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Eui-Sang Yu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Dong June Ahn
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Yong-Sang Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
253
|
Fathy YY, Jonkman LE, Bol JJ, Timmermans E, Jonker AJ, Rozemuller AJM, van de Berg WDJ. Axonal degeneration in the anterior insular cortex is associated with Alzheimer's co-pathology in Parkinson's disease and dementia with Lewy bodies. Transl Neurodegener 2022; 11:52. [PMID: 36474289 PMCID: PMC9728006 DOI: 10.1186/s40035-022-00325-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Axons, crucial for impulse transmission and cellular trafficking, are thought to be primary targets of neurodegeneration in Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Axonal degeneration occurs early, preceeding and exceeding neuronal loss, and contributes to the spread of pathology, yet is poorly described outside the nigrostriatal circuitry. The insula, a cortical brain hub, was recently discovered to be highly vulnerable to pathology and plays a role in cognitive deficits in PD and DLB. The aim of this study was to evaluate morphological features as well as burden of proteinopathy and axonal degeneration in the anterior insular sub-regions in PD, PD with dementia (PDD), and DLB. METHODS α-Synuclein, phosphorylated (p-)tau, and amyloid-β pathology load were evaluated in the anterior insular (agranular and dysgranular) subregions of post-mortem human brains (n = 27). Axonal loss was evaluated using modified Bielschowsky silver staining and quantified using stereology. Cytoskeletal damage was comprehensively studied using immunofluorescent multi-labelling and 3D confocal laser-scanning microscopy. RESULTS Compared to PD and PDD, DLB showed significantly higher α-synuclein and p-tau pathology load, argyrophilic grains, and more severe axonal loss, particularly in the anterior agranular insula. Alternatively, the dysgranular insula showed a significantly higher load of amyloid-β pathology and its axonal density correlated with cognitive performance. p-Tau contributed most to axonal loss in the DLB group, was highest in the anterior agranular insula and significantly correlated with CDR global scores for dementia. Neurofilament and myelin showed degenerative changes including swellings, demyelination, and detachment of the axon-myelin unit. CONCLUSIONS Our results highlight the selective vulnerability of the anterior insular sub-regions to various converging pathologies, leading to impaired axonal integrity in PD, PDD and DLB, disrupting their functional properties and potentially contributing to cognitive, emotional, and autonomic deficits.
Collapse
Affiliation(s)
- Yasmine Y. Fathy
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands ,grid.5645.2000000040459992XDepartment of Neurology, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, Netherlands
| | - Laura E. Jonkman
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - John J. Bol
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Evelien Timmermans
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Allert J. Jonker
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Annemieke J. M. Rozemuller
- grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands ,grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Pathology, Amsterdam Neuroscience, Vrije University Amsterdam, De Boelelaan, Amsterdam, Netherlands
| | - Wilma D. J. van de Berg
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| |
Collapse
|
254
|
Mechanistic insights into HuR inhibitor MS-444 arresting embryonic development revealed by low-input RNA-seq and STORM. Cell Biol Toxicol 2022; 38:1175-1197. [PMID: 36085230 DOI: 10.1007/s10565-022-09757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023]
Abstract
With improvements in the survival rate of patients with cancer, fertility maintenance has become a major concern in terms of cancer treatment for women of reproductive age. Thus, it is important to examine the impact on fertility of anticancer drugs that are used clinically or are undergoing trials. The HuR small-molecule inhibitor MS-444 has been used in many cancer treatment studies, but its reproductive toxicity in females is unknown. Here, we reported that MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA by inhibiting HuR dimerization, resulting in the developmental arrest of 2-cell stage embryos in mouse. Combining analysis of low-input RNA-seq for MS-444-treated 2-cell embryos and mapping binding sites of RNA-binding protein, Agbl2 was predicted to be the target gene of MS-444. For further confirmation, RNAi experiment in wild-type zygotes showed that Agbl2 knockdown reduced the proportion of embryos successfully developed to the blastocyst stage: from 71% in controls to 23%. Furthermore, RNA-FISH and luciferase reporter analyses showed that MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA and reduced its stability by inhibiting HuR dimerization. In addition, optimized stochastic optical reconstruction microscopy (STORM) imaging showed that MS-444 significantly reduced the HuR dimerization, and HuR mainly existed in cluster form in 2-cell stage embryos. In conclusion, this study provides clinical guidance for maintaining fertility during the treatment of cancer with MS-444 in women of reproductive age. And also, our research provides guidance for the application of STORM in nanometer scale studies of embryonic cells. HuR inhibitor MS-444 arrested embryonic development at 2-cell stage. Low-input RNA-seq revealed that Agbl2 was the target gene of MS-444. MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA by inhibiting HuR dimerization and reduced the stability of Agbl2 mRNA. STORM with our optimized protocol showed that HuR tended to form elliptical and dense clusters in 2-cell stage embryos.
Collapse
|
255
|
Raja WK, Neves E, Burke C, Jiang X, Xu P, Rhodes KJ, Khurana V, Scannevin RH, Chung CY. Patient-derived three-dimensional cortical neurospheres to model Parkinson's disease. PLoS One 2022; 17:e0277532. [PMID: 36454869 PMCID: PMC9714816 DOI: 10.1371/journal.pone.0277532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 10/28/2022] [Indexed: 12/03/2022] Open
Abstract
There are currently no preventive or disease-modifying therapies for Parkinson's Disease (PD). Failures in clinical trials necessitate a re-evaluation of existing pre-clinical models in order to adopt systems that better recapitulate underlying disease mechanisms and better predict clinical outcomes. In recent years, models utilizing patient-derived induced pluripotent stem cells (iPSC) have emerged as attractive models to recapitulate disease-relevant neuropathology in vitro without exogenous overexpression of disease-related pathologic proteins. Here, we utilized iPSC derived from patients with early-onset PD and dementia phenotypes that harbored either a point mutation (A53T) or multiplication at the α-synuclein/SNCA gene locus. We generated a three-dimensional (3D) cortical neurosphere culture model to better mimic the tissue microenvironment of the brain. We extensively characterized the differentiation process using quantitative PCR, Western immunoblotting and immunofluorescence staining. Differentiated and aged neurospheres revealed alterations in fatty acid profiles and elevated total and pathogenic phospho-α-synuclein levels in both A53T and the triplication lines compared to their isogenic control lines. Furthermore, treatment of the neurospheres with a small molecule inhibitor of stearoyl CoA desaturase (SCD) attenuated the protein accumulation and aberrant fatty acid profile phenotypes. Our findings suggest that the 3D cortical neurosphere model is a useful tool to interrogate targets for PD and amenable to test small molecule therapeutics.
Collapse
Affiliation(s)
- Waseem K. Raja
- Yumanity Therapeutics, Boston, MA, United States of America
- * E-mail: (CYC); (WKR)
| | - Esther Neves
- Yumanity Therapeutics, Boston, MA, United States of America
| | | | - Xin Jiang
- Yumanity Therapeutics, Boston, MA, United States of America
| | - Ping Xu
- Yumanity Therapeutics, Boston, MA, United States of America
| | | | - Vikram Khurana
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Ann Romney Center for Neurologic Disease, Boston, MA, United States of America
- Harvard Stem Cell Institute, Cambridge, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | | | - Chee Yeun Chung
- Yumanity Therapeutics, Boston, MA, United States of America
- * E-mail: (CYC); (WKR)
| |
Collapse
|
256
|
Florio D, La Manna S, Di Natale C, Leone M, Mercurio FA, Napolitano F, Malfitano AM, Marasco D. Insights into Network of Hot Spots of Aggregation in Nucleophosmin 1. Int J Mol Sci 2022; 23:14704. [PMID: 36499032 PMCID: PMC9736328 DOI: 10.3390/ijms232314704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
In a protein, point mutations associated with diseases can alter the native structure and provide loss or alteration of functional levels, and an internal structural network defines the connectivity among domains, as well as aggregate/soluble states' equilibria. Nucleophosmin (NPM)1 is an abundant nucleolar protein, which becomes mutated in acute myeloid leukemia (AML) patients. NPM1-dependent leukemogenesis, which leads to its aggregation in the cytoplasm (NPMc+), is still obscure, but the investigations have outlined a direct link between AML mutations and amyloid aggregation. Protein aggregation can be due to the cooperation among several hot spots located within the aggregation-prone regions (APR), often predictable with bioinformatic tools. In the present study, we investigated potential APRs in the entire NPM1 not yet investigated. On the basis of bioinformatic predictions and experimental structures, we designed several protein fragments and analyzed them through typical aggrsegation experiments, such as Thioflavin T (ThT), fluorescence and scanning electron microscopy (SEM) experiments, carried out at different times; in addition, their biocompatibility in SHSY5 cells was also evaluated. The presented data clearly demonstrate the existence of hot spots of aggregation located in different regions, mostly in the N-terminal domain (NTD) of the entire NPM1 protein, and provide a more comprehensive view of the molecular details potentially at the basis of NPMc+-dependent AML.
Collapse
Affiliation(s)
- Daniele Florio
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Sara La Manna
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Concetta Di Natale
- Department of Chemical, Materials and Production Engineering, University of Naples “Federico II”, 80125 Naples, Italy
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging (CNR), 80145 Naples, Italy
| | | | - Fabiana Napolitano
- Department of Translational Medical Science, University of Naples “Federico II”, 80131 Naples, Italy
| | - Anna Maria Malfitano
- Department of Translational Medical Science, University of Naples “Federico II”, 80131 Naples, Italy
| | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
- Institute of Biostructures and Bioimaging (CNR), 80145 Naples, Italy
| |
Collapse
|
257
|
Dieriks BV, Highet B, Alik A, Bellande T, Stevenson TJ, Low V, Park TIH, Correia J, Schweder P, Faull RLM, Melki R, Curtis MA, Dragunow M. Human pericytes degrade diverse α-synuclein aggregates. PLoS One 2022; 17:e0277658. [PMID: 36399706 PMCID: PMC9674377 DOI: 10.1371/journal.pone.0277658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disorder characterised by the abnormal accumulation of α-synuclein (α-syn) aggregates. Central to disease progression is the gradual spread of pathological α-syn. α-syn aggregation is closely linked to progressive neuron loss. As such, clearance of α-syn aggregates may slow the progression of PD and lead to less severe symptoms. Evidence is increasing that non-neuronal cells play a role in PD and other synucleinopathies such as Lewy body dementia and multiple system atrophy. Our previous work has shown that pericytes-vascular mural cells that regulate the blood-brain barrier-contain α-syn aggregates in human PD brains. Here, we demonstrate that pericytes efficiently internalise fibrillar α-syn irrespective of being in a monoculture or mixed neuronal cell culture. Pericytes cleave fibrillar α-syn aggregates (Fibrils, Ribbons, fibrils65, fibrils91 and fibrils110), with cleaved α-syn remaining present for up to 21 days. The number of α-syn aggregates/cell and average aggregate size depends on the type of strain, but differences disappear within 5 five hours of treatment. Our results highlight the role brain vasculature may play in reducing α-syn aggregate burden in PD.
Collapse
Affiliation(s)
- Birger Victor Dieriks
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- * E-mail: (BVD); (MD)
| | - Blake Highet
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ania Alik
- Molecular Imaging Research Center, Francois Jacob Institute, Alternative Energies and Atomic Energy Commission, and Laboratory of Neurodegenerative Diseases, National Center for Scientific Research, Fontenay‐ Aux‐Roses, France
| | - Tracy Bellande
- Molecular Imaging Research Center, Francois Jacob Institute, Alternative Energies and Atomic Energy Commission, and Laboratory of Neurodegenerative Diseases, National Center for Scientific Research, Fontenay‐ Aux‐Roses, France
| | - Taylor J. Stevenson
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Victoria Low
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Thomas I-H Park
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Jason Correia
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Auckland City Hospital, Auckland, New Zealand
| | - Richard L. M. Faull
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ronald Melki
- Molecular Imaging Research Center, Francois Jacob Institute, Alternative Energies and Atomic Energy Commission, and Laboratory of Neurodegenerative Diseases, National Center for Scientific Research, Fontenay‐ Aux‐Roses, France
| | - Maurice A. Curtis
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- * E-mail: (BVD); (MD)
| |
Collapse
|
258
|
Abstract
α-synuclein misfolding and aggregation into fibrils is a common feature of α-synucleinopathies, such as Parkinson's disease, in which α-synuclein fibrils are a characteristic hallmark of neuronal inclusions called Lewy bodies. Studies on the composition of Lewy bodies extracted postmortem from brain tissue of Parkinson's patients revealed that lipids and membranous organelles are also a significant component. Interactions between α-synuclein and lipids have been previously identified as relevant for Parkinson's disease pathology, however molecular insights into their interactions have remained elusive. Here we present cryo-electron microscopy structures of six α-synuclein fibrils in complex with lipids, revealing specific lipid-fibril interactions. We observe that phospholipids promote an alternative protofilament fold, mediate an unusual arrangement of protofilaments, and fill the central cavities of the fibrils. Together with our previous studies, these structures also indicate a mechanism for fibril-induced lipid extraction, which is likely to be involved in the development of α-synucleinopathies. Specifically, one potential mechanism for the cellular toxicity is the disruption of intracellular vesicles mediated by fibrils and oligomers, and therefore the modulation of these interactions may provide a promising strategy for future therapeutic interventions.
