1
|
Nam Y, Shin SJ, Kumar V, Won J, Kim S, Moon M. Dual modulation of amyloid beta and tau aggregation and dissociation in Alzheimer's disease: a comprehensive review of the characteristics and therapeutic strategies. Transl Neurodegener 2025; 14:15. [PMID: 40133924 PMCID: PMC11938702 DOI: 10.1186/s40035-025-00479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Alzheimer's disease (AD) is not a single-cause disease; rather, it is a complex neurodegenerative disease involving multiple pathological pathways influenced by various risk factors. Aggregation and accumulation of amyloid beta (Aβ) and tau are the most prominent features in the brains of AD patients. Aggregated Aβ and tau exert neurotoxic effects in the central nervous system, contributing to the pathogenesis and progression of AD. They also act synergistically to cause neurodegeneration, resulting in memory loss. In this context, dual inhibition of Aβ and tau aggregation, or dissociation of these two aggregates, is considered promising for AD treatment. Recently, dual inhibitors capable of simultaneously targeting the aggregation and dissociation of both Aβ and tau have been investigated. Specific amino acid domains of Aβ and tau associated with their aggregation/dissociation have been identified. Subsequently, therapeutic agents that prevent aggregation or promote disaggregation by targeting these domains have been identified/developed. In this review, we summarize the major domains and properties involved in Aβ and tau aggregation, as well as the therapeutic effects and mechanisms of agents that simultaneously regulate their aggregation and dissociation. This comprehensive review may contribute to the design and discovery of next-generation dual-targeting drugs for Aβ and tau, potentially leading to the development of more effective therapeutic strategies for AD.
Collapse
Affiliation(s)
- Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Vijay Kumar
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Jihyeon Won
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea.
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea.
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea.
| |
Collapse
|
2
|
Iwaide S, Murakami T, Sedghi Masoud N, Kobayashi N, Fortin JS, Miyahara H, Higuchi K, Chambers JK. Classification of amyloidosis and protein misfolding disorders in animals 2024: A review on pathology and diagnosis. Vet Pathol 2025; 62:117-138. [PMID: 39389927 DOI: 10.1177/03009858241283750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Amyloidosis is a group of diseases in which proteins become amyloid, an insoluble fibrillar aggregate, resulting in organ dysfunction. Amyloid deposition has been reported in various animal species. To diagnose and understand the pathogenesis of amyloidosis, it is important to identify the amyloid precursor protein involved in each disease. Although 42 amyloid precursor proteins have been reported in humans, little is known about amyloidosis in animals, except for a few well-described amyloid proteins, including amyloid A (AA), amyloid light chain (AL), amyloid β (Aβ), and islet amyloid polypeptide-derived amyloid. Recently, several types of novel amyloidosis have been identified in animals using immunohistochemistry and mass spectrometry-based proteomic analysis. Certain species are predisposed to specific types of amyloidosis, suggesting a genetic background for its pathogenesis. Age-related amyloidosis has also emerged due to the increased longevity of captive animals. In addition, experimental studies have shown that some amyloids may be transmissible. Accurate diagnosis and understanding of animal amyloidosis are necessary for appropriate therapeutic intervention and comparative pathological studies. This review provides an updated classification of animal amyloidosis, including associated protein misfolding disorders of the central nervous system, and the current understanding of their pathogenesis. Pathologic features are presented together with state-of-the-art diagnostic methods that can be applied for routine diagnosis and identification of novel amyloid proteins in animals.
Collapse
Affiliation(s)
- Susumu Iwaide
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | - Tomoaki Murakami
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | | | | | | | | | - Keiichi Higuchi
- Shinshu University, Matsumoto, Japan
- Meio University, Nago, Japan
| | | |
Collapse
|
3
|
Sakunthala A, Maji SK. Deciphering the Seed Size-Dependent Cellular Internalization Mechanism for α-Synuclein Fibrils. Biochemistry 2025; 64:377-400. [PMID: 39762762 DOI: 10.1021/acs.biochem.4c00667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Aggregation of α-synuclein (α-Syn) and Lewy body (LB) formation are the key pathological events implicated in Parkinson's disease (PD) that spread in a prion-like manner. However, biophysical and structural characteristics of toxic α-Syn species and molecular events that drive early events in the propagation of α-Syn amyloids in a prion-like manner remain elusive. We used a neuronal cell model to demonstrate the size-dependent native biological activities of α-Syn fibril seeds. Biophysical characterization of the fibril seeds generated by controlled fragmentation indicated that increased fragmentation leads to a reduction in fibril size, correlating directly with the extent of fragmentation events. Although the size-based complexity of amyloid fibrils modulates their biological activities and fibril amplification pathways, it remains unclear how the variability of fibril seed size dictates its specific uptake mechanism into the cells. The present study elucidates the mechanism of α-Syn fibril internalization and how it is regulated by the size of fibril seeds. Further, we demonstrate that size-dependent endocytic pathways (dynamin-dependent clathrin/caveolin-mediated) are more prominent for the differential uptake of short fibril seeds compared to their longer counterparts. This size-dependent preference might contribute to the enhanced uptake and transcellular propagation of short α-Syn fibril seeds in a prion-like manner. Overall, the present study suggests that the physical dimension of α-Syn amyloid fibril seeds significantly influences their cellular uptake and pathological responses in the initiation and progression of PD.
