1
|
Zeng Y, Wu T, Liang F, Long S, Guo W, Huang Y, Pei Z. Expression of human Ras-related protein Rab39B variant T168K in Caenorhabditis elegans leads to motor dysfunction and dopaminergic neuron degeneration. Heliyon 2024; 10:e26902. [PMID: 38444482 PMCID: PMC10912484 DOI: 10.1016/j.heliyon.2024.e26902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Human RAB39B gene is related to familial early-onset Parkinson disease. In early adulthood, men with the RAB39B c.503C > A (Thr168Lys, p. T168K) mutation develop typical tremor, bradykinesia, and alpha-synuclein accumulation. We investigated the pathological mechanism of RAB39B T168K in a Caenorhabditis elegans model. In early adult C. elegans, RAB39B T168K led to dopaminergic neuron degeneration that presented as disrupted dendrites and blunt neuronal cells. Abnormal dopamine secretion was inferred from a decline in motor function and a positive basal slowing phenotype. Dopamine-associated tests confirmed that synthesis and recycling of dopamine were normal. The RAB39B T168K mutation might impair dopamine vesicular transmission from the presynaptic membrane to the synaptic gap in dopaminergic neurons. The release-dependent feedback mechanism in neurotransmitters regulates the balance of receptor activities. Protein-protein interactions network analysis revealed that RAB39B may also function in lysosomal degradation and autophagy. Impaired disposal of misfolded α-synuclein eventually leads to protein aggregation. Thus, like other members of the Rab family, RAB39B may be involved in vesicular transport associated with dopamine secretion and α-synuclein clearance.
Collapse
Affiliation(s)
- Yixuan Zeng
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Tengteng Wu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengyin Liang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Simei Long
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Huang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Barker RA, Buttery PC. Disease-specific interventions: The use of cell and gene therapies for Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:171-191. [PMID: 39341654 DOI: 10.1016/b978-0-323-90120-8.00003-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Approaches to repair the brain around the loss of the nigrostriatal dopaminergic pathways in Parkinson disease (PD) are not new and have been attempted over many years. However, of late, the situation has moved forward in two main ways. In the case of cell therapies, the ability to make large numbers of authentic midbrain dopaminergic neuroblasts from human pluripotent stem cell sources has turned what was an interesting avenue of research into a major area of investment and trialing, by academics in conjunction with Pharma. In the case of gene therapies, their use around dopamine replacement has waned, as the interest in using them for disease modification targeting PD-specific pathways has grown. In this chapter, we discuss all these developments and the current status of cell and gene therapies for PD.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| | - Philip C Buttery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Bu M, Follett J, Deng I, Tatarnikov I, Wall S, Guenther D, Maczis M, Wimsatt G, Milnerwood A, Moehle MS, Khoshbouei H, Farrer MJ. Inhibition of LRRK2 kinase activity rescues deficits in striatal dopamine physiology in VPS35 p.D620N knock-in mice. NPJ Parkinsons Dis 2023; 9:167. [PMID: 38110354 PMCID: PMC10728137 DOI: 10.1038/s41531-023-00609-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 11/24/2023] [Indexed: 12/20/2023] Open
Abstract
Dysregulation of dopamine neurotransmission profoundly affects motor, motivation and learning behaviors, and can be observed during the prodromal phase of Parkinson's disease (PD). However, the mechanism underlying these pathophysiological changes remains to be elucidated. Mutations in vacuolar protein sorting 35 (VPS35) and leucine-rich repeat kinase 2 (LRRK2) both lead to autosomal dominant PD, and VPS35 and LRRK2 may physically interact to govern the trafficking of synaptic cargos within the endo-lysosomal network in a kinase-dependent manner. To better understand the functional role of VPS35 and LRRK2 on dopamine physiology, we examined Vps35 haploinsufficient (Haplo) and Vps35 p.D620N knock-in (VKI) mice and how their behavior, dopamine kinetics and biochemistry are influenced by LRRK2 kinase inhibitors. We found Vps35 p.D620N significantly elevates LRRK2-mediated phosphorylation of Rab10, Rab12 and Rab29. In contrast, Vps35 haploinsufficiency reduces phosphorylation of Rab12. While striatal dopamine transporter (DAT) expression and function is similarly impaired in both VKI and Haplo mice, that physiology is normalized in VKI by treatment with the LRRK2 kinase inhibitor, MLi-2. As a corollary, VKI animals show a significant increase in amphetamine induced hyperlocomotion, compared to Haplo mice, that is also abolished by MLi-2. Taken together, these data show Vps35 p.D620N confers a gain-of-function with respect to LRRK2 kinase activity, and that VPS35 and LRRK2 functionally interact to regulate DAT function and striatal dopamine transmission.
Collapse
Affiliation(s)
- Mengfei Bu
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jordan Follett
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Isaac Deng
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Igor Tatarnikov
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Shannon Wall
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Dylan Guenther
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Melissa Maczis
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Genevieve Wimsatt
- Department of Neurology, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Austen Milnerwood
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Mark S Moehle
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Habibeh Khoshbouei
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Matthew J Farrer
- Department of Neurology, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
4
|
Sánchez-Vidaña DI, Li J, Abokyi S, Chan JNM, Ngai SPC, Lau BWM. In vitro methods in autophagy research: Applications in neurodegenerative diseases and mood disorders. Front Mol Neurosci 2023; 16:1168948. [PMID: 37122628 PMCID: PMC10130388 DOI: 10.3389/fnmol.2023.1168948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/14/2023] [Indexed: 05/02/2023] Open
Abstract
Background Autophagy is a conserved physiological intracellular mechanism responsible for the degradation and recycling of cytoplasmic constituents (e.g., damaged organelles, and protein aggregates) to maintain cell homeostasis. Aberrant autophagy has been observed in neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Amyotrophic Lateral Sclerosis (ALS), and Huntington's Disease (HD), and recently aberrant autophagy has been associated with mood disorders, such as depression. Several in vitro methods have been developed to study the complex and tightly regulated mechanisms of autophagy. In vitro methods applied to autophagy research are used to identify molecular key players involved in dysfunctional autophagy and to screen autophagy regulators with therapeutic applications in neurological diseases and mood disorders. Therefore, the aims of this narrative review are (1) to compile information on the cell-based methods used in autophagy research, (2) to discuss their application, and (3) to create a catalog of traditional and novel in vitro methods applied in neurodegenerative diseases and depression. Methods Pubmed and Google Scholar were used to retrieve relevant in vitro studies on autophagy mechanisms in neurological diseases and depression using a combination of search terms per mechanism and disease (e.g., "macroautophagy" and "Alzheimer's disease"). A total of 37 studies were included (14 in PD, 8 in AD, 5 in ALS, 5 in %, and 5 in depression). Results A repertoire of traditional and novel approaches and techniques was compiled and discussed. The methods used in autophagy research focused on the mechanisms of macroautophagy, microautophagy, and chaperone-mediated autophagy. The in vitro tools presented in this review can be applied to explore pathophysiological mechanisms at a molecular level and to screen for potential therapeutic agents and their mechanism of action, which can be of great importance to understanding disease biology and potential therapeutic options in the context of neurodegenerative disorders and depression. Conclusion This is the first review to compile, discuss, and provide a catalog of traditional and novel in vitro models applied to neurodegenerative disorders and depression.
Collapse
Affiliation(s)
- Dalinda Isabel Sánchez-Vidaña
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Mental Health Research Centre, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Samuel Abokyi
- School of Optometry, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jackie Ngai-Man Chan
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Shirley Pui-Ching Ngai
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Benson Wui-Man Lau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Mental Health Research Centre, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
5
|
Vogiatzis S, Celestino M, Trevisan M, Magro G, Del Vecchio C, Erdengiz D, Palù G, Parolin C, Maguire-Zeiss K, Calistri A. Lentiviral Vectors Expressing Chimeric NEDD4 Ubiquitin Ligases: An Innovative Approach for Interfering with Alpha-Synuclein Accumulation. Cells 2021; 10:cells10113256. [PMID: 34831478 PMCID: PMC8624294 DOI: 10.3390/cells10113256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 11/25/2022] Open
Abstract
One of the main pathological features of Parkinson’s disease (PD) is a diffuse accumulation of alpha-synuclein (aS) aggregates in neurons. The NEDD4 E3 Ub ligase promotes aS degradation by the endosomal–lysosomal route. Interestingly, NEDD4, as well as being a small molecule able to trigger its functions, is protective against human aS toxicity in evolutionary distant models. While pharmacological activation of E3 enzymes is not easy to achieve, their flexibility and the lack of “consensus” motifs for Ub-conjugation allow the development of engineered Ub-ligases, able to target proteins of interest. We developed lentiviral vectors, encoding well-characterized anti-human aS scFvs fused in frame to the NEDD4 catalytic domain (ubiquibodies), in order to target ubiquitinate aS. We demonstrate that, while all generated ubiquibodies bind to and ubiquitinate aS, the one directed against the non-amyloid component (NAC) of aS (Nac32HECT) affects aS’s intracellular levels. Furthermore, Nac32HECT expression partially rescues aS’s overexpression or mutation toxicity in neural stem cells. Overall, our data suggest that ubiquibodies, and Nac32HECT in particular, represent a valid platform for interfering with the effects of aS’s accumulation and aggregation in neurons.
Collapse
Affiliation(s)
- Stefania Vogiatzis
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Michele Celestino
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Marta Trevisan
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Gloria Magro
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Claudia Del Vecchio
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Deran Erdengiz
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd NW, NRB, EP04, Washington, DC 20057, USA; (D.E.); (K.M.-Z.)
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Cristina Parolin
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
| | - Kathleen Maguire-Zeiss
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd NW, NRB, EP04, Washington, DC 20057, USA; (D.E.); (K.M.-Z.)
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (S.V.); (M.C.); (M.T.); (G.M.); (C.D.V.); (G.P.); (C.P.)
- Correspondence: ; Tel.: +39-049-827-2341
| |
Collapse
|
6
|
Peters JJ, Leitz J, Oses-Prieto JA, Burlingame AL, Brunger AT. Molecular Characterization of AMPA-Receptor-Containing Vesicles. Front Mol Neurosci 2021; 14:754631. [PMID: 34720876 PMCID: PMC8554035 DOI: 10.3389/fnmol.2021.754631] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Regulated delivery of AMPA receptors (AMPARs) to the postsynaptic membrane is an essential step in synaptic strength modification, and in particular, long-term potentiation (LTP). While LTP has been extensively studied using electrophysiology and light microscopy, several questions regarding the molecular mechanisms of AMPAR delivery via trafficking vesicles remain outstanding, including the gross molecular make up of AMPAR trafficking organelles and identification and location of calcium sensors required for SNARE complex-dependent membrane fusion of such trafficking vesicles with the plasma membrane. Here, we isolated AMPA-containing vesicles (ACVs) from whole mouse brains via immunoisolation and characterized them using immunoelectron microscopy, immunoblotting, and liquid chromatography–tandem mass spectrometry (LC–MS/MS). We identified several proteins on ACVs that were previously found to play a role in AMPAR trafficking, including synaptobrevin-2, Rabs, the SM protein Munc18-1, the calcium-sensor synaptotagmin-1, as well as several new candidates, including synaptophysin and synaptogyrin on ACV membranes. Additionally, we identified two populations of ACVs based on size and molecular composition: small-diameter, synaptobrevin-2- and GluA1-containing ACVs, and larger transferrin- receptor-, GluA1-, GluA2-, and GluA3-containing ACVs. The small-diameter population of ACVs may represent a fusion-capable population of vesicles due to the presence of synaptobrevin-2. Because the fusion of ACVs may be a requisite of LTP, this population could represent trafficking vesicles related to LTP.