Collapse
|
259
|
Vantaggiato L, Shaba E, Carleo A, Bezzini D, Pannuzzo G, Luddi A, Piomboni P, Bini L, Bianchi L. Neurodegenerative Disorder Risk in Krabbe Disease Carriers. Int J Mol Sci 2022; 23:13537. [PMID: 36362324 PMCID: PMC9654610 DOI: 10.3390/ijms232113537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
Abstract
Krabbe disease (KD) is a rare autosomal recessive disorder caused by mutations in the galactocerebrosidase gene (GALC). Defective GALC causes aberrant metabolism of galactolipids present almost exclusively in myelin, with consequent demyelinization and neurodegeneration of the central and peripheral nervous system (NS). KD shares some similar features with other neuropathies and heterozygous carriers of GALC mutations are emerging with an increased risk in developing NS disorders. In this work, we set out to identify possible variations in the proteomic profile of KD-carrier brain to identify altered pathways that may imbalance its homeostasis and that may be associated with neurological disorders. The differential analysis performed on whole brains from 33-day-old twitcher (galc -/-), heterozygous (galc +/-), and wild-type mice highlighted the dysregulation of several multifunctional factors in both heterozygous and twitcher mice. Notably, the KD-carrier mouse, despite its normal phenotype, presents the deregulation of vimentin, receptor of activated protein C kinase 1 (RACK1), myelin basic protein (MBP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNP), transitional endoplasmic reticulum ATPase (VCP), and N-myc downstream regulated gene 1 protein (NDRG1) as well as changes in the ubiquitinated-protein pattern. Our findings suggest the carrier may be affected by dysfunctions classically associated with neurodegeneration: (i) alteration of (mechano) signaling and intracellular trafficking, (ii) a generalized affection of proteostasis and lipid metabolism, with possible defects in myelin composition and turnover, and (iii) mitochondrion and energy supply dysfunctions.
Collapse
Affiliation(s)
- Lorenza Vantaggiato
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Enxhi Shaba
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Alfonso Carleo
- Department of Pulmonology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Daiana Bezzini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Giovanna Pannuzzo
- Department of Biochemical and Biotechnological Sciences, Section of Physiology, University of Catania, 95121 Catania, Italy
| | - Alice Luddi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Paola Piomboni
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Luca Bini
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
260
|
Iannielli A, Luoni M, Giannelli SG, Ferese R, Ordazzo G, Fossati M, Raimondi A, Opazo F, Corti O, Prehn JHM, Gambardella S, Melki R, Broccoli V. Modeling native and seeded Synuclein aggregation and related cellular dysfunctions in dopaminergic neurons derived by a new set of isogenic iPSC lines with SNCA multiplications. Cell Death Dis 2022; 13:881. [PMID: 36261424 PMCID: PMC9581971 DOI: 10.1038/s41419-022-05330-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
Abstract
Triplication of the SNCA gene, encoding the protein alpha-Synuclein (αSyn), is a rare cause of aggressive and early-onset parkinsonism. Herein, we generated iPSCs from two siblings with a recently described compact SNCA gene triplication and suffering from severe motor impairments, psychiatric symptoms, and cognitive deterioration. Using CRISPR/Cas9 gene editing, each SNCA copy was inactivated by targeted indel mutations generating a panel of isogenic iPSCs with a decremental number from 4 down to none of functional SNCA gene alleles. We differentiated these iPSC lines in midbrain dopaminergic (DA) neuronal cultures to characterize αSyn aggregation in native and seeded conditions and evaluate its associated cellular dysfunctions. Utilizing a new nanobody-based biosensor combined with super-resolved imaging, we were able to visualize and measure αSyn aggregates in early DA neurons in unstimulated conditions. Calcium dysregulation and mitochondrial alterations were the first pathological signs detectable in early differentiated DA neuronal cultures. Accelerated αSyn aggregation was induced by exposing neurons to structurally well-characterized synthetic αSyn fibrils. 4xSNCA DA neurons showed the highest vulnerability, which was associated with high levels of oxidized DA and amplified by TAX1BP1 gene disruption. Seeded DA neurons developed large αSyn deposits whose morphology and internal constituents resembled Lewy bodies commonly observed in Parkinson's disease (PD) patient brain tissues. These findings provide strong evidence that this isogenic panel of iPSCs with SNCA multiplications offers a remarkable cellular platform to investigate mechanisms of PD and validate candidate inhibitors of native and seeded αSyn aggregation.
Collapse
Affiliation(s)
- Angelo Iannielli
- grid.5326.20000 0001 1940 4177National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy ,grid.18887.3e0000000417581884Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mirko Luoni
- grid.18887.3e0000000417581884Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Serena Gea Giannelli
- grid.18887.3e0000000417581884Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Gabriele Ordazzo
- grid.18887.3e0000000417581884Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Matteo Fossati
- grid.5326.20000 0001 1940 4177National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy ,grid.417728.f0000 0004 1756 8807IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano Milan, Italy
| | - Andrea Raimondi
- grid.18887.3e0000000417581884Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Felipe Opazo
- grid.411984.10000 0001 0482 5331University Medical Center Göttingen, D-37073 Göttingen, Germany
| | - Olga Corti
- grid.425274.20000 0004 0620 5939Sorbonne Université, Institut du Cerveau (ICM), Inserm U1127, CNRS, UMR 7225 Paris, France
| | - Jochen H. M. Prehn
- grid.4912.e0000 0004 0488 7120Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Department of Physiology and Medical Physics and SFI FutureNeuro Research Centre, 123 St. Stephen’s Green, Dublin, Ireland
| | - Stefano Gambardella
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.12711.340000 0001 2369 7670Department of Biomolecular Sciences, University of Urbino “Carlo Bo,, Urbino, Italy
| | - Ronald Melki
- grid.460789.40000 0004 4910 6535Institut Francois Jacob, Molecular Imaging Center (MIRCen), Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA) and Centre National de la Recherche Scientifique (CNRS), Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Vania Broccoli
- grid.5326.20000 0001 1940 4177National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy ,grid.18887.3e0000000417581884Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
261
|
Association of Glial Activation and α-Synuclein Pathology in Parkinson's Disease. Neurosci Bull 2022; 39:479-490. [PMID: 36229715 PMCID: PMC10043108 DOI: 10.1007/s12264-022-00957-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/10/2022] [Indexed: 10/17/2022] Open
Abstract
The accumulation of pathological α-synuclein (α-syn) in the central nervous system and the progressive loss of dopaminergic neurons in the substantia nigra pars compacta are the neuropathological features of Parkinson's disease (PD). Recently, the findings of prion-like transmission of α-syn pathology have expanded our understanding of the region-specific distribution of α-syn in PD patients. Accumulating evidence suggests that α-syn aggregates are released from neurons and endocytosed by glial cells, which contributes to the clearance of α-syn. However, the activation of glial cells by α-syn species produces pro-inflammatory factors that decrease the uptake of α-syn aggregates by glial cells and promote the transmission of α-syn between neurons, which promotes the spread of α-syn pathology. In this article, we provide an overview of current knowledge on the role of glia and α-syn pathology in PD pathogenesis, highlighting the relationships between glial responses and the spread of α-syn pathology.
Collapse
|
262
|
Functional amyloid in a lipid-like environment: a merry dance of many steps. Essays Biochem 2022; 66:1035-1046. [PMID: 36205438 DOI: 10.1042/ebc20220062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022]
Abstract
Functional amyloid, which unlike its pathological counterpart serves a biological purpose, is produced in a carefully orchestrated sequence of events. In bacteria, the major amyloid component is transported over the periplasm and through the outer membrane to assemble on the bacterial cell surface. During its life time, the amyloid protein may be exposed to both membrane lipids and extracellular surfactant, making it relevant to study its interactions with these components in vitro. Particularly for charged surfactants, the interaction is quite complex and highly dependent on the surfactant:protein molar ratio. Low ratios typically promote aggregation, likely by binding the proteins to micelles and thus increasing the local concentration of proteins, while higher concentrations see an inhibition of the same process as the protein is diluted out and immobilized on individual micelles. This is particularly pronounced for strongly anionic surfactants like SDS; the naturally occurring biosurfactant rhamnolipid interacts more weakly with the protein, which still not only allows aggregation but also leads to less detrimental effects at higher ratios. Similarly, anionic vesicle-forming lipids largely stimulate aggregation likely because of weaker interactions. Anionic lysolipids, thanks to their micelle-forming properties, resemble SDS in their impact on fibrillation. There are also examples of systems where membrane binding sequesters an otherwise amyloidogenic sequence and prevents fibrillation or-quite the opposite- liberates another part of the protein to engage in self-assembly. Thus, membranes and surfactants have very varied roles to play in the biogenesis and function of bacterial amyloid.
Collapse
|
263
|
Zhaliazka K, Rizevsky S, Matveyenka M, Serada V, Kurouski D. Charge of Phospholipids Determines the Rate of Lysozyme Aggregation but Not the Structure and Toxicity of Amyloid Aggregates. J Phys Chem Lett 2022; 13:8833-8839. [PMID: 36111888 PMCID: PMC10405293 DOI: 10.1021/acs.jpclett.2c02126] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Biophysical properties of plasma membranes are determined by a chemical structure of phospholipids, including saturation of fatty acids and charge of polar heads of these molecules. Phospholipids not only determine fluidity and plasticity of membranes but also play an important role in abrupt aggregation of misfolded proteins. In this study, we investigate the role of the charge of the most abundant phospholipids in the plasma membrane on the aggregation properties of the lysozyme. We found that the charge of phospholipids determines the aggregation rate of lysozyme and the morphology of the protein aggregates. However, the secondary structure and toxicity of these protein specimens are determined by the chemical nature rather than the charge of phospholipids. These findings show that the charge of phospholipids can be a key factor that determines the stability and aggregation mechanism of amyloidogenic proteins.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biotechnology, Binh Duong University, Thu Dau Mot 820000, Vietnam
| | - Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Valeryia Serada
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
264
|
Inflammatory gut as a pathologic and therapeutic target in Parkinson’s disease. Cell Death Dis 2022; 8:396. [PMID: 36153318 PMCID: PMC9509357 DOI: 10.1038/s41420-022-01175-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 08/10/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022]
Abstract
Parkinson’s disease (PD) remains a significant unmet clinical need. Gut dysbiosis stands as a PD pathologic source and therapeutic target. Here, we assessed the role of the gut-brain axis in PD pathology and treatment. Adult transgenic (Tg) α-synuclein-overexpressing mice served as subjects and were randomly assigned to either transplantation of vehicle or human umbilical cord blood-derived stem cells and plasma. Behavioral and immunohistochemical assays evaluated the functional outcomes following transplantation. Tg mice displayed typical motor and gut motility deficits, elevated α-synuclein levels, and dopaminergic depletion, accompanied by gut dysbiosis characterized by upregulation of microbiota and cytokines associated with inflammation in the gut and the brain. In contrast, transplanted Tg mice displayed amelioration of motor deficits, improved sparing of nigral dopaminergic neurons, and downregulation of α-synuclein and inflammatory-relevant microbiota and cytokines in both gut and brain. Parallel in vitro studies revealed that cultured dopaminergic SH-SY5Y cells exposed to homogenates of Tg mouse-derived dysbiotic gut exhibited significantly reduced cell viability and elevated inflammatory signals compared to wild-type mouse-derived gut homogenates. Moreover, treatment with human umbilical cord blood-derived stem cells and plasma improved cell viability and decreased inflammation in dysbiotic gut-exposed SH-SY5Y cells. Intravenous transplantation of human umbilical cord blood-derived stem/progenitor cells and plasma reduced inflammatory microbiota and cytokine, and dampened α-synuclein overload in the gut and the brain of adult α-synuclein-overexpressing Tg mice. Our findings advance the gut-brain axis as a key pathological origin, as well as a robust therapeutic target for PD. Gut-Brain Axis as a PD Pathologic Source and Therapeutic Target. The PD murine model of α-synuclein overexpression at around 8 weeks of age manifests gut dysbiosis, characterized by inflammation-specific microbiota and cytokines, which can trigger brain neurodegeneration, especially dopaminergic depletion reminiscent of PD pathology. Targeting the dysbiotic gut via intravenous hUCB stem cell transplantation can render gut homeostasis and sequester peripheral as well as central inflammation, leading to brain repair and amelioration of PD behavioral and histological deficits.![]()
Collapse
|
265
|
Zhang H, Fang H, Liu D, Zhang Y, Adu-Amankwaah J, Yuan J, Tan R, Zhu J. Applications and challenges of rhodopsin-based optogenetics in biomedicine. Front Neurosci 2022; 16:966772. [PMID: 36213746 PMCID: PMC9537737 DOI: 10.3389/fnins.2022.966772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Optogenetics is an emerging bioengineering technology that has been rapidly developed in recent years by cross-integrating optics, genetic engineering, electrophysiology, software control, and other disciplines. Since the first demonstration of the millisecond neuromodulation ability of the channelrhodopsin-2 (ChR2), the application of optogenetic technology in basic life science research has been rapidly progressed, especially in neurobiology, which has driven the development of the discipline. As the optogenetic tool protein, microbial rhodopsins have been continuously explored, modified, and optimized, with many variants becoming available, with structural characteristics and functions that are highly diversified. Their applicability has been broadened, encouraging more researchers and clinicians to utilize optogenetics technology in research. In this review, we summarize the species and variant types of the most important class of tool proteins in optogenetic techniques, the microbial rhodopsins, and review the current applications of optogenetics based on rhodopsin qualitative light in biology and other fields. We also review the challenges facing this technology, to ultimately provide an in-depth technical reference to support the application of optogenetics in translational and clinical research.