Collapse
Affiliation(s)
- Arunima Sakunthala
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases (SCAN), Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
- Department of Biosciences& Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Samir K Maji
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases (SCAN), Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
- Department of Biosciences& Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
4
|
Wiersema AF, Rennenberg A, Smith G, Varderidou-Minasian S, Pasterkamp RJ. Shared and distinct changes in the molecular cargo of extracellular vesicles in different neurodegenerative diseases. Cell Mol Life Sci 2024; 81:479. [PMID: 39627617 PMCID: PMC11615177 DOI: 10.1007/s00018-024-05522-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/18/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024]
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD) affect millions of people worldwide. Curative treatment for these neurodegenerative disorders is still lacking and therefore a further understanding of their cause and progression is urgently needed. Extracellular vesicles (EVs) are nanosized vesicles loaded with cargo, such as proteins and miRNAs, that are released by cells and play an important role in intercellular communication. Intercellular communication through EVs can contribute to the spread of pathological proteins, such as amyloid-beta and tau, or cause pathogenesis through other mechanisms. In addition, EVs may serve as potential biomarkers for diagnosis and for monitoring disease progression. In this review, we summarize and discuss recent advances in our understanding of the role of EVs in AD, ALS an PD with an emphasis on dysregulated cargo in each disease. We highlight shared dysregulated cargo between these diseases, discuss underlying pathways, and outline future implications for therapeutic strategies.
Collapse
Affiliation(s)
- Anna F Wiersema
- Department of Translational Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Alyssa Rennenberg
- Department of Translational Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Grace Smith
- Department of Translational Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Suzy Varderidou-Minasian
- Department of Translational Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Brain Center, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
6
|
Moore K, Sengupta U, Puangmalai N, Bhatt N, Kayed R. Polymorphic Alpha-Synuclein Oligomers: Characterization and Differential Detection with Novel Corresponding Antibodies. Mol Neurobiol 2023; 60:2691-2705. [PMID: 36707462 PMCID: PMC9883140 DOI: 10.1007/s12035-023-03211-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023]
Abstract
The pathological hallmark of many neurodegenerative diseases is the accumulation of characteristic proteinaceous aggregates. Parkinson's disease and dementia with Lewy bodies can be characterized as synucleinopathies due to the abnormal accumulation of the protein alpha-synuclein (α-Syn). Studies have shown amyloidogenic proteins such as α-Syn and tau can exist as polymorphic aggregates, a theory widely studied mostly in their fibrillar morphology. It is now well understood that an intermediate state of aggregates, oligomers, are the most toxic species. We have shown α-Syn, when modified by different physiological inducers, result in distinct oligomeric conformations of α-Syn. Polymorphic α-Syn oligomers exhibit distinct properties such as aggregate size, conformation, and differentially interact with tau. In this study, we confirm α-Syn oligomeric polymorphs furthermore using in-house novel α-Syn toxic conformation monoclonal antibodies (SynTCs). It is unclear the biological relevance of α-Syn oligomeric polymorphisms. Utilizing a combination of biochemical, biophysical, and cell-based assays, we characterize α-Syn oligomeric polymorphs. We found α-Syn oligomeric polymorphs exhibit distinct immunoreactivity and SynTCs exhibit differential selectivity and binding affinity for α-Syn species. Isothermal titration calorimetry experiments suggest distinct α-Syn:SynTC binding enthalpies in a species-specific manner. Additionally, we found SynTCs differentially reduce α-Syn oligomeric polymorph-mediated neurotoxicity and propagation in primary cortical neurons in a polymorph-specific manner. These studies demonstrate the biological significance of polymorphic α-Syn oligomers along with the importance of polymorph-specific antibodies that target toxic α-Syn aggregates. Monoclonal antibodies that can target the conformational heterogeneity of α-Syn oligomeric species and reduce their mediated toxicity have promising immunotherapeutic potential.
Collapse
Affiliation(s)
- Kenya Moore
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, Neuroscience and Cell Biology, Medical Research Building Room 10.138C, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1045, USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, Neuroscience and Cell Biology, Medical Research Building Room 10.138C, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1045, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, Neuroscience and Cell Biology, Medical Research Building Room 10.138C, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1045, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, Neuroscience and Cell Biology, Medical Research Building Room 10.138C, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1045, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Neurology, Neuroscience and Cell Biology, Medical Research Building Room 10.138C, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1045, USA.