Collapse
Affiliation(s)
- John Jacob Peters
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States.,Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States.,Department of Structural Biology, Stanford University, Stanford, CA, United States.,Department of Photon Science, Stanford University, Stanford, CA, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States.,Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States.,Department of Structural Biology, Stanford University, Stanford, CA, United States.,Department of Photon Science, Stanford University, Stanford, CA, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, United States
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, United States
| | - Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States.,Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States.,Department of Structural Biology, Stanford University, Stanford, CA, United States.,Department of Photon Science, Stanford University, Stanford, CA, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
7
|
Predicting PY motif-mediated protein-protein interactions in the Nedd4 family of ubiquitin ligases. PLoS One 2021; 16:e0258315. [PMID: 34637467 PMCID: PMC8509885 DOI: 10.1371/journal.pone.0258315] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
The Nedd4 family contains several structurally related but functionally distinct HECT-type ubiquitin ligases. The members of the Nedd4 family are known to recognize substrates through their multiple WW domains, which recognize PY motifs (PPxY, LPxY) or phospho-threonine or phospho-serine residues. To better understand protein interactor recognition mechanisms across the Nedd4 family, we report the development and implementation of a python-based tool, PxYFinder, to identify PY motifs in the primary sequences of previously identified interactors of Nedd4 and related ligases. Using PxYFinder, we find that, on average, half of Nedd4 family interactions are likely PY-motif mediated. Further, we find that PPxY motifs are more prevalent than LPxY motifs and are more likely to occur in proline-rich regions and that PPxY regions are more disordered on average relative to LPxY-containing regions. Informed by consensus sequences for PY motifs across the Nedd4 interactome, we rationally designed a focused peptide library and employed a computational screen, revealing sequence- and biomolecular interaction-dependent determinants of WW-domain/PY-motif interactions. Cumulatively, our efforts provide a new bioinformatic tool and expand our understanding of sequence and structural factors that contribute to PY-motif mediated interactor recognition across the Nedd4 family.
Collapse
|
8
|
Riederer P, Monoranu C, Strobel S, Iordache T, Sian-Hülsmann J. Iron as the concert master in the pathogenic orchestra playing in sporadic Parkinson's disease. J Neural Transm (Vienna) 2021; 128:1577-1598. [PMID: 34636961 PMCID: PMC8507512 DOI: 10.1007/s00702-021-02414-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/29/2021] [Indexed: 02/07/2023]
Abstract
About 60 years ago, the discovery of a deficiency of dopamine in the nigro-striatal system led to a variety of symptomatic therapeutic strategies to supplement dopamine and to substantially improve the quality of life of patients with Parkinson's disease (PD). Since these seminal developments, neuropathological, neurochemical, molecular biological and genetic discoveries contributed to elucidate the pathology of PD. Oxidative stress, the consequences of reactive oxidative species, reduced antioxidative capacity including loss of glutathione, excitotoxicity, mitochondrial dysfunction, proteasomal dysfunction, apoptosis, lysosomal dysfunction, autophagy, suggested to be causal for ɑ-synuclein fibril formation and aggregation and contributing to neuroinflammation and neural cell death underlying this devastating disorder. However, there are no final conclusions about the triggered pathological mechanism(s) and the follow-up of pathological dysfunctions. Nevertheless, it is a fact, that iron, a major component of oxidative reactions, as well as neuromelanin, the major intraneuronal chelator of iron, undergo an age-dependent increase. And ageing is a major risk factor for PD. Iron is significantly increased in the substantia nigra pars compacta (SNpc) of PD. Reasons for this finding include disturbances in iron-related import and export mechanisms across the blood-brain barrier (BBB), localized opening of the BBB at the nigro-striatal tract including brain vessel pathology. Whether this pathology is of primary or secondary importance is not known. We assume that there is a better fit to the top-down hypotheses and pathogens entering the brain via the olfactory system, then to the bottom-up (gut-brain) hypothesis of PD pathology. Triggers for the bottom-up, the dual-hit and the top-down pathologies include chemicals, viruses and bacteria. If so, hepcidin, a regulator of iron absorption and its distribution into tissues, is suggested to play a major role in the pathogenesis of iron dyshomeostasis and risk for initiating and progressing ɑ-synuclein pathology. The role of glial components to the pathology of PD is still unknown. However, the dramatic loss of glutathione (GSH), which is mainly synthesized in glia, suggests dysfunction of this process, or GSH uptake into neurons. Loss of GSH and increase in SNpc iron concentration have been suggested to be early, may be even pre-symptomatic processes in the pathology of PD, despite the fact that they are progression factors. The role of glial ferritin isoforms has not been studied so far in detail in human post-mortem brain tissue and a close insight into their role in PD is called upon. In conclusion, "iron" is a major player in the pathology of PD. Selective chelation of excess iron at the site of the substantia nigra, where a dysfunction of the BBB is suggested, with peripherally acting iron chelators is suggested to contribute to the portfolio and therapeutic armamentarium of anti-Parkinson medications.
Collapse
Affiliation(s)
- P Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany. .,Department of Psychiatry, University of Southern Denmark, Odense, Denmark.
| | - C Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Wuerzburg, Germany
| | - S Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Wuerzburg, Germany
| | - T Iordache
- George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Târgu Mureș, Romania
| | - J Sian-Hülsmann
- Department of Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| |
Collapse
|
9
|
Schlichtmann BW, Hepker M, Palanisamy BN, John M, Anantharam V, Kanthasamy AG, Narasimhan B, Mallapragada SK. NANOTECHNOLOGY-MEDIATED THERAPEUTIC STRATEGIES AGAINST SYNUCLEINOPATHIES IN NEURODEGENERATIVE DISEASE. Curr Opin Chem Eng 2021; 31:100673. [PMID: 35419254 PMCID: PMC9004679 DOI: 10.1016/j.coche.2021.100673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Synucleinopathies are a subset of debilitating neurodegenerative disorders for which clinically approved therapeutic options to either halt or retard disease progression are currently unavailable. Multiple synergistic pathological mechanisms in combination with the characteristic misfolding of proteins are attributable to disease pathogenesis and progression. This complex interplay, as well as the difficult and multiscale nature of therapeutic delivery into the central nervous system, make finding effective treatments difficult. Nanocarriers (NCs) are a class of materials that can significantly improve therapeutic brain delivery and enable multifunctional therapies. In this review, an update on the known pathology of synucleinopathies is presented. Then, NC-enabled therapeutics designed to target the multiple mechanisms by combination therapies and multiscale targeting methods is reviewed. The implications of these strategies are synthesized and evaluated to suggest opportunities for the rational design of anti-neurodegenerative NC therapeutics.
Collapse
Affiliation(s)
- Benjamin W Schlichtmann
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011
- Nanovaccine Institute, Ames, Iowa 50011
| | - Monica Hepker
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011
| | | | - Manohar John
- PathoVacs, Incorporated, Ames, Iowa 50011
- Nanovaccine Institute, Ames, Iowa 50011
| | | | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011
- Nanovaccine Institute, Ames, Iowa 50011
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011
- Nanovaccine Institute, Ames, Iowa 50011
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011
- Nanovaccine Institute, Ames, Iowa 50011
| |
Collapse
|
10
|
Genetic Pathways Involved in the Pathogenesis of Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:195-208. [DOI: 10.1007/978-3-030-78787-5_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
11
|
Hanss Z, Larsen SB, Antony P, Mencke P, Massart F, Jarazo J, Schwamborn JC, Barbuti PA, Mellick GD, Krüger R. Mitochondrial and Clearance Impairment in p.D620N VPS35 Patient-Derived Neurons. Mov Disord 2020; 36:704-715. [PMID: 33142012 PMCID: PMC8048506 DOI: 10.1002/mds.28365] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Background VPS35 is part of the retromer complex and is responsible for the trafficking and recycling of proteins implicated in autophagy and lysosomal degradation, but also takes part in the degradation of mitochondrial proteins via mitochondria‐derived vesicles. The p.D620N mutation of VPS35 causes an autosomal‐dominant form of Parkinson's disease (PD), clinically representing typical PD. Objective Most of the studies on p.D620N VPS35 were performed on human tumor cell lines, rodent models overexpressing mutant VPS35, or in patient‐derived fibroblasts. Here, based on identified target proteins, we investigated the implication of mutant VPS35 in autophagy, lysosomal degradation, and mitochondrial function in induced pluripotent stem cell‐derived neurons from a patient harboring the p.D620N mutation. Methods We reprogrammed fibroblasts from a PD patient carrying the p.D620N mutation in the VPS35 gene and from two healthy donors in induced pluripotent stem cells. These were subsequently differentiated into neuronal precursor cells to finally generate midbrain dopaminergic neurons. Results We observed a decreased autophagic flux and lysosomal mass associated with an accumulation of α‐synuclein in patient‐derived neurons compared to controls. Moreover, patient‐derived neurons presented a mitochondrial dysfunction with decreased membrane potential, impaired mitochondrial respiration, and increased production of reactive oxygen species associated with a defect in mitochondrial quality control via mitophagy. Conclusion We describe for the first time the impact of the p.D620N VPS35 mutation on autophago‐lysosome pathway and mitochondrial function in stem cell‐derived neurons from an affected p.D620N carrier and define neuronal phenotypes for future pharmacological interventions. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Zoé Hanss
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Simone B Larsen
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Paul Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Pauline Mencke
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - François Massart
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Javier Jarazo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Peter A Barbuti
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - George D Mellick
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg.,Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg
| |
Collapse
|
12
|
Ibuchi K, Fukaya M, Shinohara T, Hara Y, Shiroshima T, Sugawara T, Sakagami H. The Vps52 subunit of the GARP and EARP complexes is a novel Arf6-interacting protein that negatively regulates neurite outgrowth of hippocampal neurons. Brain Res 2020; 1745:146905. [PMID: 32473257 DOI: 10.1016/j.brainres.2020.146905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/18/2020] [Accepted: 05/24/2020] [Indexed: 01/05/2023]
Abstract
ADP ribosylation factor 6 (Arf6) is a small GTP-binding protein implicated in neuronal morphogenesis through endosomal trafficking and actin remodeling. In this study, we identified Vps52, a core subunit of the Golgi-associated retrograde protein (GARP) and endosome-associated recycling protein (EARP) complexes, as a novel Arf6-binding protein by yeast two-hybrid screening. Vps52 interacted specifically with GTP-bound Arf6 among the Arf family. Immunohistochemical analyses of hippocampal pyramidal cells revealed that fine punctate immunolabeling for Vps52 was distributed throughout neuronal compartments, most densely in the cell body and dendritic shafts, and was largely associated with trans-Golgi network and vesicular endomembranes. In cultured hippocampal neurons, knockdown of Vps52 increased total length of axons and dendrites; these phenotypes were completely restored by co-expression of shRNA-resistant full-length Vps52. However, co-expression of a Vps52 mutant lacking the ability to interact with Arf6 restored only the Vps52-knockdown phenotype of the dendritic length. The present findings suggest that Vps52 is a novel Arf6-interacting protein that regulates neurite outgrowth in hippocampal neurons.
Collapse
Affiliation(s)
- Kanta Ibuchi
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Tetsuro Shinohara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Tomoko Shiroshima
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Takeyuki Sugawara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan.
| |
Collapse
|
13
|
Bono K, Hara-Miyauchi C, Sumi S, Oka H, Iguchi Y, Okano HJ. Endosomal dysfunction in iPSC-derived neural cells from Parkinson's disease patients with VPS35 D620N. Mol Brain 2020; 13:137. [PMID: 33032646 PMCID: PMC7542911 DOI: 10.1186/s13041-020-00675-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/22/2020] [Indexed: 11/17/2022] Open
Abstract
Mutations in the Vacuolar protein sorting 35 (VPS35) gene have been linked to familial Parkinson’s disease (PD), PARK17. VPS35 is a key component of the retromer complex, which plays a central role in endosomal trafficking. However, whether and how VPS35 deficiency or mutation contributes to PD pathogenesis remain unclear. Here, we analyzed human induced pluripotent stem cell (iPSC)-derived neurons from PD patients with the VPS35 D620N mutation and addressed relevant disease mechanisms. In the disease group, dopaminergic (DA) neurons underwent extensive apoptotic cell death. The movement of Rab5a- or Rab7a-positive endosomes was slower, and the endosome fission and fusion frequencies were lower in the PD group than in the healthy control group. Interestingly, vesicles positive for cation-independent mannose 6-phosphate receptor transported by retromers were abnormally localized in glial cells derived from patient iPSCs. Furthermore, we found α-synuclein accumulation in TH positive DA neurons. Our results demonstrate the induction of cell death, endosomal dysfunction and α -synuclein accumulation in neural cells of the PD group. PARK17 patient-derived iPSCs provide an excellent experimental tool for understanding the pathophysiology underlying PD.