Collapse
Affiliation(s)
- Hanci Zhang
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hui Fang
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Deqiang Liu
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yiming Zhang
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, China
| | - Jinxiang Yuan
- College of Life Sciences, Shandong Normal University, Jinan, China
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, China
- *Correspondence: Jinxiang Yuan,
| | - Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, China
- Rubin Tan,
| | - Jianping Zhu
- College of Life Sciences, Shandong Normal University, Jinan, China
- Jianping Zhu,
| |
Collapse
|
266
|
Small soluble α-synuclein aggregates are the toxic species in Parkinson's disease. Nat Commun 2022; 13:5512. [PMID: 36127374 PMCID: PMC9489799 DOI: 10.1038/s41467-022-33252-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Soluble α-synuclein aggregates varying in size, structure, and morphology have been closely linked to neuronal death in Parkinson's disease. However, the heterogeneity of different co-existing aggregate species makes it hard to isolate and study their individual toxic properties. Here, we show a reliable non-perturbative method to separate a heterogeneous mixture of protein aggregates by size. We find that aggregates of wild-type α-synuclein smaller than 200 nm in length, formed during an in vitro aggregation reaction, cause inflammation and permeabilization of single-liposome membranes and that larger aggregates are less toxic. Studying soluble aggregates extracted from post-mortem human brains also reveals that these aggregates are similar in size and structure to the smaller aggregates formed in aggregation reactions in the test tube. Furthermore, we find that the soluble aggregates present in Parkinson's disease brains are smaller, largely less than 100 nm, and more inflammatory compared to the larger aggregates present in control brains. This study suggests that the small non-fibrillar α-synuclein aggregates are the critical species driving neuroinflammation and disease progression.
Collapse
|
267
|
Fonseca-Ornelas L, Stricker JMS, Soriano-Cruz S, Weykopf B, Dettmer U, Muratore CR, Scherzer CR, Selkoe DJ. Parkinson-causing mutations in LRRK2 impair the physiological tetramerization of endogenous α-synuclein in human neurons. NPJ Parkinsons Dis 2022; 8:118. [PMID: 36114228 PMCID: PMC9481630 DOI: 10.1038/s41531-022-00380-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
α-Synuclein (αSyn) aggregation in Lewy bodies and neurites defines both familial and 'sporadic' Parkinson's disease. We previously identified α-helically folded αSyn tetramers, in addition to the long-known unfolded monomers, in normal cells. PD-causing αSyn mutations decrease the tetramer:monomer (T:M) ratio, associated with αSyn hyperphosphorylation and cytotoxicity in neurons and a motor syndrome of tremor and gait deficits in transgenic mice that responds in part to L-DOPA. Here, we asked whether LRRK2 mutations, the most common genetic cause of cases previously considered sporadic PD, also alter tetramer homeostasis. Patient neurons carrying G2019S, the most prevalent LRRK2 mutation, or R1441C each had decreased T:M ratios and pSer129 hyperphosphorylation of their endogenous αSyn along with increased phosphorylation of Rab10, a widely reported substrate of LRRK2 kinase activity. Two LRRK2 kinase inhibitors normalized the T:M ratio and the hyperphosphorylation in the G2019S and R1441C patient neurons. An inhibitor of stearoyl-CoA desaturase, the rate-limiting enzyme for monounsaturated fatty acid synthesis, also restored the αSyn T:M ratio and reversed pSer129 hyperphosphorylation in both mutants. Coupled with the recent discovery that PD-causing mutations of glucocerebrosidase in Gaucher's neurons also decrease T:M ratios, our findings indicate that three dominant genetic forms of PD involve life-long destabilization of αSyn physiological tetramers as a common pathogenic mechanism that can occur upstream of progressive neuronal synucleinopathy. Based on αSyn's finely-tuned interaction with certain vesicles, we hypothesize that the fatty acid composition and fluidity of membranes regulate αSyn's correct binding to highly curved membranes and subsequent assembly into metastable tetramers.
Collapse
Affiliation(s)
- Luis Fonseca-Ornelas
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jonathan M S Stricker
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Stephanie Soriano-Cruz
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Beatrice Weykopf
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Christina R Muratore
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Clemens R Scherzer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
268
|
Antonschmidt L, Matthes D, Dervişoğlu R, Frieg B, Dienemann C, Leonov A, Nimerovsky E, Sant V, Ryazanov S, Giese A, Schröder GF, Becker S, de Groot BL, Griesinger C, Andreas LB. The clinical drug candidate anle138b binds in a cavity of lipidic α-synuclein fibrils. Nat Commun 2022; 13:5385. [PMID: 36104315 PMCID: PMC9474542 DOI: 10.1038/s41467-022-32797-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Aggregation of amyloidogenic proteins is a characteristic of multiple neurodegenerative diseases. Atomic resolution of small molecule binding to such pathological protein aggregates is of interest for the development of therapeutics and diagnostics. Here we investigate the interaction between α-synuclein fibrils and anle138b, a clinical drug candidate for disease modifying therapy in neurodegeneration and a promising scaffold for positron emission tomography tracer design. We used nuclear magnetic resonance spectroscopy and the cryogenic electron microscopy structure of α-synuclein fibrils grown in the presence of lipids to locate anle138b within a cavity formed between two β-strands. We explored and quantified multiple binding modes of the compound in detail using molecular dynamics simulations. Our results reveal stable polar interactions between anle138b and backbone moieties inside the tubular cavity of the fibrils. Such cavities are common in other fibril structures as well.
Collapse
Affiliation(s)
- Leif Antonschmidt
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dirk Matthes
- Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Rıza Dervişoğlu
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Max Planck Institute for Chemical Energy Conversion, Mülheim an der Ruhr, Germany
| | - Benedikt Frieg
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
| | - Christian Dienemann
- Department of Molecular Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Andrei Leonov
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany
| | - Evgeny Nimerovsky
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Vrinda Sant
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sergey Ryazanov
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany
| | - Armin Giese
- MODAG GmbH, Mikroforum Ring 3, 55234, Wendelsheim, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Gunnar F Schröder
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
- Physics Department, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Becker
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bert L de Groot
- Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Christian Griesinger
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Loren B Andreas
- NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
269
|
Cao Q, Luo S, Yao W, Qu Y, Wang N, Hong J, Murayama S, Zhang Z, Chen J, Hashimoto K, Qi Q, Zhang JC. Suppression of abnormal α-synuclein expression by activation of BDNF transcription ameliorates Parkinson's disease-like pathology. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:1-15. [PMID: 35784012 PMCID: PMC9207554 DOI: 10.1016/j.omtn.2022.05.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/26/2022] [Indexed: 11/25/2022]
Abstract
Parkinson’s disease (PD) is characterized by the formation of Lewy bodies (LBs) in the brain. LBs are mainly composed of phosphorylated and aggregated α-synuclein (α-Syn). Thus, strategies to reduce the expression of α-Syn offer promising therapeutic avenues for PD. DNA/RNA heteroduplex oligonucleotides (HDOs) are a novel technology for gene silencing. Using an α-Syn-HDO that specifically targets α-Syn, we examined whether α-Syn-HDO attenuates pathological changes in the brain of mouse models of PD. Overexpression of α-Syn induced dopaminergic neuron degeneration through inhibition of cyclic AMP-responsive-element-binding protein (CREB) and activation of methyl CpG binding protein 2 (MeCP2), resulting in brain-derived neurotrophic factor (BDNF) downregulation. α-Syn-HDO exerted a more potent silencing effect on α-Syn than α-Syn-antisense oligonucleotides (ASOs). α-Syn-HDO attenuated abnormal α-Syn expression and ameliorated dopaminergic neuron degeneration via BDNF upregulation by activation of CREB and inhibition of MeCP2. These findings demonstrated that inhibition of α-Syn by α-Syn-HDO protected against dopaminergic neuron degeneration via activation of BDNF transcription. Therefore, α-Syn-HDO may serve as a new therapeutic agent for PD.
Collapse
Affiliation(s)
- Qianqian Cao
- Department of Physiology, School of Medicine, Jinan University, Guangzhou 510632, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shilin Luo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Wei Yao
- Guangzhou Key Laboratory of Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 260-8670 Chiba, Japan
| | - Nanbu Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510632, China
| | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shigeo Murayama
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital & Institute of Gerontology, Tokyo 173-0015, Japan
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 260-8670 Chiba, Japan
| | - Qi Qi
- MOE Key Laboratory of Tumor Molecular Biology, Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Ji-Chun Zhang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
270
|
López de Frutos L, Almeida F, Murillo-Saich J, Conceição VA, Guma M, Queheberger O, Giraldo P, Miltenberger-Miltenyi G. Serum Phospholipid Profile Changes in Gaucher Disease and Parkinson's Disease. Int J Mol Sci 2022; 23:ijms231810387. [PMID: 36142296 PMCID: PMC9499334 DOI: 10.3390/ijms231810387] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 01/30/2023] Open
Abstract
Alterations in the levels of serum sphingolipids and phospholipids have been reported in Gaucher disease and in Parkinson's disease, suggesting a potential role of these lipids as biomarkers. This project's objective is to detect novel associations and novel candidate biomarkers in the largest Spanish Gaucher and Parkinson diseases of the Iberian Peninsula. For that, 278 participants were included: 100 sporadic Parkinson's patients, 70 Gaucher patients, 15 GBA1-mutation-carrier Parkinson's patients and 93 controls. A serum lipidomics array including 10 phospholipid groups, 368 species, was performed using high-performance liquid chromatography-mass spectrometry. Lipid levels were compared between groups via multiple-regression analyses controlling for clinical and demographic parameters. Additionally, lipid levels were compared within the Gaucher and Parkinson's groups controlling for medication and/or disease severity. Results were controlled for robustness by filtering of non-detectable lipid values. There was an increase in the levels of phosphatidylcholine, with a simultaneous decrease in lyso-phosphatidylcholine, in the Gaucher, Parkinson's and GBA1-mutation-carrier Parkinson's patients vs. controls. Phosphatidylethanolamine, lyso- and plasmalogen-phosphatidylethanolamine were also increased in Gaucher and Parkinson's. Gaucher patients also showed an increase in lyso-phosphatidylserine and phosphatidylglycerol. While in the Gaucher and Parkinson's groups, velaglucerase alpha and dopamine agonists, respectively, showed positive associations with the lipid changes, miglustat treatment in Gaucher patients normalized the altered phosphatidylcholine/lyso-phosphatidylcholine ratio. In conclusion, Gaucher and Parkinson's patients showed changes in various serum phospholipid levels when compared with healthy controls, further supporting the role of such lipids in disease development and, possibly, as putative biomarkers. This hypothesis was reinforced by the normalizing effect of miglustat, and by controlling for data robustness, even though the limited number of participants, especially in the sub-distribution by treatment groups in GD requires validation in a larger number of patients.
