1
|
Ahanger IA, Dar TA. Small molecule modulators of alpha-synuclein aggregation and toxicity: Pioneering an emerging arsenal against Parkinson's disease. Ageing Res Rev 2024; 101:102538. [PMID: 39389237 DOI: 10.1016/j.arr.2024.102538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Parkinson's disease (PD) is primarily characterized by loss of dopaminergic neurons in the substantia nigra pars compacta region of the brain and accumulation of aggregated forms of alpha-synuclein (α-Syn), an intrinsically disordered protein, in the form of Lewy Bodies and Lewy Neurites. Substantial evidences point to the aggregated/fibrillar forms of α-Syn as a central event in PD pathogenesis, underscoring the modulation of α-Syn aggregation as a promising strategy for PD treatment. Consequently, numerous anti-aggregation agents, spanning from small molecules to polymers, have been scrutinized for their potential to mitigate α-Syn aggregation and its associated toxicity. Among these, small molecule modulators like osmoprotectants, polyphenols, cellular metabolites, metals, and peptides have emerged as promising candidates with significant potential in PD management. This article offers a comprehensive overview of the effects of these small molecule modulators on the aggregation propensity and associated toxicity of α-Syn and its PD-associated mutants. It serves as a valuable resource for identifying and developing potent, non-invasive, non-toxic, and highly specific small molecule-based therapeutic arsenal for combating PD. Additionally, it raises pertinent questions aimed at guiding future research endeavours in the field of α-Syn aggregation remodelling.
Collapse
Affiliation(s)
- Ishfaq Ahmad Ahanger
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| |
Collapse
|
2
|
Yamaguchi H, Nishimura Y, Matsuse D, Sekiya H, Masaki K, Tanaka T, Saiga T, Harada M, Kira YI, Dickson DW, Fujishima K, Matsuo E, Tanaka KF, Yamasaki R, Isobe N, Kira JI. A rapidly progressive multiple system atrophy-cerebellar variant model presenting marked glial reactions with inflammation and spreading of α-synuclein oligomers and phosphorylated α-synuclein aggregates. Brain Behav Immun 2024; 121:122-141. [PMID: 38986725 DOI: 10.1016/j.bbi.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 04/30/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple system atrophy (MSA) is a severe α-synucleinopathy facilitated by glial reactions; the cerebellar variant (MSA-C) preferentially involves olivopontocerebellar fibres with conspicuous demyelination. A lack of aggressive models that preferentially involve olivopontocerebellar tracts in adulthood has hindered our understanding of the mechanisms of demyelination and neuroaxonal loss, and thus the development of effective treatments for MSA. We therefore aimed to develop a rapidly progressive mouse model that recaptures MSA-C pathology. We crossed Plp1-tTA and tetO-SNCA*A53T mice to generate Plp1-tTA::tetO-SNCA*A53T bi-transgenic mice, in which human A53T α-synuclein-a mutant protein with enhanced aggregability-was specifically produced in the oligodendrocytes of adult mice using Tet-Off regulation. These bi-transgenic mice expressed mutant α-synuclein from 8 weeks of age, when doxycycline was removed from the diet. All bi-transgenic mice presented rapidly progressive motor deterioration, with wide-based ataxic gait around 22 weeks of age and death around 30 weeks of age. They also had prominent demyelination in the brainstem/cerebellum. Double immunostaining demonstrated that myelin basic protein was markedly decreased in areas in which SM132, an axonal marker, was relatively preserved. Demyelinating lesions exhibited marked ionised calcium-binding adaptor molecule 1-, arginase-1-, and toll-like receptor 2-positive microglial reactivity and glial fibrillary acidic protein-positive astrocytic reactivity. Microarray analysis revealed a strong inflammatory response and cytokine/chemokine production in bi-transgenic mice. Neuronal nuclei-positive neuronal loss and patchy microtubule-associated protein 2-positive dendritic loss became prominent at 30 weeks of age. However, a perceived decrease in tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta in bi-transgenic mice compared with wild-type mice was not significant, even at 30 weeks of age. Wild-type, Plp1-tTA, and tetO-SNCA*A53T mice developed neither motor deficits nor demyelination. In bi-transgenic mice, double immunostaining revealed human α-synuclein accumulation in neurite outgrowth inhibitor A (Nogo-A)-positive oligodendrocytes beginning at 9 weeks of age; its expression was further increased at 10 to 12 weeks, and these increased levels were maintained at 12, 24, and 30 weeks. In an α-synuclein-proximity ligation assay, α-synuclein oligomers first appeared in brainstem oligodendrocytes as early as 9 weeks of age; they then spread to astrocytes, neuropil, and neurons at 12 and 16 weeks of age. α-Synuclein oligomers in the brainstem neuropil were most abundant at 16 weeks of age and decreased thereafter; however, those in Purkinje cells successively increased until 30 weeks of age. Double immunostaining revealed the presence of phosphorylated α-synuclein in Nogo-A-positive oligodendrocytes in the brainstem/cerebellum as early as 9 weeks of age. In quantitative assessments, phosphorylated α-synuclein gradually and successively accumulated at 12, 24, and 30 weeks in bi-transgenic mice. By contrast, no phosphorylated α-synuclein was detected in wild-type, tetO-SNCA*A53T, or Plp1-tTA mice at any age examined. Pronounced demyelination and tubulin polymerisation, promoting protein-positive oligodendrocytic loss, was closely associated with phosphorylated α-synuclein aggregates at 24 and 30 weeks of age. Early inhibition of mutant α-synuclein expression by doxycycline diet at 23 weeks led to fully recovered demyelination; inhibition at 27 weeks led to persistent demyelination with glial reactions, despite resolving phosphorylated α-synuclein aggregates. In conclusion, our bi-transgenic mice exhibited progressively increasing demyelination and neuroaxonal loss in the brainstem/cerebellum, with rapidly progressive motor deterioration in adulthood. These mice showed marked microglial and astrocytic reactions with inflammation that was closely associated with phosphorylated α-synuclein aggregates. These features closely mimic human MSA-C pathology. Notably, our model is the first to suggest that α-synuclein oligomers may spread from oligodendrocytes to neurons in transgenic mice with human α-synuclein expression in oligodendrocytes. This model of MSA is therefore particularly useful for elucidating the in vivo mechanisms of α-synuclein spreading from glia to neurons, and for developing therapies that target glial reactions and/or α-synuclein oligomer spreading and aggregate formation in MSA.
Collapse
Affiliation(s)
- Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka, Japan.
| | - Yuji Nishimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Tatsunori Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Sumitomo Pharma Co., Ltd., Osaka, Japan.
| | - Toru Saiga
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaya Harada
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yuu-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | - Kei Fujishima
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Eriko Matsuo
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Translational Neuroscience Research Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka, Japan; Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, Fukuoka, Japan.
| |
Collapse
|
3
|
Xian M, Li J, Liu T, Hou K, Sun L, Wei J. β-Synuclein Intermediates α-Synuclein Neurotoxicity in Parkinson's Disease. ACS Chem Neurosci 2024; 15:2445-2453. [PMID: 38905183 DOI: 10.1021/acschemneuro.4c00263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024] Open
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disease in the world, and synuclein is closely related to the onset and progression of PD. Synuclein is considered a therapeutic target for PD. Recent studies have found that abnormal aggregation of α-synuclein (α-Syn) in the brains of PD patients leads to mitochondrial dysfunction and neuroinflammation. Research in the field of neuroscience has confirmed that β-synuclein (β-Syn) also plays a role in Parkinson's disease. However, there has been little research on the role mechanisms and interactions between β-Syn and α-Syn in PD. Therefore, the purpose of this study is to clarify the relationship between α-Syn, β-Syn, and PD and to explore the roles and interactions of β-Syn and α-Syn in PD.
Collapse
Affiliation(s)
- Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Jingwen Li
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Kaiying Hou
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, P.R. China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, P.R. China
| |
Collapse
|
4
|
Coukos R, Krainc D. Key genes and convergent pathogenic mechanisms in Parkinson disease. Nat Rev Neurosci 2024; 25:393-413. [PMID: 38600347 DOI: 10.1038/s41583-024-00812-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder marked by the preferential dysfunction and death of dopaminergic neurons in the substantia nigra. The onset and progression of PD is influenced by a diversity of genetic variants, many of which lack functional characterization. To identify the most high-yield targets for therapeutic intervention, it is important to consider the core cellular compartments and functional pathways upon which the varied forms of pathogenic dysfunction may converge. Here, we review several key PD-linked proteins and pathways, focusing on the mechanisms of their potential convergence in disease pathogenesis. These dysfunctions primarily localize to a subset of subcellular compartments, including mitochondria, lysosomes and synapses. We discuss how these pathogenic mechanisms that originate in different cellular compartments may coordinately lead to cellular dysfunction and neurodegeneration in PD.
Collapse
Affiliation(s)
- Robert Coukos
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
5
|
Galka D, Ali TT, Bast A, Niederleithinger M, Gerhardt E, Motosugi R, Sakata E, Knop M, Outeiro TF, Popova B, Braus GH. Inhibition of 26S proteasome activity by α-synuclein is mediated by the proteasomal chaperone Rpn14/PAAF1. Aging Cell 2024; 23:e14128. [PMID: 38415292 PMCID: PMC11113265 DOI: 10.1111/acel.14128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/01/2024] [Accepted: 02/11/2024] [Indexed: 02/29/2024] Open
Abstract
Parkinson's disease (PD) is characterized by aggregation of α-synuclein (α-syn) into protein inclusions in degenerating brains. Increasing amounts of aggregated α-syn species indicate significant perturbation of cellular proteostasis. Altered proteostasis depends on α-syn protein levels and the impact of α-syn on other components of the proteostasis network. Budding yeast Saccharomyces cerevisiae was used as eukaryotic reference organism to study the consequences of α-syn expression on protein dynamics. To address this, we investigated the impact of overexpression of α-syn and S129A variant on the abundance and stability of most yeast proteins using a genome-wide yeast library and a tandem fluorescent protein timer (tFT) reporter as a measure for protein stability. This revealed that the stability of in total 377 cellular proteins was altered by α-syn expression, and that the impact on protein stability was significantly enhanced by phosphorylation at Ser129 (pS129). The proteasome assembly chaperone Rpn14 was identified as one of the top candidates for increased protein stability by expression of pS129 α-syn. Elevated levels of Rpn14 enhanced the growth inhibition by α-syn and the accumulation of ubiquitin conjugates in the cell. We found that Rpn14 interacts physically with α-syn and stabilizes pS129 α-syn. The expression of α-syn along with elevated levels of Rpn14 or its human counterpart PAAF1 reduced the proteasome activity in yeast and in human cells, supporting that pS129 α-syn negatively affects the 26S proteasome through Rpn14. This comprehensive study into the alternations of protein homeostasis highlights the critical role of the Rpn14/PAAF1 in α-syn-mediated proteasome dysfunction.