Collapse
Affiliation(s)
- Keiko Bono
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.,Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Chikako Hara-Miyauchi
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Shunsuke Sumi
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hisayoshi Oka
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.,Department of Neurology, Daisan Hospital, The Jikei University School of Medicine, 4-11-1 Izumihoncho, Komae-shi, Tokyo, 201-8601, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
14
|
Farfel-Becker T, Roney JC, Cheng XT, Li S, Cuddy SR, Sheng ZH. Neuronal Soma-Derived Degradative Lysosomes Are Continuously Delivered to Distal Axons to Maintain Local Degradation Capacity. Cell Rep 2020; 28:51-64.e4. [PMID: 31269450 PMCID: PMC6696943 DOI: 10.1016/j.celrep.2019.06.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/12/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Neurons face the challenge of maintaining cellular homeostasis through lysosomal degradation. While enzymatically active degradative lysosomes are enriched in the soma, their axonal trafficking and positioning and impact on axonal physiology remain elusive. Here, we characterized axon-targeted delivery of degradative lysosomes by applying fluorescent probes that selectively label active forms of lysosomal cathepsins D, B, L, and GCase. By time-lapse imaging of cortical neurons in microfluidic devices and standard dishes, we reveal that soma-derived degradative lysosomes rapidly influx into distal axons and target to autophagosomes and Parkinson disease-related α-synuclein cargos for local degradation. Impairing lysosome axonal delivery induces an aberrant accumulation of autophagosomes and α-synuclein cargos in distal axons. Our study demonstrates that the axon is an active compartment for local degradation and reveals fundamental aspects of axonal lysosomal delivery and maintenance. Our work establishes a foundation for investigations into axonal lysosome trafficking and functionality in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tamar Farfel-Becker
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Joseph C Roney
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Xiu-Tang Cheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Sunan Li
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Sean R Cuddy
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
15
|
Marchetti B, Leggio L, L’Episcopo F, Vivarelli S, Tirolo C, Paternò G, Giachino C, Caniglia S, Serapide MF, Iraci N. Glia-Derived Extracellular Vesicles in Parkinson's Disease. J Clin Med 2020; 9:jcm9061941. [PMID: 32575923 PMCID: PMC7356371 DOI: 10.3390/jcm9061941] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
Glial cells are fundamental players in the central nervous system (CNS) development and homeostasis, both in health and disease states. In Parkinson’s disease (PD), a dysfunctional glia-neuron crosstalk represents a common final pathway contributing to the chronic and progressive death of dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNpc). Notably, glial cells communicating with each other by an array of molecules, can acquire a “beneficial” or “destructive” phenotype, thereby enhancing neuronal death/vulnerability and/or exerting critical neuroprotective and neuroreparative functions, with mechanisms that are actively investigated. An important way of delivering messenger molecules within this glia-neuron cross-talk consists in the secretion of extracellular vesicles (EVs). EVs are nano-sized membranous particles able to convey a wide range of molecular cargoes in a controlled way, depending on the specific donor cell and the microenvironmental milieu. Given the dual role of glia in PD, glia-derived EVs may deliver molecules carrying various messages for the vulnerable/dysfunctional DAergic neurons. Here, we summarize the state-of-the-art of glial-neuron interactions and glia-derived EVs in PD. Also, EVs have the ability to cross the blood brain barrier (BBB), thus acting both within the CNS and outside, in the periphery. In these regards, this review discloses the emerging applications of EVs, with a special focus on glia-derived EVs as potential carriers of new biomarkers and nanotherapeutics for PD.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy; (L.L.); (S.V.); (G.P.); (M.F.S.)
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
- Correspondence: (B.M.); (N.I.)
| | - Loredana Leggio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy; (L.L.); (S.V.); (G.P.); (M.F.S.)
| | - Francesca L’Episcopo
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy; (L.L.); (S.V.); (G.P.); (M.F.S.)
| | - Cataldo Tirolo
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy; (L.L.); (S.V.); (G.P.); (M.F.S.)
| | - Carmela Giachino
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Salvatore Caniglia
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Maria Francesca Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy; (L.L.); (S.V.); (G.P.); (M.F.S.)
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy; (L.L.); (S.V.); (G.P.); (M.F.S.)
- Correspondence: (B.M.); (N.I.)
| |
Collapse
|
16
|
Picca A, Guerra F, Calvani R, Coelho-Junior HJ, Bossola M, Landi F, Bernabei R, Bucci C, Marzetti E. Generation and Release of Mitochondrial-Derived Vesicles in Health, Aging and Disease. J Clin Med 2020; 9:jcm9051440. [PMID: 32408624 PMCID: PMC7290979 DOI: 10.3390/jcm9051440] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are intracellular organelles involved in a myriad of activities. To safeguard their vital functions, mitochondrial quality control (MQC) systems are in place to support organelle plasticity as well as physical and functional connections with other cellular compartments. In particular, mitochondrial interactions with the endosomal compartment support the shuttle of ions and metabolites across organelles, while those with lysosomes ensure the recycling of obsolete materials. The extrusion of mitochondrial components via the generation and release of mitochondrial-derived vesicles (MDVs) has recently been described. MDV trafficking is now included among MQC pathways, possibly operating via mitochondrial-lysosomal contacts. Since mitochondrial dysfunction is acknowledged as a hallmark of aging and a major pathogenic factor of multiple age-associated conditions, the analysis of MDVs and, more generally, of extracellular vesicles (EVs) is recognized as a valuable research tool. The dissection of EV trafficking may help unravel new pathophysiological pathways of aging and diseases as well as novel biomarkers to be used in research and clinical settings. Here, we discuss (1) MQC pathways with a focus on mitophagy and MDV generation; (2) changes of MQC pathways during aging and their contribution to inflamm-aging and progeroid conditions; and (3) the relevance of MQC failure to several disorders, including neurodegenerative conditions (i.e., Parkinson's disease, Alzheimer's disease) and cardiovascular disease.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (M.B.); (F.L.); (R.B.); (E.M.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, 73100 Lecce, Italy;
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (M.B.); (F.L.); (R.B.); (E.M.)
- Correspondence: (R.C.); (C.B.); Tel.: +39-06-3015-5559 (R.C.); +39-0832-29-8900 (C.B.); Fax: +39-06-305-1911 (R.C.); +39-0832-29-8941 (C.B.)
| | - Hélio José Coelho-Junior
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Maurizio Bossola
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (M.B.); (F.L.); (R.B.); (E.M.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (M.B.); (F.L.); (R.B.); (E.M.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Roberto Bernabei
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (M.B.); (F.L.); (R.B.); (E.M.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, 73100 Lecce, Italy;
- Correspondence: (R.C.); (C.B.); Tel.: +39-06-3015-5559 (R.C.); +39-0832-29-8900 (C.B.); Fax: +39-06-305-1911 (R.C.); +39-0832-29-8941 (C.B.)
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (M.B.); (F.L.); (R.B.); (E.M.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
17
|
Hopfner F, Mueller SH, Szymczak S, Junge O, Tittmann L, May S, Lohmann K, Grallert H, Lieb W, Strauch K, Müller-Nurasyid M, Berger K, Schormair B, Winkelmann J, Mollenhauer B, Trenkwalder C, Maetzler W, Berg D, Kasten M, Klein C, Höglinger GU, Gasser T, Deuschl G, Franke A, Krawczak M, Dempfle A, Kuhlenbäumer G. Rare Variants in Specific Lysosomal Genes Are Associated With Parkinson's Disease. Mov Disord 2020; 35:1245-1248. [PMID: 32267580 DOI: 10.1002/mds.28037] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Impaired lysosomal degradation of α-synuclein and other cellular constituents may play an important role in Parkinson's disease (PD). Rare genetic variants in the glucocerebrosidase (GBA) gene were consistently associated with PD. Here we examine the association between rare variants in lysosomal candidate genes and PD. METHODS We investigated the association between PD and rare genetic variants in 23 lysosomal candidate genes in 4096 patients with PD and an equal number of controls using pooled targeted next-generation DNA sequencing. Genewise association of rare variants in cases or controls was analyzed using the optimized sequence kernel association test with Bonferroni correction for the 23 tested genes. RESULTS We confirm the association of rare variants in GBA with PD and report novel associations for rare variants in ATP13A2, LAMP1, TMEM175, and VPS13C. CONCLUSION Rare variants in selected lysosomal genes, first and foremost GBA, are associated with PD. Rare variants in ATP13A2 and VPC13C previously linked to monogenic PD and more common variants in TMEM175 and VPS13C previously linked to sporadic PD in genome-wide association studies are associated with PD. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Franziska Hopfner
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Stefanie H Mueller
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany.,Institute of Health Informatics, University College London, London, United Kingdom
| | - Silke Szymczak
- Institute of Medical Informatics and Statistics, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Olaf Junge
- Institute of Medical Informatics and Statistics, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lukas Tittmann
- Institute of Epidemiology, University of Kiel, Kiel, Germany
| | - Sandra May
- Institute of Clinical Molecular Biology, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Harald Grallert
- Institute of Epidemiology II, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.,Research Unit of Molecular Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology, University of Kiel, Kiel, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.,Department of Medicine I, Ludwig-Maximilians-Universität, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Barbara Schormair
- Institute of Neurogenomics, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, Neuherberg, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, Neuherberg, Germany.,Institute of Human Genetics, Faculty of Medicine, Technical University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), München, Deutschland
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik Kassel, Kassel, Germany.,Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Claudia Trenkwalder
- Clinic for Neurosurgery, University Medical Centre, Georg August University Göttingen, Göttingen, Germany.,Paracelsus-Elena Hospital, Kassel, Germany
| | - Walter Maetzler
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Daniela Berg
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Günter U Höglinger
- Technical University of Munich, School of Medicine, Department of Neurology, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research and German Center for Neurodegenerative Diseases, University Clinic Tuebingen, Tuebingen, Germany
| | - Günther Deuschl
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - André Franke
- Institute of Clinical Molecular Biology, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Gregor Kuhlenbäumer
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
18
|
Tatullo M, Marrelli B, Zullo MJ, Codispoti B, Paduano F, Benincasa C, Fortunato F, Scacco S, Zavan B, Cocco T. Exosomes from Human Periapical Cyst-MSCs: Theranostic Application in Parkinson's Disease. Int J Med Sci 2020; 17:657-663. [PMID: 32210716 PMCID: PMC7085217 DOI: 10.7150/ijms.41515] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
The scientific community continuously strives to get new disease models, to discover early markers or novel therapeutic approaches, improving the diagnosis and prognosis of several human pathologies. Parkinson's Disease (PD) is characterized by a long asymptomatic phase, characterized by a selective loss of dopaminergic neurons. Recently, the human Periapical Cyst-Mesenchymal Stem Cells (hPCy-MSCs) have been differentiated in functional dopaminergic neurons: such oral-derived MSCs and the hPCy-MSCs-derived exosomes may represent a strategic and useful in vitro study-model, as well as intriguing therapeutic carriers. Circadian rhythm (CR) alteration variously impacts on PD pathways: an interesting research target is represented by the analysis of the exosomes released by dopaminergic neurons, derived from neural-differentiated hPCy-MSCs, after having reproduced in-vitro PD-like conditions. This review aims to describe the crosstalk among some aspects of circadian rhythm related to the onset of PD and the exosomes released by cells of PD patients. More in detail: the first part of this article will describe the main characteristics of circadian rhythm and the involvement of the exosomes found to be effective in the pathogenesis of PD. Finally, the authors will suggest how those exosomes derived from dopaminergic neurons, obtained by oral-derived stem cells (hPCy-MSCs) may represent a smart model for the in vitro research on PD, to find new biomarkers, to test new drugs or, fatally, to find new pathways applicable in future therapeutic approaches.
