101
|
Martinez JH, Alaimo A, Gorojod RM, Porte Alcon S, Fuentes F, Coluccio Leskow F, Kotler ML. Drp-1 dependent mitochondrial fragmentation and protective autophagy in dopaminergic SH-SY5Y cells overexpressing alpha-synuclein. Mol Cell Neurosci 2018; 88:107-117. [PMID: 29414102 DOI: 10.1016/j.mcn.2018.01.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/10/2018] [Accepted: 01/15/2018] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease is a neurodegenerative movement disorder caused by the loss of dopaminergic neurons from substantia nigra. It is characterized by the accumulation of aggregated α-synuclein as the major component of the Lewy bodies. Additional common features of this disease are the mitochondrial dysfunction and the activation/inhibition of autophagy both events associated to the intracellular accumulation of α-synuclein. The mechanism by which these events contribute to neural degeneration remains unknown. In the present work we investigated the effect of α-synuclein on mitochondrial dynamics and autophagy/mitophagy in SH-SY5Y cells, an in vitro model of Parkinson disease. We demonstrated that overexpression of wild type α-synuclein causes moderated toxicity, ROS generation and mitochondrial dysfunction. In addition, α-synuclein induces the mitochondrial fragmentation on a Drp-1-dependent fashion. Overexpression of the fusion protein Opa-1 prevented both mitochondrial fragmentation and cytotoxicity. On the other hand, cells expressing α-synuclein showed activated autophagy and particularly mitophagy. Employing a genetic strategy we demonstrated that autophagy is triggered in order to protect cells from α-synuclein-induced cell death. Our results clarify the role of Opa-1 and Drp-1 in mitochondrial dynamics and cell survival, a controversial α-synuclein research issue. The findings presented point to the relevance of mitochondrial homeostasis and autophagy in the pathogenesis of PD. Better understanding of the molecular interaction between these processes could give rise to novel therapeutic methods for PD prevention and amelioration.
Collapse
Affiliation(s)
- Jimena Hebe Martinez
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, Argentina.
| | - Agustina Alaimo
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Roxana Mayra Gorojod
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Soledad Porte Alcon
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| | - Federico Fuentes
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Federico Coluccio Leskow
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, Argentina.
| | - Mónica Lidia Kotler
- CONICET- Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, Buenos Aires, Argentina.
| |
Collapse
|
102
|
Tyler JK, Johnson JE. The role of autophagy in the regulation of yeast life span. Ann N Y Acad Sci 2018; 1418:31-43. [PMID: 29363766 DOI: 10.1111/nyas.13549] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/13/2017] [Accepted: 10/18/2017] [Indexed: 01/07/2023]
Abstract
The goal of the aging field is to develop novel therapeutic interventions that extend human health span and reduce the burden of age-related disease. While organismal aging is a complex, multifactorial process, a popular theory is that cellular aging is a significant contributor to the progressive decline inherent to all multicellular organisms. To explore the molecular determinants that drive cellular aging, as well as how to retard them, researchers have utilized the highly genetically tractable budding yeast Saccharomyces cerevisiae. Indeed, every intervention known to extend both cellular and organismal health span was identified in yeast, underlining the power of this approach. Importantly, a growing body of work has implicated the process of autophagy as playing a critical role in the delay of aging. This review summarizes recent reports that have identified a role for autophagy, or autophagy factors in the extension of yeast life span. These studies demonstrate (1) that yeast remains an invaluable tool for the identification and characterization of conserved mechanisms that promote cellular longevity and are likely to be relevant to humans, and (2) that the process of autophagy has been implicated in nearly all known longevity-promoting manipulations and thus represents an ideal target for interventions aimed at improving human health span.
Collapse
Affiliation(s)
- Jessica K Tyler
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Jay E Johnson
- Department of Biology, Orentreich Foundation for the Advancement of Science, Cold Spring, New York
| |
Collapse
|
103
|
Minakaki G, Menges S, Kittel A, Emmanouilidou E, Schaeffner I, Barkovits K, Bergmann A, Rockenstein E, Adame A, Marxreiter F, Mollenhauer B, Galasko D, Buzás EI, Schlötzer-Schrehardt U, Marcus K, Xiang W, Lie DC, Vekrellis K, Masliah E, Winkler J, Klucken J. Autophagy inhibition promotes SNCA/alpha-synuclein release and transfer via extracellular vesicles with a hybrid autophagosome-exosome-like phenotype. Autophagy 2018; 14:98-119. [PMID: 29198173 DOI: 10.1080/15548627.2017.1395992] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The autophagy-lysosome pathway (ALP) regulates intracellular homeostasis of the cytosolic protein SNCA/alpha-synuclein and is impaired in synucleinopathies, including Parkinson disease and dementia with Lewy bodies (DLB). Emerging evidence suggests that ALP influences SNCA release, but the underlying cellular mechanisms are not well understood. Several studies identified SNCA in exosome/extracellular vesicle (EV) fractions. EVs are generated in the multivesicular body compartment and either released upon its fusion with the plasma membrane, or cleared via the ALP. We therefore hypothesized that inhibiting ALP clearance 1) enhances SNCA release via EVs by increasing extracellular shuttling of multivesicular body contents, 2) alters EV biochemical profile, and 3) promotes SNCA cell-to-cell transfer. Indeed, ALP inhibition increased the ratio of extra- to intracellular SNCA and upregulated SNCA association with EVs in neuronal cells. Ultrastructural analysis revealed a widespread, fused multivesicular body-autophagosome compartment. Biochemical characterization revealed the presence of autophagosome-related proteins, such as LC3-II and SQSTM1. This distinct "autophagosome-exosome-like" profile was also identified in human cerebrospinal fluid (CSF) EVs. After a single intracortical injection of SNCA-containing EVs derived from CSF into mice, human SNCA colocalized with endosome and neuronal markers. Prominent SNCA immunoreactivity and a higher number of neuronal SNCA inclusions were observed after DLB patient CSF EV injections. In summary, this study provides compelling evidence that a) ALP inhibition increases SNCA in neuronal EVs, b) distinct ALP components are present in EVs, and c) CSF EVs transfer SNCA from cell to cell in vivo. Thus, macroautophagy/autophagy may regulate EV protein composition and consequently progression in synucleinopathies.
Collapse
Affiliation(s)
- Georgia Minakaki
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Stefanie Menges
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Agnes Kittel
- b Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences , Semmelweis University , Budapest , Hungary
| | - Evangelia Emmanouilidou
- c Department of Neuroscience, Center for Basic Research , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | | | - Katalin Barkovits
- e Medizinisches Proteom-Center, Medical Faculty , Ruhr University Bochum , Bochum , Germany
| | - Anna Bergmann
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Edward Rockenstein
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Anthony Adame
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Franz Marxreiter
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Brit Mollenhauer
- g Paracelsus-Elena-Klinik, Kassel and Departments of Neuropathology & Neurosurgery , & University Medical Center , Göttingen
| | - Douglas Galasko
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Edit Irén Buzás
- h Department of Genetics, Cell and Immunobiology , Semmelweis University , Budapest , Hungary
| | | | - Katrin Marcus
- e Medizinisches Proteom-Center, Medical Faculty , Ruhr University Bochum , Bochum , Germany
| | - Wei Xiang
- d Institute of Biochemistry , FAU , Erlangen , Germany
| | | | - Kostas Vekrellis
- c Department of Neuroscience, Center for Basic Research , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Eliezer Masliah
- f Department of Neurosciences , University of California , San Diego , CA USA.,j Department of Pathology , University of California , San Diego , CA USA
| | - Jürgen Winkler
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Jochen Klucken
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| |
Collapse
|
104
|
Lassen LB, Reimer L, Ferreira N, Betzer C, Jensen PH. Protein Partners of α-Synuclein in Health and Disease. Brain Pathol 2018; 26:389-97. [PMID: 26940507 DOI: 10.1111/bpa.12374] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/04/2016] [Accepted: 01/11/2016] [Indexed: 12/30/2022] Open
Abstract
α-synuclein is normally situated in the nerve terminal but it accumulates and aggregates in axons and cell bodies in synucleinopathies such as Parkinson's disease. The conformational changes occurring during α-synucleins aggregation process affects its interactions with other proteins and its subcellular localization. This review focuses on interaction partners of α-synuclein within different compartments of the cell with a focus on those preferentially binding aggregated α-synuclein. The aggregation state of α-synuclein also affects its catabolism and we hypothesize impaired macroautophagy is involved neuronal excretion of α-synuclein species responsible for the prion-like spreading of α-synuclein pathology.
Collapse
Affiliation(s)
- Louise Berkhoudt Lassen
- DANDRITE-Danish Research Institute of Translational Neuroscience & Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Lasse Reimer
- DANDRITE-Danish Research Institute of Translational Neuroscience & Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Nelson Ferreira
- DANDRITE-Danish Research Institute of Translational Neuroscience & Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Cristine Betzer
- DANDRITE-Danish Research Institute of Translational Neuroscience & Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Poul Henning Jensen
- DANDRITE-Danish Research Institute of Translational Neuroscience & Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| |
Collapse
|
105
|
Alfaro IE, Albornoz A, Molina A, Moreno J, Cordero K, Criollo A, Budini M. Chaperone Mediated Autophagy in the Crosstalk of Neurodegenerative Diseases and Metabolic Disorders. Front Endocrinol (Lausanne) 2018; 9:778. [PMID: 30766511 PMCID: PMC6365421 DOI: 10.3389/fendo.2018.00778] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Chaperone Mediated Autophagy (CMA) is a lysosomal-dependent protein degradation pathway. At least 30% of cytosolic proteins can be degraded by this process. The two major protein players of CMA are LAMP-2A and HSC70. While LAMP-2A works as a receptor for protein substrates at the lysosomal membrane, HSC70 specifically binds protein targets and takes them for CMA degradation. Because of the broad spectrum of proteins able to be degraded by CMA, this pathway has been involved in physiological and pathological processes such as lipid and carbohydrate metabolism, and neurodegenerative diseases, respectively. Both, CMA, and the mentioned processes, are affected by aging and by inadequate nutritional habits such as a high fat diet or a high carbohydrate diet. Little is known regarding about CMA, which is considered a common regulation factor that links metabolism with neurodegenerative disorders. This review summarizes what is known about CMA, focusing on its molecular mechanism, its role in protein, lipid and carbohydrate metabolism. In addition, the review will discuss how CMA could be linked to protein, lipids and carbohydrate metabolism within neurodegenerative diseases. Furthermore, it will be discussed how aging and inadequate nutritional habits can have an impact on both CMA activity and neurodegenerative disorders.