Collapse
Affiliation(s)
- Laura López de Frutos
- Fundación para el Estudio y la Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), 50006 Zaragoza, Spain
- GIIS-012, Instituto de Investigación Sanitaria Aragón (IIS Aragón), Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - Francisco Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649004 Lisbon, Portugal
| | | | - Vasco A. Conceição
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649004 Lisbon, Portugal
| | - Monica Guma
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- VA Medical Center, San Diego, CA 92093, USA
- Department of Medicine, Autonomous University of Barcelona, 08193 Bellaterra, Spain
| | - Oswald Queheberger
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Pilar Giraldo
- Fundación para el Estudio y la Terapéutica de la Enfermedad de Gaucher y Otras Lisosomales (FEETEG), 50006 Zaragoza, Spain
- Correspondence: (P.G.); (G.M.-M.); Tel.: +34-670-285-339 (P.G.); +351-21-799-9435 (G.M.-M.)
| | - Gabriel Miltenberger-Miltenyi
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649004 Lisbon, Portugal
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649004 Lisbon, Portugal
- Department of Neurology, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
- Genetics Department, Reference Center on Lysosomal Storage Disorders, Hospital Senhora da Oliveira, 4835-044 Guimarães, Portugal
- Correspondence: (P.G.); (G.M.-M.); Tel.: +34-670-285-339 (P.G.); +351-21-799-9435 (G.M.-M.)
| |
Collapse
|
271
|
Kuznetsov IA, Kuznetsov AV. An analytical solution simulating growth of Lewy bodies. MATHEMATICAL MEDICINE AND BIOLOGY : A JOURNAL OF THE IMA 2022; 39:299-312. [PMID: 35656792 DOI: 10.1093/imammb/dqac006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/24/2022] [Accepted: 04/28/2022] [Indexed: 06/15/2023]
Abstract
This paper reports a minimal model simulating the growth of a Lewy body (LB). To the best of our knowledge, this is the first model simulating LB growth. The LB is assumed to consist of a central spherical core, which is composed of membrane fragments and various dysfunctional intracellular organelles, and a halo, which is composed of alpha-synuclein (α-syn) fibrils. Membrane fragments and α-syn monomers are assumed to be produced in the soma at constant rates. The growth of the core and the halo are simulated by the Finke-Watzky model. Analytical (closed-form) solutions describing the growth of the core and the halo are obtained. A sensitivity analysis in terms of model parameters is performed.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA
| |
Collapse
|
272
|
Toomey CE, Heywood WE, Evans JR, Lachica J, Pressey SN, Foti SC, Al Shahrani M, D’Sa K, Hargreaves IP, Heales S, Orford M, Troakes C, Attems J, Gelpi E, Palkovits M, Lashley T, Gentleman SM, Revesz T, Mills K, Gandhi S. Mitochondrial dysfunction is a key pathological driver of early stage Parkinson's. Acta Neuropathol Commun 2022; 10:134. [PMID: 36076304 PMCID: PMC9461181 DOI: 10.1186/s40478-022-01424-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The molecular drivers of early sporadic Parkinson's disease (PD) remain unclear, and the presence of widespread end stage pathology in late disease masks the distinction between primary or causal disease-specific events and late secondary consequences in stressed or dying cells. However, early and mid-stage Parkinson's brains (Braak stages 3 and 4) exhibit alpha-synuclein inclusions and neuronal loss along a regional gradient of severity, from unaffected-mild-moderate-severe. Here, we exploited this spatial pathological gradient to investigate the molecular drivers of sporadic PD. METHODS We combined high precision tissue sampling with unbiased large-scale profiling of protein expression across 9 brain regions in Braak stage 3 and 4 PD brains, and controls, and verified these results using targeted proteomic and functional analyses. RESULTS We demonstrate that the spatio-temporal pathology gradient in early-mid PD brains is mirrored by a biochemical gradient of a changing proteome. Importantly, we identify two key events that occur early in the disease, prior to the occurrence of alpha-synuclein inclusions and neuronal loss: (i) a metabolic switch in the utilisation of energy substrates and energy production in the brain, and (ii) perturbation of the mitochondrial redox state. These changes may contribute to the regional vulnerability of developing alpha-synuclein pathology. Later in the disease, mitochondrial function is affected more severely, whilst mitochondrial metabolism, fatty acid oxidation, and mitochondrial respiration are affected across all brain regions. CONCLUSIONS Our study provides an in-depth regional profile of the proteome at different stages of PD, and highlights that mitochondrial dysfunction is detectable prior to neuronal loss, and alpha-synuclein fibril deposition, suggesting that mitochondrial dysfunction is one of the key drivers of early disease.
Collapse
Affiliation(s)
- Christina E. Toomey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Wendy E. Heywood
- Translational Mass Spectrometry Research Group, Genetic & Genomic Medicine, Institute of Child Health, UCL, London, UK
| | - James R. Evans
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Joanne Lachica
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah N. Pressey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Sandrine C. Foti
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Mesfer Al Shahrani
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
- College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Karishma D’Sa
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Iain P. Hargreaves
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Simon Heales
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michael Orford
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire Troakes
- London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Johannes Attems
- Newcastle Brain Tissue Resource, Institute of Neuroscience and Newcastle University Institute for Ageing, Newcastle upon Tyne, UK
| | - Ellen Gelpi
- Neurological Tissue Bank, University of Barcelona, Barcelona, Spain
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Miklos Palkovits
- Human Brain Tissue Bank, Budapest, Semmelweis University, Budapest, Hungary
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, Genetic & Genomic Medicine, Institute of Child Health, UCL, London, UK
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
273
|
Bell R, Thrush RJ, Castellana-Cruz M, Oeller M, Staats R, Nene A, Flagmeier P, Xu CK, Satapathy S, Galvagnion C, Wilson MR, Dobson CM, Kumita JR, Vendruscolo M. N-Terminal Acetylation of α-Synuclein Slows down Its Aggregation Process and Alters the Morphology of the Resulting Aggregates. Biochemistry 2022; 61:1743-1756. [PMID: 35944093 PMCID: PMC9454101 DOI: 10.1021/acs.biochem.2c00104] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/09/2022] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is associated with the aberrant aggregation of α-synuclein. Although the causes of this process are still unclear, post-translational modifications of α-synuclein are likely to play a modulatory role. Since α-synuclein is constitutively N-terminally acetylated, we investigated how this post-translational modification alters the aggregation behavior of this protein. By applying a three-pronged aggregation kinetics approach, we observed that N-terminal acetylation results in a reduced rate of lipid-induced aggregation and slows down both elongation and fibril-catalyzed aggregate proliferation. An analysis of the amyloid fibrils produced by the aggregation process revealed different morphologies for the acetylated and non-acetylated forms in both lipid-induced aggregation and seed-induced aggregation assays. In addition, we found that fibrils formed by acetylated α-synuclein exhibit a lower β-sheet content. These findings indicate that N-terminal acetylation of α-synuclein alters its lipid-dependent aggregation behavior, reduces its rate of in vitro aggregation, and affects the structural properties of its fibrillar aggregates.
Collapse
Affiliation(s)
- Rosie Bell
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Rebecca J. Thrush
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Marta Castellana-Cruz
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Marc Oeller
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Roxine Staats
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Aishwarya Nene
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Patrick Flagmeier
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Catherine K. Xu
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Sandeep Satapathy
- Department
of Cell Biology, Blavantik Institute, Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Celine Galvagnion
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Mark R. Wilson
- School
of Chemistry and Molecular Bioscience, Molecular Horizons Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Christopher M. Dobson
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Janet R. Kumita
- Department
of Pharmacology, University of Cambridge, Cambridge CB2 1PD, U.K.
| | - Michele Vendruscolo
- Centre for
Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| |
Collapse
|
274
|
Iba M, McDevitt RA, Kim C, Roy R, Sarantopoulou D, Tommer E, Siegars B, Sallin M, Kwon S, Sen JM, Sen R, Masliah E. Aging exacerbates the brain inflammatory micro-environment contributing to α-synuclein pathology and functional deficits in a mouse model of DLB/PD. Mol Neurodegener 2022; 17:60. [PMID: 36064424 PMCID: PMC9447339 DOI: 10.1186/s13024-022-00564-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/19/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Although ɑ-synuclein (ɑ-syn) spreading in age-related neurodegenerative diseases such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) has been extensively investigated, the role of aging in the manifestation of disease remains unclear. METHODS We explored the role of aging and inflammation in the pathogenesis of synucleinopathies in a mouse model of DLB/PD initiated by intrastriatal injection of ɑ-syn preformed fibrils (pff). RESULTS We found that aged mice showed more extensive accumulation of ɑ-syn in selected brain regions and behavioral deficits that were associated with greater infiltration of T cells and microgliosis. Microglial inflammatory gene expression induced by ɑ-syn-pff injection in young mice had hallmarks of aged microglia, indicating that enhanced age-associated pathologies may result from inflammatory synergy between aging and the effects of ɑ-syn aggregation. Based on the transcriptomics analysis projected from Ingenuity Pathway Analysis, we found a network that included colony stimulating factor 2 (CSF2), LPS related genes, TNFɑ and poly rl:rC-RNA as common regulators. CONCLUSIONS We propose that aging related inflammation (eg: CSF2) influences outcomes of pathological spreading of ɑ-syn and suggest that targeting neuro-immune responses might be important in developing treatments for DLB/PD.
Collapse
Affiliation(s)
- Michiyo Iba
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ross A McDevitt
- Mouse Phenotyping Unit, Comparative Medicine Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Changyoun Kim
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Roshni Roy
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Dimitra Sarantopoulou
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ella Tommer
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Byron Siegars
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michelle Sallin
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Somin Kwon
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jyoti Misra Sen
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Immunology Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| | - Ranjan Sen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Immunology Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
- Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20814, USA.
| |
Collapse
|
275
|
Choi ML, Chappard A, Singh BP, Maclachlan C, Rodrigues M, Fedotova EI, Berezhnov AV, De S, Peddie CJ, Athauda D, Virdi GS, Zhang W, Evans JR, Wernick AI, Zanjani ZS, Angelova PR, Esteras N, Vinokurov AY, Morris K, Jeacock K, Tosatto L, Little D, Gissen P, Clarke DJ, Kunath T, Collinson L, Klenerman D, Abramov AY, Horrocks MH, Gandhi S. Pathological structural conversion of α-synuclein at the mitochondria induces neuronal toxicity. Nat Neurosci 2022; 25:1134-1148. [PMID: 36042314 PMCID: PMC9448679 DOI: 10.1038/s41593-022-01140-3] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/12/2022] [Indexed: 11/08/2022]
Abstract
Aggregation of alpha-synuclein (α-Syn) drives Parkinson's disease (PD), although the initial stages of self-assembly and structural conversion have not been directly observed inside neurons. In this study, we tracked the intracellular conformational states of α-Syn using a single-molecule Förster resonance energy transfer (smFRET) biosensor, and we show here that α-Syn converts from a monomeric state into two distinct oligomeric states in neurons in a concentration-dependent and sequence-specific manner. Three-dimensional FRET-correlative light and electron microscopy (FRET-CLEM) revealed that intracellular seeding events occur preferentially on membrane surfaces, especially at mitochondrial membranes. The mitochondrial lipid cardiolipin triggers rapid oligomerization of A53T α-Syn, and cardiolipin is sequestered within aggregating lipid-protein complexes. Mitochondrial aggregates impair complex I activity and increase mitochondrial reactive oxygen species (ROS) generation, which accelerates the oligomerization of A53T α-Syn and causes permeabilization of mitochondrial membranes and cell death. These processes were also observed in induced pluripotent stem cell (iPSC)-derived neurons harboring A53T mutations from patients with PD. Our study highlights a mechanism of de novo α-Syn oligomerization at mitochondrial membranes and subsequent neuronal toxicity.