Collapse
Affiliation(s)
- Marco Tatullo
- Marrelli Health - Tecnologica Research Institute, Biomedical Section, Street E. Fermi, Crotone, Italy
- Department of Therapeutic Dentistry, Sechenov University Russia, Moscow, Russia
| | - Benedetta Marrelli
- Marrelli Health - Tecnologica Research Institute, Biomedical Section, Street E. Fermi, Crotone, Italy
- Department of Therapeutic Dentistry, Sechenov University Russia, Moscow, Russia
| | - Maria Josephine Zullo
- Department of Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Bruna Codispoti
- Marrelli Health - Tecnologica Research Institute, Biomedical Section, Street E. Fermi, Crotone, Italy
| | - Francesco Paduano
- Marrelli Health - Tecnologica Research Institute, Biomedical Section, Street E. Fermi, Crotone, Italy
| | - Caterina Benincasa
- Marrelli Health - Tecnologica Research Institute, Biomedical Section, Street E. Fermi, Crotone, Italy
| | - Francesco Fortunato
- Department of Neurological Sciences, University of Catanzaro “Magna Graecia”, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, Italy
| | - Barbara Zavan
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Tiziana Cocco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, Italy
| |
Collapse
|
19
|
Pang SYY, Ho PWL, Liu HF, Leung CT, Li L, Chang EES, Ramsden DB, Ho SL. The interplay of aging, genetics and environmental factors in the pathogenesis of Parkinson's disease. Transl Neurodegener 2019; 8:23. [PMID: 31428316 PMCID: PMC6696688 DOI: 10.1186/s40035-019-0165-9] [Citation(s) in RCA: 241] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by dopaminergic neuronal loss in the substantia nigra pars compacta and intracellular inclusions called Lewy bodies (LB). During the course of disease, misfolded α-synuclein, the major constituent of LB, spreads to different regions of the brain in a prion-like fashion, giving rise to successive non-motor and motor symptoms. Etiology is likely multifactorial, and involves interplay among aging, genetic susceptibility and environmental factors. MAIN BODY The prevalence of PD rises exponentially with age, and aging is associated with impairment of cellular pathways which increases susceptibility of dopaminergic neurons to cell death. However, the majority of those over the age of 80 do not have PD, thus other factors in addition to aging are needed to cause disease. Discovery of neurotoxins which can result in parkinsonism led to efforts in identifying environmental factors which may influence PD risk. Nevertheless, the causality of most environmental factors is not conclusively established, and alternative explanations such as reverse causality and recall bias cannot be excluded. The lack of geographic clusters and conjugal cases also go against environmental toxins as a major cause of PD. Rare mutations as well as common variants in genes such as SNCA, LRRK2 and GBA are associated with risk of PD, but Mendelian causes collectively only account for 5% of PD and common polymorphisms are associated with small increase in PD risk. Heritability of PD has been estimated to be around 30%. Thus, aging, genetics and environmental factors each alone is rarely sufficient to cause PD for most patients. CONCLUSION PD is a multifactorial disorder involving interplay of aging, genetics and environmental factors. This has implications on the development of appropriate animal models of PD which take all these factors into account. Common converging pathways likely include mitochondrial dysfunction, impaired autophagy, oxidative stress and neuroinflammation, which are associated with the accumulation and spread of misfolded α-synuclein and neurodegeneration. Understanding the mechanisms involved in the initiation and progression of PD may lead to potential therapeutic targets to prevent PD or modify its course.
Collapse
Affiliation(s)
- Shirley Yin-Yu Pang
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| | - Philip Wing-Lok Ho
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| | - Hui-Fang Liu
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| | - Chi-Ting Leung
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| | - Lingfei Li
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| | - Eunice Eun Seo Chang
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| | - David Boyer Ramsden
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Shu-Leong Ho
- Division of Neurology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People’s Republic of China
| |
Collapse
|
20
|
Yabuno Y, Uchihashi T, Sasakura T, Shimizu H, Naito Y, Fukushima K, Ota K, Kogo M, Nojima H, Yabuta N. Clathrin heavy chain phosphorylated at T606 plays a role in proper cell division. Cell Cycle 2019; 18:1976-1994. [PMID: 31272276 PMCID: PMC6681784 DOI: 10.1080/15384101.2019.1637201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/13/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022] Open
Abstract
Clathrin regulates mitotic progression, in addition to membrane trafficking. However, the detailed regulatory mechanisms of clathrin during mitosis remain elusive. Here, we demonstrate novel regulation of clathrin during mitotic phase of the cell cycle. Clathrin heavy chain (CHC) was phosphorylated at T606 by its association partner cyclin G-associated kinase (GAK). This phosphorylation was required for proper cell proliferation and tumor growth of cells implanted into nude mice. Immunofluorescence analysis showed that the localization of CHC-pT606 signals changed during mitosis. CHC-pT606 signals localized in the nucleus and at the centrosome during interphase, whereas CHC signals were mostly cytoplasmic. Co-immunoprecipitation suggested that CHC formed a complex with GAK and polo-like kinase 1 (PLK1). Depletion of GAK using siRNA induced metaphase arrest and aberrant localization of CHC-pT606, which abolished Kiz-pT379 (as a phosphorylation target of PLK1) signals on chromatin at metaphase. Taken together, we propose that the GAK_CHC-pT606_PLK1_Kiz-pT379 axis plays a role in proliferation of cancer cells.
Collapse
Affiliation(s)
- Yusuke Yabuno
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Toshihiro Uchihashi
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Towa Sasakura
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hiroyuki Shimizu
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoko Naito
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Division of Cancer Cell Regulation, Aichi Cancer Center Research Institute, Aichi, Japan
| | - Kohshiro Fukushima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kaori Ota
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Mikihiko Kogo
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroshi Nojima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Norikazu Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
21
|
Yan K, Zhang W, Han X, Chang F, Liu Y. Inhibitory role of peroxiredoxin 2 in LRRK2 kinase activity induced cellular pathogenesis. J Biomed Res 2019; 34:103-113. [PMID: 32305964 DOI: 10.7555/jbr.33.20190090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Parkinson's disease (PD) is a major neurodegenerative disease. One of the known genetic contributors to PD pathogenesis is leucine-rich repeat kinase 2 (LRRK2) whose mutations with elevated kinase activity could lead to both familial and sporadic PD. However, how the pathogenic kinase activity of LRRK2 is regulated remains largely unclear. Here we report that peroxiredoxin 2 (Prx2) was identified as a novel interacting protein to LRRK2 with preferential expression in dopaminergic neurons over other Prx proteins. We also confirmed that Prx2 interacted with LRRK2 through its COR domain and its overexpression significantly decreased the kinase activity of mutant LRRK2. Functionally, overexpressed Prx2 rescued the transfected cells from LRRK2 mutant induced apoptotic processes. Importantly, overexpressed Prx2 reversed the altered subcellular distribution of cation-independent mannose 6-phosphate receptor (CI-M6PR) induced by PD-mutant LRRK2. Our results suggest that, by interacting with LRRK2, Prx2 may play an inhibitory role in the LRRK2 mediated cellular toxicity in PD by inhibiting its kinase activity.
Collapse
Affiliation(s)
- Kang Yan
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wenfeng Zhang
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xu Han
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Fei Chang
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yongjian Liu
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
22
|
Harischandra DS, Ghaisas S, Zenitsky G, Jin H, Kanthasamy A, Anantharam V, Kanthasamy AG. Manganese-Induced Neurotoxicity: New Insights Into the Triad of Protein Misfolding, Mitochondrial Impairment, and Neuroinflammation. Front Neurosci 2019; 13:654. [PMID: 31293375 PMCID: PMC6606738 DOI: 10.3389/fnins.2019.00654] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
Abstract
Occupational or environmental exposure to manganese (Mn) can lead to the development of "Manganism," a neurological condition showing certain motor symptoms similar to Parkinson's disease (PD). Like PD, Mn toxicity is seen in the central nervous system mainly affecting nigrostriatal neuronal circuitry and subsequent behavioral and motor impairments. Since the first report of Mn-induced toxicity in 1837, various experimental and epidemiological studies have been conducted to understand this disorder. While early investigations focused on the impact of high concentrations of Mn on the mitochondria and subsequent oxidative stress, current studies have attempted to elucidate the cellular and molecular pathways involved in Mn toxicity. In fact, recent reports suggest the involvement of Mn in the misfolding of proteins such as α-synuclein and amyloid, thus providing credence to the theory that environmental exposure to toxicants can either initiate or propagate neurodegenerative processes by interfering with disease-specific proteins. Besides manganism and PD, Mn has also been implicated in other neurological diseases such as Huntington's and prion diseases. While many reviews have focused on Mn homeostasis, the aim of this review is to concisely synthesize what we know about its effect primarily on the nervous system with respect to its role in protein misfolding, mitochondrial dysfunction, and consequently, neuroinflammation and neurodegeneration. Based on the current evidence, we propose a 'Mn Mechanistic Neurotoxic Triad' comprising (1) mitochondrial dysfunction and oxidative stress, (2) protein trafficking and misfolding, and (3) neuroinflammation.
Collapse
Affiliation(s)
- Dilshan S Harischandra
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Shivani Ghaisas
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Gary Zenitsky
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Laboratory, Iowa State University, Ames, IA, United States
| |
Collapse
|
23
|
Riederer P, Berg D, Casadei N, Cheng F, Classen J, Dresel C, Jost W, Krüger R, Müller T, Reichmann H, Rieß O, Storch A, Strobel S, van Eimeren T, Völker HU, Winkler J, Winklhofer KF, Wüllner U, Zunke F, Monoranu CM. α-Synuclein in Parkinson's disease: causal or bystander? J Neural Transm (Vienna) 2019; 126:815-840. [PMID: 31240402 DOI: 10.1007/s00702-019-02025-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) comprises a spectrum of disorders with differing subtypes, the vast majority of which share Lewy bodies (LB) as a characteristic pathological hallmark. The process(es) underlying LB generation and its causal trigger molecules are not yet fully understood. α-Synuclein (α-syn) is a major component of LB and SNCA gene missense mutations or duplications/triplications are causal for rare hereditary forms of PD. As typical sporadic PD is associated with LB pathology, a factor of major importance is the study of the α-syn protein and its pathology. α-Syn pathology is, however, also evident in multiple system atrophy (MSA) and Lewy body disease (LBD), making it non-specific for PD. In addition, there is an overlap of these α-synucleinopathies with other protein-misfolding diseases. It has been proven that α-syn, phosphorylated tau protein (pτ), amyloid beta (Aβ) and other proteins show synergistic effects in the underlying pathogenic mechanisms. Multiple cell death mechanisms can induce pathological protein-cascades, but this can also be a reverse process. This holds true for the early phases of the disease process and especially for the progression of PD. In conclusion, while rare SNCA gene mutations are causal for a minority of familial PD patients, in sporadic PD (where common SNCA polymorphisms are the most consistent genetic risk factor across populations worldwide, accounting for 95% of PD patients) α-syn pathology is an important feature. Conversely, with regard to the etiopathogenesis of α-synucleinopathies PD, MSA and LBD, α-syn is rather a bystander contributing to multiple neurodegenerative processes, which overlap in their composition and individual strength. Therapeutic developments aiming to impact on α-syn pathology should take this fact into consideration.