Collapse
Affiliation(s)
- Iván E. Alfaro
- Fundación Ciencia & Vida, Santiago, Chile
- *Correspondence: Iván E. Alfaro
| | | | - Alfredo Molina
- Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
| | - José Moreno
- Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
| | - Karina Cordero
- Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
| | - Alfredo Criollo
- Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
- Autophagy Research Center (ARC), Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Mauricio Budini
- Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
- Autophagy Research Center (ARC), Santiago, Chile
- Mauricio Budini
| |
Collapse
|
106
|
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by Lewy body pathology of which the primary constituent is aggregated misfolded alpha-synuclein protein. Currently, there are no clinical therapies for treatment of the underlying alpha-synuclein dysfunction and accumulation, and the standard of care for patients with Parkinson's disease focuses only on symptom management, creating an immense therapeutic gap that needs to be filled. Defects in autophagy have been strongly implicated in Parkinson's disease. Here, we review evidence from human, mouse, and cell culture studies to briefly explain these defects in autophagy in Parkinson's disease and the necessity for autophagy to be carefully and precisely tuned to maintain neuron survival. We summarize recent experimental agents for treating alpha-synuclein accumulation in α-synuclein Parkinson's disease and related synucleinopathies. Most of the efforts for developing experimental agents have focused on immunotherapeutic strategies, but we discuss why those efforts are misplaced. Finally, we emphasize why increasing autophagy flux for alpha-synuclein clearance is the most promising therapeutic strategy. Activating autophagy has been successful in preclinical models of Parkinson's disease and yields promising results in clinical trials.
Collapse
Affiliation(s)
- Alan J Fowler
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA.
| |
Collapse
|
107
|
Autophagy impairment in Parkinson's disease. Essays Biochem 2017; 61:711-720. [PMID: 29233880 DOI: 10.1042/ebc20170023] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/14/2017] [Accepted: 10/24/2017] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) is a debilitating movement disorder typically associated with the accumulation of intracytoplasmic aggregate prone protein deposits. Over recent years, increasing evidence has led to the suggestion that the mutations underlying certain forms of PD impair autophagy. Autophagy is a degradative pathway that delivers cytoplasmic content to lysosomes for degradation and represents a major route for degradation of aggregated cellular proteins and dysfunctional organelles. Autophagy up-regulation is a promising therapeutic strategy that is being explored for its potential to protect cells against the toxicity of aggregate-prone proteins in neurodegenerative diseases. Here, we describe how the mutations in different subtypes of PD can affect different stages of autophagy.
Collapse
|
108
|
Kaur K, Gill JS, Bansal PK, Deshmukh R. Neuroinflammation - A major cause for striatal dopaminergic degeneration in Parkinson's disease. J Neurol Sci 2017; 381:308-314. [DOI: 10.1016/j.jns.2017.08.3251] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 07/25/2017] [Accepted: 08/23/2017] [Indexed: 12/14/2022]
|
109
|
Manecka DL, Vanderperre B, Fon EA, Durcan TM. The Neuroprotective Role of Protein Quality Control in Halting the Development of Alpha-Synuclein Pathology. Front Mol Neurosci 2017; 10:311. [PMID: 29021741 PMCID: PMC5623686 DOI: 10.3389/fnmol.2017.00311] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022] Open
Abstract
Synucleinopathies are a family of neurodegenerative disorders that comprises Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Each of these disorders is characterized by devastating motor, cognitive, and autonomic consequences. Current treatments for synucleinopathies are not curative and are limited to improvement of quality of life for affected individuals. Although the underlying causes of these diseases are unknown, a shared pathological hallmark is the presence of proteinaceous inclusions containing the α-synuclein (α-syn) protein in brain tissue. In the past few years, it has been proposed that these inclusions arise from the self-templated, prion-like spreading of misfolded and aggregated forms of α-syn throughout the brain, leading to neuronal dysfunction and death. In this review, we describe how impaired protein homeostasis is a prominent factor in the α-syn aggregation cascade, with alterations in protein quality control (PQC) pathways observed in the brains of patients. We discuss how PQC modulates α-syn accumulation, misfolding and aggregation primarily through chaperoning activity, proteasomal degradation, and lysosome-mediated degradation. Finally, we provide an overview of experimental data indicating that targeting PQC pathways is a promising avenue to explore in the design of novel neuroprotective approaches that could impede the spreading of α-syn pathology and thus provide a curative treatment for synucleinopathies.
Collapse
Affiliation(s)
| | | | | | - Thomas M. Durcan
- Neurodegenerative Diseases Group and iPSC-CRISPR Core Facility, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
110
|
Loos B, Klionsky DJ, Wong E. Augmenting brain metabolism to increase macro- and chaperone-mediated autophagy for decreasing neuronal proteotoxicity and aging. Prog Neurobiol 2017; 156:90-106. [DOI: 10.1016/j.pneurobio.2017.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/06/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
|
111
|
Autophagy enhancement is rendered ineffective in presence of α-synuclein in melanoma cells. J Cell Commun Signal 2017; 11:381-394. [PMID: 28748508 DOI: 10.1007/s12079-017-0402-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022] Open
Abstract
Increased cellular concentration of α-synuclein (α-syn) predisposes it to misfolding and aggregation that in turn impair the degradation pathways. This poses a limitation to the use of overexpression models for studies on α-syn clearance by autophagy, which is widely investigated for its therapeutic potential. This limitation can be overcome with the use of endogenous models. In this study, SK-MEL-28, a melanoma cell model with endogenous α-syn expression, was employed to study α-syn clearance through autophagy. We demonstrated the dual localization of α-syn to nucleus and cytoplasm that varied in response to changes in cellular environment. Autophagy inhibition and exposure to dopamine favored cytoplasmic localization of α-syn, while autophagy induction favored increased localization to the nucleus. The inhibitory effect of dopamine on autophagy was heightened in presence of α-syn. Additionally, because α-syn had a regulatory effect on autophagy, cells showed an increased resistance to autophagy induction in presence of α-syn. This resistance prevented effective induction of autophagy even under conditions of prolonged autophagy inhibition. These results highlight alternate physiological roles of α-syn, particularly in non-neuronal cells. Because autophagy enhancement could reverse neither the increase in α-syn levels nor the autophagy inhibition, there arises a need to evaluate the efficacy of autophagy-based therapeutic strategies.
Collapse
|
112
|
Liu Y, Song Y, Zhu X. MicroRNA-181a Regulates Apoptosis and Autophagy Process in Parkinson's Disease by Inhibiting p38 Mitogen-Activated Protein Kinase (MAPK)/c-Jun N-Terminal Kinases (JNK) Signaling Pathways. Med Sci Monit 2017; 23:1597-1606. [PMID: 28365714 PMCID: PMC5386441 DOI: 10.12659/msm.900218] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background microRNA (miR)-181a has been reported to be downregulated in Parkinson’s disease (PD), but the regulatory mechanism of miR-181a on neuron apoptosis and autophagy is still poorly understood. We aimed to investigate the neuroprotective effects of miR-181a on PD in vitro. Material/Methods Human SK-N-SH neuroblastoma cells were incubated with different concentrations of 1-methyl-4-phenylpyridinium ion (MPP+) to induce the PD model. The expression of miR-181a was then analyzed. After transfection with miR-181a mimic or scramble following MPP+ treatment, the expression of autophagy protein markers (LC3II, LC3I, and Beclin 1) and p38 mitogen-activated protein kinase (MAPK)/c-Jun N-terminal kinases (JNK) signaling proteins (p-p38, p38, p-JNK, and JNK) and cell apoptosis were detected. Furthermore, the cells were transfected with miR-181a inhibitor and cultured in the presence or absence of p38 inhibitor SB203582 or JNK inhibitor SP600125, and the cell apoptosis was tested again. Results The expression of miR-181a was gradually decreased with the increase of MPP+ concentration (P<0.05, P<0.01, or P<0.001). Overexpression of miR-181a significantly decreased the LC3II/LC3I ratio, Beclin 1 expression, cell apoptosis, and the expression of p-p38 and p-JNK compared to the MPP+ + miR-181a scramble group (all P<0.05). In addition, we observed that SB203582 or SP600125 showed no effects on cell apoptosis, but the effects of miR-181a inhibitor on cell apoptosis were reversed by administration of SB203582 or SP600125 compared to the scramble group (P<0.05). Conclusions Our results suggest that miR-181a regulates apoptosis and autophagy in PD by inhibiting the p38 MAPK/JNK pathway.
Collapse
Affiliation(s)
- Ying Liu
- Department of Internal Medicine Neurology, HuaMei Branch of the 2nd People's Hospital of Liaocheng, Liaocheng, Shandong, China (mainland)
| | - Yanfeng Song
- Department of Internal Medicine Neurology, HuaMei Branch of the 2nd People's Hospital of Liaocheng, Liaocheng, Shandong, China (mainland)
| | - Xiaotun Zhu
- Department of Internal Medicine Neurology, HuaMei Branch of the 2nd People's Hospital of Liaocheng, Liaocheng, Shandong, China (mainland)
| |
Collapse
|
113
|
Abstract
Parkinson disease is the second-most common neurodegenerative disorder that affects 2-3% of the population ≥65 years of age. Neuronal loss in the substantia nigra, which causes striatal dopamine deficiency, and intracellular inclusions containing aggregates of α-synuclein are the neuropathological hallmarks of Parkinson disease. Multiple other cell types throughout the central and peripheral autonomic nervous system are also involved, probably from early disease onwards. Although clinical diagnosis relies on the presence of bradykinesia and other cardinal motor features, Parkinson disease is associated with many non-motor symptoms that add to overall disability. The underlying molecular pathogenesis involves multiple pathways and mechanisms: α-synuclein proteostasis, mitochondrial function, oxidative stress, calcium homeostasis, axonal transport and neuroinflammation. Recent research into diagnostic biomarkers has taken advantage of neuroimaging in which several modalities, including PET, single-photon emission CT (SPECT) and novel MRI techniques, have been shown to aid early and differential diagnosis. Treatment of Parkinson disease is anchored on pharmacological substitution of striatal dopamine, in addition to non-dopaminergic approaches to address both motor and non-motor symptoms and deep brain stimulation for those developing intractable L-DOPA-related motor complications. Experimental therapies have tried to restore striatal dopamine by gene-based and cell-based approaches, and most recently, aggregation and cellular transport of α-synuclein have become therapeutic targets. One of the greatest current challenges is to identify markers for prodromal disease stages, which would allow novel disease-modifying therapies to be started earlier.