Collapse
Affiliation(s)
- Minee L Choi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | - Bhanu P Singh
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh, UK
- School of Physics, University of Edinburgh, Edinburgh, UK
| | | | - Margarida Rodrigues
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Dementia Research institute at University of Cambridge, Cambridge, UK
| | - Evgeniya I Fedotova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
- Cell Physiology and Pathology Laboratory, Orel State University, Orel, Russia
| | - Alexey V Berezhnov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
- Cell Physiology and Pathology Laboratory, Orel State University, Orel, Russia
| | - Suman De
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Dementia Research institute at University of Cambridge, Cambridge, UK
| | | | - Dilan Athauda
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Gurvir S Virdi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Weijia Zhang
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - James R Evans
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Anna I Wernick
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zeinab Shadman Zanjani
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Plamena R Angelova
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Noemi Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Andrey Y Vinokurov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel, Russia
| | - Katie Morris
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh, UK
| | - Kiani Jeacock
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh, UK
| | - Laura Tosatto
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Istituto di Biofisica, National Council of Research, Trento, Italy
| | - Daniel Little
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - David J Clarke
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh, UK
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | | | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Dementia Research institute at University of Cambridge, Cambridge, UK
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
- Cell Physiology and Pathology Laboratory, Orel State University, Orel, Russia.
| | - Mathew H Horrocks
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh, UK.
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK.
- The Francis Crick Institute, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
276
|
Solomon M, Loeck M, Silva-Abreu M, Moscoso R, Bautista R, Vigo M, Muro S. Altered blood-brain barrier transport of nanotherapeutics in lysosomal storage diseases. J Control Release 2022; 349:1031-1044. [PMID: 35901858 PMCID: PMC10550198 DOI: 10.1016/j.jconrel.2022.07.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/02/2022] [Accepted: 07/19/2022] [Indexed: 12/24/2022]
Abstract
Treatment of neurological lysosomal storage disorders (LSDs) are limited because of impermeability of the blood-brain barrier (BBB) to macromolecules. Nanoformulations targeting BBB transcytosis are being explored, but the status of these routes in LSDs is unknown. We studied nanocarriers (NCs) targeted to the transferrin receptor (TfR), ganglioside GM1 or ICAM1, associated to the clathrin, caveolar or cell adhesion molecule (CAM) routes, respectively. We used brain endothelial cells and mouse models of acid sphingomyelinase-deficient Niemann Pick disease (NPD), and postmortem LSD patients' brains, all compared to respective controls. NC transcytosis across brain endothelial cells and brain distribution in mice were affected, yet through different mechanisms. Reduced TfR and clathrin expression were found, along with decreased transcytosis in cells and mouse brain distribution. Caveolin-1 expression and GM1 transcytosis were also reduced, yet increased GM1 levels seemed to compensate, providing similar NC brain distribution in NPD vs. control mice. A tendency to lower NHE-1 levels was seen, but highly increased ICAM1 expression in cells and human brains correlated with increased transcytosis and brain distribution in mice. Thus, transcytosis-related alterations in NPD and likely other LSDs may impact therapeutic access to the brain, illustrating the need for these mechanistic studies.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA.
| | - Maximilian Loeck
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marcelle Silva-Abreu
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ronaldo Moscoso
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Ronelle Bautista
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | - Marco Vigo
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA; Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain; Institute of Catalonia for Research and Advanced Studies, Barcelona, Spain.
| |
Collapse
|
277
|
Dasari AKR, Dillard L, Yi S, Viverette E, Hojjatian A, Sengupta U, Kayed R, Taylor KA, Borgnia MJ, Lim KH. Untwisted α-Synuclein Filaments Formed in the Presence of Lipid Vesicles. Biochemistry 2022; 61:1766-1773. [PMID: 36001818 DOI: 10.1021/acs.biochem.2c00283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Accumulation of filamentous aggregates of α-synuclein is a pathological hallmark of several neurodegenerative diseases, including Parkinson's disease (PD). The interaction between α-synuclein and phospholipids has been shown to play a critical role in the aggregation of α-synuclein. Most structural studies have, however, been focused on α-synuclein filaments formed in the absence of lipids. Here, we report the structural investigation of α-synuclein filaments assembled under the quiescent condition in the presence of anionic lipid vesicles using electron microscopy (EM), including cryogenic electron microscopy (cryo-EM). Our transmission electron microscopy (TEM) analyses reveal that α-synuclein forms curly protofilaments at an early stage of aggregation. The flexible protofilaments were then converted to long filaments after a longer incubation of 30 days. More detailed structural analyses using cryo-EM reveal that the long filaments adopt untwisted structures with different diameters, which have not been observed in previous α-synuclein fibrils formed in vitro. The untwisted filaments are rather similar to straight filaments with no observable twist that are extracted from patients with dementia with Lewy bodies. Our structural studies highlight the conformational diversity of α-synuclein filaments, requiring additional structural investigation of not only more ex vivo α-synuclein filaments but also in vitro α-synuclein filaments formed in the presence of diverse cofactors to better understand the molecular basis of diverse molecular conformations of α-synuclein filaments.
Collapse
Affiliation(s)
- Anvesh K R Dasari
- Department of Chemistry, East Carolina University, Greenville, North Carolina 27858, United States
| | - Lucas Dillard
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, United States
| | - Sujung Yi
- Department of Chemistry, East Carolina University, Greenville, North Carolina 27858, United States
| | - Elizabeth Viverette
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, United States
| | - Alimohammad Hojjatian
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4380, United States
| | - Urmi Sengupta
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Rakez Kayed
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Kenneth A Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4380, United States
| | - Mario Juan Borgnia
- Department of Health and Human Services, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, United States
| | - Kwang Hun Lim
- Department of Chemistry, East Carolina University, Greenville, North Carolina 27858, United States
| |
Collapse
|
278
|
Dubackic M, Liu Y, Kelley EG, Hetherington C, Haertlein M, Devos JM, Linse S, Sparr E, Olsson U. α-Synuclein Interaction with Lipid Bilayer Discs. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:10216-10224. [PMID: 35952001 PMCID: PMC9404543 DOI: 10.1021/acs.langmuir.2c01368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/18/2022] [Indexed: 06/15/2023]
Abstract
α-Synuclein (aSyn) is a 140 residue long protein present in presynaptic termini of nerve cells. The protein is associated with Parkinson's disease, in which case it has been found to self-assemble into long amyloid fibrils forming intracellular inclusions that are also rich in lipids. Furthermore, its synaptic function is proposed to involve interaction with lipid membranes, and hence, it is of interest to understand aSyn-lipid membrane interactions in detail. In this paper we report on the interaction of aSyn with model membranes in the form of lipid bilayer discs. Using a combination of cryogenic transmission electron microscopy and small-angle neutron scattering, we show that circular discs undergo a significant shape transition after the adsorption of aSyn. When aSyn self-assembles into fibrils, aSyn molecules desorb from the bilayer discs, allowing them to recover to their original shape. Interestingly, the desorption process has an all-or-none character, resulting in a binary coexistence of circular bilayer discs with no adsorbed aSyn and deformed bilayer discs having a maximum amount of adsorbed protein. The observed coexistence is consistent with the recent finding of cooperative aSyn adsorption to anionic lipid bilayers.
Collapse
Affiliation(s)
- Marija Dubackic
- Physical
Chemistry, Department of Chemistry, Lund
University, SE-22100 Lund, Sweden
| | - Yun Liu
- Center
for Neutron Research, National Institute
of Standards and Technology, Gaithersburg, Maryland 20878, United States
- Chemical
and Biomolecular Engineering Department, University of Delaware, Newark, Delaware 19716, United States
| | - Elizabeth G. Kelley
- Center
for Neutron Research, National Institute
of Standards and Technology, Gaithersburg, Maryland 20878, United States
| | - Crispin Hetherington
- National
Center for High Resolution Electron Microscopy, Centre for Analysis
and Synthesis, Chemistry Centre, Lund University, SE-22100 Lund, Sweden
| | | | | | - Sara Linse
- Biochemistry
and Structural Biology, Department of Chemistry, Lund University, SE-22100 Lund, Sweden
| | - Emma Sparr
- Physical
Chemistry, Department of Chemistry, Lund
University, SE-22100 Lund, Sweden
| | - Ulf Olsson
- Physical
Chemistry, Department of Chemistry, Lund
University, SE-22100 Lund, Sweden
| |
Collapse
|
279
|
Eteläinen TS, Kilpeläinen TP, Ignatius A, Auno S, De Lorenzo F, Uhari-Väänänen JK, Julku UH, Myöhänen TT. Removal of proteinase K resistant αSyn species does not correlate with cell survival in a virus vector-based Parkinson's disease mouse model. Neuropharmacology 2022; 218:109213. [PMID: 35964686 DOI: 10.1016/j.neuropharm.2022.109213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 10/31/2022]
Abstract
Parkinson's disease (PD) is characterized by degeneration of nigrostriatal dopaminergic neurons and accumulation of α-synuclein (αSyn) as Lewy bodies. Currently, there is no disease-modifying therapy available for PD. We have shown that a small molecular inhibitor for prolyl oligopeptidase (PREP), KYP-2047, relieves αSyn-induced toxicity in various PD models by inducing autophagy and preventing αSyn aggregation. In this study, we wanted to study the effects of PREP inhibition on different αSyn species by using cell culture and in vivo models. We used Neuro2A cells with transient αSyn overexpression and oxidative stress or proteasomal inhibition-induced αSyn aggregation to assess the effect of KYP-2047 on soluble αSyn oligomers and on cell viability. Here, the levels of soluble αSyn were measured by using ELISA, and the impact of KYP-2047 was compared to anle138b, nilotinib and deferiprone. To evaluate the effect of KYP-2047 on αSyn fibrillization in vivo, we used unilateral nigral AAV1/2-A53T-αSyn mouse model, where the KYP-2047 treatment was initiated two- or four-weeks post injection. KYP-2047 and anle138b protected cells from αSyn toxicity but interestingly, KYP-2047 did not reduce soluble αSyn oligomers. In AAV-A53T-αSyn mouse model, KYP-2047 reduced significantly proteinase K-resistant αSyn oligomers and oxidative damage related to αSyn aggregation. However, the KYP-2047 treatment that was initiated at the time of symptom onset, failed to protect the nigrostriatal dopaminergic neurons. Our results emphasize the importance of whole αSyn aggregation process in the pathology of PD and raise an important question about the forms of αSyn that are reasonable targets for PD drug therapy.
Collapse
Affiliation(s)
- Tony S Eteläinen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Tommi P Kilpeläinen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Adele Ignatius
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Samuli Auno
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Francesca De Lorenzo
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Johanna K Uhari-Väänänen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Ulrika H Julku
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland
| | - Timo T Myöhänen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Finland; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Finland.
| |
Collapse
|
280
|
Mori F, Nakamura Y, Miki Y, Tanji K, Kon T, Tomiyama M, Kakita A, Wakabayashi K. Alteration of Vesicle-Associated Membrane Protein-Binding Protein B in α-Synuclein Aggregates in Lewy Body Disease. J Neuropathol Exp Neurol 2022; 81:807-815. [PMID: 35950959 DOI: 10.1093/jnen/nlac070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
α-Synuclein (α-Syn) binds to vesicle-associated membrane protein-binding protein B (VAPB) in the endoplasmic reticulum membrane. Recent studies have shown that α-Syn-immunoreactive Lewy pathology is characterized by membrane crowding, including vesicular structures. To elucidate the role of VAPB and vesicular structures in Parkinson's disease (PD) and in dementia with Lewy bodies (DLB), the relationships among VAPB, vesicular structures, and Lewy pathology were investigated by immunohistochemistry and immunoelectron microscopy in 8 PD and 4 DLB autopsy cases. The proportions of VAPB-negative neurons in the substantia nigra in PD and in the temporal cortex in DLB were significantly higher than those in 5 controls. In PD, the incidence of α-Syn inclusions in VAPB-negative neurons was significantly higher (77.4%) than in VAPB-positive neurons (1.6%) in the substantia nigra. In DLB, the incidence of α-Syn inclusions in VAPB-negative neurons was also significantly higher (65.3%) than in VAPB-positive neurons (2.8%) in the temporal cortex. Immunoelectron microscopy revealed that α-Syn and VAPB were localized to filamentous structures of Lewy bodies (LBs). However, only a few vesicular structures labeled with anti-α-Syn were observed within LBs. These findings suggest that reduction of VAPB is involved in the disease processes of PD and DLB, although vesicular structures may not directly contribute to the formation of LBs.