| |
Collapse
|
7
|
Sabari SS, Balasubramani K, Iyer M, Sureshbabu HW, Venkatesan D, Gopalakrishnan AV, Narayanaswamy A, Senthil Kumar N, Vellingiri B. Type 2 Diabetes (T2DM) and Parkinson's Disease (PD): a Mechanistic Approach. Mol Neurobiol 2023:10.1007/s12035-023-03359-y. [PMID: 37118323 PMCID: PMC10144908 DOI: 10.1007/s12035-023-03359-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
Growing evidence suggest that there is a connection between Parkinson's disease (PD) and insulin dysregulation in the brain, whilst the connection between PD and type 2 diabetes mellitus (T2DM) is still up for debate. Insulin is widely recognised to play a crucial role in neuronal survival and brain function; any changes in insulin metabolism and signalling in the central nervous system (CNS) can lead to the development of various brain disorders. There is accumulating evidence linking T2DM to PD and other neurodegenerative diseases. In fact, they have a lot in common patho-physiologically, including insulin dysregulation, oxidative stress resulting in mitochondrial dysfunction, microglial activation, and inflammation. As a result, initial research should focus on the role of insulin and its molecular mechanism in order to develop therapeutic outcomes. In this current review, we will look into the link between T2DM and PD, the function of insulin in the brain, and studies related to impact of insulin in causing T2DM and PD. Further, we have also highlighted the role of various insulin signalling pathway in both T2DM and PD. We have also suggested that T2DM-targeting pharmacological strategies as potential therapeutic approach for individuals with cognitive impairment, and we have demonstrated the effectiveness of T2DM-prescribed drugs through current PD treatment trials. In conclusion, this investigation would fill a research gap in T2DM-associated Parkinson's disease (PD) with a potential therapy option.
Collapse
Affiliation(s)
- S Sri Sabari
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Kiruthika Balasubramani
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to Be University), Coimbatore, 641021, Tamil Nadu, India
| | - Harysh Winster Sureshbabu
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014, India
| | - Arul Narayanaswamy
- Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Nachimuthu Senthil Kumar
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796004, Mizoram, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India.
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| |
Collapse
|
8
|
Zhang Y, Zhang C, He XZ, Li ZH, Meng JC, Mao RT, Li X, Xue R, Gui Q, Zhang GX, Wang LH. Interaction Between the Glymphatic System and α-Synuclein in Parkinson's Disease. Mol Neurobiol 2023; 60:2209-2222. [PMID: 36637746 DOI: 10.1007/s12035-023-03212-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023]
Abstract
The glymphatic system contributes to the clearance of amyloid-β from the brain and is disrupted in Alzheimer's disease. However, whether the system is involved in the removal of α-synuclein (α-syn) and whether it is suppressed in Parkinson's disease (PD) remain largely unknown. In mice receiving the intranigral injection of recombinant human α-syn, we found that the glymphatic suppression via aquaporin-4 (AQP4) gene deletion or acetazolamide treatment reduced the clearance of injected α-syn from the brain. In mice overexpressing the human A53T-α-syn, we revealed that AQP4 deficiency accelerated the accumulation of α-syn, facilitated the loss of dopaminergic neurons, and accelerated PD-like symptoms. We also found that the overexpression of A53T-α-syn reduced the expression/polarization of AQP4 and suppressed the glymphatic activity of mice. The study demonstrates a close interaction between the AQP4-mediated glymphatic system and parenchymal α-syn, indicating that restoring the glymphatic activity is a potential therapeutic target to delay PD progression.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Cui Zhang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Xu-Zhong He
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Zhen-Hua Li
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Jing-Cai Meng
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Rui-Ting Mao
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Xin Li
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Rong Xue
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Qian Gui
- Department of Neurology, Suzhou Municipal Hospital, 26 Dao-Qian Street, Suzhou, 215002, People's Republic of China
| | - Guo-Xing Zhang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Lin-Hui Wang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
9
|
Kim J, Daadi EW, Oh T, Daadi ES, Daadi MM. Human Induced Pluripotent Stem Cell Phenotyping and Preclinical Modeling of Familial Parkinson's Disease. Genes (Basel) 2022; 13:1937. [PMID: 36360174 PMCID: PMC9689743 DOI: 10.3390/genes13111937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 12/05/2022] Open
Abstract
Parkinson's disease (PD) is primarily idiopathic and a highly heterogenous neurodegenerative disease with patients experiencing a wide array of motor and non-motor symptoms. A major challenge for understanding susceptibility to PD is to determine the genetic and environmental factors that influence the mechanisms underlying the variations in disease-associated traits. The pathological hallmark of PD is the degeneration of dopaminergic neurons in the substantia nigra pars compacta region of the brain and post-mortem Lewy pathology, which leads to the loss of projecting axons innervating the striatum and to impaired motor and cognitive functions. While the cause of PD is still largely unknown, genome-wide association studies provide evidence that numerous polymorphic variants in various genes contribute to sporadic PD, and 10 to 15% of all cases are linked to some form of hereditary mutations, either autosomal dominant or recessive. Among the most common mutations observed in PD patients are in the genes LRRK2, SNCA, GBA1, PINK1, PRKN, and PARK7/DJ-1. In this review, we cover these PD-related mutations, the use of induced pluripotent stem cells as a disease in a dish model, and genetic animal models to better understand the diversity in the pathogenesis and long-term outcomes seen in PD patients.