Collapse
|
114
|
Ferroptosis and cell death mechanisms in Parkinson's disease. Neurochem Int 2017; 104:34-48. [DOI: 10.1016/j.neuint.2017.01.004] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/18/2016] [Accepted: 01/06/2017] [Indexed: 01/18/2023]
|
115
|
Sharma SK, Priya S. Expanding role of molecular chaperones in regulating α-synuclein misfolding; implications in Parkinson's disease. Cell Mol Life Sci 2017; 74:617-629. [PMID: 27522545 PMCID: PMC11107554 DOI: 10.1007/s00018-016-2340-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 12/19/2022]
Abstract
Protein misfolding under stressful environmental conditions cause several cellular problems owing to the disturbed cellular protein homeostasis, which may further lead to neurological disorders like Parkinson's disease (PD), Alzheimer's disease (AD), Amyloid lateral sclerosis and Huntington disease (HD). The presence of cellular defense mechanisms like molecular chaperones and proteasomal degradation systems prevent protein misfolding and aggregation. Molecular chaperones plays primary role in preventing protein misfolding by mediating proper native folding, unfolding and refolding of the polypeptides along with vast number of cellular functions. In past few years, the understanding of molecular chaperone mechanisms has been expanded enormously although implementation to prevent protein aggregation diseases is still deficient. We in this review evaluated major classes of molecular chaperones and their mechanisms relevant for preventing protein aggregation, specific case of α-synuclein aggregation. We also evaluate the molecular chaperone function as a novel therapeutic approach and the chaperone inhibitors or activators as small molecular drug targets.
Collapse
Affiliation(s)
- Sandeep K Sharma
- Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, 226001, Uttar Pradesh, India
- Nanotherapeutics and Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, 226001, Uttar Pradesh, India
| | - Smriti Priya
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
116
|
Moors TE, Hoozemans JJM, Ingrassia A, Beccari T, Parnetti L, Chartier-Harlin MC, van de Berg WDJ. Therapeutic potential of autophagy-enhancing agents in Parkinson's disease. Mol Neurodegener 2017; 12:11. [PMID: 28122627 PMCID: PMC5267440 DOI: 10.1186/s13024-017-0154-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 01/18/2017] [Indexed: 01/07/2023] Open
Abstract
Converging evidence from genetic, pathological and experimental studies have increasingly suggested an important role for autophagy impairment in Parkinson’s Disease (PD). Genetic studies have identified mutations in genes encoding for components of the autophagy-lysosomal pathway (ALP), including glucosidase beta acid 1 (GBA1), that are associated with increased risk for developing PD. Observations in PD brain tissue suggest an aberrant regulation of autophagy associated with the aggregation of α-synuclein (α-syn). As autophagy is one of the main systems involved in the proteolytic degradation of α-syn, pharmacological enhancement of autophagy may be an attractive strategy to combat α-syn aggregation in PD. Here, we review the potential of autophagy enhancement as disease-modifying therapy in PD based on preclinical evidence. In particular, we provide an overview of the molecular regulation of autophagy and targets for pharmacological modulation within the ALP. In experimental models, beneficial effects on multiple pathological processes involved in PD, including α-syn aggregation, cell death, oxidative stress and mitochondrial dysfunction, have been demonstrated using the autophagy enhancers rapamycin and lithium. However, selectivity of these agents is limited, while upstream ALP signaling proteins are involved in many other pathways than autophagy. Broad stimulation of autophagy may therefore cause a wide spectrum of dose-dependent side-effects, suggesting that its clinical applicability is limited. However, recently developed agents selectively targeting core ALP components, including Transcription Factor EB (TFEB), lysosomes, GCase as well as chaperone-mediated autophagy regulators, exert more specific effects on molecular pathogenetic processes causing PD. To conclude, the targeted manipulation of downstream ALP components, rather than broad autophagy stimulation, may be an attractive strategy for the development of novel pharmacological therapies in PD. Further characterization of dysfunctional autophagy in different stages and molecular subtypes of PD in combination with the clinical translation of downstream autophagy regulation offers exciting new avenues for future drug development.
Collapse
Affiliation(s)
- Tim E Moors
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands.
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Angela Ingrassia
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Department of Medicine, Section of Neurology, University of Perugia, Perugia, Italy
| | - Marie-Christine Chartier-Harlin
- UMR-S 1172-JPArc-Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, University of Lille, Lille, F-59000, France.,Inserm, UMR-S 1172, Team "Early stages of Parkinson's disease", F-59000, Lille, France
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
117
|
Ottolini D, Calí T, Szabò I, Brini M. Alpha-synuclein at the intracellular and the extracellular side: functional and dysfunctional implications. Biol Chem 2017; 398:77-100. [DOI: 10.1515/hsz-2016-0201] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022]
Abstract
Abstract
Alpha-synuclein (α-syn) is an abundant neuronal protein whose physiological function, even if still not completely understood, has been consistently related to synaptic function and vesicle trafficking. A group of disorders known as synucleinopathies, among which Parkinson’s disease (PD), is deeply associated with the misfolding and aggregation of α-syn, which can give rise to proteinaceous inclusion known as Lewy bodies (LB). Proteostasis stress is a relevant aspect in these diseases and, currently, the presence of oligomeric α-syn species rather than insoluble aggregated forms, appeared to be associated with cytotoxicity. Many observations suggest that α-syn is responsible for neurodegeneration by interfering with multiple signaling pathways. α-syn protein can directly form plasma membrane channels or modify with their activity, thus altering membrane permeability to ions, abnormally associate with mitochondria and cause mitochondrial dysfunction (i.e. mitochondrial depolarization, Ca2+ dys-homeostasis, cytochrome c release) and interfere with autophagy regulation. The picture is further complicated by the fact that single point mutations, duplications and triplication in α-syn gene are linked to autosomal dominant forms of PD. In this review we discuss the multi-faced aspect of α-syn biology and address the main hypothesis at the basis of its involvement in neuronal degeneration.
Collapse
|
118
|
van Diggelen F, Tepper AWJW, Apetri MM, Otzen DE. α-Synuclein Oligomers: A Study in Diversity. Isr J Chem 2016. [DOI: 10.1002/ijch.201600116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Femke van Diggelen
- Crossbeta Biosciences; Padualaan 8 3584CH Utrecht The Netherlands
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| | | | | | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| |
Collapse
|
119
|
Sala G, Marinig D, Arosio A, Ferrarese C. Role of Chaperone-Mediated Autophagy Dysfunctions in the Pathogenesis of Parkinson's Disease. Front Mol Neurosci 2016; 9:157. [PMID: 28066181 PMCID: PMC5179559 DOI: 10.3389/fnmol.2016.00157] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/08/2016] [Indexed: 12/20/2022] Open
Abstract
Chaperone-mediated autophagy (CMA) represents a selective form of autophagy involved in the degradation of specific soluble proteins containing a pentapeptide motif that is recognized by a cytosolic chaperone able to deliver proteins to the lysosomes for degradation. Physiologically, CMA contributes to maintain crucial cellular functions including energetic balance and protein quality control. Dysfunctions in CMA have been associated to the pathogenesis of several neurodegenerative diseases characterized by accumulation and aggregation of proteins identified as CMA substrates. In particular, increasing evidence highlights the existence of a strong relationship between CMA defects and Parkinson’s disease (PD). Several mutations associated with familial forms of PD (SNCA, LRRK2, UCHL1 and DJ-1) have been demonstrated to block or reduce the activity of CMA, the main catabolic pathway for alpha-synuclein (asyn). CMA dysfunctions also leads to a mislocalization and inactivation of the transcription factor MEF2D that plays a key-role in the survival of dopaminergic neurons. Furthermore, reduced levels of CMA markers have been observed in post mortem brain samples from PD patients. The aim of this review article is to provide an organic revision of evidence for the involvement of CMA dysfunctions in the pathogenesis of PD. Updated findings obtained in patient’s specimens will be resumed, and results deriving from in vivo and in vitro studies will be discussed to evidence the current knowledge on the molecular mechanisms underlying CMA alterations in PD. Finally, the possibility of up-regulating CMA pathway as promising neuroprotective strategy will be considered.
Collapse
Affiliation(s)
- Gessica Sala
- Laboratory of Neurobiology, School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca Monza, Italy
| | - Daniele Marinig
- Laboratory of Neurobiology, School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-BicoccaMonza, Italy; PhD Program in Neuroscience, University of Milano-BicoccaMonza, Italy
| | - Alessandro Arosio
- Laboratory of Neurobiology, School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca Monza, Italy
| | - Carlo Ferrarese
- Laboratory of Neurobiology, School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-BicoccaMonza, Italy; Department of Neurology, San Gerardo Hospital, University of Milano-BicoccaMonza, Italy
| |
Collapse
|
120
|
Roosen DA, Cookson MR. LRRK2 at the interface of autophagosomes, endosomes and lysosomes. Mol Neurodegener 2016; 11:73. [PMID: 27927216 PMCID: PMC5142374 DOI: 10.1186/s13024-016-0140-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023] Open
Abstract
Over the past 20 years, substantial progress has been made in identifying the underlying genetics of Parkinson's disease (PD). Of the known genes, LRRK2 is a major genetic contributor to PD. However, the exact function of LRRK2 remains to be elucidated. In this review, we discuss how familial forms of PD have led us to hypothesize that alterations in endomembrane trafficking play a role in the pathobiology of PD. We will discuss the major observations that have been made to elucidate the role of LRRK2 in particular, including LRRK2 animal models and high-throughput proteomics approaches. Taken together, these studies strongly support a role of LRRK2 in vesicular dynamics. We also propose that targeting these pathways may not only be beneficial for developing therapeutics for LRRK2-driven PD, but also for other familial and sporadic cases.