Collapse
Affiliation(s)
- Dajana Galka
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and GeneticsUniversity of GöttingenGöttingenGermany
| | - Tariq T. Ali
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and GeneticsUniversity of GöttingenGöttingenGermany
| | - Alexander Bast
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and GeneticsUniversity of GöttingenGöttingenGermany
| | - Marie Niederleithinger
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and GeneticsUniversity of GöttingenGöttingenGermany
| | - Ellen Gerhardt
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
| | - Ryo Motosugi
- Institute for Auditory NeuroscienceUniversity Medical Center GöttingenGöttingenGermany
- Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC)University of GöttingenGöttingenGermany
| | - Eri Sakata
- Institute for Auditory NeuroscienceUniversity Medical Center GöttingenGöttingenGermany
- Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC)University of GöttingenGöttingenGermany
| | - Michael Knop
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ‐ZMBH AllianceHeidelberg UniversityHeidelbergGermany
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)GöttingenGermany
| | - Blagovesta Popova
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and GeneticsUniversity of GöttingenGöttingenGermany
| | - Gerhard H. Braus
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and GeneticsUniversity of GöttingenGöttingenGermany
| |
Collapse
|
6
|
Hsieh Y, Augur ZM, Arbery M, Ashour N, Barrett K, Pearse RV, Tio ES, Duong DM, Felsky D, De Jager PL, Bennett DA, Seyfried NT, Young‐Pearse TL. Person-specific differences in ubiquitin-proteasome mediated proteostasis in human neurons. Alzheimers Dement 2024; 20:2952-2967. [PMID: 38470006 PMCID: PMC11032531 DOI: 10.1002/alz.13680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND Impairment of the ubiquitin-proteasome system (UPS) has been implicated in abnormal protein accumulation in Alzheimer's disease. It remains unclear if genetic variation affects the intrinsic properties of neurons that render some individuals more vulnerable to UPS impairment. METHODS Induced pluripotent stem cell (iPSC)-derived neurons were generated from over 50 genetically variant and highly characterized participants of cohorts of aging. Proteomic profiling, proteasome activity assays, and Western blotting were employed to examine neurons at baseline and in response to UPS perturbation. RESULTS Neurons with lower basal UPS activity were more vulnerable to tau accumulation following mild UPS inhibition. Chronic reduction in proteasome activity in human neurons induced compensatory elevation of regulatory proteins involved in proteostasis and several proteasome subunits. DISCUSSION These findings reveal that genetic variation influences basal UPS activity in human neurons and differentially sensitizes them to external factors perturbing the UPS, leading to the accumulation of aggregation-prone proteins such as tau. HIGHLIGHTS Polygenic risk score for AD is associated with the ubiquitin-proteasome system (UPS) in neurons. Basal proteasome activity correlates with aggregation-prone protein levels in neurons. Genetic variation affects the response to proteasome inhibition in neurons. Neuronal proteasome perturbation induces an elevation in specific proteins involved in proteostasis. Low basal proteasome activity leads to enhanced tau accumulation with UPS challenge.
Collapse
Affiliation(s)
- Yi‐Chen Hsieh
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Zachary M. Augur
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Mason Arbery
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Nancy Ashour
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Katharine Barrett
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Richard V. Pearse
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Earvin S. Tio
- Department of Psychiatry and Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
| | - Duc M. Duong
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Daniel Felsky
- Department of Psychiatry and Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Krembil Centre for NeuroinformaticsCentre for Addiction and Mental HealthTorontoOntarioCanada
| | - Philip L. De Jager
- Center for Translational and Computational NeuroimmunologyDepartment of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Nicholas T. Seyfried
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Tracy L. Young‐Pearse
- Ann Romney Centerfor Neurologic DiseasesDepartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Harvard Stem Cell InstituteHarvard UniversityCambridgeMassachusettsUSA
| |
Collapse
|
7
|
Kandel R, Jung J, Neal S. Proteotoxic stress and the ubiquitin proteasome system. Semin Cell Dev Biol 2024; 156:107-120. [PMID: 37734998 PMCID: PMC10807858 DOI: 10.1016/j.semcdb.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/01/2023] [Accepted: 08/20/2023] [Indexed: 09/23/2023]
Abstract
The ubiquitin proteasome system maintains protein homeostasis by regulating the breakdown of misfolded proteins, thereby preventing misfolded protein aggregates. The efficient elimination is vital for preventing damage to the cell by misfolded proteins, known as proteotoxic stress. Proteotoxic stress can lead to the collapse of protein homeostasis and can alter the function of the ubiquitin proteasome system. Conversely, impairment of the ubiquitin proteasome system can also cause proteotoxic stress and disrupt protein homeostasis. This review examines two impacts of proteotoxic stress, 1) disruptions to ubiquitin homeostasis (ubiquitin stress) and 2) disruptions to proteasome homeostasis (proteasome stress). Here, we provide a mechanistic description of the relationship between proteotoxic stress and the ubiquitin proteasome system. This relationship is illustrated by findings from several protein misfolding diseases, mainly neurodegenerative diseases, as well as from basic biology discoveries from yeast to mammals. In addition, we explore the importance of the ubiquitin proteasome system in endoplasmic reticulum quality control, and how proteotoxic stress at this organelle is alleviated. Finally, we highlight how cells utilize the ubiquitin proteasome system to adapt to proteotoxic stress and how the ubiquitin proteasome system can be genetically and pharmacologically manipulated to maintain protein homeostasis.
Collapse
Affiliation(s)
- Rachel Kandel
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, United States
| | - Jasmine Jung
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, United States
| | - Sonya Neal
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, United States; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
8
|
Lee M, Barnes J, Vermilyea S, Meints J, Martinez H. Soluble and insoluble lysates from the human A53T mutant α-synuclein transgenic mouse model induces α-synucleinopathy independent of injection site. RESEARCH SQUARE 2024:rs.3.rs-3982325. [PMID: 38496623 PMCID: PMC10942550 DOI: 10.21203/rs.3.rs-3982325/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Pathological aggregation of a-synuclein (aS) is implicated in the pathogenesis of Parkinson's disease (PD) and other a-synucleinopathies. The current view is that neuron-to-neuron spreading of aS pathology contributes to the progression of a-synucleinopathy. We used an A53T mutant human aS transgenic mouse model (TgA53T) to examine whether the site of pathogenic aS inoculation affects the pattern of neuropathology and whether soluble and insoluble fractions derived from crude pathogenic tissue lysates exhibit differential capacities to initiate aS pathology. To test whether the inoculation site impacts the ultimate spatial/temporal patterns of aS pathology, aS preformed fibrils (PFF), or brain homogenates from TgA53T mice with a-synucleinopathy, were injected into the cortex/striatum, brain stem, or skeletal muscle. In all cases, inoculation of pathogenic aS induced end-stage motor dysfunction within ~100 days post-inoculation (dpi). Significantly, irrespective of the inoculation sites, ultimate distribution of the aS pathology was like that seen in normally aged TgA53T mice at end-stage, indicating that the intrinsic neuronal vulnerability is a significant determinant in the induction of aS pathology, even when initiated by inoculation of pathogenic aS. Temporal analysis of brain stem injected TgA53T mice show that initial aS pathology was seen by 30 days post-inoculation and inflammatory changes occur at later stages. To determine if the aS species with differential solubility are differentially pathogenic, brain lysates from end-stage TgA53Tmice were fractionated into highly soluble (S150) and insoluble (P150) fractions, as well as the endoplasmic reticulum (ER)-enriched fraction (P100). Significantly, all fractions were able to seed de novo aS pathology in vivo, when injected unilaterally into TgA53Tmice with the ER fractions being most pathogenic. Our results suggest that multiple aS species from brain can initiate the development of progressive aS pathology.
Collapse
|
9
|
Liu HN, Wang T, Hu JJ, Chen L, Shi X, Li YM, Luo SZ. The disordered protein SERF promotes α-Synuclein aggregation through liquid-liquid phase separation. J Biol Chem 2024; 300:105667. [PMID: 38272228 PMCID: PMC10877630 DOI: 10.1016/j.jbc.2024.105667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
The aggregation of α-Synuclein (α-Syn) into amyloid fibrils is the hallmark of Parkinson's disease. Under stress or other pathological conditions, the accumulation of α-Syn oligomers is the main contributor to the cytotoxicity. A potential approach for treating Parkinson's disease involves preventing the accumulation of these α-Syn oligomers. In this study, we present a novel mechanism involving a conserved group of disorderly proteins known as small EDRK-rich factor (SERF), which promotes the aggregation of α-Syn through a cophase separation process. Using diverse methods like confocal microscopy, fluorescence recovery after photobleaching assays, solution-state NMR spectroscopy, and Western blot, we determined that the N-terminal domain of SERF1a plays a role in the interactions that occur during cophase separation. Within these droplets, α-Syn undergoes a gradual transformation from solid condensates to amyloid fibrils, while SERF1a is excluded from the condensates and dissolves into the solution. Notably, in vivo experiments show that SERF1a cophase separation with α-Syn significantly reduces the deposition of α-Syn oligomers and decreases its cellular toxicity under stress. These findings suggest that SERF1a accelerates the conversion of α-Syn from highly toxic oligomers to less toxic fibrils through cophase separation, thereby mitigating the biological damage of α-Syn aggregation.
Collapse
Affiliation(s)
- He-Ning Liu
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ting Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jin-Jian Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Long Chen
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xiangyan Shi
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, China.
| | - Yan-Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China.
| | - Shi-Zhong Luo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
10
|
Brash-Arias D, García LI, Pérez-Estudillo CA, Rojas-Durán F, Aranda-Abreu GE, Herrera-Covarrubias D, Chi-Castañeda D. The Role of Astrocytes and Alpha-Synuclein in Parkinson's Disease: A Review. NEUROSCI 2024; 5:71-86. [PMID: 39483813 PMCID: PMC11523690 DOI: 10.3390/neurosci5010005] [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: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 11/03/2024] Open
Abstract
The search for new therapies to reduce symptoms and find a cure for Parkinson's disease has focused attention on two key points: the accumulation of alpha-synuclein aggregates and astrocytes. The former is a hallmark of the disease, while the latter corresponds to a type of glial cell with an important role in both the prevention and development of this neurodegenerative disorder. Traditionally, research has focused on therapies targeting dopaminergic neurons. Currently, as more is known about the genetic and molecular factors and the neuroglial interaction in the disease, great emphasis has been placed on the neuroprotective role of astrocytes in the early stages of the disease and on the astrocytic capture of alpha-synuclein under both physiological and pathological conditions. This review aims to analyze the contribution of alpha-synuclein and astrocytes to the development and progression of Parkinson's disease, as well as to evaluate recent therapeutic proposals specifically focused on synucleopathies and astroglial cells as potential therapies for the disease.
Collapse
Affiliation(s)
- David Brash-Arias
- Doctorado en Investigaciones Cerebrales, Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico;
| | - Luis I. García
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico
| | | | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico
| | | | | | - Donaji Chi-Castañeda
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91190, Mexico
| |
Collapse
|
11
|
Carreras Mascaro A, Grochowska MM, Boumeester V, Dits NFJ, Bilgiҫ EN, Breedveld GJ, Vergouw L, de Jong FJ, van Royen ME, Bonifati V, Mandemakers W. LRP10 and α-synuclein transmission in Lewy body diseases. Cell Mol Life Sci 2024; 81:75. [PMID: 38315424 PMCID: PMC10844361 DOI: 10.1007/s00018-024-05135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/13/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024]
Abstract
Autosomal dominant variants in LRP10 have been identified in patients with Lewy body diseases (LBDs), including Parkinson's disease (PD), Parkinson's disease-dementia (PDD), and dementia with Lewy bodies (DLB). Nevertheless, there is little mechanistic insight into the role of LRP10 in disease pathogenesis. In the brains of control individuals, LRP10 is typically expressed in non-neuronal cells like astrocytes and neurovasculature, but in idiopathic and genetic cases of PD, PDD, and DLB, it is also present in α-synuclein-positive neuronal Lewy bodies. These observations raise the questions of what leads to the accumulation of LRP10 in Lewy bodies and whether a possible interaction between LRP10 and α-synuclein plays a role in disease pathogenesis. Here, we demonstrate that wild-type LRP10 is secreted via extracellular vesicles (EVs) and can be internalised via clathrin-dependent endocytosis. Additionally, we show that LRP10 secretion is highly sensitive to autophagy inhibition, which induces the formation of atypical LRP10 vesicular structures in neurons in human-induced pluripotent stem cells (iPSC)-derived brain organoids. Furthermore, we show that LRP10 overexpression leads to a strong induction of monomeric α-synuclein secretion, together with time-dependent, stress-sensitive changes in intracellular α-synuclein levels. Interestingly, patient-derived astrocytes carrying the c.1424 + 5G > A LRP10 variant secrete aberrant high-molecular-weight species of LRP10 in EV-free media fractions. Finally, we show that this truncated patient-derived LRP10 protein species (LRP10splice) binds to wild-type LRP10, reduces LRP10 wild-type levels, and antagonises the effect of LRP10 on α-synuclein levels and distribution. Together, this work provides initial evidence for a possible functional role of LRP10 in LBDs by modulating intra- and extracellular α-synuclein levels, and pathogenic mechanisms linked to the disease-associated c.1424 + 5G > A LRP10 variant, pointing towards potentially important disease mechanisms in LBDs.