Collapse
Affiliation(s)
- Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
| | - Etienne W. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Elyas S. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Marcel M. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
- Department of Radiology, Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
10
|
A light-inducible protein clustering system for in vivo analysis of α-synuclein aggregation in Parkinson disease. PLoS Biol 2022; 20:e3001578. [PMID: 35263320 PMCID: PMC8936469 DOI: 10.1371/journal.pbio.3001578] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 03/21/2022] [Accepted: 02/18/2022] [Indexed: 11/19/2022] Open
Abstract
Neurodegenerative disorders refer to a group of diseases commonly associated with abnormal protein accumulation and aggregation in the central nervous system. However, the exact role of protein aggregation in the pathophysiology of these disorders remains unclear. This gap in knowledge is due to the lack of experimental models that allow for the spatiotemporal control of protein aggregation, and the investigation of early dynamic events associated with inclusion formation. Here, we report on the development of a light-inducible protein aggregation (LIPA) system that enables spatiotemporal control of α-synuclein (α-syn) aggregation into insoluble deposits called Lewy bodies (LBs), the pathological hallmark of Parkinson disease (PD) and other proteinopathies. We demonstrate that LIPA-α-syn inclusions mimic key biochemical, biophysical, and ultrastructural features of authentic LBs observed in PD-diseased brains. In vivo, LIPA-α-syn aggregates compromise nigrostriatal transmission, induce neurodegeneration and PD-like motor impairments. Collectively, our findings provide a new tool for the generation, visualization, and dissection of the role of α-syn aggregation in PD.
Collapse
|
11
|
Jiang J, Liu Y, Wu Q. Revisit the Cellular Transmission and Emerging Techniques in Understanding the Mechanisms of Proteinopathies. Front Neurosci 2021; 15:781722. [PMID: 34867177 PMCID: PMC8636772 DOI: 10.3389/fnins.2021.781722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s and Parkinson’s diseases (AD and PD) are amongst top of the prevalent neurodegenerative disease. One-third of PD patients are diagnosed with dementia, a pre-symptom of AD, but the underlying mechanism is elusive. Amyloid beta (Aβ) and α-synuclein are two of the most investigated proteins, whose pathological aggregation and spreading are crucial to the pathogenesis of AD and PD, respectively. Transcriptomic studies of the mammalian central nervous system shed light on gene expression profiles at molecular levels, regarding the complexity of neuronal morphologies and electrophysiological inputs/outputs. In the last decade, the booming of the single-cell RNA sequencing technique helped to understand gene expression patterns, alternative splicing, novel transcripts, and signal pathways in the nervous system at single-cell levels, providing insight for molecular taxonomy and mechanistic targets of the degenerative nervous system. Here, we re-visited the cell-cell transmission mechanisms of Aβ and α-synuclein in mediating disease propagation, and summarized recent single-cell transcriptome sequencing from different perspectives and discussed its understanding of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jinwen Jiang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yu Liu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Qihui Wu
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Gadhe L, Sakunthala A, Mukherjee S, Gahlot N, Bera R, Sawner AS, Kadu P, Maji SK. Intermediates of α-synuclein aggregation: Implications in Parkinson's disease pathogenesis. Biophys Chem 2021; 281:106736. [PMID: 34923391 DOI: 10.1016/j.bpc.2021.106736] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/27/2021] [Accepted: 11/27/2021] [Indexed: 12/11/2022]
Abstract
Cytoplasmic deposition of aberrantly misfolded α-synuclein (α-Syn) is a common feature of synucleinopathies, including Parkinson's disease (PD). However, the precise pathogenic mechanism of α-Syn in synucleinopathies remains elusive. Emerging evidence has suggested that α-Syn may contribute to PD pathogenesis in several ways; wherein the contribution of fibrillar species, for exerting toxicity and disease transmission, cannot be neglected. Further, the oligomeric species could be the most plausible neurotoxic species causing neuronal cell death. However, understanding the structural and molecular insights of these oligomers are very challenging due to the heterogeneity and transient nature of the species. In this review, we discuss the recent advancements in understanding the formation and role of α-Syn oligomers in PD pathogenesis. We also summarize the different types of α-Syn oligomeric species and potential mechanisms to exert neurotoxicity. Finally, we address the possible ways to target α-Syn as a promising approach against PD and the possible future directions.
Collapse
Affiliation(s)
- Laxmikant Gadhe
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Riya Bera
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
13
|
Jellinger KA, Wenning GK, Stefanova N. Is Multiple System Atrophy a Prion-like Disorder? Int J Mol Sci 2021; 22:10093. [PMID: 34576255 PMCID: PMC8472631 DOI: 10.3390/ijms221810093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive, fatal neurodegenerative disease of uncertain aetiology that belongs to the family of α-synucleinopathies. It clinically presents with parkinsonism, cerebellar, autonomic, and motor impairment in variable combinations. Pathological hallmarks are fibrillary α-synuclein (αSyn)-rich glial cytoplasmic inclusions (GCIs) mainly involving oligodendroglia and to a lesser extent neurons, inducing a multisystem neurodegeneration, glial activation, and widespread demyelinization. The neuronal αSyn pathology of MSA has molecular properties different from Lewy bodies in Parkinson's disease (PD), both of which could serve as a pool of αSyn (prion) seeds that could initiate and drive the pathogenesis of synucleinopathies. The molecular cascade leading to the "prion-like" transfer of "strains" of aggregated αSyn contributing to the progression of the disease is poorly understood, while some presented evidence that MSA is a prion disease. However, this hypothesis is difficult to reconcile with postmortem analysis of human brains and the fact that MSA-like pathology was induced by intracerebral inoculation of human MSA brain homogenates only in homozygous mutant 53T mice, without production of disease-specific GCIs, or with replication of MSA prions in primary astrocyte cultures from transgenic mice expressing human αSyn. Whereas recent intrastriatal injection of Lewy body-derived or synthetic human αSyn fibrils induced PD-like pathology including neuronal αSyn aggregates in macaques, no such transmission of αSyn pathology in non-human primates by MSA brain lysate has been reported until now. Given the similarities between αSyn and prions, there is a considerable debate whether they should be referred to as "prions", "prion-like", "prionoids", or something else. Here, the findings supporting the proposed nature of αSyn as a prion and its self-propagation through seeding as well as the transmissibility of neurodegenerative disorders are discussed. The proof of disease causation rests on the concordance of scientific evidence, none of which has provided convincing evidence for the classification of MSA as a prion disease or its human transmission until now.