Collapse
Affiliation(s)
- Dorien A. Roosen
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707 USA
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP UK
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707 USA
| |
Collapse
|
121
|
Figueira I, Fernandes A, Mladenovic Djordjevic A, Lopez-Contreras A, Henriques CM, Selman C, Ferreiro E, Gonos ES, Trejo JL, Misra J, Rasmussen LJ, Xapelli S, Ellam T, Bellantuono I. Interventions for age-related diseases: Shifting the paradigm. Mech Ageing Dev 2016; 160:69-92. [DOI: 10.1016/j.mad.2016.09.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/18/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
|
122
|
Xilouri M, Stefanis L. Chaperone mediated autophagy in aging: Starve to prosper. Ageing Res Rev 2016; 32:13-21. [PMID: 27484893 DOI: 10.1016/j.arr.2016.07.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/07/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
The major lysosomal proteolytic pathways essential for maintaining proper cellular homeostasis are macroautophagy, chaperone-mediated autophagy (CMA) and microautophagy. What differentiates CMA from the other types of autophagy is the fact that it does not involve vesicle formation; the unique feature of this pathway is the selective targeting of substrate proteins containing a CMA-targeting motif and the direct translocation into the lysosomal lumen, through the aid of chaperones/co-chaperones localized both at the cytosol and the lysosomes. CMA operates at basal conditions in most mammalian cell models analyzed so far, but it is mostly activated in response to stressors, such as trophic deprivation or oxidative stress. The activity of CMA has been shown to decline with age and such decline, correlating with accumulation of damaged/oxidized/aggregated proteins, may contribute to tissue dysfunction and, possibly, neurodegeneration. Herein, we review the recent knowledge regarding the molecular components, regulation and physiology of the CMA pathway, the contribution of impaired CMA activity to poor cellular homeostasis and inefficient response to stress during aging, and discuss the therapeutic opportunities offered by the restoration of CMA-dependent proteolysis in age-associated degenerative diseases.
Collapse
|
123
|
Colacurcio DJ, Nixon RA. Disorders of lysosomal acidification-The emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res Rev 2016; 32:75-88. [PMID: 27197071 DOI: 10.1016/j.arr.2016.05.004] [Citation(s) in RCA: 322] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis deliver unneeded cellular materials to lysosomes for degradation. Beyond processing cellular waste, lysosomes release metabolites and ions that serve signaling and nutrient sensing roles, linking the functions of the lysosome to various pathways for intracellular metabolism and nutrient homeostasis. Each of these lysosomal behaviors is influenced by the intraluminal pH of the lysosome, which is maintained in the low acidic range by a proton pump, the vacuolar ATPase (v-ATPase). New reports implicate altered v-ATPase activity and lysosomal pH dysregulation in cellular aging, longevity, and adult-onset neurodegenerative diseases, including forms of Parkinson disease and Alzheimer disease. Genetic defects of subunits composing the v-ATPase or v-ATPase-related proteins occur in an increasingly recognized group of familial neurodegenerative diseases. Here, we review the expanding roles of the v-ATPase complex as a platform regulating lysosomal hydrolysis and cellular homeostasis. We discuss the unique vulnerability of neurons to persistent low level lysosomal dysfunction and review recent clinical and experimental studies that link dysfunction of the v-ATPase complex to neurodegenerative diseases across the age spectrum.
Collapse
|
124
|
Ntsapi C, Loos B. Caloric restriction and the precision-control of autophagy: A strategy for delaying neurodegenerative disease progression. Exp Gerontol 2016; 83:97-111. [DOI: 10.1016/j.exger.2016.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 01/07/2023]
|
125
|
Huang L, Deng M, He Y, Lu S, Liu S, Fang Y. β-asarone increases MEF2D and TH levels and reduces α-synuclein level in 6-OHDA-induced rats via regulating the HSP70/MAPK/MEF2D/Beclin-1 pathway: Chaperone-mediated autophagy activation, macroautophagy inhibition and HSP70 up-expression. Behav Brain Res 2016; 313:370-379. [DOI: 10.1016/j.bbr.2016.07.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/13/2016] [Accepted: 07/17/2016] [Indexed: 12/31/2022]
|
126
|
Tyson T, Steiner JA, Brundin P. Sorting out release, uptake and processing of alpha-synuclein during prion-like spread of pathology. J Neurochem 2016; 139 Suppl 1:275-289. [PMID: 26617280 PMCID: PMC4958606 DOI: 10.1111/jnc.13449] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/17/2022]
Abstract
Parkinson's disease is a progressive neurological disorder that is characterized by the formation of intracellular protein inclusion bodies composed primarily of a misfolded and aggregated form of the protein α-synuclein. There is growing evidence that supports the prion-like hypothesis of α-synuclein progression. This hypothesis postulates that α-synuclein is a prion-like pathological agent and is responsible for the progression of Parkinson pathology in the brain. Potential misfolding or aggregation of α-synuclein that might occur in the peripheral nervous system as a result of some insult, environmental or genetic (or more likely a combination of both) that might spread into the midbrain, eventually causing degeneration of the neurons in the substantia nigra. As the disease progresses further, it is likely that α-synuclein pathology continues to spread throughout the brain, including the cortex, leading to deterioration of cognition and higher brain functions. While it is unknown why α-synuclein initially misfolds and aggregates, a great deal has been learned about how the cell handles aberrant α-synuclein assemblies. In this review, we focus on these mechanisms and discuss them in an attempt to define the role that they might play in the propagation of misfolded α-synuclein from cell-to-cell. The prion-like hypothesis of α-synuclein pathology suggests a method for the transmission of misfolded α-synuclein from one neuron to another. This hypothesis postulates that misfolded α-synuclein becomes aggregation prone and when released and taken up by neighboring cells, seeds further misfolding and aggregation. In this review we examine the cellular mechanisms that are involved in the processing of α-synuclein and how these may contribute to the prion-like propagation of α-synuclein pathology. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Trevor Tyson
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jennifer A Steiner
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA.
| |
Collapse
|
127
|
Xilouri M, Brekk OR, Polissidis A, Chrysanthou-Piterou M, Kloukina I, Stefanis L. Impairment of chaperone-mediated autophagy induces dopaminergic neurodegeneration in rats. Autophagy 2016; 12:2230-2247. [PMID: 27541985 DOI: 10.1080/15548627.2016.1214777] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Chaperone-mediated autophagy (CMA) involves the selective lysosomal degradation of cytosolic proteins such as SNCA (synuclein α), a protein strongly implicated in Parkinson disease (PD) pathogenesis. However, the physiological role of CMA and the consequences of CMA failure in the living brain remain elusive. Here we show that CMA inhibition in the adult rat substantia nigra via adeno-associated virus-mediated delivery of short hairpin RNAs targeting the LAMP2A receptor, involved in CMA's rate limiting step, was accompanied by intracellular accumulation of SNCA-positive puncta, which were also positive for UBIQUITIN, and in accumulation of autophagic vacuoles within LAMP2A-deficient nigral neurons. Strikingly, LAMP2A downregulation resulted in progressive loss of nigral dopaminergic neurons, severe reduction in striatal dopamine levels/terminals, increased astro- and microgliosis and relevant motor deficits. Thus, this study highlights for the first time the importance of the CMA pathway in the dopaminergic system and suggests that CMA impairment may underlie PD pathogenesis.
Collapse
Affiliation(s)
- Maria Xilouri
- a Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Oeystein Roed Brekk
- a Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens , Athens , Greece.,b University of Crete, School of Medicine , Heraklion , Crete , Greece
| | - Alexia Polissidis
- a Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Margarita Chrysanthou-Piterou
- c Research Unit of Histochemistry and Electron Microscopy , 1st Department of Psychiatry, University of Athens Medical School, Eginition Hospital , Athens , Greece
| | - Ismini Kloukina
- d Center of Basic Research, Biomedical Research Foundation of the Academy of Athens , Athens , Greece.,e Neurobiology Research Institute, Theodor Theohari Cozzika Foundation , Athens , Greece
| | - Leonidas Stefanis
- a Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens , Athens , Greece.,f Second Department of Neurology , National and Kapodistrian University of Athens Medical School , Athens , Greece
| |
Collapse
|
128
|
Smethurst DGJ, Cooper KF. ER fatalities-The role of ER-mitochondrial contact sites in yeast life and death decisions. Mech Ageing Dev 2016; 161:225-233. [PMID: 27507669 DOI: 10.1016/j.mad.2016.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/22/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
Following extracellular stress signals, all eukaryotic cells choose whether to elicit a pro-survival or pro-death response. The decision over which path to take is governed by the severity and duration of the damage. In response to mild stress, pro-survival programs are initiated (unfolded protein response, autophagy, mitophagy) whereas severe or chronic stress forces the cell to abandon these adaptive programs and shift towards regulated cell death to remove irreversibly damaged cells. Both pro-survival and pro-death programs involve regulated communication between the endoplasmic reticulum (ER) and mitochondria. In yeast, recent data suggest this inter-organelle contact is facilitated by the endoplasmic reticulum mitochondria encounter structure (ERMES). These membrane contacts are not only important for the exchange of cellular signals, but also play a role in mitochondrial tethering during mitophagy, mitochondrial fission and mitochondrial inheritance. This review focuses on recent findings in yeast that shed light on how ER-mitochondrial communication mediates critical cell fate decisions.