Collapse
Affiliation(s)
- Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany. .,Department of Psychiatry, University of South Denmark, Odense, Denmark.
| | - Daniela Berg
- Department of Neurology, UKHS, Christian-Albrechts-Universität, Campus Kiel, Kiel, Germany
| | - Nicolas Casadei
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Fubo Cheng
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Joseph Classen
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Christian Dresel
- Department of Neurology, Center for Movement Disorders, Neuroimaging Center Mainz, Clinical Neurophysiology, Forschungszentrum Translationale Neurowissenschaften (FTN), Rhein-Main-Neuronetz, Mainz, Germany
| | | | - Rejko Krüger
- Clinical and Experimental Neuroscience, LCSB (Luxembourg Centre for Systems, Biomedicine), University of Luxembourg, Esch-sur-Alzette and Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg.,National Center for Excellence in Research, Parkinson's disease (NCER-PD), Parkinson Research Clinic, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Thomas Müller
- Department of Neurology, Alexianer St. Joseph Berlin-Weißensee, Berlin, Germany
| | - Heinz Reichmann
- Department of Neurology, University of Dresden, Dresden, Germany
| | - Olaf Rieß
- Institute of Medical Genetics and Applied Genomics, Tübingen, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany.,German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Sabrina Strobel
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Thilo van Eimeren
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | | | - Jürgen Winkler
- Department Kopfkliniken, Molekulare Neurologie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Konstanze F Winklhofer
- Institute of Biochemistry and Pathobiochemistry, Ruhr-Universität Bochum, Bochum, Germany
| | - Ullrich Wüllner
- Department of Neurology, University of Bonn, German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
| | - Friederike Zunke
- Department of Biochemistry, Medical Faculty, University of Kiel, Kiel, Germany
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Lee JC, Yang JS, Moon MH. Simultaneous Relative Quantification of Various Polyglycerophospholipids with Isotope-Labeled Methylation by Nanoflow Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectrometry. Anal Chem 2019; 91:6716-6723. [PMID: 31008580 DOI: 10.1021/acs.analchem.9b00800] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Herein, we introduce a comprehensive analytical method for the separation and relative quantification of polyglycerophospholipids (PGPLs), including phosphatidylglycerol (PG), bis(monoacylglycero)phosphate (BMP), bis(diacylglycero)phosphate (BDP), Hemi BDP, cardiolipin (CL), monolysocardiolipin (MLCL), and dilysocardiolipin (DLCL), using isotope-labeled methylation (ILM) with nanoflow ultrahigh performance liquid chromatography-electrospray ionization-tandem mass spectrometry (nUHPLC-ESI-MS/MS). Abnormal levels of BMP and CL have been associated with the pathology of lysosomal storage and neurodegenerative diseases. Thus, simultaneous analysis of all PGPLs is important to understand the mechanisms and pathologies of such diseases. In this study, improved separation and MS detection of PGPLs, including their regioisomers, was achieved by the methylation of PGPL. ILM-based relative quantification was applied to lipid extracts from a dopaminergic cell line (SH-SY5Y) treated with drugs commonly used for Parkinson's disease (PD), resulting in the identification of 229 unique PGPLs, including 121 CLs, 71 MLCLs, and 16 Hemi BDP species. The drug treatment induced significant increases in the amount of CLs containing polyunsaturated fatty acyl chains, including 20:4 and 22:6, as well as decreased levels of BMP, Hemi BDP, and BDP species, demonstrating the feasibility of using ILM for the comprehensive and high-speed relative quantification of PGPLs.
Collapse
Affiliation(s)
- Jong Cheol Lee
- Department of Chemistry , Yonsei University , 50 Yonsei-ro , Seodaemun-gu, Seoul 03722 , Korea
| | - Joon Seon Yang
- Department of Chemistry , Yonsei University , 50 Yonsei-ro , Seodaemun-gu, Seoul 03722 , Korea
| | - Myeong Hee Moon
- Department of Chemistry , Yonsei University , 50 Yonsei-ro , Seodaemun-gu, Seoul 03722 , Korea
| |
Collapse
|
25
|
Koh JY, Kim HN, Hwang JJ, Kim YH, Park SE. Lysosomal dysfunction in proteinopathic neurodegenerative disorders: possible therapeutic roles of cAMP and zinc. Mol Brain 2019; 12:18. [PMID: 30866990 PMCID: PMC6417073 DOI: 10.1186/s13041-019-0439-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
A number of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, share intra- and/or extracellular deposition of protein aggregates as a common core pathology. While the species of accumulating proteins are distinct in each disease, an increasing body of evidence indicates that defects in the protein clearance system play a crucial role in the gradual accumulation of protein aggregates. Among protein degradation systems, the endosome-autophagosome-lysosome pathway (EALP) is the main degradation machinery, especially for large protein aggregates. Lysosomal dysfunction or defects in fusion with vesicles containing cargo are commonly observed abnormalities in proteinopathic neurodegenerative diseases. In this review, we discuss the available evidence for a mechanistic connection between components of the EALP-especially lysosomes-and neurodegenerative diseases. We also focus on lysosomal pH regulation and its significance in maintaining flux through the EALP. Finally, we suggest that raising cAMP and free zinc levels in brain cells may be beneficial in normalizing lysosomal pH and EALP flux.
Collapse
Affiliation(s)
- Jae-Young Koh
- Department of Neurology, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Ha Na Kim
- Neural Injury Lab, Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Jung Jin Hwang
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yang-Hee Kim
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, South Korea
| | - Sang Eun Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| |
Collapse
|
26
|
Bandres-Ciga S, Saez-Atienzar S, Bonet-Ponce L, Billingsley K, Vitale D, Blauwendraat C, Gibbs JR, Pihlstrøm L, Gan-Or Z, Cookson MR, Nalls MA, Singleton AB. The endocytic membrane trafficking pathway plays a major role in the risk of Parkinson's disease. Mov Disord 2019; 34:460-468. [PMID: 30675927 DOI: 10.1002/mds.27614] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/05/2018] [Accepted: 12/23/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND PD is a complex polygenic disorder. In recent years, several genes from the endocytic membrane-trafficking pathway have been suggested to contribute to disease etiology. However, a systematic analysis of pathway-specific genetic risk factors is yet to be performed. OBJECTIVES To comprehensively study the role of the endocytic membrane-trafficking pathway in the risk of PD. METHODS Linkage disequilibrium score regression was used to estimate PD heritability explained by 252 genes involved in the endocytic membrane-trafficking pathway including genome-wide association studies data from 18,869 cases and 22,452 controls. We used pathway-specific single-nucleotide polymorphisms to construct a polygenic risk score reflecting the cumulative risk of common variants. To prioritize genes for follow-up functional studies, summary-data based Mendelian randomization analyses were applied to explore possible functional genomic associations with expression or methylation quantitative trait loci. RESULTS The heritability estimate attributed to endocytic membrane-trafficking pathway was 3.58% (standard error = 1.17). Excluding previously nominated PD endocytic membrane-trafficking pathway genes, the missing heritability was 2.21% (standard error = 0.42). Random heritability simulations were estimated to be 1.44% (standard deviation = 0.54), indicating that the unbiased total heritability explained by the endocytic membrane-trafficking pathway was 2.14%. Polygenic risk score based on endocytic membrane-trafficking pathway showed a 1.25 times increase of PD risk per standard deviation of genetic risk. Finally, Mendelian randomization identified 11 endocytic membrane-trafficking pathway genes showing functional consequence associated to PD risk. CONCLUSIONS We provide compelling genetic evidence that the endocytic membrane-trafficking pathway plays a relevant role in disease etiology. Further research on this pathway is warranted given that critical effort should be made to identify potential avenues within this biological process suitable for therapeutic interventions. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sara Bandres-Ciga
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Sara Saez-Atienzar
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Luis Bonet-Ponce
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Kimberley Billingsley
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA.,Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.,Department of Pathophysiology, University of Tartu, Tartu, Estonia
| | - Dan Vitale
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Jesse Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Ziv Gan-Or
- Department of Neurology and Neurosurgery, Department of Human Genetics, McGill University, Montréal, Quebec, Canada.,Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | | | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Mike A Nalls
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA.,Data Tecnica International, Glen Echo, Maryland, USA
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Yoshida S, Hasegawa T, Suzuki M, Sugeno N, Kobayashi J, Ueyama M, Fukuda M, Ido-Fujibayashi A, Sekiguchi K, Ezura M, Kikuchi A, Baba T, Takeda A, Mochizuki H, Nagai Y, Aoki M. Parkinson's disease-linked DNAJC13 mutation aggravates alpha-synuclein-induced neurotoxicity through perturbation of endosomal trafficking. Hum Mol Genet 2019; 27:823-836. [PMID: 29309590 DOI: 10.1093/hmg/ddy003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 12/29/2017] [Indexed: 01/23/2023] Open
Abstract
Mutations in DNAJC13 gene have been linked to familial form of Parkinson's disease (PD) with Lewy pathology. DNAJC13 is an endosome-related protein and believed to regulate endosomal membrane trafficking. However, the mechanistic link between DNAJC13 mutation and α-synuclein (αSYN) pathology toward neurodegeneration remains poorly understood. In this study, we showed that PD-linked N855S-mutant DNAJC13 caused αSYN accumulation in the endosomal compartment, presumably due to defective cargo trafficking from the early endosome to the late and/or recycling endosome. In vivo experiments using human αSYN transgenic flies showed that mutant DNAJC13 not only increased the amount of insoluble αSYN in fly head but also induced dopaminergic neurodegeneration, rough eye phenotype and age-dependent locomotor impairment. Together, these findings suggest that DNAJC13 mutation perturbs multi-directional endosomal trafficking, resulting in the aberrant endosomal retention of αSYN, which might predispose to the neurodegenerative process that leads to PD.
Collapse
Affiliation(s)
- Shun Yoshida
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Takafumi Hasegawa
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Mari Suzuki
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan.,Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Naoto Sugeno
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Junpei Kobayashi
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Morio Ueyama
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Akemi Ido-Fujibayashi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Michinori Ezura
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Akio Kikuchi
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Toru Baba
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai-Nishitaga Hospital, Sendai 982-8555, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshitaka Nagai
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masashi Aoki
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| |
Collapse
|
28
|
Lin AW, Gill KK, Castañeda MS, Matucci I, Eder N, Claxton S, Flynn H, Snijders AP, George R, Ultanir SK. Chemical genetic identification of GAK substrates reveals its role in regulating Na +/K +-ATPase. Life Sci Alliance 2018; 1:e201800118. [PMID: 30623173 PMCID: PMC6312924 DOI: 10.26508/lsa.201800118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/15/2022] Open
Abstract
Novel GAK phosphorylation targets are identified using chemical genetic methods. One of the substrates is the α subunit of the Na+/K+-ATPase, phosphorylation of which is necessary for its surface trafficking from endosomes. Conserved functions of NAK family kinases are described. Cyclin G–associated kinase (GAK) is a ubiquitous serine/threonine kinase that facilitates clathrin uncoating during vesicle trafficking. GAK phosphorylates a coat adaptor component, AP2M1, to help achieve this function. GAK is also implicated in Parkinson's disease through genome-wide association studies. However, GAK's role in mammalian neurons remains unclear, and insight may come from identification of further substrates. Employing a chemical genetics method, we show here that the sodium potassium pump (Na+/K+-ATPase) α-subunit Atp1a3 is a GAK target and that GAK regulates Na+/K+-ATPase trafficking to the plasma membrane. Whole-cell patch clamp recordings from CA1 pyramidal neurons in GAK conditional knockout mice show a larger change in resting membrane potential when exposed to the Na+/K+-ATPase blocker ouabain, indicating compromised Na+/K+-ATPase function in GAK knockouts. Our results suggest a modulatory role for GAK via phosphoregulation of substrates such as Atp1a3 during cargo trafficking.
Collapse
Affiliation(s)
- Amy W Lin
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Kalbinder K Gill
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | | | - Irene Matucci
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Noreen Eder
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom.,Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | - Suzanne Claxton
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| | - Helen Flynn
- Mass Spectrometry Platform, The Francis Crick Institute, London, United Kingdom
| | | | - Roger George
- Protein Purification Facility, The Francis Crick Institute, London, United Kingdom
| | - Sila K Ultanir
- Kinase and Brain Development Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
29
|
van der Lienden MJC, Gaspar P, Boot R, Aerts JMFG, van Eijk M. Glycoprotein Non-Metastatic Protein B: An Emerging Biomarker for Lysosomal Dysfunction in Macrophages. Int J Mol Sci 2018; 20:E66. [PMID: 30586924 PMCID: PMC6337583 DOI: 10.3390/ijms20010066] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Several diseases are caused by inherited defects in lysosomes, the so-called lysosomal storage disorders (LSDs). In some of these LSDs, tissue macrophages transform into prominent storage cells, as is the case in Gaucher disease. Here, macrophages become the characteristic Gaucher cells filled with lysosomes laden with glucosylceramide, because of their impaired enzymatic degradation. Biomarkers of Gaucher cells were actively searched, particularly after the development of costly therapies based on enzyme supplementation and substrate reduction. Proteins selectively expressed by storage macrophages and secreted into the circulation were identified, among which glycoprotein non-metastatic protein B (GPNMB). This review focusses on the emerging potential of GPNMB as a biomarker of stressed macrophages in LSDs as well as in acquired pathologies accompanied by an excessive lysosomal substrate load in macrophages.