Collapse
Affiliation(s)
| | - Gregor K. Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| |
Collapse
|
14
|
Beekes M. The Neural Gut-Brain Axis of Pathological Protein Aggregation in Parkinson's Disease and Its Counterpart in Peroral Prion Infections. Viruses 2021; 13:1394. [PMID: 34372600 PMCID: PMC8310171 DOI: 10.3390/v13071394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/17/2022] Open
Abstract
A neuropathological hallmark of Parkinson's disease (PD) is the cerebral deposition of abnormally aggregated α-synuclein (αSyn). PD-associated αSyn (αSynPD) aggregates are assumed to act, in a prion-like manner, as proteinaceous nuclei ("seeds") capable of self-templated propagation. Braak and colleagues put forward the idea of a neural gut-brain axis mediating the centripetal spread of αSynPD pathology from the enteric nervous system (ENS) to the brain in PD. This has sparked great interest and initiated passionate discussions both in support of and opposing the suggested hypothesis. A precedent for the spread of protein seeds or seeding from the gastro-intestinal (GI) tract to the central nervous system (CNS) had been previously revealed for pathological prion protein in peroral prion infections. This article scrutinizes the similarities and dissimilarities between the pathophysiological spread of disease-associated protein aggregation along the neural gut-brain axis in peroral prion infections and PD. On this basis, evidence supporting the proposed neural gut-brain axis in PD is concluded to be not as robust as that established for peroral prion infections. New tools for the ultrasensitive detection of αSynPD-associated seeding activity in archived or fresh human tissue samples such as real-time quaking induced conversion (RT-QuIC) or protein misfolding cyclic amplification (PMCA) assays can possibly help to address this deficit in the future.
Collapse
Affiliation(s)
- Michael Beekes
- Prion and Prionoid Research Unit, ZBS 6-Proteomics and Spectroscopy, ZBS-Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany
| |
Collapse
|
15
|
Marsal-García L, Urbizu A, Arnaldo L, Campdelacreu J, Vilas D, Ispierto L, Gascón-Bayarri J, Reñé R, Álvarez R, Beyer K. Expression Levels of an Alpha-Synuclein Transcript in Blood May Distinguish between Early Dementia with Lewy Bodies and Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22020725. [PMID: 33450872 PMCID: PMC7828374 DOI: 10.3390/ijms22020725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 01/30/2023] Open
Abstract
Lewy body diseases (LBD) including dementia with Lewy bodies (DLB) and Parkinson disease (PD) are characterized by alpha-synuclein pathology. DLB is difficult to diagnose and peripheral biomarkers are urgently needed. Therefore, we analyzed the expression of five alpha-synuclein gene (SNCA) transcripts, SNCAtv1, SNCAtv2, SNCAtv3, SNCA126, and SNCA112, in 45 LBD and control temporal cortex samples and in the blood of 72 DLB, 59 PD, and 54 control subjects. The results revealed overexpression of SNCAtv1 and SNCA112 in DLB, and SNCAtv2 in PD temporal cortices. In DLB blood, diminution of all SNCA transcripts was observed. SNCAtv1 and SNCAtv2 were diminished in PD with disease onset before 70 years. SNCAtv3, driven by its own promoter, showed opposite expression in early DLB and PD, suggesting that its amount may be an early, DLB specific biomarker. Correlation between blood transcript levels and disease duration was positive in DLB and negative in PD, possibly reflecting differences in brain alpha-synuclein aggregation rates associated with differences in disease courses. In conclusion, SNCA transcripts showed a disease-specific increase in the brain and were diminished in blood of LBD patients. SNCAtv3 expression was decreased in early DLB and increased in early PD and could be a biomarker for early DLB diagnosis.