Collapse
Affiliation(s)
- Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yukino Nakamura
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasuo Miki
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tomoya Kon
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Masahiko Tomiyama
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan (AK)
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
281
|
Stykel MG, Ryan SD. Nitrosative stress in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:104. [PMID: 35953517 PMCID: PMC9372037 DOI: 10.1038/s41531-022-00370-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s Disease (PD) is a neurodegenerative disorder characterized, in part, by the loss of dopaminergic neurons within the nigral-striatal pathway. Multiple lines of evidence support a role for reactive nitrogen species (RNS) in degeneration of this pathway, specifically nitric oxide (NO). This review will focus on how RNS leads to loss of dopaminergic neurons in PD and whether RNS accumulation represents a central signal in the degenerative cascade. Herein, we provide an overview of how RNS accumulates in PD by considering the various cellular sources of RNS including nNOS, iNOS, nitrate, and nitrite reduction and describe evidence that these sources are upregulating RNS in PD. We document that over 1/3 of the proteins that deposit in Lewy Bodies, are post-translationally modified (S-nitrosylated) by RNS and provide a broad description of how this elicits deleterious effects in neurons. In doing so, we identify specific proteins that are modified by RNS in neurons which are implicated in PD pathogenesis, with an emphasis on exacerbation of synucleinopathy. How nitration of alpha-synuclein (aSyn) leads to aSyn misfolding and toxicity in PD models is outlined. Furthermore, we delineate how RNS modulates known PD-related phenotypes including axo-dendritic-, mitochondrial-, and dopamine-dysfunctions. Finally, we discuss successful outcomes of therapeutics that target S-nitrosylation of proteins in Parkinson’s Disease related clinical trials. In conclusion, we argue that targeting RNS may be of therapeutic benefit for people in early clinical stages of PD.
Collapse
Affiliation(s)
- Morgan G Stykel
- The Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON N1G 2W1, ON, Canada
| | - Scott D Ryan
- The Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON N1G 2W1, ON, Canada. .,Neurodegenerative Disease Center, Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
282
|
Zhang C, Pei Y, Zhang Z, Xu L, Liu X, Jiang L, Pielak GJ, Zhou X, Liu M, Li C. C-terminal truncation modulates α-Synuclein's cytotoxicity and aggregation by promoting the interactions with membrane and chaperone. Commun Biol 2022; 5:798. [PMID: 35945337 PMCID: PMC9363494 DOI: 10.1038/s42003-022-03768-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/25/2022] [Indexed: 12/20/2022] Open
Abstract
α-Synuclein (α-syn) is the main protein component of Lewy bodies, the major pathological hallmarks of Parkinson's disease (PD). C-terminally truncated α-syn is found in the brain of PD patients, reduces cell viability and tends to form fibrils. Nevertheless, little is known about the mechanisms underlying the role of C-terminal truncation on the cytotoxicity and aggregation of α-syn. Here, we use nuclear magnetic resonance spectroscopy to show that the truncation alters α-syn conformation, resulting in an attractive interaction of the N-terminus with membranes and molecular chaperone, protein disulfide isomerase (PDI). The truncated protein is more toxic to mitochondria than full-length protein and diminishes the effect of PDI on α-syn fibrillation. Our findings reveal a modulatory role for the C-terminus in the cytotoxicity and aggregation of α-syn by interfering with the N-terminus binding to membranes and chaperone, and provide a molecular basis for the pathological role of C-terminal truncation in PD pathogenesis.
Collapse
Affiliation(s)
- Cai Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- Graduate University of Chinese Academy of Science, 100049, Beijing, China
| | - Yunshan Pei
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- Graduate University of Chinese Academy of Science, 100049, Beijing, China
| | - Zeting Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China.
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, 430071, Wuhan, China.
| | - Lingling Xu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Xiaoli Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Ling Jiang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, 430071, Wuhan, China
| | - Gary J Pielak
- Department of Chemistry, Department of Biochemistry and Biophysics, Lineberger Comprehensive Cancer Center, Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, 430071, Wuhan, China
| | - Maili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China
- Graduate University of Chinese Academy of Science, 100049, Beijing, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, 430071, Wuhan, China
| | - Conggang Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, 430071, Wuhan, China.
- Graduate University of Chinese Academy of Science, 100049, Beijing, China.
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, 430071, Wuhan, China.
| |
Collapse
|
283
|
Increased Expression of Alpha-, Beta-, and Gamma-Synucleins in Brainstem Regions of a Non-Human Primate Model of Parkinson’s Disease. Int J Mol Sci 2022; 23:ijms23158586. [PMID: 35955716 PMCID: PMC9369189 DOI: 10.3390/ijms23158586] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by cell loss in the substantia nigra and the presence of alpha-synuclein (α-syn)-containing neuronal Lewy bodies. While α-syn has received major interest in the pathogenesis of PD, the function of beta- and gamma-synucleins (β-syn and γ-syn, respectively) is not really known. Yet, these proteins are members of the same family and also concentrated in neuronal terminals. The current preclinical study investigated the expression levels of α-, β-, and γ-synucleins in brainstem regions involved in PD physiopathology. We analyzed synuclein expression in the substantia nigra, raphe nuclei, pedunculopontine nucleus, and locus coeruleus from control and parkinsonian (by MPTP) macaques. MPTP-intoxicated monkeys developed a more or less severe parkinsonian score and were sacrificed after a variable post-MPTP period ranging from 1 to 20 months. The expression of the three synucleins was increased in the substantia nigra after MPTP, and this increase correlates positively, although not very strongly, with cell loss and motor score and not with the time elapsed after intoxication. In the dorsal raphe nucleus, the expression of the three synucleins was also increased, but only α- and γ-Syn are linked to the motor score and associated cell loss. Finally, although no change in synuclein expression was demonstrated in the locus coeruleus after MPTP, we found increased expression levels of γ-Syn, which are only correlated with cell loss in the pedunculopontine nucleus. Altogether, our data suggest that these proteins may play a key role in brainstem regions and mesencephalic tegmentum. Given the involvement of these brain regions in non-motor symptoms of PD, these data also strengthen the relevance of the MPTP macaque model of PD, which exhibits pathological changes beyond nigral DA cell loss and α-synucleinopathy.
Collapse
|
284
|
Mehra S, Ahlawat S, Kumar H, Datta D, Navalkar A, Singh N, Patel K, Gadhe L, Kadu P, Kumar R, Jha NN, Sakunthala A, Sawner AS, Padinhateeri R, Udgaonkar JB, Agarwal V, Maji SK. α-Synuclein aggregation intermediates form fibril polymorphs with distinct prion-like properties. J Mol Biol 2022; 434:167761. [PMID: 35907572 DOI: 10.1016/j.jmb.2022.167761] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 11/26/2022]
Abstract
α-Synuclein (α-Syn) amyloids in synucleinopathies are suggested to be structurally and functionally diverse, reminiscent of prion-like strains. But how the aggregation of the same precursor protein results in the formation of fibril polymorphs remains elusive. Here, we demonstrate the structure-function relationship of two polymorphs, pre-matured fibrils (PMFs) and helix-matured fibrils (HMFs), based on α-Syn aggregation intermediates. These polymorphs display the structural differences as demonstrated by solid-state NMR and mass spectrometry studies and also possess different cellular activities such as seeding, internalization, and cell-to-cell transfer of aggregates. HMFs with a compact core structure exhibit low seeding potency but readily internalize and transfer from one cell to another. The less structured PMFs lack transcellular transfer ability but induce abundant α-Syn pathology and trigger the formation of aggresomes in cells. Overall, the study highlights that the conformational heterogeneity in the aggregation pathway may lead to fibril polymorphs with distinct prion-like behavior.
Collapse
Affiliation(s)
- Surabhi Mehra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Sahil Ahlawat
- Tata Institute of Fundamental Research, Sy. No. 36/P, Gopanpally, Hyderabad-500 046, India
| | - Harish Kumar
- Indian Institute of Science Education and Research, Pune- 411 008, India; National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065, India
| | - Debalina Datta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Nitu Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Laxmikant Gadhe
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Narendra N Jha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Ajay S Sawner
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Ranjith Padinhateeri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Jayant B Udgaonkar
- Indian Institute of Science Education and Research, Pune- 411 008, India; National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065, India
| | - Vipin Agarwal
- Tata Institute of Fundamental Research, Sy. No. 36/P, Gopanpally, Hyderabad-500 046, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India.
| |
Collapse
|
285
|
Paradis M, Kucharowski N, Edwards Faret G, Maya Palacios SJ, Meyer C, Stümpges B, Jamitzky I, Kalinowski J, Thiele C, Bauer R, Paululat A, Sellin J, Bülow MH. The ER protein Creld regulates ER-mitochondria contact dynamics and respiratory complex 1 activity. SCIENCE ADVANCES 2022; 8:eabo0155. [PMID: 35867795 PMCID: PMC9307246 DOI: 10.1126/sciadv.abo0155] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/07/2022] [Indexed: 05/26/2023]
Abstract
Dynamic contacts are formed between endoplasmic reticulum (ER) and mitochondria that enable the exchange of calcium and phospholipids. Disturbed contacts between ER and mitochondria impair mitochondrial dynamics and are a molecular hallmark of Parkinson's disease, which is also characterized by impaired complex I activity and dopaminergic neuron degeneration. Here, we analyzed the role of cysteine-rich with EGF-like domain (Creld), a poorly characterized risk gene for Parkinson's disease, in the regulation of mitochondrial dynamics and function. We found that loss of Creld leads to mitochondrial hyperfusion and reduced ROS signaling in Drosophila melanogaster, Xenopus tropicalis, and human cells. Creld fly mutants show differences in ER-mitochondria contacts and reduced respiratory complex I activity. The resulting low-hydrogen peroxide levels are linked to disturbed neuronal activity and lead to impaired locomotion, but not neurodegeneration, in Creld mutants. We conclude that Creld regulates ER-mitochondria communication and thereby hydrogen peroxide formation, which is required for normal neuron function.
Collapse
Affiliation(s)
- Marie Paradis
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Nicole Kucharowski
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Gabriela Edwards Faret
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | | | - Christian Meyer
- Department of Zoology and Developmental Biology, University of Osnabrück, Barbarastr. 11, 49076 Osnabrück, Germany
| | - Birgit Stümpges
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Isabell Jamitzky
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Julia Kalinowski
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Christoph Thiele
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Reinhard Bauer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Achim Paululat
- Department of Zoology and Developmental Biology, University of Osnabrück, Barbarastr. 11, 49076 Osnabrück, Germany
| | - Julia Sellin
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
- Institute for Digitalization and General Medicine, University Hospital Aachen, Pauwelsstr. 30, 52074 Aachen
| | - Margret Helene Bülow
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| |
Collapse
|
286
|
Cavalheiro EKFF, da Silva LE, Oliveira MP, Silva MG, Damiani AP, Ribeiro CB, Magenis ML, Cucker L, Michels M, Joaquim L, Machado RS, Vilela TC, Bitencourt RM, Andrade VM, Dal-Pizzol F, Petronilho F, Tuon T, Rezin GT. Effects of obesity on neuroinflammatory and neurochemical parameters in an animal model of reserpine-induced Parkinson's disease. Behav Brain Res 2022; 434:114019. [PMID: 35872330 DOI: 10.1016/j.bbr.2022.114019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/30/2022] [Accepted: 07/19/2022] [Indexed: 12/06/2022]
Abstract
Obesity is associated with low-grade chronic inflammation and oxidative stress, affecting the brain's reward system by decreasing dopaminergic neurotransmission. It is known that dopaminergic neurotransmission is also reduced in Parkinson's disease (PD), and high adiposity is considered a risk factor for the development of several neurodegenerative diseases, including PD. This study aimed to assess the effects of obesity on neuroinflammatory and neurochemical parameters in an animal model of reserpine-induced PD. The obese group showed increased inflammation and oxidative damage as well as inhibition of mitochondrial respiratory chain complexes I and II and DNA damage in the evaluated structures. The PD group did not show inflammation or mitochondrial dysfunction but exhibited oxidative damage in the hippocampus. The combination group (obesity + PD) showed reduced inflammation and oxidative stress and increased activity of complexes I and II of the mitochondrial respiratory chain in most of the analyzed structures. On the other hand, obesity + PD caused oxidative damage to proteins in the liver, prefrontal cortex, striatum, and cerebral cortex and oxidative stress in the hypothalamus, resulting in reduced catalase activity. Furthermore, the combination group showed DNA damage in blood, liver, and cerebral cortex. In conclusion, it was observed that the association of obesity and PD did not increase inflammation, oxidative stress, or mitochondrial dysfunction in most of the evaluated structures but increased oxidative damage and induced mechanisms that led to DNA damage in peripheral tissues and brain structures.