Collapse
Affiliation(s)
| | - Paulo Gaspar
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Rolf Boot
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Johannes M F G Aerts
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Marco van Eijk
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
30
|
Domingo-Fernández D, Hoyt CT, Bobis-Álvarez C, Marín-Llaó J, Hofmann-Apitius M. ComPath: an ecosystem for exploring, analyzing, and curating mappings across pathway databases. NPJ Syst Biol Appl 2018; 5:3. [PMID: 30564458 PMCID: PMC6292919 DOI: 10.1038/s41540-018-0078-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 11/09/2022] Open
Abstract
Although pathways are widely used for the analysis and representation of biological systems, their lack of clear boundaries, their dispersion across numerous databases, and the lack of interoperability impedes the evaluation of the coverage, agreements, and discrepancies between them. Here, we present ComPath, an ecosystem that supports curation of pathway mappings between databases and fosters the exploration of pathway knowledge through several novel visualizations. We have curated mappings between three of the major pathway databases and present a case study focusing on Parkinson’s disease that illustrates how ComPath can generate new biological insights by identifying pathway modules, clusters, and cross-talks with these mappings. The ComPath source code and resources are available at https://github.com/ComPath and the web application can be accessed at https://compath.scai.fraunhofer.de/.
Collapse
Affiliation(s)
- Daniel Domingo-Fernández
- 1Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, 53754 Sankt Augustin, Germany.,2Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Charles Tapley Hoyt
- 1Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, 53754 Sankt Augustin, Germany.,2Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Carlos Bobis-Álvarez
- 3Faculty of Medicine and Health Sciences, University of Oviedo, 33006 Oviedo, Spain
| | - Josep Marín-Llaó
- 1Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, 53754 Sankt Augustin, Germany.,4Rovira i Virgili University, 43003 Tarragona, Spain
| | - Martin Hofmann-Apitius
- 1Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, 53754 Sankt Augustin, Germany.,2Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| |
Collapse
|
31
|
Kaur G, Gauthier SA, Perez-Gonzalez R, Pawlik M, Singh AB, Cosby B, Mohan PS, Smiley JF, Levy E. Cystatin C prevents neuronal loss and behavioral deficits via the endosomal pathway in a mouse model of down syndrome. Neurobiol Dis 2018; 120:165-173. [PMID: 30176349 DOI: 10.1016/j.nbd.2018.08.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/14/2018] [Accepted: 08/30/2018] [Indexed: 01/18/2023] Open
Abstract
Cystatin C (CysC) plays diverse protective roles under conditions of neuronal challenge. We investigated whether CysC protects from trisomy-induced pathologies in a mouse model of Down syndrome (DS), the most common cause of developmental cognitive and behavioral impairments in humans. We have previously shown that the segmental trisomy mouse model, Ts[Rb(12.1716)]2Cje (Ts2) has DS-like neuronal and behavioral deficiencies. The current study reveals that transgene-mediated low levels of human CysC overexpression has a preventive effect on numerous neuropathologies in the brains of Ts2 mice, including reducing early and late endosome enlargement in cortical neurons and decreasing loss of basal forebrain cholinergic neurons (BFCNs). Consistent with these cellular benefits, behavioral dysfunctions were also prevented, including deficits in nesting behavior and spatial memory. We determined that the CysC-induced neuroprotective mechanism involves activation of the phosphotidylinositol kinase (PI3K)/AKT pathway. Activating this pathway leads to enhanced clearance of accumulated endosomal substrates, protecting cells from DS-mediated dysfunctions in the endosomal system and, for BFCNs, from neurodegeneration. Our findings suggest that modulation of the PI3/AKT pathway offers novel therapeutic interventions for patients with DS.
Collapse
Affiliation(s)
| | | | | | - Monika Pawlik
- Nathan S. Kline Institute, Orangeburg, NY, USA 10962
| | | | | | | | - John F Smiley
- Nathan S. Kline Institute, Orangeburg, NY, USA 10962; Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA 10016
| | - Efrat Levy
- Nathan S. Kline Institute, Orangeburg, NY, USA 10962; Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA 10016; Department of Biochemistry and Molecular Pharmacology, NYU Langone School of Medicine, New York, NY, USA 10016; Neuroscience Institute, NYU Langone School of Medicine, New York, NY, USA 10016.
| |
Collapse
|
32
|
Tofaris GK, Buckley NJ. Convergent molecular defects underpin diverse neurodegenerative diseases. J Neurol Neurosurg Psychiatry 2018; 89:962-969. [PMID: 29459380 DOI: 10.1136/jnnp-2017-316988] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/24/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022]
Abstract
In our ageing population, neurodegenerative disorders carry an enormous personal, societal and economic burden. Although neurodegenerative diseases are often thought of as clinicopathological entities, increasing evidence suggests a considerable overlap in the molecular underpinnings of their pathogenesis. Such overlapping biological processes include the handling of misfolded proteins, defective organelle trafficking, RNA processing, synaptic health and neuroinflammation. Collectively but in different proportions, these biological processes in neurons or non-neuronal cells lead to regionally distinct patterns of neuronal vulnerability and progression of pathology that could explain the disease symptomology. With the advent of patient-derived cellular models and novel genetic manipulation tools, we are now able to interrogate this commonality despite the cellular complexity of the brain in order to develop novel therapeutic strategies to prevent or arrest neurodegeneration. Here, we describe broadly these concepts and their relevance across neurodegenerative diseases.
Collapse
Affiliation(s)
- George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Noel J Buckley
- Department of Psychiatry, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Tofaris GK. A Critical Assessment of Exosomes in the Pathogenesis and Stratification of Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2018; 7:569-576. [PMID: 28922170 PMCID: PMC5676982 DOI: 10.3233/jpd-171176] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Extracellular vesicles including exosomes are released by a variety of cell types including neurons and exhibit molecular profiles that reflect normal and disease states. As their content represents a snapshot of the intracellular milieu, they could be exploited as biomarkers of the otherwise inaccessible brain microenvironment. In addition they may contribute to the progression of neurodegenerative disorders by facilitating the spread of misfolded proteins at distant sites or activating immune cells. This review summarizes recent advances in the study of exosomes in Parkinson’s disease pathophysiology and their potential as disease biomarkers.
Collapse
Affiliation(s)
- George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
34
|
Abstract
Recently, a new form of autosomal recessive early-onset parkinsonism (PARK20), due to mutations in the gene encoding the phosphoinositide phosphatase, Synaptojanin 1 (Synj1), has been reported. Several genes responsible for hereditary forms of Parkinson’s disease are implicated in distinct steps of the endolysosomal pathway. However, the nature and the degree of endocytic membrane trafficking impairment in early-onset parkinsonism remains elusive. Here, we show that depletion of Synj1 causes drastic alterations of early endosomes, which become enlarged and more numerous, while it does not affect the morphology of late endosomes both in non-neuronal and neuronal cells. Moreover, Synj1 loss impairs the recycling of transferrin, while it does not alter the trafficking of the epidermal growth factor receptor. The ectopic expression of Synj1 restores the functions of early endosomes, and rescues these trafficking defects in depleted cells. Importantly, the same alterations of early endosomal compartments and trafficking defects occur in fibroblasts of PARK20 patients. Our data indicate that Synj1 plays a crucial role in regulating the homeostasis and functions of early endosomal compartments in different cell types, and highlight defective cellular pathways in PARK20. In addition, they strengthen the link between endosomal trafficking and Parkinson’s disease.
Collapse
|
35
|
Microarray Analysis of the Molecular Mechanism Involved in Parkinson's Disease. PARKINSONS DISEASE 2018; 2018:1590465. [PMID: 29686831 PMCID: PMC5852864 DOI: 10.1155/2018/1590465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/21/2017] [Accepted: 10/18/2017] [Indexed: 02/03/2023]
Abstract
Purpose This study aimed to investigate the underlying molecular mechanisms of Parkinson's disease (PD) by bioinformatics. Methods Using the microarray dataset GSE72267 from the Gene Expression Omnibus database, which included 40 blood samples from PD patients and 19 matched controls, differentially expressed genes (DEGs) were identified after data preprocessing, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Protein-protein interaction (PPI) network, microRNA- (miRNA-) target regulatory network, and transcription factor- (TF-) target regulatory networks were constructed. Results Of 819 DEGs obtained, 359 were upregulated and 460 were downregulated. Two GO terms, “rRNA processing” and “cytoplasm,” and two KEGG pathways, “metabolic pathways” and “TNF signaling pathway,” played roles in PD development. Intercellular adhesion molecule 1 (ICAM1) was the hub node in the PPI network; hsa-miR-7-5p, hsa-miR-433-3p, and hsa-miR-133b participated in PD pathogenesis. Six TFs, including zinc finger and BTB domain-containing 7A, ovo-like transcriptional repressor 1, GATA-binding protein 3, transcription factor dp-1, SMAD family member 1, and quiescin sulfhydryl oxidase 1, were related to PD. Conclusions “rRNA processing,” “cytoplasm,” “metabolic pathways,” and “TNF signaling pathway” were key pathways involved in PD. ICAM1, hsa-miR-7-5p, hsa-miR-433-3p, hsa-miR-133b, and the abovementioned six TFs might play important roles in PD development.
Collapse
|
36
|
Genetic analysis of TMEM230 in Japanese patients with familial Parkinson's disease. Parkinsonism Relat Disord 2018; 48:107-108. [DOI: 10.1016/j.parkreldis.2017.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 11/21/2022]
|
37
|
Shaw AE, Hughes J, Gu Q, Behdenna A, Singer JB, Dennis T, Orton RJ, Varela M, Gifford RJ, Wilson SJ, Palmarini M. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol 2017; 15:e2004086. [PMID: 29253856 PMCID: PMC5747502 DOI: 10.1371/journal.pbio.2004086] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/29/2017] [Accepted: 11/22/2017] [Indexed: 12/31/2022] Open
Abstract
The host innate immune response mediated by type I interferon (IFN) and the resulting up-regulation of hundreds of interferon-stimulated genes (ISGs) provide an immediate barrier to virus infection. Studies of the type I ‘interferome’ have mainly been carried out at a single species level, often lacking the power necessary to understand key evolutionary features of this pathway. Here, using a single experimental platform, we determined the properties of the interferomes of multiple vertebrate species and developed a webserver to mine the dataset. This approach revealed a conserved ‘core’ of 62 ISGs, including genes not previously associated with IFN, underscoring the ancestral functions associated with this antiviral host response. We show that gene expansion contributes to the evolution of the IFN system and that interferomes are shaped by lineage-specific pressures. Consequently, each mammal possesses a unique repertoire of ISGs, including genes common to all mammals and others unique to their specific species or phylogenetic lineages. An analysis of genes commonly down-regulated by IFN suggests that epigenetic regulation of transcription is a fundamental aspect of the IFN response. Our study provides a resource for the scientific community highlighting key paradigms of the type I IFN response. The type I interferon (IFN) response is triggered upon sensing of an incoming pathogen in an infected cell and results in the expression of hundreds of IFN-stimulated genes (ISGs, collectively referred to as ‘the interferome’). Studies on the interferome have been carried out mainly in human cells and therefore often lack the power to understand comparative evolutionary aspects of this critical pathway. In this study, we characterized the interferome in several animal species (including humans) using a single experimental framework. This approach allowed us to identify fundamental properties of the innate immune system. In particular, we revealed 62 ‘core’ ISGs, up-regulated in response to IFN in all vertebrates, highlighting the ancestral functions of the IFN system. In addition, we show that many genes repressed by the IFN response normally function as regulators of cell transcription. ISGs shared by multiple species have a higher propensity than other genes to exist as multiple copies in the genome. Importantly, we observed that genes have arisen as ISGs throughout evolution. Hence, every animal species possesses a unique repertoire of ISGs that includes core and lineage-specific genes. Collectively, our data provide a framework on which it will be possible to test the role of the IFN response in pathogen emergence and cross-species transmission.