Collapse
Affiliation(s)
- Laura Marsal-García
- Department of Pathology, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (L.M.-G.); (A.U.); (L.A.)
| | - Aintzane Urbizu
- Department of Pathology, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (L.M.-G.); (A.U.); (L.A.)
| | - Laura Arnaldo
- Department of Pathology, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (L.M.-G.); (A.U.); (L.A.)
| | - Jaume Campdelacreu
- Servei de Neurologia, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (J.C.); (J.G.-B.); (R.R.)
| | - Dolores Vilas
- Servei de Neurologia, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (D.V.); (L.I.); (R.Á.)
| | - Lourdes Ispierto
- Servei de Neurologia, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (D.V.); (L.I.); (R.Á.)
| | - Jordi Gascón-Bayarri
- Servei de Neurologia, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (J.C.); (J.G.-B.); (R.R.)
| | - Ramón Reñé
- Servei de Neurologia, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (J.C.); (J.G.-B.); (R.R.)
| | - Ramiro Álvarez
- Servei de Neurologia, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (D.V.); (L.I.); (R.Á.)
| | - Katrin Beyer
- Department of Pathology, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain; (L.M.-G.); (A.U.); (L.A.)
- Correspondence: ; Tel.: +34-93-497-8355
| |
Collapse
|
16
|
Pinder P, Thomzig A, Schulz-Schaeffer WJ, Beekes M. Alpha-synuclein seeds of Parkinson's disease show high prion-exceeding resistance to steam sterilization. J Hosp Infect 2020; 108:25-32. [PMID: 33137444 DOI: 10.1016/j.jhin.2020.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Cerebral deposition of abnormally misfolded and aggregated alpha-synuclein (αSyn) is a neuropathological hallmark of Parkinson's disease (PD). Pathologically aggregated αSyn species of PD (αSynPD) can act, in a 'prion-like' manner, as proteinaceous nuclei ('seeds') which are capable of self-templated propagation. This has raised concerns that αSynPD seeds transmitted iatrogenically between humans may stimulate αSyn pathologies or clinically harmful effects in the recipients. Effective decontamination when reprocessing medical devices could significantly counteract such risks. Steam sterilization at 134°C is recommended as an essential pathogen inactivation step in many reprocessing guidelines for medical devices, and also shows effectiveness against prions, the self-propagating biological agents long thought to exhibit the highest resistance to steam sterilization. METHODS This study examined the reduction in αSynPD seeding activity in brain tissue homogenates from patients with PD after steam sterilization at 134°C using a specifically adapted real-time quaking induced conversion assay. FINDINGS Titres of approximately 1010 50% seeding doses per gram were detected in non-steam-sterilized caudate nucleus tissue of patients with PD by endpoint titration. Five minutes of steam sterilization reduced this titre by only 2.25 ± 0.15 decadic-logarithmic units, with an extension of the sterilization time to 90 min not causing additional inactivation. These findings reveal that αSynPD species are disease-associated biological agents with seeding activity that has higher resistance to steam sterilization than prions. CONCLUSION The remarkable heat resistance of αSynPD seeds calls for thoroughly validated cleaning and disinfection methods that reliably remove or inactivate possible contaminations of seeding-active αSyn aggregates when reprocessing medical devices.
Collapse
Affiliation(s)
- P Pinder
- Prion and Prionoid Research Unit, ZBS 6 - Proteomics and Spectroscopy, ZBS - Centre for Biological Threads and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - A Thomzig
- Prion and Prionoid Research Unit, ZBS 6 - Proteomics and Spectroscopy, ZBS - Centre for Biological Threads and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - W J Schulz-Schaeffer
- Institute of Neuropathology, Saarland University Medical Centre, Homburg, Germany
| | - M Beekes
- Prion and Prionoid Research Unit, ZBS 6 - Proteomics and Spectroscopy, ZBS - Centre for Biological Threads and Special Pathogens, Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
17
|
Henríquez G, Mendez L, Schmid AN, Guerrero ED, Collins SA, Castañeda E, Narayan M. Testing Amyloid Cross-Toxicity in the Vertebrate Brain. ACS OMEGA 2020; 5:15586-15591. [PMID: 32637834 PMCID: PMC7331027 DOI: 10.1021/acsomega.0c01819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/28/2020] [Indexed: 05/05/2023]
Abstract
While amyloid proteins such as amyloid β (Aβ),α-synuclein, tau, and lysozyme are known to be prion-like; emerging data have revealed that they are also able to seed the misfolding of prion-like proteins differing in sequence. In the present study, we have developed a tool designed to test neurohistochemical outcomes associated with the entry of an amyloid protein into heterotypic neurons, i.e., neurons that do not express the invading amyloid and, instead, endogenously express amyloids differing in sequence. The stereotaxic introduction of Aβ into the rodent tegmental area of the mid-brain revealed that the foreign amyloid had infiltrated into nigral neurons. Furthermore, Aβ was found colocalized with α-synuclein, an amyloid endogenous to the substantia nigra and differing in sequence relative to Aβ. Disruption of α-synuclein status in the substantia nigra is associated with Parkinson's disease onset and progress. In addition to the study findings, a significant inroad to future neurodegenerative research was made via the stereotaxic introduction of the foreign amyloid. This technique limits the presence of confounding neurometabolic variables that may be prevalent in transgenic animal models of cross-toxicity and, thereby, better addresses the role of individual neuronal factors in cross-toxicity. Finally, the data from this work may help reconcile the high frequency of clinical comorbidity seen in neurodegenerative diseases.