Collapse
Affiliation(s)
- Daniel G J Smethurst
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08055 USA
| | - Katrina F Cooper
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08055 USA.
| |
Collapse
|
129
|
|
130
|
Sala G, Marinig D, Riva C, Arosio A, Stefanoni G, Brighina L, Formenti M, Alberghina L, Colangelo AM, Ferrarese C. Rotenone down-regulates HSPA8/hsc70 chaperone protein in vitro: A new possible toxic mechanism contributing to Parkinson's disease. Neurotoxicology 2016; 54:161-169. [PMID: 27133439 DOI: 10.1016/j.neuro.2016.04.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/30/2016] [Accepted: 04/26/2016] [Indexed: 12/21/2022]
Abstract
HSPA8/hsc70 (70-kDa heat shock cognate) chaperone protein exerts multiple protective roles. Beside its ability to confer to the cells a generic resistance against several metabolic stresses, it is also involved in at least two critical processes whose activity is essential in preventing Parkinson's disease (PD) pathology. Actually, hsc70 protein acts as the main carrier of chaperone-mediated autophagy (CMA), a selective catabolic pathway for alpha-synuclein, the main pathogenic protein that accumulates in degenerating dopaminergic neurons in PD. Furthermore, hsc70 efficiently fragments alpha-synuclein fibrils in vitro and promotes depolymerization into non-toxic alpha-synuclein monomers. Considering that the mitochondrial complex I inhibitor rotenone, used to generate PD animal models, induces alpha-synuclein aggregation, this study was designed in order to verify whether rotenone exposure leads to hsc70 alteration possibly contributing to alpha-synuclein aggregation. To this aim, human SH-SY5Y neuroblastoma cells were treated with rotenone and hsc70 mRNA and protein expression were assessed; the effect of rotenone on hsc70 was compared with that exerted by hydrogen peroxide, a generic oxidative stress donor with no inhibitory activity on mitochondrial complex I. Furthermore, the effect of rotenone on hsc70 was verified in primary mouse cortical neurons. The possible contribution of macroautophagy to rotenone-induced hsc70 modulation was explored and the influence of hsc70 gene silencing on neurotoxicity was assessed. We demonstrated that rotenone, but not hydrogen peroxide, induced a significant reduction of hsc70 mRNA and protein expression. We also observed that the toxic effect of rotenone on alpha-synuclein levels was amplified when macroautophagy was inhibited, although rotenone-induced hsc70 reduction was independent from macroautophagy. Finally, we demonstrated that hsc70 gene silencing up-regulated alpha-synuclein mRNA and protein levels without affecting cell viability and without altering rotenone- and hydrogen peroxide-induced cytotoxicity. These findings demonstrate the existence of a novel mechanism of rotenone toxicity mediated by hsc70 and indicate that dysfunction of both CMA and macroautophagy can synergistically exacerbate alpha-synuclein toxicity, suggesting that hsc70 up-regulation may represent a valuable therapeutic strategy for PD.
Collapse
Affiliation(s)
- Gessica Sala
- Lab. of Neurobiology, School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy.
| | - Daniele Marinig
- Lab. of Neurobiology, School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy; PhD Program in Neuroscience, University of Milano-Bicocca, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Chiara Riva
- Lab. of Neurobiology, School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Alessandro Arosio
- Lab. of Neurobiology, School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Giovanni Stefanoni
- Lab. of Neurobiology, School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy; Dept. of Neurology, San Gerardo Hospital, Monza, Milano, Italy
| | - Laura Brighina
- Dept. of Neurology, San Gerardo Hospital, Monza, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Matteo Formenti
- Lab. of Neuroscience R. Levi-Montalcini, Dept. of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy; SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Milano, Italy
| | - Lilia Alberghina
- Lab. of Neuroscience R. Levi-Montalcini, Dept. of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy; SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Anna Maria Colangelo
- Lab. of Neuroscience R. Levi-Montalcini, Dept. of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy; SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| | - Carlo Ferrarese
- Lab. of Neurobiology, School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy; Dept. of Neurology, San Gerardo Hospital, Monza, Milano, Italy; NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
131
|
Vidoni C, Follo C, Savino M, Melone MAB, Isidoro C. The Role of Cathepsin D in the Pathogenesis of Human Neurodegenerative Disorders. Med Res Rev 2016; 36:845-70. [DOI: 10.1002/med.21394] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Chiara Vidoni
- Laboratory of Molecular Pathology, Department of Health Sciences; Università del Piemonte Orientale “A. Avogadro,”; Novara Italy
| | - Carlo Follo
- Laboratory of Molecular Pathology, Department of Health Sciences; Università del Piemonte Orientale “A. Avogadro,”; Novara Italy
| | - Miriam Savino
- Laboratory of Molecular Pathology, Department of Health Sciences; Università del Piemonte Orientale “A. Avogadro,”; Novara Italy
| | - Mariarosa A. B. Melone
- Division of Neurology, Department of Clinic and Experimental Medicine and Surgery; Second University of Naples; Naples Italy
- InterUniversity Center for Research in Neurosciences; Second University of Naples; Naples Italy
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences; Università del Piemonte Orientale “A. Avogadro,”; Novara Italy
- InterUniversity Center for Research in Neurosciences; Second University of Naples; Naples Italy
| |
Collapse
|
132
|
Caloric restriction alleviates alpha-synuclein toxicity in aged yeast cells by controlling the opposite roles of Tor1 and Sir2 on autophagy. Mech Ageing Dev 2016; 161:270-276. [PMID: 27109470 DOI: 10.1016/j.mad.2016.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/11/2016] [Accepted: 04/18/2016] [Indexed: 01/28/2023]
Abstract
Alpha-synuclein (syn) is the main component of proteinaceous inclusions known as Lewy bodies (LBs), which are implicated in the pathogenesis of the neurodegenerative diseases known as synucleinopathies, like Parkinson's disease (PD). Aging is a major risk factor for PD and thus, interventions that delay aging will have promising effects in PD and other synucleinopathies. Caloric restriction (CR) is the only non-genetic intervention shown to promote lifespan extension in several model organisms. CR has been shown to alleviate syn toxicity and herein we confirmed the same effect on the yeast model for synucleinopathies during chronological lifespan. The data gathered showed that TOR1 deletion also results in similar longevity extension and abrogation of syn toxicity. Intriguingly, these interventions were associated with decreased autophagy, which was maintained at homeostatic levels. Autophagy maintenance at homeostatic levels promoted by CR or TOR1 abrogation in syn-expressing cells was achieved by decreasing Sir2 levels and activity. Furthermore, the opposite function of Tor1 and Sir2 in autophagy is probably associated with the maintenance of autophagy activity at homeostatic levels, a central event linked to abrogation of syn toxicity promoted by CR.
Collapse
|
133
|
Migdalska-Richards A, Schapira AHV. The relationship between glucocerebrosidase mutations and Parkinson disease. J Neurochem 2016; 139 Suppl 1:77-90. [PMID: 26860875 PMCID: PMC5111601 DOI: 10.1111/jnc.13385] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/08/2015] [Accepted: 10/02/2015] [Indexed: 01/12/2023]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder after Alzheimer disease, whereas Gaucher disease (GD) is the most frequent lysosomal storage disorder caused by homozygous mutations in the glucocerebrosidase (GBA1) gene. Increased risk of developing PD has been observed in both GD patients and carriers. It has been estimated that GBA1 mutations confer a 20‐ to 30‐fold increased risk for the development of PD, and that at least 7–10% of PD patients have a GBA1 mutation. To date, mutations in the GBA1 gene constitute numerically the most important risk factor for PD. The type of PD associated with GBA1 mutations (PD‐GBA1) is almost identical to idiopathic PD, except for a slightly younger age of onset and a tendency to more cognitive impairment. Importantly, the pathology of PD‐GBA1 is identical to idiopathic PD, with nigral dopamine cell loss, Lewy bodies, and neurites containing alpha‐synuclein. The mechanism by which GBA1 mutations increase the risk for PD is still unknown. However, given that clinical manifestation and pathological findings in PD‐GBA1 patients are almost identical to those in idiopathic PD individuals, it is likely that, as in idiopathic PD, alpha‐synuclein accumulation, mitochondrial dysfunction, autophagic impairment, oxidative and endoplasmic reticulum stress may contribute to the development and progression of PD‐GBA1. Here, we review the GBA1 gene, its role in GD, and its link with PD.
The impact of glucocerebrosidase 1 (GBA1) mutations on functioning of endoplasmic reticulum (ER), lysosomes, and mitochondria. GBA1 mutations resulting in production of misfolded glucocerebrosidase (GCase) significantly affect the ER functioning. Misfolded GCase trapped in the ER leads to both an increase in the ubiquitin–proteasome system (UPS) and the ER stress. The presence of ER stress triggers the unfolded protein response (UPR) and/or endoplasmic reticulum‐associated degradation (ERAD). The prolonged activation of UPR and ERAD subsequently leads to increased apoptosis. The presence of misfolded GCase in the lysosomes together with a reduction in wild‐type GCase levels lead to a retardation of alpha‐synuclein degradation via chaperone‐mediated autophagy (CMA), which subsequently results in alpha‐synuclein accumulation and aggregation. Impaired lysosomal functioning also causes a decrease in the clearance of autophagosomes, and so their accumulation. GBA1 mutations perturb normal mitochondria functioning by increasing generation of free radical species (ROS) and decreasing adenosine triphosphate (ATP) production, oxygen consumption, and membrane potential. GBA1 mutations also lead to accumulation of dysfunctional and fragmented mitochondria.
This article is part of a special issue on Parkinson disease.
Collapse
|
134
|
Xilouri M, Brekk OR, Stefanis L. Autophagy and Alpha-Synuclein: Relevance to Parkinson's Disease and Related Synucleopathies. Mov Disord 2016; 31:178-92. [PMID: 26813776 DOI: 10.1002/mds.26477] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/21/2015] [Accepted: 10/25/2015] [Indexed: 12/14/2022] Open
Abstract
Evidence from human postmortem material, transgenic mice, and cellular/animal models of PD link alpha-synuclein accumulation to alterations in the autophagy lysosomal pathway. Conversely, alpha-synuclein mutations related to PD pathogenesis, as well as post-translational modifications of the wild-type protein, result in the generation of aberrant species that may impair further the function of the autophagy lysosomal pathway, thus generating a vicious cycle leading to neuronal death. Moreover, PD-linked mutations in lysosomal-related genes, such as glucocerebrosidase, have been also shown to contribute to alpha-synuclein accumulation and related toxicity, indicating that lysosomal dysfunction may, in part, account for the neurodegeneration observed in synucleinopathies. In the current review, we summarize findings related to the inter-relationship between alpha-synuclein and lysosomal proteolytic pathways, focusing especially on recent experimental strategies based on the manipulation of the autophagy lysosomal pathway to counteract alpha-synuclein-mediated neurotoxicity in vivo. Pinpointing the factors that regulate alpha-synuclein association to the lysosome may represent potential targets for therapeutic interventions in PD and related synucleinopathies.