Collapse
Affiliation(s)
- Ana Carreras Mascaro
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martyna M Grochowska
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Valerie Boumeester
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Natasja F J Dits
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ece Naz Bilgiҫ
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guido J Breedveld
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Leonie Vergouw
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frank Jan de Jong
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim Mandemakers
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
12
|
Kowalski A, Betzer C, Larsen ST, Gregersen E, Newcombe EA, Bermejo MC, Bendtsen VW, Diemer J, Ernstsen CV, Jain S, Bou AE, Langkilde AE, Nejsum LN, Klipp E, Edwards R, Kragelund BB, Jensen PH, Nissen P. Monomeric α-synuclein activates the plasma membrane calcium pump. EMBO J 2023; 42:e111122. [PMID: 37916890 PMCID: PMC10690453 DOI: 10.15252/embj.2022111122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/19/2023] [Accepted: 10/11/2023] [Indexed: 11/03/2023] Open
Abstract
Alpha-synuclein (aSN) is a membrane-associated and intrinsically disordered protein, well known for pathological aggregation in neurodegeneration. However, the physiological function of aSN is disputed. Pull-down experiments have pointed to plasma membrane Ca2+ -ATPase (PMCA) as a potential interaction partner. From proximity ligation assays, we find that aSN and PMCA colocalize at neuronal synapses, and we show that calcium expulsion is activated by aSN and PMCA. We further show that soluble, monomeric aSN activates PMCA at par with calmodulin, but independent of the autoinhibitory domain of PMCA, and highly dependent on acidic phospholipids and membrane-anchoring properties of aSN. On PMCA, the key site is mapped to the acidic lipid-binding site, located within a disordered PMCA-specific loop connecting the cytosolic A domain and transmembrane segment 3. Our studies point toward a novel physiological role of monomeric aSN as a stimulator of calcium clearance in neurons through activation of PMCA.
Collapse
Affiliation(s)
- Antoni Kowalski
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
- REPIN and Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Department of Molecular NeurochemistryMedical University of LodzLodzPoland
- Present address:
ImmunAware ApSHørsholmDenmark
| | - Cristine Betzer
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Present address:
Region Midtjylland, Regionshospitalet GødstrupHerningDenmark
| | - Sigrid Thirup Larsen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
| | - Emil Gregersen
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Present address:
Department of Clinical MedicineAarhus UniversityAarhus NDenmark
| | - Estella A Newcombe
- REPIN and Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Montaña Caballero Bermejo
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
- Department Biochemistry and Molecular Biology and Genetics, IBMPUniversity of ExtremaduraBadajozSpain
| | - Viktor Wisniewski Bendtsen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
| | - Jorin Diemer
- Theoretical BiophysicsHumboldt‐Universität zu BerlinBerlinGermany
| | | | - Shweta Jain
- Departments of Neurology and PhysiologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Alicia Espiña Bou
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
| | | | - Lene N Nejsum
- Department of Clinical MedicineAarhus UniversityAarhus NDenmark
| | - Edda Klipp
- Theoretical BiophysicsHumboldt‐Universität zu BerlinBerlinGermany
| | - Robert Edwards
- Departments of Neurology and PhysiologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Birthe B Kragelund
- REPIN and Structural Biology and NMR Laboratory, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Poul Henning Jensen
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Poul Nissen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Danish Research Institute of Translational Neuroscience – DANDRITEAarhus UniversityAarhusDenmark
| |
Collapse
|
13
|
Tsalenchuk M, Gentleman SM, Marzi SJ. Linking environmental risk factors with epigenetic mechanisms in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:123. [PMID: 37626097 PMCID: PMC10457362 DOI: 10.1038/s41531-023-00568-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Sporadic Parkinson's disease (PD) is a progressive neurodegenerative disease, with a complex risk structure thought to be influenced by interactions between genetic variants and environmental exposures, although the full aetiology is unknown. Environmental factors, including pesticides, have been reported to increase the risk of developing the disease. Growing evidence suggests epigenetic changes are key mechanisms by which these environmental factors act upon gene regulation, in disease-relevant cell types. We present a systematic review critically appraising and summarising the current body of evidence of the relationship between epigenetic mechanisms and environmental risk factors in PD to inform future research in this area. Epigenetic studies of relevant environmental risk factors in animal and cell models have yielded promising results, however, research in humans is just emerging. While published studies in humans are currently relatively limited, the importance of the field for the elucidation of molecular mechanisms of pathogenesis opens clear and promising avenues for the future of PD research. Carefully designed epidemiological studies carried out in PD patients hold great potential to uncover disease-relevant gene regulatory mechanisms. Therefore, to advance this burgeoning field, we recommend broadening the scope of investigations to include more environmental exposures, increasing sample sizes, focusing on disease-relevant cell types, and recruiting more diverse cohorts.
Collapse
Affiliation(s)
- Maria Tsalenchuk
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Sarah J Marzi
- UK Dementia Research Institute, Imperial College London, London, UK.
- Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
14
|
Lee J, Sung KW, Bae EJ, Yoon D, Kim D, Lee JS, Park DH, Park DY, Mun SR, Kwon SC, Kim HY, Min JO, Lee SJ, Suh YH, Kwon YT. Targeted degradation of ⍺-synuclein aggregates in Parkinson's disease using the AUTOTAC technology. Mol Neurodegener 2023; 18:41. [PMID: 37355598 PMCID: PMC10290391 DOI: 10.1186/s13024-023-00630-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/31/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND There are currently no disease-modifying therapeutics for Parkinson's disease (PD). Although extensive efforts were undertaken to develop therapeutic approaches to delay the symptoms of PD, untreated α-synuclein (α-syn) aggregates cause cellular toxicity and stimulate further disease progression. PROTAC (Proteolysis-Targeting Chimera) has drawn attention as a therapeutic modality to target α-syn. However, no PROTACs have yet shown to selectively degrade α-syn aggregates mainly owing to the limited capacity of the proteasome to degrade aggregates, necessitating the development of novel approaches to fundamentally eliminate α-syn aggregates. METHODS We employed AUTOTAC (Autophagy-Targeting Chimera), a macroautophagy-based targeted protein degradation (TPD) platform developed in our earlier studies. A series of AUTOTAC chemicals was synthesized as chimeras that bind both α-syn aggregates and p62/SQSTM1/Sequestosome-1, an autophagic receptor. The efficacy of Autotacs was evaluated to target α-syn aggregates to phagophores and subsequently lysosomes for hydrolysis via p62-dependent macroautophagy. The target engagement was monitored by oligomerization and localization of p62 and autophagic markers. The therapeutic efficacy to rescue PD symptoms was characterized in cultured cells and mice. The PK/PD (pharmacokinetics/pharmacodynamics) profiles were investigated to develop an oral drug for PD. RESULTS ATC161 induced selective degradation of α-syn aggregates at DC50 of ~ 100 nM. No apparent degradation was observed with monomeric α-syn. ATC161 mediated the targeting of α-syn aggregates to p62 by binding the ZZ domain and accelerating p62 self-polymerization. These p62-cargo complexes were delivered to autophagic membranes for lysosomal degradation. In PD cellular models, ATC161 exhibited therapeutic efficacy to reduce cell-to-cell transmission of α-syn and to rescue cells from the damages in DNA and mitochondria. In PD mice established by injecting α-syn preformed fibrils (PFFs) into brain striata via stereotaxic surgery, oral administration of ATC161 at 10 mg/kg induced the degradation of α-syn aggregates and reduced their propagation. ATC161 also mitigated the associated glial inflammatory response and improved muscle strength and locomotive activity. CONCLUSION AUTOTAC provides a platform to develop drugs for PD. ATC161, an oral drug with excellent PK/PD profiles, induces selective degradation of α-syn aggregates in vitro and in vivo. We suggest that ATC161 is a disease-modifying drug that degrades the pathogenic cause of PD.
Collapse
Affiliation(s)
- Jihoon Lee
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- AUTOTAC Bio Inc., Changkyunggung-Ro 254, Jongno-Gu, Seoul, 03077, Republic of Korea
| | - Ki Woon Sung
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- AUTOTAC Bio Inc., Changkyunggung-Ro 254, Jongno-Gu, Seoul, 03077, Republic of Korea
| | - Eun-Jin Bae
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Dabin Yoon
- AUTOTAC Bio Inc., Changkyunggung-Ro 254, Jongno-Gu, Seoul, 03077, Republic of Korea
- Department of Physical Education, Sejong University, Seoul, 05006, Republic of Korea
| | - Dasarang Kim
- AUTOTAC Bio Inc., Changkyunggung-Ro 254, Jongno-Gu, Seoul, 03077, Republic of Korea
| | - Jin Saem Lee
- AUTOTAC Bio Inc., Changkyunggung-Ro 254, Jongno-Gu, Seoul, 03077, Republic of Korea
| | - Da-Ha Park
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Daniel Youngjae Park
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Su Ran Mun
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Soon Chul Kwon
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hye Yeon Kim
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Joo-Ok Min
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Neuramedy Co. Ltd, Seoul, 04796, Republic of Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| | - Yong Tae Kwon
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- AUTOTAC Bio Inc., Changkyunggung-Ro 254, Jongno-Gu, Seoul, 03077, Republic of Korea.
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
15
|
Carceles-Cordon M, Weintraub D, Chen-Plotkin AS. Cognitive heterogeneity in Parkinson's disease: A mechanistic view. Neuron 2023; 111:1531-1546. [PMID: 37028431 PMCID: PMC10198897 DOI: 10.1016/j.neuron.2023.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/22/2022] [Accepted: 03/13/2023] [Indexed: 04/09/2023]
Abstract
Cognitive impairment occurs in most individuals with Parkinson's disease (PD), exacting a high toll on patients, their caregivers, and the healthcare system. In this review, we begin by summarizing the current clinical landscape surrounding cognition in PD. We then discuss how cognitive impairment and dementia may develop in PD based on the spread of the pathological protein alpha-synuclein (aSyn) from neurons in brainstem regions to those in the cortical regions of the brain responsible for higher cognitive functions, as first proposed in the Braak hypothesis. We appraise the Braak hypothesis from molecular (conformations of aSyn), cell biological (cell-to-cell spread of pathological aSyn), and organ-level (region-to-region spread of aSyn pathology at the whole brain level) viewpoints. Finally, we argue that individual host factors may be the most poorly understood aspect of this pathological process, accounting for substantial heterogeneity in the pattern and pace of cognitive decline in PD.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dan Weintraub
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Rosado-Ramos R, Poças GM, Marques D, Foito A, M Sevillano D, Lopes-da-Silva M, Gonçalves LG, Menezes R, Ottens M, Stewart D, Ibáñez de Opakua A, Zweckstetter M, Seabra MC, Mendes CS, Outeiro TF, Domingos PM, Santos CN. Genipin prevents alpha-synuclein aggregation and toxicity by affecting endocytosis, metabolism and lipid storage. Nat Commun 2023; 14:1918. [PMID: 37024503 PMCID: PMC10079842 DOI: 10.1038/s41467-023-37561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Parkinson's Disease (PD) is a common neurodegenerative disorder affecting millions of people worldwide for which there are only symptomatic therapies. Small molecules able to target key pathological processes in PD have emerged as interesting options for modifying disease progression. We have previously shown that a (poly)phenol-enriched fraction (PEF) of Corema album L. leaf extract modulates central events in PD pathogenesis, namely α-synuclein (αSyn) toxicity, aggregation and clearance. PEF was now subjected to a bio-guided fractionation with the aim of identifying the critical bioactive compound. We identified genipin, an iridoid, which relieves αSyn toxicity and aggregation. Furthermore, genipin promotes metabolic alterations and modulates lipid storage and endocytosis. Importantly, genipin was able to prevent the motor deficits caused by the overexpression of αSyn in a Drosophila melanogaster model of PD. These findings widens the possibility for the exploitation of genipin for PD therapeutics.