Collapse
Affiliation(s)
- Eulla Keimili Fernandes Ferreira Cavalheiro
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Mariana P Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Marina G Silva
- Laboratory of Behavioral Neuroscience, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Adriani P Damiani
- Laboratório de Biologia Celular e Molecular, Universidade do Extremo Sul Catarinense, UNESC, Avenida Universitária, 1105, Criciúma, SC, Brazil
| | - Catharina B Ribeiro
- Laboratório de Biologia Celular e Molecular, Universidade do Extremo Sul Catarinense, UNESC, Avenida Universitária, 1105, Criciúma, SC, Brazil
| | - Marina L Magenis
- Laboratório de Biologia Celular e Molecular, Universidade do Extremo Sul Catarinense, UNESC, Avenida Universitária, 1105, Criciúma, SC, Brazil
| | - Luana Cucker
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Larissa Joaquim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Richard Simon Machado
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Thais C Vilela
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Rafael M Bitencourt
- Laboratory of Behavioral Neuroscience, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Vanessa M Andrade
- Laboratório de Biologia Celular e Molecular, Universidade do Extremo Sul Catarinense, UNESC, Avenida Universitária, 1105, Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Fabrícia Petronilho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Talita Tuon
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil.
| |
Collapse
|
287
|
Galectin-3, a rising star in modulating microglia activation under conditions of neurodegeneration. Cell Death Dis 2022; 13:628. [PMID: 35859075 PMCID: PMC9300700 DOI: 10.1038/s41419-022-05058-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 01/21/2023]
Abstract
The advent of high-throughput single-cell transcriptomic analysis of microglia has revealed different phenotypes that are inherently associated with disease conditions. A common feature of some of these activated phenotypes is the upregulation of galectin-3. Representative examples of these phenotypes include disease-associated microglia (DAM) and white-associated microglia (WAM), whose role(s) in neuroprotection/neurotoxicity is a matter of high interest in the microglia community. In this review, we summarise the main findings that demonstrate the ability of galectin-3 to interact with key pattern recognition receptors, including, among others, TLR4 and TREM2 and the importance of galectin-3 in the regulation of microglia activation. Finally, we discuss increasing evidence supporting the involvement of this lectin in the main neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, traumatic brain injury, and stroke.
Collapse
|
288
|
Bayati A, Banks E, Han C, Luo W, Reintsch WE, Zorca CE, Shlaifer I, Del Cid Pellitero E, Vanderperre B, McBride HM, Fon EA, Durcan TM, McPherson PS. Rapid macropinocytic transfer of α-synuclein to lysosomes. Cell Rep 2022; 40:111102. [PMID: 35858558 DOI: 10.1016/j.celrep.2022.111102] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/10/2022] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
The nervous system spread of alpha-synuclein fibrils is thought to cause Parkinson's disease (PD) and other synucleinopathies; however, the mechanisms underlying internalization and cellular spread are enigmatic. Here, we use confocal and superresolution microscopy, subcellular fractionation, and electron microscopy (EM) of immunogold-labeled α-synuclein preformed fibrils (PFFs) to demonstrate that this form of the protein undergoes rapid internalization and is targeted directly to lysosomes in as little as 2 min. Uptake of PFFs is disrupted by macropinocytic inhibitors and circumvents classical endosomal pathways. Immunogold-labeled PFFs are seen at the highly curved inward edge of membrane ruffles, in newly formed macropinosomes, in multivesicular bodies and in lysosomes. While most fibrils remain in lysosomes, a portion is transferred to neighboring naive cells along with markers of exosomes. These data indicate that PFFs use a unique internalization mechanism as a component of cell-to-cell propagation.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Emily Banks
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Chanshuai Han
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Wen Luo
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Wolfgang E Reintsch
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Cornelia E Zorca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Irina Shlaifer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Esther Del Cid Pellitero
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Benoit Vanderperre
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Heidi M McBride
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
289
|
α-Synuclein Conformational Plasticity: Physiologic States, Pathologic Strains, and Biotechnological Applications. Biomolecules 2022; 12:biom12070994. [PMID: 35883550 PMCID: PMC9313095 DOI: 10.3390/biom12070994] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 02/08/2023] Open
Abstract
α-Synuclein (αS) is remarkable for both its extensive conformational plasticity and pathologic prion-like properties. Physiologically, αS may populate disordered monomeric, helically folded tetrameric, or membrane-bound oligomeric states. Pathologically, αS may assemble into toxic oligomers and subsequently fibrils, the prion-like transmission of which is implicated in a class of neurodegenerative disorders collectively termed α-synucleinopathies. Notably, αS does not adopt a single "amyloid fold", but rather exists as structurally distinct amyloid-like conformations referred to as "strains". The inoculation of animal models with different strains induces distinct pathologies, and emerging evidence suggests that the propagation of disease-specific strains underlies the differential pathologies observed in patients with different α-synucleinopathies. The characterization of αS strains has provided insight into the structural basis for the overlapping, yet distinct, symptoms of Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. In this review, we first explore the physiological and pathological differences between conformational states of αS. We then discuss recent studies on the influence of micro-environmental factors on αS species formation, propagation, and the resultant pathological characteristics. Lastly, we review how an understanding of αS conformational properties has been translated to emerging strain amplification technologies, which have provided further insight into the role of specific strains in distinct α-synucleinopathies, and show promise for the early diagnosis of disease.
Collapse
|
290
|
Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther 2022; 7:218. [PMID: 35798726 PMCID: PMC9261907 DOI: 10.1038/s41392-022-01080-1] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Molecular oxygen (O2) is essential for most biological reactions in mammalian cells. When the intracellular oxygen content decreases, it is called hypoxia. The process of hypoxia is linked to several biological processes, including pathogenic microbe infection, metabolic adaptation, cancer, acute and chronic diseases, and other stress responses. The mechanism underlying cells respond to oxygen changes to mediate subsequent signal response is the central question during hypoxia. Hypoxia-inducible factors (HIFs) sense hypoxia to regulate the expressions of a series of downstream genes expression, which participate in multiple processes including cell metabolism, cell growth/death, cell proliferation, glycolysis, immune response, microbe infection, tumorigenesis, and metastasis. Importantly, hypoxia signaling also interacts with other cellular pathways, such as phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-B (NF-κB) pathway, extracellular signal-regulated kinases (ERK) signaling, and endoplasmic reticulum (ER) stress. This paper systematically reviews the mechanisms of hypoxia signaling activation, the control of HIF signaling, and the function of HIF signaling in human health and diseases. In addition, the therapeutic targets involved in HIF signaling to balance health and diseases are summarized and highlighted, which would provide novel strategies for the design and development of therapeutic drugs.
Collapse
|
291
|
Peddie CJ, Genoud C, Kreshuk A, Meechan K, Micheva KD, Narayan K, Pape C, Parton RG, Schieber NL, Schwab Y, Titze B, Verkade P, Aubrey A, Collinson LM. Volume electron microscopy. NATURE REVIEWS. METHODS PRIMERS 2022; 2:51. [PMID: 37409324 PMCID: PMC7614724 DOI: 10.1038/s43586-022-00131-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 07/07/2023]
Abstract
Life exists in three dimensions, but until the turn of the century most electron microscopy methods provided only 2D image data. Recently, electron microscopy techniques capable of delving deep into the structure of cells and tissues have emerged, collectively called volume electron microscopy (vEM). Developments in vEM have been dubbed a quiet revolution as the field evolved from established transmission and scanning electron microscopy techniques, so early publications largely focused on the bioscience applications rather than the underlying technological breakthroughs. However, with an explosion in the uptake of vEM across the biosciences and fast-paced advances in volume, resolution, throughput and ease of use, it is timely to introduce the field to new audiences. In this Primer, we introduce the different vEM imaging modalities, the specialized sample processing and image analysis pipelines that accompany each modality and the types of information revealed in the data. We showcase key applications in the biosciences where vEM has helped make breakthrough discoveries and consider limitations and future directions. We aim to show new users how vEM can support discovery science in their own research fields and inspire broader uptake of the technology, finally allowing its full adoption into mainstream biological imaging.
Collapse
Affiliation(s)
- Christopher J. Peddie
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Christel Genoud
- Electron Microscopy Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Kimberly Meechan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Present address: Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Constantin Pape
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Robert G. Parton
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicole L. Schieber
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Yannick Schwab
- Cell Biology and Biophysics Unit/ Electron Microscopy Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Paul Verkade
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Aubrey Aubrey
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| |
Collapse
|
292
|
Ghane M, Ang MC, Nilashi M, Sorooshian S. Enhanced decision tree induction using evolutionary techniques for Parkinson's disease classification. Biocybern Biomed Eng 2022. [DOI: 10.1016/j.bbe.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
293
|
Lafora Disease and Alpha-Synucleinopathy in Two Adult Free-Ranging Moose (Alces alces) Presenting with Signs of Blindness and Circling. Animals (Basel) 2022; 12:ani12131633. [PMID: 35804532 PMCID: PMC9264765 DOI: 10.3390/ani12131633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Reports of behavioral signs, such as blindness and circling in free-ranging moose from different parts of the world, have spurred comprehensive pathological investigation to find the causes of the disease that have clinical relevance. In this case study, brains collected from two adult free-ranging moose (Alces alces) cows that were seemingly blind and found walking in circles were examined by light and electron microscopy with further ancillary testing. Here, we report for the first time Lafora disease and alpha-synucleinopathy in two wild free-ranging moose cows who presented with abnormal behavior and blindness, with similar neuronal polyglucosan body (PGB) accumulations identified in humans and other animals. Microscopic analysis of the hippocampus of brain revealed inclusion bodies resembling PGBs (Lafora disease) in the neurons with ultrastructural findings of aggregates of branching filaments, consistent with polyglucosan bodies. Furthermore, α-synuclein immunopositivity was noted in the hippocampus, with accumulations of small granules ultrastructurally distinct from PGBs and morphologically compatible with alpha-synucleinopathy (Lewy body). The apparent blindness found in these moose could be related to an injury associated with secondary bacterial invasion; however, an accumulation of neurotoxicants (PGBs and α-synucleins) in retinal ganglion cells could also be the cause. Lafora disease and alpha-synucleinopathy were considered in the differential diagnosis of the young adult moose who presented with signs of blindness and behavioral signs such as circling. Abstract Lafora disease is an autosomal recessive glycogen-storage disorder resulting from an accumulation of toxic polyglucosan bodies (PGBs) in the central nervous system, which causes behavioral and neurologic symptoms in humans and other animals. In this case study, brains collected from two young adult free-ranging moose (Alces alces) cows that were seemingly blind and found walking in circles were examined by light and electron microscopy. Microscopic analysis of the hippocampus of the brain revealed inclusion bodies resembling PGBs in the neuronal perikaryon, neuronal processes, and neuropil. These round inclusions measuring up to 30 microns in diameter were predominantly confined to the hippocampus region of the brain in both animals. The inclusions tested α-synuclein-negative by immunohistochemistry, α-synuclein-positive with PAS, GMS, and Bielschowsky’s staining; and diastase-resistant with central basophilic cores and faintly radiating peripheral lines. Ultrastructural examination of the affected areas of the hippocampus showed non-membrane-bound aggregates of asymmetrically branching filaments that bifurcated regularly, consistent with PGBs in both animals. Additionally, α-synuclein immunopositivity was noted in the different regions of the hippocampus with accumulations of small granules ultrastructurally distinct from PGBs and morphologically compatible with alpha-synucleinopathy (Lewy body). The apparent blindness found in these moose could be related to an injury associated with secondary bacterial invasion; however, an accumulation of neurotoxicants (PGBs and α-synuclein) in retinal ganglions cells could also be the cause. This is the first report demonstrating Lafora disease with concurrent alpha-synucleinopathy (Lewy body neuropathy) in a non-domesticated animal.