Collapse
Affiliation(s)
- Maria Xilouri
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Oeystein Roed Brekk
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Leonidas Stefanis
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Second Department of Neurology, University of Athens Medical School, Athens, Greece
| |
Collapse
|
135
|
Spencer B, Kim C, Gonzalez T, Bisquertt A, Patrick C, Rockenstein E, Adame A, Lee SJ, Desplats P, Masliah E. α-Synuclein interferes with the ESCRT-III complex contributing to the pathogenesis of Lewy body disease. Hum Mol Genet 2016; 25:1100-15. [PMID: 26740557 DOI: 10.1093/hmg/ddv633] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/30/2015] [Indexed: 01/17/2023] Open
Abstract
α-Synuclein (α-syn) has been implicated in neurological disorders with parkinsonism, including Parkinson's disease and Dementia with Lewy body. Recent studies have shown α-syn oligomers released from neurons can propagate from cell-to-cell in a prion-like fashion exacerbating neurodegeneration. In this study, we examined the role of the endosomal sorting complex required for transport (ESCRT) pathway on the propagation of α-syn. α-syn, which is transported via the ESCRT pathway through multivesicular bodies for degradation, can also target the degradation of the ESCRT protein-charged multivesicular body protein (CHMP2B), thus generating a roadblock of endocytosed α-syn. Disruption of the ESCRT transport system also resulted in increased exocytosis of α-syn thus potentially increasing cell-to-cell propagation of synuclein. Conversely, delivery of a lentiviral vector overexpressing CHMP2B rescued the neurodegeneration in α-syn transgenic mice. Better understanding of the mechanisms of intracellular trafficking of α-syn might be important for understanding the pathogenesis and developing new treatments for synucleinopathies.
Collapse
Affiliation(s)
| | - Changyoun Kim
- Department of Neuroscience and Department of Medicine, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | | | | | | | | | | | - Seung-Jae Lee
- Department of Medicine, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | | | - Eliezer Masliah
- Department of Neuroscience and Department of Pathology, University of California, San Diego, San Diego, CA 92103, USA and
| |
Collapse
|
136
|
Poehler AM, Xiang W, Spitzer P, May VEL, Meixner H, Rockenstein E, Chutna O, Outeiro TF, Winkler J, Masliah E, Klucken J. Autophagy modulates SNCA/α-synuclein release, thereby generating a hostile microenvironment. Autophagy 2015; 10:2171-92. [PMID: 25484190 DOI: 10.4161/auto.36436] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
SNCA/α-synuclein aggregation plays a crucial role in synucleinopathies such as Parkinson disease and dementia with Lewy bodies. Aggregating and nonaggregating SNCA species are degraded by the autophagy-lysosomal pathway (ALP). Previously, we have shown that the ALP is not only responsible for SNCA degradation but is also involved in the intracellular aggregation process of SNCA. An additional role of extracellular SNCA in the pathology of synucleinopathies substantiating a prion-like propagation hypothesis has been suggested since released SNCA species and spreading of SNCA pathology throughout neural cells have been observed. However, the molecular interplay between intracellular pathways, SNCA aggregation, release, and response of the local microenvironment remains unknown. Here, we attributed SNCA-induced toxicity mainly to secreted species in a cell culture model of SNCA aggregation and in SNCA transgenic mice: We showed that ALP inhibition by bafilomycinA1 reduced intracellular SNCA aggregation but increased secretion of smaller oligomers that exacerbated microenvironmental response including uptake, inflammation, and cellular damage. Low-aggregated SNCA was predominantly released by exosomes and RAB11A-associated pathways whereas high-aggregated SNCA was secreted by membrane shedding. In summary, our study revealed a novel role of the ALP by linking protein degradation to nonclassical secretion for toxic SNCA species. Thus, impaired ALP in the diseased brain not only limits intracellular degradation of misfolded proteins, but also leads to a detrimental microenvironmental response due to enhanced SNCA secretion. These findings suggest that the major toxic role of SNCA is related to its extracellular species and further supports a protective role of intracellular SNCA aggregation.
Collapse
Key Words
- ACTB/bAct, actin, β
- AIF1/Iba1, allograft inflammatory factor 1
- AK, adenylate kinase
- ALP, autophagy-lysosomal pathway
- ANXA5, annexin A5
- BafA1, bafilomycinA1
- CA1, cornu ammonis field1
- CASP3/aCasp3, caspase-3
- CD63, CD63 molecule
- CM, conditioned medium
- CMA, chaperone-mediated autophagy
- CSF, cerebrospinal fluid
- DLB, dementia with Lewy bodies
- ER, endoplasmatic reticulum
- ESCRT, endosomal sorting complex required for transport
- EV, empty vector
- GFAP, glial fibrillary acidic protein
- HRP, horseradish peroxidase
- HSPA8/Hsc70, heat shock 70kDa protein 8
- Hippo, hippocampus
- IL6/IL-6, interleukin-6
- ILVs, intraluminal vesicles
- LAMP2A/Lamp2a, lysosomal-associated membrane protein 2, isoform A
- LB, Lewy bodies
- LN, Lewy neuritis
- MAP2, microtubule-associated protein 2
- ML, molecular layer
- MVBs, multivesicular bodies
- N, neuron
- Neoctx, neocortex
- PD, Parkinson disease
- PDGFB/PDGFb, platelet-derived growth factor subunit b
- PF, particle fraction
- PS, phosphatidylserine
- Parkinson disease
- RAB11A/rab11, member RAS oncogene family
- RBFOX3/NeuN, RNA binding protein, fox-1 homolog (C. elegans) 3
- RT, room temperature
- S100B/S100b, S100 calcium-binding protein B
- SL, stratum lacunosum; SNCA/aSyn
- SNCA-T, tagged α-synuclein
- SNCAIP/Sph1, synphilin-1
- SYP, synaptophysin
- TNF/TNFa, tumor necrosis factor α
- TUBB3/b-III-Tub, tubulin, β 3 class III
- UPS, ubiquitin proteasome system
- WT-SNCA, wild-type α-synuclein
- inflammation
- lysosomal degradation
- protein aggregation
- secretion
- synucleinopathies
- tg, transgenic
- α-synuclein
Collapse
Affiliation(s)
- Anne-Maria Poehler
- a Department of Molecular Neurology ; Friedrich-Alexander-University Erlangen-Nürnberg (FAU) ; Erlangen , Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Wu H, Chen S, Ammar AB, Xu J, Wu Q, Pan K, Zhang J, Hong Y. Crosstalk Between Macroautophagy and Chaperone-Mediated Autophagy: Implications for the Treatment of Neurological Diseases. Mol Neurobiol 2015; 52:1284-1296. [PMID: 25330936 PMCID: PMC4586010 DOI: 10.1007/s12035-014-8933-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/09/2014] [Indexed: 12/19/2022]
Abstract
Macroautophagy and chaperone-mediated autophagy (CMA) are two important subtypes of autophagy that play a critical role in cellular quality control under physiological and pathological conditions. Despite the marked differences between these two autophagic pathways, macroautophagy and CMA are intimately connected with each other during the autophagy-lysosomal degradation process, in particular, in the setting of neurological illness. Macroautophagy serves as a backup mechanism to removal of malfunctioning proteins (i.e., aberrant α-synuclein) from the cytoplasm when CMA is compromised, and vice versa. The molecular mechanisms underlying the conversation between macroautophagy and CMA are being clarified. Herein, we survey current overviews concentrating on the complex interactions between macroautophagy and CMA, and present therapeutic potentials through utilization and manipulation of macroautophagy-CMA crosstalk in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Al-Baadani Ammar
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kum Pan
- Department of Neurological Surgery, Weill Cornell Medical College, New York, USA
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan Hong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
138
|
Vieira BDM, Radford RA, Chung RS, Guillemin GJ, Pountney DL. Neuroinflammation in Multiple System Atrophy: Response to and Cause of α-Synuclein Aggregation. Front Cell Neurosci 2015; 9:437. [PMID: 26778958 PMCID: PMC4700780 DOI: 10.3389/fncel.2015.00437] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disease presenting with combinations of autonomic dysfunction, parkinsonism, cerebellar ataxia and/or pyramidal signs. Oligodendroglial cytoplasmic inclusions (GCIs) rich in α-synuclein (α-syn) constitute the disease hallmark, accompanied by neuronal loss and activation of glial cells which indicate neuroinflammation. Recent studies demonstrate that α-syn may be released from degenerating neurons to mediate formation of abnormal inclusion bodies and to induce neuroinflammation which, interestingly, might also favor the formation of intracellular α-syn aggregates as a consequence of cytokine release and the shift to a pro-inflammatory environment. Here, we critically review the relationships between α-syn and astrocytic and microglial activation in MSA to explore the potential of therapeutics which target neuroinflammation.
Collapse
Affiliation(s)
| | - Rowan A Radford
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Roger S Chung
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Dean L Pountney
- Menzies Health Institute Queensland, Griffith University Gold Coast, QLD, Australia
| |
Collapse
|
139
|
Papagiannakis N, Xilouri M, Koros C, Stamelou M, Antonelou R, Maniati M, Papadimitriou D, Moraitou M, Michelakakis H, Stefanis L. Lysosomal alterations in peripheral blood mononuclear cells of Parkinson's disease patients. Mov Disord 2015; 30:1830-4. [PMID: 26769460 DOI: 10.1002/mds.26433] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/21/2015] [Accepted: 08/23/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Reduced expression of lysosomal-associated membrane protein 2a and heatshock-cognate 70 proteins, involved in chaperone-mediated autophagy and of glucocerebrosidase, is reported in PD brains. The aim of this study was to identify systemic alterations in lysosomal-associated membrane protein 2a, heatshock cognate-70, and glucocerebrosidase levels/activity in peripheral blood mononuclear cells from PD patients. METHODS Protein/mRNA levels were assessed in PD patients from genetically undetermined background, alpha-synuclein (G209A/A53T), or glucocerebrosidase mutation carriers and age-/sex-matched controls. RESULTS Heatshock cognate 70 protein levels were reduced in all PD groups, whereas its mRNA levels were decreased only in the genetically undetermined group. Glucocerebrosidase protein levels were decreased only in the genetic PD groups, whereas increased mRNA levels and decreased activity were detected only in the glucocerebrosidase mutation group. CONCLUSIONS Reduced heatshock cognate-70 levels are suggestive of an apparent systemic chaperone-mediated autophagy dysfunction irrespective of genetic background. Glucocerebrosidase activity may serve as a screening tool to identify glucocerebrosidase mutation carriers with PD.