Collapse
Affiliation(s)
- Rita Rosado-Ramos
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Gonçalo M Poças
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Daniela Marques
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Alexandre Foito
- Environmental and Biochemical Sciences, The James Hutton Institute, DD2 5DA, Dundee, Scotland
| | - David M Sevillano
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Mafalda Lopes-da-Silva
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Luís G Gonçalves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Regina Menezes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal
- CBIOS - Universidade Lusófona's Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024, Lisboa, Portugal
| | - Marcel Ottens
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Derek Stewart
- Environmental and Biochemical Sciences, The James Hutton Institute, DD2 5DA, Dundee, Scotland
| | | | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), 37075, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Department of NMR-based Structural Biology, Am Fassberg 11, 37077, Göttingen, Germany
| | - Miguel C Seabra
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - César S Mendes
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Tiago Fleming Outeiro
- German Center for Neurodegenerative Diseases (DZNE), 37075, Göttingen, Germany
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, NE2 4HH, UK
- Scientific employee with an honorary contract at German Center for Neurodegenerative Diseases (DZNE), 37075, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Pedro M Domingos
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Cláudia N Santos
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal.
- iNOVA4Health, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
17
|
Kohler V, Andréasson C. Reversible protein assemblies in the proteostasis network in health and disease. Front Mol Biosci 2023; 10:1155521. [PMID: 37021114 PMCID: PMC10067754 DOI: 10.3389/fmolb.2023.1155521] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
While proteins populating their native conformations constitute the functional entities of cells, protein aggregates are traditionally associated with cellular dysfunction, stress and disease. During recent years, it has become clear that large aggregate-like protein condensates formed via liquid-liquid phase separation age into more solid aggregate-like particles that harbor misfolded proteins and are decorated by protein quality control factors. The constituent proteins of the condensates/aggregates are disentangled by protein disaggregation systems mainly based on Hsp70 and AAA ATPase Hsp100 chaperones prior to their handover to refolding and degradation systems. Here, we discuss the functional roles that condensate formation/aggregation and disaggregation play in protein quality control to maintain proteostasis and why it matters for understanding health and disease.
Collapse
Affiliation(s)
- Verena Kohler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Claes Andréasson
- Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| |
Collapse
|
18
|
Jagtap YA, Kumar P, Kinger S, Dubey AR, Choudhary A, Gutti RK, Singh S, Jha HC, Poluri KM, Mishra A. Disturb mitochondrial associated proteostasis: Neurodegeneration and imperfect ageing. Front Cell Dev Biol 2023; 11:1146564. [PMID: 36968195 PMCID: PMC10036443 DOI: 10.3389/fcell.2023.1146564] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
The disturbance in mitochondrial functions and homeostasis are the major features of neuron degenerative conditions, like Parkinson’s disease, Amyotrophic Lateral Sclerosis, and Alzheimer’s disease, along with protein misfolding. The aberrantly folded proteins are known to link with impaired mitochondrial pathways, further contributing to disease pathogenesis. Despite their central significance, the implications of mitochondrial homeostasis disruption on other organelles and cellular processes remain insufficiently explored. Here, we have reviewed the dysfunction in mitochondrial physiology, under neuron degenerating conditions. The disease misfolded proteins impact quality control mechanisms of mitochondria, such as fission, fusion, mitophagy, and proteasomal clearance, to the detriment of neuron. The adversely affected mitochondrial functional roles, like oxidative phosphorylation, calcium homeostasis, and biomolecule synthesis as well as its axes and contacts with endoplasmic reticulum and lysosomes are also discussed. Mitochondria sense and respond to multiple cytotoxic stress to make cell adapt and survive, though chronic dysfunction leads to cell death. Mitochondria and their proteins can be candidates for biomarkers and therapeutic targets. Investigation of internetworking between mitochondria and neurodegeneration proteins can enhance our holistic understanding of such conditions and help in designing more targeted therapies.
Collapse
Affiliation(s)
- Yuvraj Anandrao Jagtap
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Prashant Kumar
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sumit Kinger
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Ankur Rakesh Dubey
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Akash Choudhary
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Ravi Kumar Gutti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Sarika Singh
- Division of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Simrol, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
- *Correspondence: Amit Mishra,
| |
Collapse
|
19
|
Dabiri S, Ramírez Ruiz MI, Jean-Louis G, Ntekim OE, Obisesan TO, Campbell AL, Mwendwa DT. The Mediating Role of Inflammation in the Relationship Between α-Synuclein and Cognitive Functioning. J Gerontol A Biol Sci Med Sci 2023; 78:206-212. [PMID: 36269624 PMCID: PMC10215981 DOI: 10.1093/gerona/glac217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidence suggests that α-synuclein plays a role in the pathophysiology of Alzheimer's disease (AD). This study examined whether α-synuclein level in cerebrospinal fluid (CSF) was associated with cognitive functioning among older adults. We also explored whether this relationship was mediated by proinflammatory cytokines TNF-α and IL-6, along with sIL-6R and vascular endothelial growth factor (VEGF). Using a cross-sectional Alzheimer's Disease Neuroimaging Initiative (ADNI; N = 148) sample, we examined the relationship between α-synuclein and participants' performance on Mini-Mental State Examination (MMSE) and Alzheimer's Disease Assessment Scale Cognitive Subscale (ADAS-Cog 13) at baseline. Mediation analyses were utilized, adjusting for age, education, APOEe4, and Geriatric Depression Scale scores. All biological markers were measured in CSF. Participants in the current sample were 58.3% males, 41.7% females, and Caucasian (95.5%); their average education and age were 15.5 (standard deviation [SD] = 2.97) and 74.4 (SD = 7.51) years, respectively. Higher accumulation of α-synuclein was associated with poorer MMSE scores (β = -0.41, standard error [SE] = 1.54, p < .001). This relationship appeared to be mediated by VEGF (β = 0.27, SE = 2.15, p = .025) and IL-6r (β = 0.22, SE = 1.66, p < .026). In addition, α-synuclein was associated with poorer performance on the ADAS-Cog 13 (β = 0.34, p = .005) and mediated by VEGF (β = -0.19, SE = 4.13, p = .025) after adjusting for age, education, APOEe4, and depressive symptoms. α-Synuclein may serve as an additional biomarker for determining poor cognitive functioning. VEGF and IL-6 soluble receptors were significant mediators of the relationship between α-synuclein and cognitive functioning. If confirmed in prospective analyses, these findings can further inform the pathologic cascade and early diagnosis of AD.
Collapse
Affiliation(s)
- Sanaz Dabiri
- Department of Psychology, Howard University, Washington, District of Columbia, Washington, DC, USA
| | - Mara I Ramírez Ruiz
- Department of Psychology, Howard University, Washington, District of Columbia, Washington, DC, USA
| | - Girardin Jean-Louis
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Oyonumo E Ntekim
- Department of Graduate Nutritional Sciences, Howard University, Washington, District of Columbia, Washington, DC, USA
| | - Thomas O Obisesan
- Division of Geriatrics, Department of Medicine, Howard University Hospital, Washington, District of Columbia, Washington, DC, USA
| | - Alfonso L Campbell
- Department of Psychology, Howard University, Washington, District of Columbia, Washington, DC, USA
| | - Denée T Mwendwa
- Department of Psychology, Howard University, Washington, District of Columbia, Washington, DC, USA
| | | |
Collapse
|
20
|
Khan AN, Khan RH. Protein misfolding and related human diseases: A comprehensive review of toxicity, proteins involved, and current therapeutic strategies. Int J Biol Macromol 2022; 223:143-160. [PMID: 36356861 DOI: 10.1016/j.ijbiomac.2022.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Most of the cell's chemical reactions and structural components are facilitated by proteins. But proteins are highly dynamic molecules, where numerous modifications or changes in the cellular environment can affect their native conformational fold leading to protein aggregation. Various stress conditions, such as oxidative stress, mutations and metal toxicity may cause protein misfolding and aggregation by shifting the conformational equilibrium towards more aggregation-prone states. Most of the protein misfolding diseases (PMDs) involve aggregation of protein. We have discussed such proteins like Aβ peptide, α-synuclein, amylin and lysozyme involved in Alzheimer's, Parkinson's, type II diabetes and non-neuropathic systemic amyloidosis respectively. Till date, all advances in PMDs therapeutics help symptomatically but do not prevent the root cause of the disease, i.e., the aggregation of protein involved in the diseases. Current efforts focused on developing therapies for PMDs have employed diverse strategies; repositioning pre-existing drugs as it saves time and money; natural compounds that are touted as potential drug candidates have an advantage of being taken in diet normally and will induce lesser side effects. This review also covers recently developed therapeutic strategies like antisense drugs and disaggregases which has yielded therapeutic agents that have transitioned from preclinical studies into human clinical trials.
Collapse
Affiliation(s)
- Asra Nasir Khan
- Interdisciplinary Biotechnology Unit, AMU, Aligarh 202002, India
| | | |
Collapse
|
21
|
Meszka I, Polanowska J, Xirodimas DP. Mixed in chains: NEDD8 polymers in the Protein Quality Control system. Semin Cell Dev Biol 2022; 132:27-37. [PMID: 35078718 DOI: 10.1016/j.semcdb.2022.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022]
Abstract
Post-translational modification of proteins with the Ubiquitin-like molecule NEDD8 is a critical regulatory mechanism for several biological processes and a potential target for therapeutic intervention. The role of NEDD8 has been mainly characterised through its modification as single moiety on the cullin family of proteins and control of Cullin-Ring-Ligases, but also on non-cullin substrates. In addition to monoNEDDylation, recent studies have now revealed that NEDD8 can also generate diverse polymers. This is either through modification of the 9 available lysines in NEDD8 and the formation of polyNEDD8 chains, or NEDDylation of Ubiquitin and SUMO-2 for the generation of hybrid NEDD8 chains. Here, we review recent findings that characterise the formation of NEDD8 polymers under distinct modes of protein NEDDylation (canonical/atypical) and their potential role as regulatory signals of the proteotoxic stress response and the Protein Quality Control system.
Collapse
Affiliation(s)
- Igor Meszka
- CRBM, Univ. Montpellier, CNRS, Montpellier, France
| | | | | |
Collapse
|
22
|
Padilla-Godínez FJ, Ruiz-Ortega LI, Guerra-Crespo M. Nanomedicine in the Face of Parkinson's Disease: From Drug Delivery Systems to Nanozymes. Cells 2022; 11:3445. [PMID: 36359841 PMCID: PMC9657131 DOI: 10.3390/cells11213445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 01/02/2024] Open
Abstract
The complexity and overall burden of Parkinson's disease (PD) require new pharmacological approaches to counteract the symptomatology while reducing the progressive neurodegeneration of affected dopaminergic neurons. Since the pathophysiological signature of PD is characterized by the loss of physiological levels of dopamine (DA) and the misfolding and aggregation of the alpha-synuclein (α-syn) protein, new proposals seek to restore the lost DA and inhibit the progressive damage derived from pathological α-syn and its impact in terms of oxidative stress. In this line, nanomedicine (the medical application of nanotechnology) has achieved significant advances in the development of nanocarriers capable of transporting and delivering basal state DA in a controlled manner in the tissues of interest, as well as highly selective catalytic nanostructures with enzyme-like properties for the elimination of reactive oxygen species (responsible for oxidative stress) and the proteolysis of misfolded proteins. Although some of these proposals remain in their early stages, the deepening of our knowledge concerning the pathological processes of PD and the advances in nanomedicine could endow for the development of potential treatments for this still incurable condition. Therefore, in this paper, we offer: (i) a brief summary of the most recent findings concerning the physiology of motor regulation and (ii) the molecular neuropathological processes associated with PD, together with (iii) a recapitulation of the current progress in controlled DA release by nanocarriers and (iv) the design of nanozymes, catalytic nanostructures with oxidoreductase-, chaperon, and protease-like properties. Finally, we conclude by describing the prospects and knowledge gaps to overcome and consider as research into nanotherapies for PD continues, especially when clinical translations take place.