Collapse
Affiliation(s)
- Gabriela Henríquez
- Department
of Environmental Science and Engineering, The University of Texas at El Paso (UTEP), 500 W. University Avenue, El Paso, Texas 79968, United States
| | - Lois Mendez
- Department
of Chemistry and Biochemistry, The University
of Texas at El Paso (UTEP), 500 W. University Avenue, El Paso, Texas 79968, United
States
| | - Ariel N. Schmid
- Department
of Chemistry and Biochemistry, The University
of Texas at El Paso (UTEP), 500 W. University Avenue, El Paso, Texas 79968, United
States
| | - Erick D. Guerrero
- Department
of Chemistry and Biochemistry, The University
of Texas at El Paso (UTEP), 500 W. University Avenue, El Paso, Texas 79968, United
States
| | - Stephen A. Collins
- Department
of Psychology, The University of Texas at
El Paso (UTEP), 500 W.
University Avenue, El Paso, Texas 79968, United
States
| | - Edward Castañeda
- Department
of Psychology, The University of Texas at
El Paso (UTEP), 500 W.
University Avenue, El Paso, Texas 79968, United
States
| | - Mahesh Narayan
- Department
of Chemistry and Biochemistry, The University
of Texas at El Paso (UTEP), 500 W. University Avenue, El Paso, Texas 79968, United
States
| |
Collapse
|
18
|
Lucas HR, Fernández RD. Navigating the dynamic landscape of alpha-synuclein morphology: a review of the physiologically relevant tetrameric conformation. Neural Regen Res 2020; 15:407-415. [PMID: 31571649 PMCID: PMC6921358 DOI: 10.4103/1673-5374.265792] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
N-acetylated α-synuclein (αSyn) has long been established as an intrinsically disordered protein associated with a dysfunctional role in Parkinson's disease. In recent years, a physiologically relevant, higher order conformation has been identified as a helical tetramer that is tailored by buried hydrophobic interactions and is distinctively aggregation resistant. The canonical mechanism by which the tetramer assembles remains elusive. As novel biochemical approaches, computational methods, pioneering purification platforms, and powerful imaging techniques continue to develop, puzzling information that once sparked debate as to the veracity of the tetramer has now shed light upon this new counterpart in αSyn neurobiology. Nuclear magnetic resonance and computational studies on multimeric αSyn structure have revealed that the protein folding propensity is controlled by small energy barriers that enable large scale reconfiguration. Alternatively, familial mutations ablate tetramerization and reconfigure polymorphic fibrillization. In this review, we will discuss the dynamic landscape of αSyn quaternary structure with a focus on the tetrameric conformation.
Collapse
Affiliation(s)
- Heather R Lucas
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Ricardo D Fernández
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
19
|
Analysis of the Relationship between Type II Diabetes Mellitus and Parkinson's Disease: A Systematic Review. PARKINSONS DISEASE 2019; 2019:4951379. [PMID: 31871617 PMCID: PMC6906831 DOI: 10.1155/2019/4951379] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 12/31/2022]
Abstract
In the early sixties, a discussion started regarding the association between Parkinson's disease (PD) and type II diabetes mellitus (T2DM). Today, this potential relationship is still a matter of debate. This review aims to analyze both diseases concerning causal relationships and treatments. A total of 104 articles were found, and studies on animal and “in vitro” models showed that T2DM causes neurological alterations that may be associated with PD, such as deregulation of the dopaminergic system, a decrease in the expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), an increase in the expression of phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes 15 (PED/PEA-15), and neuroinflammation, as well as acceleration of the formation of alpha-synuclein amyloid fibrils. In addition, clinical studies described that Parkinson's symptoms were notably worse after the onset of T2DM, and seven deregulated genes were identified in the DNA of T2DM and PD patients. Regarding treatment, the action of antidiabetic drugs, especially incretin mimetic agents, seems to confer certain degree of neuroprotection to PD patients. In conclusion, the available evidence on the interaction between T2DM and PD justifies more robust clinical trials exploring this interaction especially the clinical management of patients with both conditions.
Collapse
|
20
|
Herrera-Vaquero M, Bouquio D, Kallab M, Biggs K, Nair G, Ochoa J, Heras-Garvin A, Heid C, Hadrovic I, Poewe W, Wenning GK, Klärner FG, Schrader T, Bitan G, Stefanova N. The molecular tweezer CLR01 reduces aggregated, pathologic, and seeding-competent α-synuclein in experimental multiple system atrophy. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165513. [PMID: 31319154 DOI: 10.1016/j.bbadis.2019.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/09/2019] [Accepted: 07/12/2019] [Indexed: 11/26/2022]
Abstract
Multiple system atrophy (MSA) is a fatal, adult-onset neurodegenerative disorder that has no cure and very limited treatment options. MSA is characterized by deposition of fibrillar α-synuclein (α-syn) in glial cytoplasmic inclusions in oligodendrocytes. Similar to other synucleinopathies, α-syn self-assembly is thought to be a key pathologic event and a prominent target for disease modification in MSA. Molecular tweezers are broad-spectrum nanochaperones that prevent formation of toxic protein assemblies and enhance their clearance. The current lead compound, CLR01, has been shown to inhibit α-syn aggregation but has not yet been tested in the context of MSA. To fill this gap, here, we conducted a proof-of-concept study to assess the efficacy of CLR01 in remodeling MSA-like α-syn pathology in the PLP-α-syn mouse model of MSA. Six-month-old mice received intracerebroventricular CLR01 (0.3 or 1 mg/kg per day) or vehicle for 32 days. Open-field test revealed a significant, dose-dependent amelioration of an anxiety-like phenotype. Subsequently, immunohistochemical and biochemical analyses showed dose-dependent reduction of pathological and seeding-competent forms of α-syn, which correlated with the behavioral phenotype. CLR01 treatment also promoted dopaminergic neuron survival in the substantia nigra. To our knowledge, this is the first demonstration of an agent that reduces formation of putative high-molecular-weight oligomers and seeding-competent α-syn in a mouse model of MSA, supporting the view that these species are key to the neurodegenerative process and its cell-to-cell progression in MSA. Our study suggests that CLR01 is an attractive therapeutic candidate for disease modification in MSA and related synucleinopathies, supporting further preclinical development.