Collapse
Affiliation(s)
- Nikolaos Papagiannakis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Second Department of Neurology, University of Athens Medical School, Athens, Greece
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Christos Koros
- Second Department of Neurology, University of Athens Medical School, Athens, Greece
| | - Maria Stamelou
- Second Department of Neurology, University of Athens Medical School, Athens, Greece
| | - Roubina Antonelou
- Second Department of Neurology, University of Athens Medical School, Athens, Greece
| | - Matina Maniati
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Marina Moraitou
- Department of Enzymology and Cellular Function, Institute of Child Health, Athens, Greece
| | - Helen Michelakakis
- Department of Enzymology and Cellular Function, Institute of Child Health, Athens, Greece
| | - Leonidas Stefanis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Second Department of Neurology, University of Athens Medical School, Athens, Greece
| |
Collapse
|
140
|
Xiong R, Zhou W, Siegel D, Kitson RRA, Freed CR, Moody CJ, Ross D. A Novel Hsp90 Inhibitor Activates Compensatory Heat Shock Protein Responses and Autophagy and Alleviates Mutant A53T α-Synuclein Toxicity. Mol Pharmacol 2015; 88:1045-54. [PMID: 26405178 DOI: 10.1124/mol.115.101451] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/16/2015] [Indexed: 12/20/2022] Open
Abstract
A potential cause of neurodegenerative diseases, including Parkinson's disease (PD), is protein misfolding and aggregation that in turn leads to neurotoxicity. Targeting Hsp90 is an attractive strategy to halt neurodegenerative diseases, and benzoquinone ansamycin (BQA) Hsp90 inhibitors such as geldanamycin (GA) and 17-(allylamino)-17-demethoxygeldanamycin have been shown to be beneficial in mutant A53T α-synuclein PD models. However, current BQA inhibitors result in off-target toxicities via redox cycling and/or arylation of nucleophiles at the C19 position. We developed novel 19-substituted BQA (19BQA) as a means to prevent arylation. In this study, our data demonstrated that 19-phenyl-GA, a lead 19BQA in the GA series, was redox stable and exhibited little toxicity relative to its parent quinone GA in human dopaminergic SH-SY5Y cells as examined by oxygen consumption, trypan blue, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT), and apoptosis assays. Meanwhile, 19-phenyl-GA retained the ability to induce autophagy and potentially protective heat shock proteins (HSPs) such as Hsp70 and Hsp27. We found that transduction of A53T, but not wild type (WT) α-synuclein, induced toxicity in SH-SY5Y cells. 19-Phenyl-GA decreased oligomer formation and toxicity of A53T α-synuclein in transduced cells. Mechanistic studies indicated that mammalian target of rapamycin (mTOR)/p70 ribosomal S6 kinase signaling was activated by A53T but not WT α-synuclein, and 19-phenyl-GA decreased mTOR activation that may be associated with A53T α-synuclein toxicity. In summary, our results indicate that 19BQAs such as 19-phenyl-GA may provide a means to modulate protein-handling systems including HSPs and autophagy, thereby reducing the aggregation and toxicity of proteins such as mutant A53T α-synuclein.
Collapse
Affiliation(s)
- Rui Xiong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Wenbo Zhou
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Russell R A Kitson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Curt R Freed
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Christopher J Moody
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| |
Collapse
|
141
|
Villar-Piqué A, Lopes da Fonseca T, Outeiro TF. Structure, function and toxicity of alpha-synuclein: the Bermuda triangle in synucleinopathies. J Neurochem 2015; 139 Suppl 1:240-255. [PMID: 26190401 DOI: 10.1111/jnc.13249] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 06/29/2015] [Accepted: 07/14/2015] [Indexed: 12/11/2022]
Abstract
Parkinson's disease belongs to a group of currently incurable neurodegenerative disorders characterized by the misfolding and accumulation of alpha-synuclein aggregates that are commonly known as synucleinopathies. Clinically, synucleinopathies are heterogeneous, reflecting the somewhat selective neuronal vulnerability characteristic of each disease. The precise molecular underpinnings of synucleinopathies remain unclear, but the process of aggregation of alpha-synuclein appears as a central event. However, there is still no consensus with respect to the toxic forms of alpha-synuclein, hampering our ability to use the protein as a target for therapeutic intervention. To decipher the molecular bases of synucleinopathies, it is essential to understand the complex triangle formed between the structure, function and toxicity of alpha-synuclein. Recently, important steps have been undertaken to elucidate the role of the protein in both physiological and pathological conditions. Here, we provide an overview of recent findings in the field of alpha-synuclein research, and put forward a new perspective over paradigms that persist in the field. Establishing whether alpha-synuclein has a causative role in all synucleinopathies will enable the identification of targets for the development of novel therapeutic strategies for this devastating group of disorders. Alpha-synuclein is the speculated cornerstone of several neurodegenerative disorders known as Synucleinopathies. Nevertheless, the mechanisms underlying the pathogenic effects of this protein remain unknown. Here, we review the recent findings in the three corners of alpha-synuclein biology - structure, function and toxicity - and discuss the enigmatic roads that have accompanied alpha-synuclein from the beginning. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Anna Villar-Piqué
- Department of NeuroDegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| | - Tomás Lopes da Fonseca
- Department of NeuroDegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany.,Instituto de Fisiologia, Faculty of Medicine, University of Lisbon, Lisboa, Portugal
| | - Tiago Fleming Outeiro
- Department of NeuroDegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany. .,Instituto de Fisiologia, Faculty of Medicine, University of Lisbon, Lisboa, Portugal. .,CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
142
|
Büttner S, Broeskamp F, Sommer C, Markaki M, Habernig L, Alavian-Ghavanini A, Carmona-Gutierrez D, Eisenberg T, Michael E, Kroemer G, Tavernarakis N, Sigrist SJ, Madeo F. Spermidine protects against α-synuclein neurotoxicity. Cell Cycle 2015; 13:3903-8. [PMID: 25483063 PMCID: PMC4614020 DOI: 10.4161/15384101.2014.973309] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
As our society ages, neurodegenerative disorders like Parkinson`s disease (PD) are increasing in pandemic proportions. While mechanistic understanding of PD is advancing, a treatment with well tolerable drugs is still elusive. Here, we show that administration of the naturally occurring polyamine spermidine, which declines continuously during aging in various species, alleviates a series of PD-related degenerative processes in the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans, two established model systems for PD pathology. In the fruit fly, simple feeding with spermidine inhibited loss of climbing activity and early organismal death upon heterologous expression of human α-synuclein, which is thought to be the principal toxic trigger of PD. In this line, administration of spermidine rescued α-synuclein-induced loss of dopaminergic neurons, a hallmark of PD, in nematodes. Alleviation of PD-related neurodegeneration by spermidine was accompanied by induction of autophagy, suggesting that this cytoprotective process may be responsible for the beneficial effects of spermidine administration.
Collapse
Affiliation(s)
- Sabrina Büttner
- a Institute of Molecular Biosciences ; University of Graz ; Graz , Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Beilina A, Cookson MR. Genes associated with Parkinson's disease: regulation of autophagy and beyond. J Neurochem 2015. [PMID: 26223426 DOI: 10.1111/jnc.13266] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Substantial progress has been made in the genetic basis of Parkinson's disease (PD). In particular, by identifying genes that segregate with inherited PD or show robust association with sporadic disease, and by showing the same genes are found on both lists, we have generated an outline of the cause of this condition. Here, we will discuss what those genes tell us about the underlying biology of PD. We specifically discuss the relationships between protein products of PD genes and show that common links include regulation of the autophagy-lysosome system, an important way by which cells recycle proteins and organelles. We also discuss whether all PD genes should be considered to be in the same pathway and propose that in some cases the relationships are closer, whereas in other cases the interactions are more distant and might be considered separate. Beilina and Cookson review the links between genes for Parkinson's disease (red) and the autophagy-lysosomal system. They propose the hypothesis that many of the known PD genes can be assigned to pathways that affect (I) turnover of mitochondria via mitophagy (II) turnover of several vesicular structures via macroautophagy or chaperone-mediated autophagy or (III) general lysosome function. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Alexandra Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA.
| |
Collapse
|
144
|
Yap YW, Llanos RM, La Fontaine S, Cater MA, Beart PM, Cheung NS. Comparative Microarray Analysis Identifies Commonalities in Neuronal Injury: Evidence for Oxidative Stress, Dysfunction of Calcium Signalling, and Inhibition of Autophagy-Lysosomal Pathway. Neurochem Res 2015; 41:554-67. [PMID: 26318862 DOI: 10.1007/s11064-015-1666-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/14/2015] [Accepted: 07/14/2015] [Indexed: 12/21/2022]
Abstract
Mitochondrial dysfunction, ubiquitin-proteasomal system impairment and excitotoxicity occur during the injury and death of neurons in neurodegenerative conditions. The aim of this work was to elucidate the cellular mechanisms that are universally altered by these conditions. Through overlapping expression profiles of rotenone-, lactacystin- and N-methyl-D-aspartate-treated cortical neurons, we have identified three affected biological processes that are commonly affected; oxidative stress, dysfunction of calcium signalling and inhibition of the autophagic-lysosomal pathway. These data provides many opportunities for therapeutic intervention in neurodegenerative conditions, where mitochondrial dysfunction, proteasomal inhibition and excitotoxicity are evident.
Collapse
Affiliation(s)
- Yann Wan Yap
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Roxana M Llanos
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Sharon La Fontaine
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, VIC, 3125, Australia.,Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Michael A Cater
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Nam Sang Cheung
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, VIC, 3125, Australia.
| |
Collapse
|
145
|
Passeri E, Mocchetti I, Moussa C. Is human immunodeficiency virus-mediated dementia an autophagic defect that leads to neurodegeneration? CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2015; 13:1571-9. [PMID: 25106633 DOI: 10.2174/1871527313666140806125841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/04/2014] [Accepted: 06/06/2014] [Indexed: 11/22/2022]
Abstract
Autophagy is a cellular process that mediates selective degradation of cellular components in lysosomes. Autophagy may protect against neuronal apoptosis, which is induced in a number of neurodegenerative diseases. Thus, compounds that modulate autophagy could be beneficial to treat neurological disorders characterized by apoptosis such as Parkinson's and Alzheimer's diseases, as well as human-immunodeficiency virus-dementia complex. In this paper, we review new and old evidence on the role of autophagy in neuronal cell survival and we present evidence that humanimmunodeficiency virus may have adapted strategies to alter autophagic pathways in neurons. Moreover, we discuss the usefulness of drugs that facilitate autophagic clearance of proteins that are associated with neurodegeneration.