Collapse
Affiliation(s)
- Francisco J. Padilla-Godínez
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
- Regenerative Medicine Laboratory, Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
| | - Leonardo I. Ruiz-Ortega
- Institute for Physical Sciences, National Autonomous University of Mexico, Cuernavaca 62210, Mexico
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Magdalena Guerra-Crespo
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
- Regenerative Medicine Laboratory, Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Coyoacan, Mexico City 04510, Mexico
| |
Collapse
|
23
|
Mandel N, Agarwal N. Role of SUMOylation in Neurodegenerative Diseases. Cells 2022; 11:3395. [PMID: 36359791 PMCID: PMC9654019 DOI: 10.3390/cells11213395] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 09/26/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are irreversible, progressive diseases with no effective treatment. The hallmark of NDDs is the aggregation of misfolded, modified proteins, which impair neuronal vulnerability and cause brain damage. The loss of synaptic connection and the progressive loss of neurons result in cognitive defects. Several dysregulated proteins and overlapping molecular mechanisms contribute to the pathophysiology of NDDs. Post-translational modifications (PTMs) are essential regulators of protein function, trafficking, and maintaining neuronal hemostasis. The conjugation of a small ubiquitin-like modifier (SUMO) is a reversible, dynamic PTM required for synaptic and cognitive function. The onset and progression of neurodegenerative diseases are associated with aberrant SUMOylation. In this review, we have summarized the role of SUMOylation in regulating critical proteins involved in the onset and progression of several NDDs.
Collapse
Affiliation(s)
| | - Nitin Agarwal
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Quantitative super-resolution imaging of pathological aggregates reveals distinct toxicity profiles in different synucleinopathies. Proc Natl Acad Sci U S A 2022; 119:e2205591119. [PMID: 36206368 PMCID: PMC9573094 DOI: 10.1073/pnas.2205591119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Protein aggregation is a hallmark of major neurodegenerative disorders. Increasing data suggest that smaller aggregates cause higher toxic response than filamentous aggregates (fibrils). However, the size of small aggregates has challenged their detection within biologically relevant environments. Here, we report approaches to quantitatively super-resolve aggregates in live cells and ex vivo brain tissues. We show that Amytracker 630 (AT630), a commercial aggregate-activated fluorophore, has outstanding photophysical properties that enable super-resolution imaging of α-synuclein, tau, and amyloid-β aggregates, achieving ∼4 nm precision. Applying AT630 to AppNL-G-F mouse brain tissues or aggregates extracted from a Parkinson's disease donor, we demonstrate excellent agreement with antibodies specific for amyloid-β or α-synuclein, respectively, confirming the specificity of AT630. Subsequently, we use AT630 to reveal a linear relationship between α-synuclein aggregate size and cellular toxicity and discovered that aggregates smaller than 450 ± 60 nm (aggregate450nm) readily penetrated the plasma membrane. We determine aggregate450nm concentrations in six Parkinson's disease and dementia with Lewy bodies donor samples and show that aggregates in different synucleinopathies demonstrate distinct potency in toxicity. We further show that cell-penetrating aggregates are surrounded by proteasomes, which assemble into foci to gradually process aggregates. Our results suggest that the plasma membrane effectively filters out fibrils but is vulnerable to penetration by aggregates of 450 ± 60 nm. Together, our findings present an exciting strategy to determine specificity of aggregate toxicity within heterogeneous samples. Our approach to quantitatively measure these toxic aggregates in biological environments opens possibilities to molecular examinations of disease mechanisms under physiological conditions.
Collapse
|
25
|
Kim S, Choi JG, Kim SW, Park SC, Kang YR, Park DS, Son M, Lee CH. Inhibition of α-synuclein aggregation by MT101-5 is neuroprotective in mouse models of Parkinson's disease. Biomed Pharmacother 2022; 154:113637. [PMID: 36058149 DOI: 10.1016/j.biopha.2022.113637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, after Alzheimer's disease, and becomes increasingly prevalent with age. α-Synuclein (α-syn) forms the major filamentous component of Lewy bodies, which are pathological hallmarks of α-synucleinopathies such as PD. We evaluated the neuroprotective effects of MT101-5, a standardized herbal formula that consists of an ethanolic extract of Genkwae Flos, Clematidis Radix, and Gastrodiae Rhizoma, against α-synuclein-induced cytotoxicity in vivo. MT101-5 protected against behavioral deficits and loss of dopaminergic neurons in human α-syn-overexpressing transgenic mice after treatment with 30 mg/kg/day for 5 months. We investigated transcriptomic changes within MT101-5 mechanisms of action (MOA) suppressing α-syn aggregation in an α-synuclein preformed fibril (α-syn PFF) mouse model of sporadic PD. We found that inhibition of α-syn fibril formation was associated with changes in transcripts in mitochondrial biogenesis, electron transport, chaperones, and proteasomes following treatment with MT101-5. These results suggest that the mixed herbal formula MT101-5 may be used as a pharmaceutical agent for preventing or improving PD.
Collapse
Affiliation(s)
- Sinyeon Kim
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea
| | - Jin Gyu Choi
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea
| | - Se Woong Kim
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea
| | - Sang Cheol Park
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea
| | - Yu-Ra Kang
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea
| | - Dong Seok Park
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea
| | - Miwon Son
- MtheraPharma Co., Ltd., 38, Magokjungang 8-ro 1-gil, Gangseo-gu, Seoul, the Republic of Korea.
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, the Republic of Korea.
| |
Collapse
|
26
|
Thellung S, Corsaro A, Dellacasagrande I, Nizzari M, Zambito M, Florio T. Proteostasis unbalance in prion diseases: Mechanisms of neurodegeneration and therapeutic targets. Front Neurosci 2022; 16:966019. [PMID: 36148145 PMCID: PMC9485628 DOI: 10.3389/fnins.2022.966019] [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: 06/10/2022] [Accepted: 08/05/2022] [Indexed: 01/18/2023] Open
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, are progressive neurodegenerative disorders of the central nervous system that affect humans and animals as sporadic, inherited, and infectious forms. Similarly to Alzheimer's disease and other neurodegenerative disorders, any attempt to reduce TSEs' lethality or increase the life expectancy of affected individuals has been unsuccessful. Typically, the onset of symptoms anticipates the fatal outcome of less than 1 year, although it is believed to be the consequence of a decades-long process of neuronal death. The duration of the symptoms-free period represents by itself a major obstacle to carry out effective neuroprotective therapies. Prions, the infectious entities of TSEs, are composed of a protease-resistant protein named prion protein scrapie (PrPSc) from the prototypical TSE form that afflicts ovines. PrPSc misfolding from its physiological counterpart, cellular prion protein (PrPC), is the unifying pathogenic trait of all TSEs. PrPSc is resistant to intracellular turnover and undergoes amyloid-like fibrillation passing through the formation of soluble dimers and oligomers, which are likely the effective neurotoxic entities. The failure of PrPSc removal is a key pathogenic event that defines TSEs as proteopathies, likewise other neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, characterized by alteration of proteostasis. Under physiological conditions, protein quality control, led by the ubiquitin-proteasome system, and macroautophagy clears cytoplasm from improperly folded, redundant, or aggregation-prone proteins. There is evidence that both of these crucial homeostatic pathways are impaired during the development of TSEs, although it is still unclear whether proteostasis alteration facilitates prion protein misfolding or, rather, PrPSc protease resistance hampers cytoplasmic protein quality control. This review is aimed to critically analyze the most recent advancements in the cause-effect correlation between PrPC misfolding and proteostasis alterations and to discuss the possibility that pharmacological restoring of ubiquitin-proteasomal competence and stimulation of autophagy could reduce the intracellular burden of PrPSc and ameliorate the severity of prion-associated neurodegeneration.
Collapse
Affiliation(s)
- Stefano Thellung
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Alessandro Corsaro
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Irene Dellacasagrande
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Mario Nizzari
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Martina Zambito
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- *Correspondence: Tullio Florio
| |
Collapse
|
27
|
Xu J, Ao YL, Huang C, Song X, Zhang G, Cui W, Wang Y, Zhang XQ, Zhang Z. Harmol promotes α-synuclein degradation and improves motor impairment in Parkinson's models via regulating autophagy-lysosome pathway. NPJ Parkinsons Dis 2022; 8:100. [PMID: 35933473 PMCID: PMC9357076 DOI: 10.1038/s41531-022-00361-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/18/2022] [Indexed: 11/10/2022] Open
Abstract
The abnormal accumulation of α-synuclein (α-syn) is a crucial factor for the onset and pathogenesis of Parkinson's disease (PD), and the autophagy-lysosome pathway (ALP) contributes to α-syn turnover. AMP-activated protein kinase (AMPK) and the mammalian target of rapamycin (mTOR) regulate autophagy by initiating the macroautophagy cascade and promoting lysosomal biogenesis via increased transcription factor EB (TFEB) activity. Hence, activation of AMPK-mTOR-TFEB axis-mediated autophagy might promote α-syn clearance in PD. Harmol is a β-carboline alkaloid that has been extensively studied in a variety of diseases but rarely in PD models. In this study, we aimed to evaluate the effect and underlying mechanism of harmol in PD models in vitro and in vivo. We show that harmol reduces α-syn via ALP in a dose- and time-dependent manner in cell model that overexpressed human A53T mutant α-syn. We also demonstrate that harmol promotes the translocation of TFEB into the nucleus and accompanies the restoration of autophagic flux and lysosomal biogenesis. Importantly, harmol improves motor impairment and down-regulates α-syn levels in the substantia nigra and prefrontal cortex in the α-syn transgenic mice model. Further studies revealed that harmol might activate ALP through AMPK-mTOR-TFEB to promote α-syn clearance. These in vitro and in vivo improvements demonstrate that harmol activates the AMPK-mTOR-TFEB mediated ALP pathway, resulting in reduced α-syn, and suggesting the potential benefit of harmol in the treatment of PD.
Collapse
Affiliation(s)
- Jie Xu
- Guangdong Provincial Engineering Research Center for Modernization of TCM, Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Yun-Lin Ao
- Guangdong Provincial Engineering Research Center for Modernization of TCM, Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Chunhui Huang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Xiubao Song
- Department of Rehabilitation, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Guiliang Zhang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Wei Cui
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Yuqiang Wang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China
| | - Xiao-Qi Zhang
- Guangdong Provincial Engineering Research Center for Modernization of TCM, Guangdong Provincial Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China.
| | - Zaijun Zhang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, P. R. China.
| |
Collapse
|
28
|
Won SY, Park JJ, You ST, Hyeun JA, Kim HK, Jin BK, McLean C, Shin EY, Kim EG. p21-activated kinase 4 controls the aggregation of α-synuclein by reducing the monomeric and aggregated forms of α-synuclein: involvement of the E3 ubiquitin ligase NEDD4-1. Cell Death Dis 2022; 13:575. [PMID: 35773260 PMCID: PMC9247077 DOI: 10.1038/s41419-022-05030-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/10/2022] [Accepted: 06/17/2022] [Indexed: 01/21/2023]
Abstract
Aggregation of misfolded alpha-synuclein (α-synuclein) is a central player in the pathogenesis of neurodegenerative diseases. Therefore, the regulatory mechanism underlying α-synuclein aggregation has been intensively studied in Parkinson's disease (PD) but remains poorly understood. Here, we report p21-activated kinase 4 (PAK4) as a key regulator of α-synuclein aggregation. Immunohistochemical analysis of human PD brain tissues revealed an inverse correlation between PAK4 activity and α-synuclein aggregation. To investigate their causal relationship, we performed loss-of-function and gain-of-function studies using conditional PAK4 depletion in nigral dopaminergic neurons and the introduction of lentivirus expressing a constitutively active form of PAK4 (caPAK4; PAK4S445N/S474E), respectively. For therapeutic relevance in the latter setup, we injected lentivirus into the striatum following the development of motor impairment and analyzed the effects 6 weeks later. In the loss-of-function study, Cre-driven PAK4 depletion in dopaminergic neurons enhanced α-synuclein aggregation, intracytoplasmic Lewy body-like inclusions and Lewy-like neurites, and reduced dopamine levels in PAK4DAT-CreER mice compared to controls. Conversely, caPAK4 reduced α-synuclein aggregation, as assessed by a marked decrease in both proteinase K-resistant and Triton X100-insoluble forms of α-synuclein in the AAV-α-synuclein-induced PD model. Mechanistically, PAK4 specifically interacted with the NEDD4-1 E3 ligase, whose pharmacological inhibition and knockdown suppressed the PAK4-mediated downregulation of α-synuclein. Collectively, these results provide new insights into the pathogenesis of PD and suggest PAK4-based gene therapy as a potential disease-modifying therapy in PD.