Collapse
Affiliation(s)
- Andrew E. Shaw
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Abdelkader Behdenna
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Joshua B. Singer
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Tristan Dennis
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Richard J. Orton
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Mariana Varela
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Robert J. Gifford
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Sam J. Wilson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
- * E-mail: (SJW); (MP)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
- * E-mail: (SJW); (MP)
| |
Collapse
|
38
|
Differential Alterations in Metabolism and Proteolysis-Related Proteins in Human Parkinson's Disease Substantia Nigra. Neurotox Res 2017; 33:560-568. [PMID: 29218503 DOI: 10.1007/s12640-017-9843-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 12/31/2022]
Abstract
Parkinson's disease is the most common neurodegenerative disorder after Alzheimer's disease, with the majority of cases being sporadic or "idiopathic". The aetiology of the sporadic form is still unknown, but there is a broad consensus that Parkinson's disease involves multiple pathways. In previous human post-mortem studies investigating substantia nigra of parkinsonian subjects, gene expression alterations in various metabolic pathways including protein folding, trafficking, aggregation, ubiquitination and oxidative stress were found. These studies revealed transcriptomic dysregulation of various genes, amongst others Skp1A and PSMC4 (part of ubiquitin-proteasome system), HSC70 (belonging to the chaperone family) and ALDH1A1 (an enzyme involved in the catabolism of dopamine). To investigate whether these alterations are manifested at the protein level, we performed immunohistochemical analysis in the substantia nigra of Parkinson's disease and compared them to Alzheimer's disease and non-neurological post-mortem controls. We were able to confirm cell-specific reductions in the protein content of ALHD1A1 and Skp1A in the dopaminergic neurons of the substantia nigra of Parkinsonian patients compared to Alzheimer's and control subjects. Furthermore, we observed particular distribution for HSC70 and PSMC4 in the cytoplasm and accumulation within Lewy body in the dopaminergic neurons of the substantia nigra in Parkinson patients. These findings, together with previous evidence, suggest a malfunction of the ubiquitin-proteasome and possible autophagy systems as major players in protein misfolding and aggregation in Parkinson's disease. Nevertheless, this needs further proof, possibly with trajectory time line.
Collapse
|
39
|
Bai Y, Dong L, Huang X, Zheng S, Qiu P, Lan F. Associations of rs823128, rs1572931, and rs823156 polymorphisms with reduced Parkinson's disease risks. Neuroreport 2017; 28:936-941. [PMID: 28749816 PMCID: PMC5585133 DOI: 10.1097/wnr.0000000000000846] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/29/2017] [Indexed: 12/14/2022]
Abstract
The PARK16 locus is considered to play a protective role in Parkinson's disease (PD). However, the epidemiological evidence on the relationships between PARK16 single-nucleotide polymorphisms (rs823128, rs1572931, and rs823156) and PD is inconsistent. Therefore, we carried out a meta-analysis to validate the relationships and performed a bioinformatic analysis to explore putative regulation mechanisms of the single-nucleotide polymorphisms in PD. Through meta-analysis, we confirmed that minor variants of rs823128A>G, rs1572931C>T, and rs823156A>G played protective roles in PD. Through bioinformatic analysis, we predicted that rs823128, rs1572931, and rs823156 as noncoding variants of NUCKS1, RAB29, and SLC41A1, respectively, might affect PD risk by altering the transcription factor-binding capability of the genes. These findings suggest new clues for PD research and potential targets for PD prevention and treatment.
Collapse
Affiliation(s)
- Ye Bai
- Department of Clinical Genetics and Experimental Medicine, Fuzhou General Hospital
| | - Lihong Dong
- Department of Clinical Genetics and Experimental Medicine, Fuzhou General Hospital
| | - Xinghua Huang
- Department of Clinical Genetics and Experimental Medicine, Fuzhou General Hospital
| | - Shuanglin Zheng
- Department of Clinical Genetics and Experimental Medicine, Affiliated Dongfang Hospital of Xiamen University, Fuzhou, China
| | - Ping Qiu
- Department of Clinical Genetics and Experimental Medicine, Affiliated Dongfang Hospital of Xiamen University, Fuzhou, China
| | - Fenghua Lan
- Department of Clinical Genetics and Experimental Medicine, Fuzhou General Hospital
| |
Collapse
|
40
|
Tofaris GK, Goedert M, Spillantini MG. The Transcellular Propagation and Intracellular Trafficking of α-Synuclein. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a024380. [PMID: 27920026 DOI: 10.1101/cshperspect.a024380] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Parkinson's disease is the second most common neurodegenerative disorder, with only partial symptomatic therapy and no mechanism-based therapies. The accumulation and aggregation of α-synuclein is causatively linked to the sporadic form of the disease, which accounts for 95% of cases. The pathology is a result of a gain of toxic function of misfolded α-synuclein conformers, which can template the aggregation of soluble monomers and lead to cellular dysfunction, at least partly by interfering with membrane fusion events at synaptic terminals. Here, we discuss the transcellular propagation and intracellular trafficking of α-synuclein and posit that endosomal processing could be a point of convergence between these two routes. Understanding these events will clarify the therapeutic potential of enzymes that regulate protein trafficking and degradation in synucleinopathies.
Collapse
Affiliation(s)
- George K Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
41
|
Reducing Endogenous α-Synuclein Mitigates the Degeneration of Selective Neuronal Populations in an Alzheimer's Disease Transgenic Mouse Model. J Neurosci 2017; 36:7971-84. [PMID: 27466341 DOI: 10.1523/jneurosci.0775-16.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/13/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Alzheimer's disease (AD) is characterized by the progressive accumulation of amyloid β (Aβ) and microtubule associate protein tau, leading to the selective degeneration of neurons in the neocortex, limbic system, and nucleus basalis, among others. Recent studies have shown that α-synuclein (α-syn) also accumulates in the brains of patients with AD and interacts with Aβ and tau, forming toxic hetero-oligomers. Although the involvement of α-syn has been investigated extensively in Lewy body disease, less is known about the role of this synaptic protein in AD. Here, we found that reducing endogenous α-syn in an APP transgenic mouse model of AD prevented the degeneration of cholinergic neurons, ameliorated corresponding deficits, and recovered the levels of Rab3a and Rab5 proteins involved in intracellular transport and sorting of nerve growth factor and brain-derived neurotrophic factor. Together, these results suggest that α-syn might participate in mechanisms of vulnerability of selected neuronal populations in AD and that reducing α-syn might be a potential approach to protecting these populations from the toxic effects of Aβ. SIGNIFICANCE STATEMENT Reducing endogenous α-synuclein (α-syn) in an APP transgenic mouse model of Alzheimer's disease (AD) prevented the degeneration of cholinergic neurons, ameliorated corresponding deficits, and recovered the levels of Rab3a and Rab5 proteins involved in intracellular transport and sorting of nerve growth factor and brain-derived neurotrophic factor. These results suggest that α-syn might participate in mechanisms of vulnerability of selected neuronal populations in AD and that reducing α-syn might be a potential approach to protecting these populations from the toxic effects of amyloid β.
Collapse
|
42
|
Dysregulation of the causative genes for hereditary parkinsonism in the midbrain in Parkinson's disease. Mov Disord 2017; 32:1211-1220. [DOI: 10.1002/mds.27019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 02/26/2017] [Accepted: 03/17/2017] [Indexed: 11/07/2022] Open
|
43
|
Gorenberg EL, Chandra SS. The Role of Co-chaperones in Synaptic Proteostasis and Neurodegenerative Disease. Front Neurosci 2017; 11:248. [PMID: 28579939 PMCID: PMC5437171 DOI: 10.3389/fnins.2017.00248] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/18/2017] [Indexed: 12/14/2022] Open
Abstract
Synapses must be preserved throughout an organism's lifespan to allow for normal brain function and behavior. Synapse maintenance is challenging given the long distances between the termini and the cell body, reliance on axonal transport for delivery of newly synthesized presynaptic proteins, and high rates of synaptic vesicle exo- and endocytosis. Hence, synapses rely on efficient proteostasis mechanisms to preserve their structure and function. To this end, the synaptic compartment has specific chaperones to support its functions. Without proper synaptic chaperone activity, local proteostasis imbalances lead to neurotransmission deficits, dismantling of synapses, and neurodegeneration. In this review, we address the roles of four synaptic chaperones in the maintenance of the nerve terminal, as well as their genetic links to neurodegenerative disease. Three of these are Hsp40 co-chaperones (DNAJs): Cysteine String Protein alpha (CSPα; DNAJC5), auxilin (DNAJC6), and Receptor-Mediated Endocytosis 8 (RME-8; DNAJC13). These co-chaperones contain a conserved J domain through which they form a complex with heat shock cognate 70 (Hsc70), enhancing the chaperone's ATPase activity. CSPα is a synaptic vesicle protein known to chaperone the t-SNARE SNAP-25 and the endocytic GTPase dynamin-1, thereby regulating synaptic vesicle exocytosis and endocytosis. Auxilin binds assembled clathrin cages, and through its interactions with Hsc70 leads to the uncoating of clathrin-coated vesicles, a process necessary for the regeneration of synaptic vesicles. RME-8 is a co-chaperone on endosomes and may have a role in clathrin-coated vesicle endocytosis on this organelle. These three co-chaperones maintain client function by preserving folding and assembly to prevent client aggregation, but they do not break down aggregates that have already formed. The fourth synaptic chaperone we will discuss is Heat shock protein 110 (Hsp110), which interacts with Hsc70, DNAJAs, and DNAJBs to constitute a disaggregase. Hsp110-related disaggregase activity is present at the synapse and is known to protect against aggregation of proteins such as α-synuclein. Congruent with their importance in the nervous system, mutations of these co-chaperones lead to familial neurodegenerative disease. CSPα mutations cause adult neuronal ceroid lipofuscinosis, while auxilin mutations result in early-onset Parkinson's disease, demonstrating their significance in preservation of the nervous system.
Collapse
Affiliation(s)
- Erica L Gorenberg
- Interdepartmental Neuroscience Program, Yale UniversityNew Haven, CT, United States
| | - Sreeganga S Chandra
- Department of Neurology, Yale UniversityNew Haven, CT, United States.,Department of Neuroscience, Yale UniversityNew Haven, CT, United States
| |
Collapse
|
44
|
Martinez A, Lectez B, Ramirez J, Popp O, Sutherland JD, Urbé S, Dittmar G, Clague MJ, Mayor U. Quantitative proteomic analysis of Parkin substrates in Drosophila neurons. Mol Neurodegener 2017; 12:29. [PMID: 28399880 PMCID: PMC5387213 DOI: 10.1186/s13024-017-0170-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/30/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Parkin (PARK2) is an E3 ubiquitin ligase that is commonly mutated in Familial Parkinson's Disease (PD). In cell culture models, Parkin is recruited to acutely depolarised mitochondria by PINK1. PINK1 activates Parkin activity leading to ubiquitination of multiple proteins, which in turn promotes clearance of mitochondria by mitophagy. Many substrates have been identified using cell culture models in combination with depolarising drugs or proteasome inhibitors, but not in more physiological settings. METHODS Here we utilized the recently introduced BioUb strategy to isolate ubiquitinated proteins in flies. Following Parkin Wild-Type (WT) and Parkin Ligase dead (LD) expression we analysed by mass spectrometry and stringent bioinformatics analysis those proteins differentially ubiquitinated to provide the first survey of steady state Parkin substrates using an in vivo model. We further used an in vivo ubiquitination assay to validate one of those substrates in SH-SY5Y cells. RESULTS We identified 35 proteins that are more prominently ubiquitinated following Parkin over-expression. These include several mitochondrial proteins and a number of endosomal trafficking regulators such as v-ATPase sub-units, Syx5/STX5, ALiX/PDCD6IP and Vps4. We also identified the retromer component, Vps35, another PD-associated gene that has recently been shown to interact genetically with parkin. Importantly, we validated Parkin-dependent ubiquitination of VPS35 in human neuroblastoma cells. CONCLUSIONS Collectively our results provide new leads to the possible physiological functions of Parkin activity that are not overtly biased by acute mitochondrial depolarisation.