Collapse
Affiliation(s)
| | | | - Charbel Moussa
- Georgetown University Medical Center, Department of Neuroscience, NRB WP13, 3970 Reservoir Rd, NW, Washington, DC 20057, USA.
| |
Collapse
|
146
|
Latge C, Cabral KMS, de Oliveira GAP, Raymundo DP, Freitas JA, Johanson L, Romão LF, Palhano FL, Herrmann T, Almeida MS, Foguel D. The Solution Structure and Dynamics of Full-length Human Cerebral Dopamine Neurotrophic Factor and Its Neuroprotective Role against α-Synuclein Oligomers. J Biol Chem 2015; 290:20527-40. [PMID: 26149686 PMCID: PMC4536457 DOI: 10.1074/jbc.m115.662254] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/02/2015] [Indexed: 11/06/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is a promising therapeutic agent for Parkinson disease. As such, there has been great interest in studying its mode of action, which remains unknown. The three-dimensional crystal structure of the N terminus (residues 9-107) of CDNF has been determined, but there have been no published structural studies on the full-length protein due to proteolysis of its C-terminal domain, which is considered intrinsically disordered. An improved purification protocol enabled us to obtain active full-length CDNF and to determine its three-dimensional structure in solution. CDNF contains two well folded domains (residues 10-100 and 111-157) that are linked by a loop of intermediate flexibility. We identified two surface patches on the N-terminal domain that were characterized by increased conformational dynamics that should allow them to embrace active sites. One of these patches is formed by residues Ser-33, Leu-34, Ala-66, Lys-68, Ile-69, Leu-70, Ser-71, and Glu-72. The other includes a flexibly disordered N-terminal tail (residues 1-9), followed by the N-terminal portion of α-helix 1 (residues Cys-11, Glu-12, Val-13, Lys-15, and Glu-16) and residue Glu-88. The surface of the C-terminal domain contains two conserved active sites, which have previously been identified in mesencephalic astrocyte-derived neurotrophic factor, a CDNF paralog, which corresponds to its intracellular mode of action. We also showed that CDNF was able to protect dopaminergic neurons against injury caused by α-synuclein oligomers. This advises its use against physiological damages caused by α-synuclein oligomers, as observed in Parkinson disease and several other neurodegenerative diseases.
Collapse
Affiliation(s)
- Cristiane Latge
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Katia M S Cabral
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Guilherme A P de Oliveira
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Diana P Raymundo
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Julia A Freitas
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Laizes Johanson
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Luciana F Romão
- the UFRJ/Pólo Xerém, Universidade Federal do Rio de Janeiro 25245-390, Rio de Janeiro, Brazil
| | - Fernando L Palhano
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Torsten Herrmann
- the Institut des Sciences Analytiques, UMR 5280 CNRS, 5 rue de la Doua, 69100 Villeurbanne, France, and
| | - Marcius S Almeida
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil, the Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Debora Foguel
- From the Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil,
| |
Collapse
|
147
|
Jia J, Le W. Molecular network of neuronal autophagy in the pathophysiology and treatment of depression. Neurosci Bull 2015; 31:427-34. [PMID: 26254058 PMCID: PMC5563719 DOI: 10.1007/s12264-015-1548-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/17/2015] [Indexed: 12/14/2022] Open
Abstract
Major depressive disorder (MDD) is a complicated multifactorial induced disease, characterized by depressed mood, anhedonia, fatigue, and altered cognitive function. Recently, many studies have shown that antidepressants regulate autophagy. In fact, autophagy, a conserved lysosomal degradation pathway, is essential for the central nervous system. Dysregulation of autophagic pathways, such as the mammalian target of rapamycin (mTOR) signaling pathway and the beclin pathway, has been studied in neurodegenerative diseases. However, autophagy in MDD has not been fully studied. Here, we discuss whether the dysregulation of autophagy contributes to the pathophysiology and treatment of MDD and summarize the current evidence that shows the involvement of autophagy in MDD.
Collapse
Affiliation(s)
- Jack Jia
- Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA USA
- New Jersey Medical Institute, Trenton, NJ USA
| | - Weidong Le
- Center for Translational Research on Neurological Disease, First Affiliated Hospital, Dalian, 116011 China
| |
Collapse
|
148
|
Chaperone-mediated autophagy and neurodegeneration: connections, mechanisms, and therapeutic implications. Neurosci Bull 2015. [PMID: 26206600 DOI: 10.1007/s12264-015-1542-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Lysosomes degrade dysfunctional intracellular components via three pathways: macroautophagy, microautophagy, and chaperone-mediated autophagy (CMA). Unlike the other two, CMA degrades cytosolic proteins with a recognized KFERQ-like motif in lysosomes and is important for cellular homeostasis. CMA activity declines with age and is altered in neurodegenerative diseases. Its impairment leads to the accumulation of aggregated proteins, some of which may be directly tied to the pathogenic processes of neurodegenerative diseases. Its induction may accelerate the clearance of pathogenic proteins and promote cell survival, representing a potential therapeutic approach for the treatment of neurodegenerative diseases. In this review, we summarize the current findings on how CMA is involved in neurodegenerative diseases, especially in Parkinson's disease.
Collapse
|
149
|
Alpha-Synuclein affects neurite morphology, autophagy, vesicle transport and axonal degeneration in CNS neurons. Cell Death Dis 2015; 6:e1811. [PMID: 26158517 PMCID: PMC4650722 DOI: 10.1038/cddis.2015.169] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 12/23/2022]
Abstract
Many neuropathological and experimental studies suggest that the degeneration of dopaminergic terminals and axons precedes the demise of dopaminergic neurons in the substantia nigra, which finally results in the clinical symptoms of Parkinson disease (PD). The mechanisms underlying this early axonal degeneration are, however, still poorly understood. Here, we examined the effects of overexpression of human wildtype alpha-synuclein (αSyn-WT), a protein associated with PD, and its mutant variants αSyn-A30P and -A53T on neurite morphology and functional parameters in rat primary midbrain neurons (PMN). Moreover, axonal degeneration after overexpression of αSyn-WT and -A30P was analyzed by live imaging in the rat optic nerve in vivo. We found that overexpression of αSyn-WT and of its mutants A30P and A53T impaired neurite outgrowth of PMN and affected neurite branching assessed by Sholl analysis in a variant-dependent manner. Surprisingly, the number of primary neurites per neuron was increased in neurons transfected with αSyn. Axonal vesicle transport was examined by live imaging of PMN co-transfected with EGFP-labeled synaptophysin. Overexpression of all αSyn variants significantly decreased the number of motile vesicles and decelerated vesicle transport compared with control. Macroautophagic flux in PMN was enhanced by αSyn-WT and -A53T but not by αSyn-A30P. Correspondingly, colocalization of αSyn and the autophagy marker LC3 was reduced for αSyn-A30P compared with the other αSyn variants. The number of mitochondria colocalizing with LC3 as a marker for mitophagy did not differ among the groups. In the rat optic nerve, both αSyn-WT and -A30P accelerated kinetics of acute axonal degeneration following crush lesion as analyzed by in vivo live imaging. We conclude that αSyn overexpression impairs neurite outgrowth and augments axonal degeneration, whereas axonal vesicle transport and autophagy are severely altered.
Collapse
|
150
|
Jang W, Kim HJ, Li H, Jo KD, Lee MK, Yang HO. The Neuroprotective Effect of Erythropoietin on Rotenone-Induced Neurotoxicity in SH-SY5Y Cells Through the Induction of Autophagy. Mol Neurobiol 2015; 53:3812-3821. [PMID: 26156288 DOI: 10.1007/s12035-015-9316-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/23/2015] [Indexed: 12/25/2022]
Abstract
Currently, the autophagy pathway is thought to be important for the pathogenesis of Parkinson's disease (PD), and the modulation of autophagy may be a novel strategy for the treatment of this disease. Erythropoietin (EPO) has been reported to have neuroprotective effects through anti-oxidative, anti-apoptotic, and anti-inflammatory mechanisms, and it has also been shown to modulate autophagy signaling in an oxygen toxicity model. Therefore, we investigated the effects of EPO on autophagy markers and evaluated its neuroprotective effect on rotenone-induced neurotoxicity. We adapted the rotenone-induced neurotoxicity model to SH-SY5Y cells as an in vitro model of PD. We measured cell viability using MTT and annexin V/propidium iodide assays and measured intracellular levels of reactive oxygen species. Immunofluorescence analysis was performed to measure the expression of LC3 and α-synuclein. Intracellular signaling proteins associated with autophagy were examined by immunoblot analysis. EPO mono-treatment increased the levels of mammalian target of rapamycin (mTOR)-independent/upstream autophagy markers, including Beclin-1, AMPK, and ULK-1. Rotenone treatment of SH-SY5Y cells reduced their viability, increased reactive oxygen species levels, and induced apoptosis and α-synuclein expression, and simultaneous exposure to EPO significantly reduced these effects. Rotenone enhanced mTOR expression and suppressed Beclin-1 expression, indicating suppression of the autophagy system. However, combined treatment with EPO restored Beclin-1 expression and decreased mTOR expression. EPO protects against rotenone-induced neurotoxicity in SH-SY5Y cells by enhancing autophagy-related signaling pathways. The experimental evidence for the EPO-induced neuroprotection against rotenone-induced dopaminergic neurotoxicity may significantly impact the development of future PD treatment strategies.
Collapse
Affiliation(s)
- Wooyoung Jang
- Department of Neurology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Bangdong-ri, Sacheon-myeon, Gangneung, Gangwon-do, 210-711, Republic of Korea. .,Biomedical Research Center, Gangneung Asan Hospital, Bangdong-ri, Sacheon-myeon, Gangneung, 210-711, Gangwon-do, Republic of Korea.
| | - Hee Ju Kim
- Natural Products Research Center, Korea Institute of Science and Technology, 679 Saimdang-ro, Gangneung, 210-340, Republic of Korea.,Biomedical Research Center, Gangneung Asan Hospital, Bangdong-ri, Sacheon-myeon, Gangneung, 210-711, Gangwon-do, Republic of Korea
| | - Huan Li
- Natural Products Research Center, Korea Institute of Science and Technology, 679 Saimdang-ro, Gangneung, 210-340, Republic of Korea
| | - Kwang Deog Jo
- Department of Neurology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Bangdong-ri, Sacheon-myeon, Gangneung, Gangwon-do, 210-711, Republic of Korea
| | - Moon Kyu Lee
- Department of Neurology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Bangdong-ri, Sacheon-myeon, Gangneung, Gangwon-do, 210-711, Republic of Korea
| | - Hyun Ok Yang
- Natural Products Research Center, Korea Institute of Science and Technology, 679 Saimdang-ro, Gangneung, 210-340, Republic of Korea.
| |
Collapse
|