Collapse
Affiliation(s)
- So-Yoon Won
- grid.289247.20000 0001 2171 7818Department of Biochemistry & Molecular Biology, Department of Neuroscience, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 02447 South Korea
| | - Jung-Jin Park
- grid.254229.a0000 0000 9611 0917Department of Biochemistry and Medical Research Center, Chungbuk National University College of Medicine, Cheongju, 28644 South Korea
| | - Soon-Tae You
- grid.416965.90000 0004 0647 774XDepartment of Neurosurgery, the Catholic University of Korea, St. Vincent’s Hospital, Suwon, Gyeonggi-do 16247 South Korea
| | - Jong-A Hyeun
- grid.254229.a0000 0000 9611 0917Department of Biochemistry and Medical Research Center, Chungbuk National University College of Medicine, Cheongju, 28644 South Korea
| | - Hyong-Kyu Kim
- grid.254229.a0000 0000 9611 0917Department of Medicine and Microbiology, Chungbuk National University College of Medicine, Cheongju, 28644 South Korea
| | - Byung Kwan Jin
- grid.289247.20000 0001 2171 7818Department of Biochemistry & Molecular Biology, Department of Neuroscience, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, 02447 South Korea
| | - Catriona McLean
- grid.1623.60000 0004 0432 511XDepartment of Pathology, The Alfred Hospital, Melbourne, VIC 3004 Australia
| | - Eun-Young Shin
- grid.254229.a0000 0000 9611 0917Department of Biochemistry and Medical Research Center, Chungbuk National University College of Medicine, Cheongju, 28644 South Korea
| | - Eung-Gook Kim
- grid.254229.a0000 0000 9611 0917Department of Biochemistry and Medical Research Center, Chungbuk National University College of Medicine, Cheongju, 28644 South Korea
| |
Collapse
|
29
|
Sahoo S, Padhy AA, Kumari V, Mishra P. Role of Ubiquitin-Proteasome and Autophagy-Lysosome Pathways in α-Synuclein Aggregate Clearance. Mol Neurobiol 2022; 59:5379-5407. [PMID: 35699874 DOI: 10.1007/s12035-022-02897-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/21/2022] [Indexed: 11/26/2022]
Abstract
Synuclein aggregation in neuronal cells is the primary underlying cause of synucleinopathies. Changes in gene expression patterns, structural modifications, and altered interactions with other cellular proteins often trigger aggregation of α-synuclein, which accumulates as oligomers or fibrils in Lewy bodies. Although fibrillar forms of α-synuclein are primarily considered pathological, recent studies have revealed that even the intermediate states of aggregates are neurotoxic, complicating the development of therapeutic interventions. Autophagy and ubiquitin-proteasome pathways play a significant role in maintaining the soluble levels of α-synuclein inside cells; however, the heterogeneous nature of the aggregates presents a significant bottleneck to its degradation by these cellular pathways. With studies focused on identifying the proteins that modulate synuclein aggregation and clearance, detailed mechanistic insights are emerging about the individual and synergistic effects of these degradation pathways in regulating soluble α-synuclein levels. In this article, we discuss the impact of α-synuclein aggregation on autophagy-lysosome and ubiquitin-proteasome pathways and the therapeutic strategies that target various aspects of synuclein aggregation or degradation via these pathways. Additionally, we also highlight the natural and synthetic compounds that have shown promise in alleviating the cellular damage caused due to synuclein aggregation.
Collapse
Affiliation(s)
- Subhashree Sahoo
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Amrita Arpita Padhy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Varsha Kumari
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Parul Mishra
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
30
|
Mao H, Ye Y, Sun X, Qian C, Wang B, Xie L, Zhang S. Quiescent Elongation of α-Synuclein Pre-form Fibrils Under Different Solution Conditions. Front Neurosci 2022; 16:902077. [PMID: 35692426 PMCID: PMC9175570 DOI: 10.3389/fnins.2022.902077] [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: 03/22/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
The intracellular aggregation of α-synuclein in neurons/glia is considered to be a key step in the pathogenesis of synucleinopathy [including Parkinson’s disease (PD), dementia with Lewy body (DLB), multiple system atrophy (MSA), etc.]. Increasing evidence indicates that the initial pathological α-synuclein aggregates can replicate themselves and propagate in a “seeding” manner to multiple areas of the brain and even to peripheral tissue, which makes it the most important biomarker for the diagnosis of synucleinopathies in recent years. The amplification and propagation capabilities of α-synuclein aggregates are very similar to those of prion-like diseases, which are based on the inherent self-recruitment capabilities of existing misfolded proteins. In vitro, the rapid recruitment process can be reproduced in a simplified model by adding a small amount of α-synuclein pre-formed fibrils to the monomer solution as fibril seeds, which may partially reveal the properties of α-synuclein aggregates. In this study, we explored the elongation rate of α-synuclein pre-formed fibrils under a quiescent incubation condition (rather than shaking/agitating). By using the ThT fluorescence assay, we compared and quantified the elongation fluorescence curves to explore the factors that affect fibril elongation. These factors include proteins’ concentration, temperature, NaCl strength, SDS, temperature pretreatment, and so on. Our work further describes the elongation of α-synuclein fibrils under quiescent incubation conditions. This may have important implications for the in vitro amplification and preservation of α-synuclein aggregates to further understand the prion-like transmission mechanism of PD.
Collapse
Affiliation(s)
- Hengxu Mao
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yongyi Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Sun
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Qian
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Baoyan Wang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linghai Xie
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shizhong Zhang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Shizhong Zhang,
| |
Collapse
|
31
|
Effects of oligomer toxicity, fibril toxicity and fibril spreading in synucleinopathies. Cell Mol Life Sci 2022; 79:174. [PMID: 35244787 PMCID: PMC8897347 DOI: 10.1007/s00018-022-04166-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 12/18/2022]
Abstract
Protein misfolding is a general hallmark of protein deposition diseases, such as Alzheimer’s disease or Parkinson’s disease, in which different types of aggregated species (oligomers, protofibrils and fibrils) are generated by the cells. Despite widespread interest, the relationship between oligomers and fibrils in the aggregation process and spreading remains elusive. A large variety of experimental evidences supported the idea that soluble oligomeric species of different proteins might be more toxic than the larger fibrillar forms. Furthermore, the lack of correlation between the presence of the typical pathological inclusions and disease sustained this debate. However, recent data show that the β-sheet core of the α-Synuclein (αSyn) fibrils is unable to establish persistent interactions with the lipid bilayers, but they can release oligomeric species responsible for an immediate dysfunction of the recipient neurons. Reversibly, such oligomeric species could also contribute to pathogenesis via neuron-to-neuron spreading by their direct cell-to-cell transfer or by generating new fibrils, following their neuronal uptake. In this Review, we discuss the various mechanisms of cellular dysfunction caused by αSyn, including oligomer toxicity, fibril toxicity and fibril spreading.
Collapse
|
32
|
Georgiou M, Yang C, Atkinson R, Pan K, Buskin A, Molina MM, Collin J, Al‐Aama J, Goertler F, Ludwig SEJ, Davey T, Lührmann R, Nagaraja‐Grellscheid S, Johnson CA, Ali R, Armstrong L, Korolchuk V, Urlaub H, Mozaffari‐Jovin S, Lako M. Activation of autophagy reverses progressive and deleterious protein aggregation in PRPF31 patient-induced pluripotent stem cell-derived retinal pigment epithelium cells. Clin Transl Med 2022; 12:e759. [PMID: 35297555 PMCID: PMC8926896 DOI: 10.1002/ctm2.759] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Mutations in pre-mRNA processing factor 31 (PRPF31), a core protein of the spliceosomal tri-snRNP complex, cause autosomal-dominant retinitis pigmentosa (adRP). It has remained an enigma why mutations in ubiquitously expressed tri-snRNP proteins result in retina-specific disorders, and so far, the underlying mechanism of splicing factors-related RP is poorly understood. METHODS We used the induced pluripotent stem cell (iPSC) technology to generate retinal organoids and RPE models from four patients with severe and very severe PRPF31-adRP, unaffected individuals and a CRISPR/Cas9 isogenic control. RESULTS To fully assess the impacts of PRPF31 mutations, quantitative proteomics analyses of retinal organoids and RPE cells were carried out showing RNA splicing, autophagy and lysosome, unfolded protein response (UPR) and visual cycle-related pathways to be significantly affected. Strikingly, the patient-derived RPE and retinal cells were characterised by the presence of large amounts of cytoplasmic aggregates containing the mutant PRPF31 and misfolded, ubiquitin-conjugated proteins including key visual cycle and other RP-linked tri-snRNP proteins, which accumulated progressively with time. The mutant PRPF31 variant was not incorporated into splicing complexes, but reduction of PRPF31 wild-type levels led to tri-snRNP assembly defects in Cajal bodies of PRPF31 patient retinal cells, altered morphology of nuclear speckles and reduced formation of active spliceosomes giving rise to global splicing dysregulation. Moreover, the impaired waste disposal mechanisms further exacerbated aggregate formation, and targeting these by activating the autophagy pathway using Rapamycin reduced cytoplasmic aggregates, leading to improved cell survival. CONCLUSIONS Our data demonstrate that it is the progressive aggregate accumulation that overburdens the waste disposal machinery rather than direct PRPF31-initiated mis-splicing, and thus relieving the RPE cells from insoluble cytoplasmic aggregates presents a novel therapeutic strategy that can be combined with gene therapy studies to fully restore RPE and retinal cell function in PRPF31-adRP patients.
Collapse
Affiliation(s)
- Maria Georgiou
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | - Chunbo Yang
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | - Robert Atkinson
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | - Kuan‐Ting Pan
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Adriana Buskin
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | | | - Joseph Collin
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | - Jumana Al‐Aama
- Faculty of MedicineKing Abdulaziz UniversitySaudi Arabia
| | | | | | - Tracey Davey
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | | | | | | | | | - Lyle Armstrong
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| | | | - Henning Urlaub
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Bioanalytics, Department of Clinical ChemistryUniversity Medical CenterGoettingenGermany
| | - Sina Mozaffari‐Jovin
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Medical Genetics Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Majlinda Lako
- Newcastle University Biosciences InstituteNewcastle upon TyneUK
| |
Collapse
|
33
|
Cheng A, Fukunaga K. [Role of fatty acid-binding protein 7 and novel therapeutic approach in synucleinopathies]. Nihon Yakurigaku Zasshi 2022; 157:396-400. [PMID: 36328545 DOI: 10.1254/fpj.22056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The synucleinopathies are neurodegenerative disease caused by abnormal accumulation of the 140-amino acid-containing protein α-synuclein (αSyn), including Parkinson's disease (PD), diffuse Lewy body dementia (DLBD), and multiple system atrophy (MSA). In patients with PD and DLBD, αSyn is misfolded in neurons, and its aggregation forms Lewy bodies (LB) and Lewy neurites (LN). On the other hand, in patients with MSA, αSyn accumulates primarily in oligodendrocytes (OLGs) and forms glial inclusion bodies (GCIs), a typical pathological feature of MSA. We recently demonstrated a making complex between αSyn and fatty acid-binding proteins (FABPs) in synucleinopathies and received wide attention. Fatty acid-binding protein 3 (FABP3) in dopamine nerves, and fatty acid-binding protein 7 (FABP7) in glial cells promoted αSyn accumulation and aggregation, respectively and caused cell death. Here, we introduced the current studies about the role of αSyn and FABP7 in MSA and novel therapeutic approach targeting for FABP7.