Collapse
Affiliation(s)
- Aitor Martinez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,Functional Genomics Unit, CIC bioGUNE, Derio, Spain.,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Benoit Lectez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,Functional Genomics Unit, CIC bioGUNE, Derio, Spain
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Sylvie Urbé
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Gunnar Dittmar
- Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Michael J Clague
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain. .,Functional Genomics Unit, CIC bioGUNE, Derio, Spain. .,Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain.
| |
Collapse
|
45
|
Redenšek S, Trošt M, Dolžan V. Genetic Determinants of Parkinson's Disease: Can They Help to Stratify the Patients Based on the Underlying Molecular Defect? Front Aging Neurosci 2017; 9:20. [PMID: 28239348 PMCID: PMC5301007 DOI: 10.3389/fnagi.2017.00020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/25/2017] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a sporadic progressive neurodegenerative brain disorder with a relatively strong genetic background. We have reviewed the current literature about the genetic factors that could be indicative of pathophysiological pathways of PD and their applications in everyday clinical practice. Information on novel risk genes is coming from several genome-wide association studies (GWASs) and their meta-analyses. GWASs that have been performed so far enabled the identification of 24 loci as PD risk factors. These loci take part in numerous cellular processes that may contribute to PD pathology: protein aggregation, protein, and membrane trafficking, lysosomal autophagy, immune response, synaptic function, endocytosis, inflammation, and metabolic pathways are among the most important ones. The identified single nucleotide polymorphisms are usually located in the non-coding regions and their functionality remains to be determined, although they presumably influence gene expression. It is important to be aware of a very low contribution of a single genetic risk factor to PD development; therefore, novel prognostic indices need to account for the cumulative nature of genetic risk factors. A better understanding of PD pathophysiology and its genetic background will help to elucidate the underlying pathological processes. Such knowledge may help physicians to recognize subjects with the highest risk for the development of PD, and provide an opportunity for the identification of novel potential targets for neuroprotective treatment. Moreover, it may enable stratification of the PD patients according to their genetic fingerprint to properly personalize their treatment as well as supportive measures.
Collapse
Affiliation(s)
- Sara Redenšek
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana Ljubljana, Slovenia
| | - Maja Trošt
- Department of Neurology, University Medical Centre Ljubljana Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana Ljubljana, Slovenia
| |
Collapse
|
46
|
Domert J, Sackmann C, Severinsson E, Agholme L, Bergström J, Ingelsson M, Hallbeck M. Aggregated Alpha-Synuclein Transfer Efficiently between Cultured Human Neuron-Like Cells and Localize to Lysosomes. PLoS One 2016; 11:e0168700. [PMID: 28030591 PMCID: PMC5193351 DOI: 10.1371/journal.pone.0168700] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 12/04/2016] [Indexed: 01/06/2023] Open
Abstract
Parkinson's disease and other alpha-synucleinopathies are progressive neurodegenerative diseases characterized by aggregates of misfolded alpha-synuclein spreading throughout the brain. Recent evidence suggests that the pathological progression is likely due to neuron-to-neuron transfer of these aggregates between neuroanatomically connected areas of the brain. As the impact of this pathological spreading mechanism is currently debated, we aimed to investigate the transfer and subcellular location of alpha-synuclein species in a novel 3D co-culture human cell model based on highly differentiated SH-SY5Y cells. Fluorescently-labeled monomeric, oligomeric and fibrillar species of alpha-synuclein were introduced into a donor cell population and co-cultured with an EGFP-expressing acceptor-cell population of differentiated neuron-like cells. Subsequent transfer and colocalization of the different species were determined with confocal microscopy. We could confirm cell-to-cell transfer of all three alpha-synuclein species investigated. Interestingly the level of transferred oligomers and fibrils and oligomers were significantly higher than monomers, which could affect the probability of seeding and pathology in the recipient cells. Most alpha-synuclein colocalized with the lysosomal/endosomal system, both pre- and postsynaptically, suggesting its importance in the processing and spreading of alpha-synuclein.
Collapse
Affiliation(s)
- Jakob Domert
- Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Christopher Sackmann
- Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Emelie Severinsson
- Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Lotta Agholme
- Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Joakim Bergström
- Department of Public Health/Geriatrics, Rudbeck Laboratory, Uppsala University,Uppsala,Sweden
| | - Martin Ingelsson
- Department of Public Health/Geriatrics, Rudbeck Laboratory, Uppsala University,Uppsala,Sweden
| | - Martin Hallbeck
- Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
47
|
Determining the Roles of Inositol Trisphosphate Receptors in Neurodegeneration: Interdisciplinary Perspectives on a Complex Topic. Mol Neurobiol 2016; 54:6870-6884. [PMID: 27771899 DOI: 10.1007/s12035-016-0205-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023]
Abstract
It is well known that calcium (Ca2+) is involved in the triggering of neuronal death. Ca2+ cytosolic levels are regulated by Ca2+ release from internal stores located in organelles, such as the endoplasmic reticulum. Indeed, Ca2+ transit from distinct cell compartments follows complex dynamics that are mediated by specific receptors, notably inositol trisphosphate receptors (IP3Rs). Ca2+ release by IP3Rs plays essential roles in several neurological disorders; however, details of these processes are poorly understood. Moreover, recent studies have shown that subcellular location, molecular identity, and density of IP3Rs profoundly affect Ca2+ transit in neurons. Therefore, regulation of IP3R gene products in specific cellular vicinities seems to be crucial in a wide range of cellular processes from neuroprotection to neurodegeneration. In this regard, microRNAs seem to govern not only IP3Rs translation levels but also subcellular accumulation. Combining new data from molecular cell biology with mathematical modelling, we were able to summarize the state of the art on this topic. In addition to presenting how Ca2+ dynamics mediated by IP3R activation follow a stochastic regimen, we integrated a theoretical approach in an easy-to-apply, cell biology-coherent fashion. Following the presented premises and in contrast to previously tested hypotheses, Ca2+ released by IP3Rs may play different roles in specific neurological diseases, including Alzheimer's disease and Parkinson's disease.
Collapse
|
48
|
Analysis of DNAJC13 mutations in French-Canadian/French cohort of Parkinson's disease. Neurobiol Aging 2016; 45:212.e13-212.e17. [DOI: 10.1016/j.neurobiolaging.2016.04.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/29/2016] [Accepted: 04/26/2016] [Indexed: 12/14/2022]
|
49
|
Domingo A, Amar D, Grütz K, Lee LV, Rosales R, Brüggemann N, Jamora RD, Cutiongco-Dela Paz E, Rolfs A, Dressler D, Walter U, Krainc D, Lohmann K, Shamir R, Klein C, Westenberger A. Evidence of TAF1 dysfunction in peripheral models of X-linked dystonia-parkinsonism. Cell Mol Life Sci 2016; 73:3205-15. [PMID: 26879577 PMCID: PMC11108471 DOI: 10.1007/s00018-016-2159-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/30/2016] [Accepted: 02/04/2016] [Indexed: 11/30/2022]
Abstract
The molecular dysfunction in X-linked dystonia-parkinsonism is not completely understood. Thus far, only noncoding alterations have been found in genetic analyses, located in or nearby the TATA-box binding protein-associated factor 1 (TAF1) gene. Given that this gene is ubiquitously expressed and is a critical component of the cellular transcription machinery, we sought to study differential gene expression in peripheral models by performing microarray-based expression profiling in blood and fibroblasts, and comparing gene expression in affected individuals vs. ethnically matched controls. Validation was performed via quantitative polymerase chain reaction in discovery and independent replication sets. We observed consistent downregulation of common TAF1 transcripts in samples from affected individuals in gene-level and high-throughput experiments. This signal was accompanied by a downstream effect in the microarray, reflected by the dysregulation of 307 genes in the disease group. Gene Ontology and network analyses revealed enrichment of genes involved in RNA polymerase II-dependent transcription, a pathway relevant to TAF1 function. Thus, the results converge on TAF1 dysfunction in peripheral models of X-linked dystonia-parkinsonism, and provide evidence of altered expression of a canonical gene in this disease. Furthermore, our study illustrates a link between the previously described genetic alterations and TAF1 dysfunction at the transcriptome level.
Collapse
Affiliation(s)
- Aloysius Domingo
- Institute of Neurogenetics, University of Lübeck, Maria Goeppert Str. 1, 23562, Lübeck, Germany
- Graduate School Lübeck, University of Lübeck, Lübeck, Germany
| | - David Amar
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Karen Grütz
- Institute of Neurogenetics, University of Lübeck, Maria Goeppert Str. 1, 23562, Lübeck, Germany
| | - Lillian V Lee
- XDP Study Group, Philippine Children's Medical Center, Quezon City, Philippines
| | - Raymond Rosales
- Department of Neurology and Psychiatry, University of Santo Tomas, Manila, Philippines
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, Maria Goeppert Str. 1, 23562, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Roland Dominic Jamora
- Department of Neurosciences, College of Medicine, Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Eva Cutiongco-Dela Paz
- National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Philippine Genome Center, University of the Philippines, Diliman, Quezon City, Philippines
| | - Arndt Rolfs
- Albrecht-Kossel-Institute for Neuroregeneration, University of Rostock, Rostock, Germany
| | - Dirk Dressler
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Uwe Walter
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Dimitri Krainc
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Maria Goeppert Str. 1, 23562, Lübeck, Germany
| | - Ron Shamir
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv, Israel
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Maria Goeppert Str. 1, 23562, Lübeck, Germany.
| | - Ana Westenberger
- Institute of Neurogenetics, University of Lübeck, Maria Goeppert Str. 1, 23562, Lübeck, Germany
| |
Collapse
|
50
|
Deubiquitinase Usp8 regulates α-synuclein clearance and modifies its toxicity in Lewy body disease. Proc Natl Acad Sci U S A 2016; 113:E4688-97. [PMID: 27444016 PMCID: PMC4987833 DOI: 10.1073/pnas.1523597113] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In Parkinson's disease, misfolded α-synuclein accumulates, often in a ubiquitinated form, in neuronal inclusions termed Lewy bodies. An important outstanding question is whether ubiquitination in Lewy bodies is directly relevant to α-synuclein trafficking or turnover and Parkinson's pathogenesis. By comparative analysis in human postmortem brains, we found that ubiquitin immunoreactivity in Lewy bodies is largely due to K63-linked ubiquitin chains and markedly reduced in the substantia nigra compared with the neocortex. The ubiquitin staining in cells with Lewy bodies inversely correlated with the content and pathological localization of the deubiquitinase Usp8. Usp8 interacted and partly colocalized with α-synuclein in endosomal membranes and, both in cells and after purification, it deubiquitinated K63-linked chains on α-synuclein. Knockdown of Usp8 in the Drosophila eye reduced α-synuclein levels and α-synuclein-induced eye toxicity. Accordingly, in human cells, Usp8 knockdown increased the lysosomal degradation of α-synuclein. In the dopaminergic neurons of the Drosophila model, unlike knockdown of other deubiquitinases, Usp8 protected from α-synuclein-induced locomotor deficits and cell loss. These findings strongly suggest that removal of K63-linked ubiquitin chains on α-synuclein by Usp8 is a critical mechanism that reduces its lysosomal degradation in dopaminergic neurons and may contribute to α-synuclein accumulation in Lewy body disease.
Collapse
|