Collapse
|
294
|
Parkinson's disease-risk protein TMEM175 is a proton-activated proton channel in lysosomes. Cell 2022; 185:2292-2308.e20. [PMID: 35750034 DOI: 10.1016/j.cell.2022.05.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/07/2021] [Accepted: 05/24/2022] [Indexed: 02/06/2023]
Abstract
Lysosomes require an acidic lumen between pH 4.5 and 5.0 for effective digestion of macromolecules. This pH optimum is maintained by proton influx produced by the V-ATPase and efflux through an unidentified "H+ leak" pathway. Here we show that TMEM175, a genetic risk factor for Parkinson's disease (PD), mediates the lysosomal H+ leak by acting as a proton-activated, proton-selective channel on the lysosomal membrane (LyPAP). Acidification beyond the normal range potently activated LyPAP to terminate further acidification of lysosomes. An endogenous polyunsaturated fatty acid and synthetic agonists also activated TMEM175 to trigger lysosomal proton release. TMEM175 deficiency caused lysosomal over-acidification, impaired proteolytic activity, and facilitated α-synuclein aggregation in vivo. Mutational and pH normalization analyses indicated that the channel's H+ conductance is essential for normal lysosome function. Thus, modulation of LyPAP by cellular cues may dynamically tune the pH optima of endosomes and lysosomes to regulate lysosomal degradation and PD pathology.
Collapse
|
295
|
Sinnige T. Molecular mechanisms of amyloid formation in living systems. Chem Sci 2022; 13:7080-7097. [PMID: 35799826 PMCID: PMC9214716 DOI: 10.1039/d2sc01278b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/14/2022] [Indexed: 12/28/2022] Open
Abstract
Fibrillar protein aggregation is a hallmark of a variety of human diseases. Examples include the deposition of amyloid-β and tau in Alzheimer's disease, and that of α-synuclein in Parkinson's disease. The molecular mechanisms by which soluble proteins form amyloid fibrils have been extensively studied in the test tube. These investigations have revealed the microscopic steps underlying amyloid formation, and the role of factors such as chaperones that modulate these processes. This perspective explores the question to what extent the mechanisms of amyloid formation elucidated in vitro apply to human disease. The answer is not yet clear, and may differ depending on the protein and the associated disease. Nevertheless, there are striking qualitative similarities between the aggregation behaviour of proteins in vitro and the development of the related diseases. Limited quantitative data obtained in model organisms such as Caenorhabditis elegans support the notion that aggregation mechanisms in vivo can be interpreted using the same biophysical principles established in vitro. These results may however be biased by the high overexpression levels typically used in animal models of protein aggregation diseases. Molecular chaperones have been found to suppress protein aggregation in animal models, but their mechanisms of action have not yet been quantitatively analysed. Several mechanisms are proposed by which the decline of protein quality control with organismal age, but also the intrinsic nature of the aggregation process may contribute to the kinetics of protein aggregation observed in human disease.
Collapse
Affiliation(s)
- Tessa Sinnige
- Bijvoet Centre for Biomolecular Research, Utrecht University Padualaan 8 3584 CH Utrecht The Netherlands
| |
Collapse
|
296
|
Vrijsen S, Vrancx C, Del Vecchio M, Swinnen JV, Agostinis P, Winderickx J, Vangheluwe P, Annaert W. Inter-organellar Communication in Parkinson's and Alzheimer's Disease: Looking Beyond Endoplasmic Reticulum-Mitochondria Contact Sites. Front Neurosci 2022; 16:900338. [PMID: 35801175 PMCID: PMC9253489 DOI: 10.3389/fnins.2022.900338] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/05/2022] [Indexed: 01/13/2023] Open
Abstract
Neurodegenerative diseases (NDs) are generally considered proteinopathies but whereas this may initiate disease in familial cases, onset in sporadic diseases may originate from a gradually disrupted organellar homeostasis. Herein, endolysosomal abnormalities, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and altered lipid metabolism are commonly observed in early preclinical stages of major NDs, including Parkinson's disease (PD) and Alzheimer's disease (AD). Among the multitude of underlying defective molecular mechanisms that have been suggested in the past decades, dysregulation of inter-organellar communication through the so-called membrane contact sites (MCSs) is becoming increasingly apparent. Although MCSs exist between almost every other type of subcellular organelle, to date, most focus has been put on defective communication between the ER and mitochondria in NDs, given these compartments are critical in neuronal survival. Contributions of other MCSs, notably those with endolysosomes and lipid droplets are emerging, supported as well by genetic studies, identifying genes functionally involved in lysosomal homeostasis. In this review, we summarize the molecular identity of the organelle interactome in yeast and mammalian cells, and critically evaluate the evidence supporting the contribution of disturbed MCSs to the general disrupted inter-organellar homeostasis in NDs, taking PD and AD as major examples.
Collapse
Affiliation(s)
- Stephanie Vrijsen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Mara Del Vecchio
- Laboratory of Functional Biology, Department of Biology, KU Leuven, Heverlee, Belgium
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, VIB-Center for Cancer Research, KU Leuven, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joris Winderickx
- Laboratory of Functional Biology, Department of Biology, KU Leuven, Heverlee, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
297
|
Henderson MX, Henrich MT, Geibl FF, Oertel WH, Brundin P, Surmeier DJ. The roles of connectivity and neuronal phenotype in determining the pattern of α-synuclein pathology in Parkinson's disease. Neurobiol Dis 2022; 168:105687. [PMID: 35283326 PMCID: PMC9610381 DOI: 10.1016/j.nbd.2022.105687] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, and motor dysfunction has been attributed to loss of dopaminergic neurons. However, motor dysfunction is only one of many symptoms experienced by patients. A neuropathological hallmark of PD is intraneuronal protein aggregates called Lewy pathology (LP). Neuropathological staging studies have shown that dopaminergic neurons are only one of the many cell types prone to manifest LP. Progressive appearance of LP in multiple brain regions, as well as peripheral nerves, has led to the popular hypothesis that LP and misfolded forms of one of its major components - α-synuclein (aSYN) - can spread through synaptically connected circuits. However, not all brain regions or neurons within connected circuits develop LP, suggesting that cell autonomous factors modulate the development of pathology. Here, we review studies about how LP develops and progressively engages additional brain regions. We focus on how connectivity constrains progression and discuss cell autonomous factors that drive pathology development. We propose a mixed model of cell autonomous factors and trans-synaptic spread as mediators of pathology progression and put forward this model as a framework for future experiments exploring PD pathophysiology.
Collapse
Affiliation(s)
- Michael X Henderson
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America.
| | - Martin T Henrich
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany; Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg 35043, Germany; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| | - Fanni F Geibl
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany; Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg 35043, Germany; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| | - Wolfgang H Oertel
- Department of Neurology, Philipps-University Marburg, Marburg 35043, Germany
| | - Patrik Brundin
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America
| |
Collapse
|
298
|
Beton JG, Monistrol J, Wentink A, Johnston EC, Roberts AJ, Bukau BG, Hoogenboom BW, Saibil HR. Cooperative amyloid fibre binding and disassembly by the Hsp70 disaggregase. EMBO J 2022; 41:e110410. [PMID: 35698800 PMCID: PMC9379549 DOI: 10.15252/embj.2021110410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 01/05/2023] Open
Abstract
Although amyloid fibres are highly stable protein aggregates, a specific combination of human Hsp70 system chaperones can disassemble them, including fibres formed of α-synuclein, huntingtin, or Tau. Disaggregation requires the ATPase activity of the constitutively expressed Hsp70 family member, Hsc70, together with the J domain protein DNAJB1 and the nucleotide exchange factor Apg2. Clustering of Hsc70 on the fibrils appears to be necessary for disassembly. Here we use atomic force microscopy to show that segments of in vitro assembled α-synuclein fibrils are first coated with chaperones and then undergo bursts of rapid, unidirectional disassembly. Cryo-electron tomography and total internal reflection fluorescence microscopy reveal fibrils with regions of densely bound chaperones, preferentially at one end of the fibre. Sub-stoichiometric amounts of Apg2 relative to Hsc70 dramatically increase recruitment of Hsc70 to the fibres, creating localised active zones that then undergo rapid disassembly at a rate of ~ 4 subunits per second. The observed unidirectional bursts of Hsc70 loading and unravelling may be explained by differences between the two ends of the polar fibre structure.
Collapse
Affiliation(s)
- Joseph George Beton
- Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck University of London, London, UK
| | - Jim Monistrol
- Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck University of London, London, UK
| | - Anne Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Erin C Johnston
- Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck University of London, London, UK
| | - Anthony John Roberts
- Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck University of London, London, UK
| | - Bernd Gerhard Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Bart W Hoogenboom
- London Centre for Nanotechnology, University College London, London, UK.,Department of Physics & Astronomy, University College London, London, UK
| | - Helen R Saibil
- Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck University of London, London, UK
| |
Collapse
|
299
|
Fanning S, Cirka H, Thies JL, Jeong J, Niemi SM, Yoon J, Ho GPH, Pacheco JA, Dettmer U, Liu L, Clish CB, Hodgetts KJ, Hutchinson JN, Muratore CR, Caldwell GA, Caldwell KA, Selkoe D. Lipase regulation of cellular fatty acid homeostasis as a Parkinson's disease therapeutic strategy. NPJ Parkinsons Dis 2022; 8:74. [PMID: 35680956 PMCID: PMC9184586 DOI: 10.1038/s41531-022-00335-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
Synucleinopathy (Parkinson's disease (PD); Lewy body dementia) disease-modifying treatments represent a huge unmet medical need. Although the PD-causing protein α-synuclein (αS) interacts with lipids and fatty acids (FA) physiologically and pathologically, targeting FA homeostasis for therapeutics is in its infancy. We identified the PD-relevant target stearoyl-coA desaturase: inhibiting monounsaturated FA synthesis reversed PD phenotypes. However, lipid degradation also generates FA pools. Here, we identify the rate-limiting lipase enzyme, LIPE, as a candidate target. Decreasing LIPE in human neural cells reduced αS inclusions. Patient αS triplication vs. corrected neurons had increased pSer129 and insoluble αS and decreased αS tetramer:monomer ratios. LIPE inhibition rescued all these and the abnormal unfolded protein response. LIPE inhibitors decreased pSer129 and restored tetramer:monomer equilibrium in αS E46K-expressing human neurons. LIPE reduction in vivo alleviated αS-induced dopaminergic neurodegeneration in Caenorhabditis elegans. Co-regulating FA synthesis and degradation proved additive in rescuing PD phenotypes, signifying co-targeting as a therapeutic strategy.
Collapse
Affiliation(s)
- Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Haley Cirka
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer L Thies
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Jooyoung Jeong
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah M Niemi
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Joon Yoon
- Department of Biostatistics, The Harvard Chan School of Public Health, Boston, MA, 02115, USA
| | - Gary P H Ho
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Kevin J Hodgetts
- Laboratory for Drug Discovery in Neuroscience, Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - John N Hutchinson
- Department of Biostatistics, The Harvard Chan School of Public Health, Boston, MA, 02115, USA
| | - Christina R Muratore
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Dennis Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
300
|
Lazarevic V, Yang Y, Paslawski W, Svenningsson P. α-Synuclein induced cholesterol lowering increases tonic and reduces depolarization-evoked synaptic vesicle recycling and glutamate release. NPJ Parkinsons Dis 2022; 8:71. [PMID: 35672421 PMCID: PMC9174203 DOI: 10.1038/s41531-022-00334-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
α-Synuclein (α-syn) is a key molecule linked to Parkinson's disease pathology. Physiologically, the monomeric α-syn in the presynaptic termini is involved in regulation of neurotransmission, but the pathophysiology of extracellular monomeric α-syn is still unknown. Utilizing both in vivo and in vitro approaches, we investigated how extracellular α-syn impact presynaptic structure and function. Our data revealed that treatment with exogenous α-syn leads to increased tonic and decreased depolarization-evoked synaptic vesicle (SV) recycling and glutamate release. This was associated with mobilization of molecularly distinct SV pools and reorganization of active zone components. Our study also showed that exogenous α-syn impaired neuronal cholesterol level and that the cholesterol binding domain of α-syn was sufficient to exert the same presynaptic phenotype as the full-length protein. The present study sheds new light on physiological functions of extracellular α-syn in overall maintenance of presynaptic activity that involves the reorganization of both presynaptic compartment and cholesterol-rich plasma membrane domains.
Collapse
Affiliation(s)
- Vesna Lazarevic
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Yunting Yang
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|