Collapse
Affiliation(s)
- An Cheng
- Department of CNS drug innovation, Graduate School of Pharmaceutical Sciences, Tohoku University
| | - Kohji Fukunaga
- Department of CNS drug innovation, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
34
|
Defective transcription elongation in human cancers imposes targetable proteotoxic vulnerability. Transl Oncol 2021; 16:101323. [PMID: 34954455 PMCID: PMC8718660 DOI: 10.1016/j.tranon.2021.101323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/12/2021] [Accepted: 12/19/2021] [Indexed: 11/24/2022] Open
Abstract
Successful cancer therapy is contingent on identifying cancer-specific aberrant phenotypes and their associated vulnerabilities. We recently reported that a subset of almost every cancer type contains a genome-wide defect in RNA Polymerase II-mediated transcription elongation (TEdef), which impairs the expression of long genes and confers resistance to anti-tumor immune attack. Using a combination of computational analysis and laboratory experiments, we report that tumor cells with TEdef have widespread overexpression of the components of the protein homeostasis machinery (mostly composed of short genes), including protein folding and clearance. Accordingly, TEdef cells were characterized by abnormally high levels of insoluble protein aggregates in the cytoplasm and autophagy influx. We present evidence that TEdef cells exhibit impaired clearance of misfolded protein aggregates through the ubiquitin-proteasome system, and thus rely on autophagy for their degradation. As such, while these cells were highly resistant to proteasome inhibitors, they were acutely sensitive to inhibitors of autophagy in vitro and in vivo. This study reveals a major aberrant phenotype that is observed in ∼15-25% of all cancers and characterizes a unique cellular vulnerability that can be readily exploited in the clinic to improve treatment efficacy.
Collapse
|
35
|
Gadhe L, Sakunthala A, Mukherjee S, Gahlot N, Bera R, Sawner AS, Kadu P, Maji SK. Intermediates of α-synuclein aggregation: Implications in Parkinson's disease pathogenesis. Biophys Chem 2021; 281:106736. [PMID: 34923391 DOI: 10.1016/j.bpc.2021.106736] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/27/2021] [Accepted: 11/27/2021] [Indexed: 12/11/2022]
Abstract
Cytoplasmic deposition of aberrantly misfolded α-synuclein (α-Syn) is a common feature of synucleinopathies, including Parkinson's disease (PD). However, the precise pathogenic mechanism of α-Syn in synucleinopathies remains elusive. Emerging evidence has suggested that α-Syn may contribute to PD pathogenesis in several ways; wherein the contribution of fibrillar species, for exerting toxicity and disease transmission, cannot be neglected. Further, the oligomeric species could be the most plausible neurotoxic species causing neuronal cell death. However, understanding the structural and molecular insights of these oligomers are very challenging due to the heterogeneity and transient nature of the species. In this review, we discuss the recent advancements in understanding the formation and role of α-Syn oligomers in PD pathogenesis. We also summarize the different types of α-Syn oligomeric species and potential mechanisms to exert neurotoxicity. Finally, we address the possible ways to target α-Syn as a promising approach against PD and the possible future directions.
Collapse
Affiliation(s)
- Laxmikant Gadhe
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Riya Bera
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
36
|
George DE, Tepe JJ. Advances in Proteasome Enhancement by Small Molecules. Biomolecules 2021; 11:1789. [PMID: 34944433 PMCID: PMC8699248 DOI: 10.3390/biom11121789] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 01/11/2023] Open
Abstract
The proteasome system is a large and complex molecular machinery responsible for the degradation of misfolded, damaged, and redundant cellular proteins. When proteasome function is impaired, unwanted proteins accumulate, which can lead to several diseases including age-related and neurodegenerative diseases. Enhancing proteasome-mediated substrate degradation with small molecules may therefore be a valuable strategy for the treatment of various neurodegenerative diseases such as Parkinson's, Alzheimer's, and Huntington's diseases. In this review, we discuss the structure of proteasome and how proteasome's proteolytic activity is associated with aging and various neurodegenerative diseases. We also summarize various classes of compounds that are capable of enhancing, directly or indirectly, proteasome-mediated protein degradation.
Collapse
Affiliation(s)
| | - Jetze J. Tepe
- Department of Chemistry and Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
37
|
Memou A, Dimitrakopoulos L, Kedariti M, Kentros M, Lamprou A, Petropoulou-Vathi L, Valkimadi PE, Rideout HJ. Defining (and blocking) neuronal death in Parkinson's disease: Does it matter what we call it? Brain Res 2021; 1771:147639. [PMID: 34492263 DOI: 10.1016/j.brainres.2021.147639] [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: 02/25/2021] [Revised: 07/29/2021] [Accepted: 08/24/2021] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, comprised of both familial and idiopathic forms, behind only Alzheimer's disease (AD). The disease is characterized, regardless of the pathogenesis, primarily by a loss of DA neurons in the ventral midbrain as well as noradrenergic neurons of the locus coeruleus; however, by the time symptoms manifest, considerable neuronal loss in both areas has occurred. Neuroprotective strategies thus have to be paired with more sensitive and specific biomarker assays that can identify early at-risk patients in order to initiate disease-modifying therapies at an earlier stage in the disease. Complicating this is the fact that multiple forms of cell death mediate the neuronal loss; however, with a common underlying element that the cell death is considered a "regulated" form of cell death, in contrast to an un-controlled necrotic cell death process. In this review we focus our discussion on several categories of regulated cell death in the context of PD: apoptosis, necroptosis, pyroptosis, and autophagic cell death. In clinical studies as well as experimental in vivo models of PD, there is evidence for a role of each of these forms of cell death in the loss of midbrain DA neurons, and specific therapeutic strategies have been proposed and tested. What remains unclear however is the relative contributions of these distinct forms of cell death to the overall loss of DA neurons, whether they occur at different stages of the disease, or whether specific sub-regions within the midbrain are more susceptible to specific death triggers and pathways.
Collapse
Affiliation(s)
- Anna Memou
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Lampros Dimitrakopoulos
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Kedariti
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Michalis Kentros
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Andriana Lamprou
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Lilian Petropoulou-Vathi
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Polytimi-Eleni Valkimadi
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Hardy J Rideout
- Laboratory of Neurodegenerative Diseases, Center for Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
38
|
Menon S, Kofoed RH, Nabbouh F, Xhima K, Al-Fahoum Y, Langman T, Mount HTJ, Shihabuddin LS, Sardi SP, Fraser PE, Watts JC, Aubert I, Tandon A. Viral alpha-synuclein knockdown prevents spreading synucleinopathy. Brain Commun 2021; 3:fcab247. [PMID: 34761222 PMCID: PMC8576194 DOI: 10.1093/braincomms/fcab247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/14/2021] [Accepted: 07/17/2021] [Indexed: 12/19/2022] Open
Abstract
The accumulation of aggregated alpha-synuclein (α-syn) in Parkinson's disease, dementia with Lewy bodies and multiple system atrophy is thought to involve a common prion-like mechanism, whereby misfolded α-syn provides a conformational template for further accumulation of pathological α-syn. We tested whether silencing α-syn gene expression could reduce native non-aggregated α-syn substrate and thereby disrupt the propagation of pathological α-syn initiated by seeding with synucleinopathy-affected mouse brain homogenates. Unilateral intracerebral injections of adeno-associated virus serotype-1 encoding microRNA targeting the α-syn gene reduced the extent and severity of both the α-syn pathology and motor deficits. Importantly, a moderate 50% reduction in α-syn was sufficient to prevent the spread of α-syn pathology to distal brain regions. Our study combines behavioural, immunohistochemical and biochemical data that strongly support α-syn knockdown gene therapy for synucleinopathies.
Collapse
Affiliation(s)
- Sindhu Menon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Rikke H Kofoed
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Fadl Nabbouh
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Kristiana Xhima
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Yasmeen Al-Fahoum
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Tammy Langman
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Howard T J Mount
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Lamya S Shihabuddin
- Sanofi, Framingham, MA 01701, USA
- Present address: 5AM Ventures, Boston, MA, USA
| | | | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Isabelle Aubert
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence to: Anurag Tandon, PhD Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower Rm. 4KD481, 60 Leonard Ave, Toronto, ON M5T 0S8, Canada E-mail:
| |
Collapse
|
39
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
40
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
41
|
Raha S, Dutta D, Roy A, Pahan K. Reduction of Lewy Body Pathology by Oral Cinnamon. J Neuroimmune Pharmacol 2021; 16:592-608. [PMID: 32889602 PMCID: PMC7933354 DOI: 10.1007/s11481-020-09955-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022]
Abstract
α-Synucleinopathies in a broader sense comprise of several neurodegenerative disorders that primarily include Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). These disorders are well characterized by the accumulation of aggregated insoluble α-synuclein (α-syn) protein known as Lewy bodies. Till date no effective cure is available to reduce the burden of Lewy body. The present investigation underlines the importance of a naturally used spice and flavoring agent viz. cinnamon in reducing α-syn deposits in transgenic mice expressing mutant A53T human α-syn. Upon oral administration, cinnamon markedly reduced the level of insoluble α-syn in nigra, hippocampus and brain stem of A53T mice. We also demonstrated that sodium benzoate (NaB), a metabolite of cinnamon, a widely used food additive and a FDA-approved drug for glycine encephalopathy, was also capable of reducing α-syn deposits in A53T mice. In addition, both cinnamon and NaB treatments showed improvement in their motor and cognitive functions. Glial activation plays an important role in the pathogenesis of various neurodegenerative disorders including PD, DLB and MSA, and we found suppression of microglial and astroglial activation in the nigra of A53T mice upon cinnamon treatment. Moreover, neuroprotective proteins like DJ-1 and Parkin are known to reduce the formation of Lewy bodies in the CNS. Accordingly, we observed upregulation and/or normalization of DJ-1 and Parkin in the nigra of A53T mice by treatment with cinnamon and NaB. Together, these results highlight a new therapeutic property of cinnamon and suggest that cinnamon and NaB may be used to halt the progression of α-synucleinopathies.Graphical Abstract.
Collapse
Affiliation(s)
- Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA.
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue, Chicago, IL, 60612, USA.
| |
Collapse
|
42
|
Popova B, Galka D, Häffner N, Wang D, Schmitt K, Valerius O, Knop M, Braus GH. α-Synuclein Decreases the Abundance of Proteasome Subunits and Alters Ubiquitin Conjugates in Yeast. Cells 2021; 10:cells10092229. [PMID: 34571878 PMCID: PMC8468666 DOI: 10.3390/cells10092229] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 01/18/2023] Open
Abstract
Parkinson’s disease (PD) is the most prevalent movement disorder characterized with loss of dopaminergic neurons in the brain. One of the pathological hallmarks of the disease is accumulation of aggregated α-synuclein (αSyn) in cytoplasmic Lewy body inclusions that indicates significant dysfunction of protein homeostasis in PD. Accumulation is accompanied with highly elevated S129 phosphorylation, suggesting that this posttranslational modification is linked to pathogenicity and altered αSyn inclusion dynamics. To address the role of S129 phosphorylation on protein dynamics further we investigated the wild type and S129A variants using yeast and a tandem fluorescent timer protein reporter approach to monitor protein turnover and stability. Overexpression of both variants leads to inhibited yeast growth. Soluble S129A is more stable and additional Y133F substitution permits αSyn degradation in a phosphorylation-independent manner. Quantitative cellular proteomics revealed significant αSyn-dependent disturbances of the cellular protein homeostasis, which are increased upon S129 phosphorylation. Disturbances are characterized by decreased abundance of the ubiquitin-dependent protein degradation machinery. Biotin proximity labelling revealed that αSyn interacts with the Rpt2 base subunit. Proteasome subunit depletion by reducing the expression of the corresponding genes enhances αSyn toxicity. Our studies demonstrate that turnover of αSyn and depletion of the proteasome pool correlate in a complex relationship between altered proteasome composition and increased αSyn toxicity.
Collapse
Affiliation(s)
- Blagovesta Popova
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
- Correspondence: (B.P.); (G.H.B.)
| | - Dajana Galka
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
| | - Nicola Häffner
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
| | - Dan Wang
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
| | - Kerstin Schmitt
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
| | - Oliver Valerius
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
| | - Michael Knop
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany;
| | - Gerhard H. Braus
- Department of Molecular Microbiology and Genetics, Institute for Microbiology and Genetics, University of Göttingen, 37077 Göttingen, Germany; (D.G.); (N.H.); (D.W.); (K.S.); (O.V.)
- Correspondence: (B.P.); (G.H.B.)
| |
Collapse
|
43
|
Maresh ME, Chen P, Hazbun TR, Trader DJ. A Yeast Chronological Lifespan Assay to Assess Activity of Proteasome Stimulators. Chembiochem 2021; 22:2553-2560. [PMID: 34043860 PMCID: PMC8478123 DOI: 10.1002/cbic.202100117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/26/2021] [Indexed: 11/10/2022]
Abstract
Aging is characterized by changes in several cellular processes, including dysregulation of proteostasis. Current research has shown long-lived rodents display elevated proteasome activity throughout life and proteasome dysfunction is linked to shorter lifespans in a transgenic mouse model. The ubiquitin proteasome system (UPS) is one of the main pathways leading to cellular protein clearance and quality maintenance. Reduction in proteasome activity is associated with aging and its related pathologies. Small molecule stimulators of the proteasome have been proposed to help alleviate cellular stress related to unwanted protein accumulation. Here we have described the development of techniques to monitor the impact of proteasome stimulation in wild-type yeast and a strain that has impaired proteasome expression. We validated our chronological lifespan assay using both types of yeast with a variety of small molecule stimulators at different concentrations. By modifying the media conditions for the yeast, molecules can be evaluated for their potential to increase chronological lifespan in five days. Additionally, our assay conditions can be used to monitor the activity of proteasome stimulators in modulating the degradation of a YFP-α-synuclein fusion protein produced by yeast. We anticipate these methods to be valuable for those wishing to study the impact of increasing proteasome-mediated degradation of proteins in a eukaryotic model organism.
Collapse
Affiliation(s)
- Marianne E. Maresh
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907
| | - Panyue Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907
| | - Tony R. Hazbun
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907
| | - Darci J. Trader
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907
| |
Collapse
|
44
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
45
|
Ferreira N, Gram H, Sorrentino ZA, Gregersen E, Schmidt SI, Reimer L, Betzer C, Perez-Gozalbo C, Beltoja M, Nagaraj M, Wang J, Nowak JS, Dong M, Willén K, Cholak E, Bjerregaard-Andersen K, Mendez N, Rabadia P, Shahnawaz M, Soto C, Otzen DE, Akbey Ü, Meyer M, Giasson BI, Romero-Ramos M, Jensen PH. Multiple system atrophy-associated oligodendroglial protein p25α stimulates formation of novel α-synuclein strain with enhanced neurodegenerative potential. Acta Neuropathol 2021; 142:87-115. [PMID: 33978813 PMCID: PMC8217051 DOI: 10.1007/s00401-021-02316-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 01/08/2023]
Abstract
Pathology consisting of intracellular aggregates of alpha-Synuclein (α-Syn) spread through the nervous system in a variety of neurodegenerative disorders including Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The discovery of structurally distinct α-Syn polymorphs, so-called strains, supports a hypothesis where strain-specific structures are templated into aggregates formed by native α-Syn. These distinct strains are hypothesised to dictate the spreading of pathology in the tissue and the cellular impact of the aggregates, thereby contributing to the variety of clinical phenotypes. Here, we present evidence of a novel α-Syn strain induced by the multiple system atrophy-associated oligodendroglial protein p25α. Using an array of biophysical, biochemical, cellular, and in vivo analyses, we demonstrate that compared to α-Syn alone, a substoichiometric concentration of p25α redirects α-Syn aggregation into a unique α-Syn/p25α strain with a different structure and enhanced in vivo prodegenerative properties. The α-Syn/p25α strain induced larger inclusions in human dopaminergic neurons. In vivo, intramuscular injection of preformed fibrils (PFF) of the α-Syn/p25α strain compared to α-Syn PFF resulted in a shortened life span and a distinct anatomical distribution of inclusion pathology in the brain of a human A53T transgenic (line M83) mouse. Investigation of α-Syn aggregates in brain stem extracts of end-stage mice demonstrated that the more aggressive phenotype of the α-Syn/p25α strain was associated with an increased load of α-Syn aggregates based on a Förster resonance energy transfer immunoassay and a reduced α-Syn aggregate seeding activity based on a protein misfolding cyclic amplification assay. When injected unilaterally into the striata of wild-type mice, the α-Syn/p25α strain resulted in a more-pronounced motoric phenotype than α-Syn PFF and exhibited a "tropism" for nigro-striatal neurons compared to α-Syn PFF. Overall, our data support a hypothesis whereby oligodendroglial p25α is responsible for generating a highly prodegenerative α-Syn strain in multiple system atrophy.
Collapse
|
46
|
Manzanza NDO, Sedlackova L, Kalaria RN. Alpha-Synuclein Post-translational Modifications: Implications for Pathogenesis of Lewy Body Disorders. Front Aging Neurosci 2021; 13:690293. [PMID: 34248606 PMCID: PMC8267936 DOI: 10.3389/fnagi.2021.690293] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Lewy Body Disorders (LBDs) lie within the spectrum of age-related neurodegenerative diseases now frequently categorized as the synucleinopathies. LBDs are considered to be among the second most common form of neurodegenerative dementias after Alzheimer's disease. They are progressive conditions with variable clinical symptoms embodied within specific cognitive and behavioral disorders. There are currently no effective treatments for LBDs. LBDs are histopathologically characterized by the presence of abnormal neuronal inclusions commonly known as Lewy Bodies (LBs) and extracellular Lewy Neurites (LNs). The inclusions predominantly comprise aggregates of alpha-synuclein (aSyn). It has been proposed that post-translational modifications (PTMs) such as aSyn phosphorylation, ubiquitination SUMOylation, Nitration, o-GlcNacylation, and Truncation play important roles in the formation of toxic forms of the protein, which consequently facilitates the formation of these inclusions. This review focuses on the role of different PTMs in aSyn in the pathogenesis of LBDs. We highlight how these PTMs interact with aSyn to promote misfolding and aggregation and interplay with cell membranes leading to the potential functional and pathogenic consequences detected so far, and their involvement in the development of LBDs.
Collapse
Affiliation(s)
- Nelson de Oliveira Manzanza
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lucia Sedlackova
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Raj N. Kalaria
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
47
|
Super-resolution imaging reveals α-synuclein seeded aggregation in SH-SY5Y cells. Commun Biol 2021; 4:613. [PMID: 34021258 PMCID: PMC8139990 DOI: 10.1038/s42003-021-02126-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Aggregation of α-synuclein (α-syn) is closely linked to Parkinson's disease (PD) and the related synucleinopathies. Aggregates spread through the brain during the progression of PD, but the mechanism by which this occurs is still not known. One possibility is a self-propagating, templated-seeding mechanism, but this cannot be established without quantitative information about the efficiencies and rates of the key steps in the cellular process. To address this issue, we imaged the uptake and seeding of unlabeled exogenous α-syn fibrils by SH-SY5Y cells and the resulting secreted aggregates, using super-resolution microscopy. Externally-applied fibrils very inefficiently induced self-assembly of endogenous α-syn in a process accelerated by the proteasome. Seeding resulted in the increased secretion of nanoscopic aggregates (mean 35 nm diameter), of both α-syn and Aβ. Our results suggest that cells respond to seed-induced disruption of protein homeostasis predominantly by secreting nanoscopic aggregates; this mechanism may therefore be an important protective response by cells to protein aggregation.
Collapse
|
48
|
Kabir MT, Uddin MS, Abdeen A, Ashraf GM, Perveen A, Hafeez A, Bin-Jumah MN, Abdel-Daim MM. Evidence Linking Protein Misfolding to Quality Control in Progressive Neurodegenerative Diseases. Curr Top Med Chem 2021; 20:2025-2043. [PMID: 32552649 DOI: 10.2174/1568026620666200618114924] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/25/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022]
Abstract
Several proteolytic systems including ubiquitin (Ub)-proteasome system (UPS), chaperonemediated autophagy (CMA), and macroautophagy are used by the mammalian cells to remove misfolded proteins (MPs). UPS mediates degradation of most of the MPs, where Ub-conjugated substrates are deubiquitinated, unfolded, and passed through the proteasome's narrow chamber, and eventually break into smaller peptides. It has been observed that the substrates that show a specific degradation signal, the KFERQ sequence motif, can be delivered to and go through CMA-mediated degradation in lysosomes. Macroautophagy can help in the degradation of substrates that are prone to aggregation and resistant to both the CMA and UPS. In the aforesaid case, cargoes are separated into autophagosomes before lysosomal hydrolase-mediated degradation. Even though the majority of the aggregated and MPs in the human proteome can be removed via cellular protein quality control (PQC), some mutant and native proteins tend to aggregate into β-sheet-rich oligomers that exhibit resistance to all identified proteolytic processes and can, therefore, grow into extracellular plaques or inclusion bodies. Indeed, the buildup of protease-resistant aggregated and MPs is a usual process underlying various protein misfolding disorders, including neurodegenerative diseases (NDs) for example Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and prion diseases. In this article, we have focused on the contribution of PQC in the degradation of pathogenic proteins in NDs.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.,Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
49
|
Cheng A, Jia W, Kawahata I, Fukunaga K. Impact of Fatty Acid-Binding Proteins in α-Synuclein-Induced Mitochondrial Injury in Synucleinopathy. Biomedicines 2021; 9:biomedicines9050560. [PMID: 34067791 PMCID: PMC8156290 DOI: 10.3390/biomedicines9050560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Synucleinopathies are diverse diseases with motor and cognitive dysfunction due to progressive neuronal loss or demyelination, due to oligodendrocyte loss in the brain. While the etiology of neurodegenerative disorders (NDDs) is likely multifactorial, mitochondrial injury is one of the most vital factors in neuronal loss and oligodendrocyte dysfunction, especially in Parkinson’s disease, dementia with Lewy body, multiple system atrophy, and Krabbe disease. In recent years, the abnormal accumulation of highly neurotoxic α-synuclein in the mitochondrial membrane, which leads to mitochondrial dysfunction, was well studied. Furthermore, fatty acid-binding proteins (FABPs), which are members of a superfamily and are essential in fatty acid trafficking, were reported to trigger α-synuclein oligomerization in neurons and glial cells and to target the mitochondrial outer membrane, thereby causing mitochondrial loss. Here, we provide an updated overview of recent findings on FABP and α-synuclein interactions and mitochondrial injury in NDDs.
Collapse
Affiliation(s)
- An Cheng
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
| | - Wenbin Jia
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
| | - Ichiro Kawahata
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
| | - Kohji Fukunaga
- Departments of Pharmacology, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan; (A.C.); (W.J.); (I.K.)
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Science, Tohoku University, Sendai 980-8578, Japan
- Correspondence: ; Tel.: +81-(22)-795-6837
| |
Collapse
|
50
|
Pinto MJ, Tomé D, Almeida RD. The Ubiquitinated Axon: Local Control of Axon Development and Function by Ubiquitin. J Neurosci 2021; 41:2796-2813. [PMID: 33789876 PMCID: PMC8018891 DOI: 10.1523/jneurosci.2251-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 02/01/2023] Open
Abstract
Ubiquitin tagging sets protein fate. With a wide range of possible patterns and reversibility, ubiquitination can assume many shapes to meet specific demands of a particular cell across time and space. In neurons, unique cells with functionally distinct axons and dendrites harboring dynamic synapses, the ubiquitin code is exploited at the height of its power. Indeed, wide expression of ubiquitination and proteasome machinery at synapses, a diverse brain ubiquitome, and the existence of ubiquitin-related neurodevelopmental diseases support a fundamental role of ubiquitin signaling in the developing and mature brain. While special attention has been given to dendritic ubiquitin-dependent control, how axonal biology is governed by this small but versatile molecule has been considerably less discussed. Herein, we set out to explore the ubiquitin-mediated spatiotemporal control of an axon's lifetime: from its differentiation and growth through presynaptic formation, function, and pruning.
Collapse
Affiliation(s)
- Maria J Pinto
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Diogo Tomé
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ramiro D Almeida
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|