Collapse
Affiliation(s)
- Marcos Herrera-Vaquero
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | - Danielle Bouquio
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Martin Kallab
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | - Karl Biggs
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Gayatri Nair
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jessica Ochoa
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Antonio Heras-Garvin
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | - Christian Heid
- Institute of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Inesa Hadrovic
- Institute of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Werner Poewe
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | - Gregor K Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | | | - Thomas Schrader
- Institute of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria.
| |
Collapse
|
21
|
|
22
|
Limatola A, Eichmann C, Jacob RS, Ben-Nissan G, Sharon M, Binolfi A, Selenko P. Time-Resolved NMR Analysis of Proteolytic α-Synuclein Processing in vitro and in cellulo. Proteomics 2018; 18:e1800056. [PMID: 30260559 DOI: 10.1002/pmic.201800056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/14/2018] [Indexed: 11/07/2022]
Abstract
Targeted proteolysis of the disordered Parkinson's disease protein alpha-synuclein (αSyn) constitutes an important event under physiological and pathological cell conditions. In this work, site-specific αSyn cleavage by different endopeptidases in vitro and by endogenous proteases in extracts of challenged and unchallenged cells was studied by time-resolved NMR spectroscopy. Specifically, proteolytic processing was monitored under neutral and low pH conditions and in response to Rotenone-induced oxidative stress. Further, time-dependent degradation of electroporation-delivered αSyn in intact SH-SY5Y and A2780 cells was analyzed. Results presented here delineate a general framework for NMR-based proteolysis studies in vitro and in cellulo, and confirm earlier reports pertaining to the exceptional proteolytic stability of αSyn under physiological cell conditions. However, experimental findings also reveal altered protease susceptibilities in selected mammalian cell lines and upon induced cell stress.
Collapse
Affiliation(s)
- Antonio Limatola
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Department of Biology, Stanford University, Stanford, CA, 94305-5430, USA
| | - Cédric Eichmann
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany
| | - Reeba Susan Jacob
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Department of Biological Regulation, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| | - Andres Binolfi
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET) and Plataforma Argentina de Biología Estructural y Metabolómica (PLABEM), Ocampo y Esmeralda, 2000, Rosario, Argentina
| | - Philipp Selenko
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Department of Biological Regulation, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| |
Collapse
|
23
|
Jiang T, Li G, Xu J, Gao S, Chen X. The Challenge of the Pathogenesis of Parkinson's Disease: Is Autoimmunity the Culprit? Front Immunol 2018; 9:2047. [PMID: 30319601 PMCID: PMC6170625 DOI: 10.3389/fimmu.2018.02047] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022] Open
Abstract
The role of autoimmunity in Parkinson's disease (PD), as one of the most popular research subjects, has been intensively investigated in recent years. Although the ultimate cause of PD is unknown, one major area of interest remains identifying new therapeutic targets and options for patients suffering from PD. Herein, we present a comprehensive review of the impacts of autoimmunity in neurodegenerative diseases, especially PD, and we have composed a logical argument to substantiate that autoimmunity is actively involved in the pathogenesis of PD through several proteins, including α-synuclein, DJ-1, PINK1, and Parkin, as well as immune cells, such as dendritic cells, microglia, T cells, and B cells. Furthermore, a detailed analysis of the relevance of autoimmunity to the clinical symptoms of PD provides strong evidence for the close correlation of autoimmunity with PD. In addition, the previously identified relationships between other autoimmune diseases and PD help us to better understand the disease pattern, laying the foundation for new therapeutic solutions to PD. In summary, this review aims to integrate and present currently available data to clarify the pathogenesis of PD and discuss some controversial but innovative research perspectives on the involvement of autoimmunity in PD, as well as possible novel diagnostic methods and treatments based on autoimmunity targets.
Collapse
Affiliation(s)
- Tianfang Jiang
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiang Su University, Shanghai, China
| | - Gen Li
- Department of Neurology & Institute of Neurology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Xu
- East Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Shane Gao
- East Hospital, Tong Ji University School of Medicine, Shanghai, China
| | - Xu Chen
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiang Su University, Shanghai, China
| |
Collapse
|
24
|
Terada M, Suzuki G, Nonaka T, Kametani F, Tamaoka A, Hasegawa M. The effect of truncation on prion-like properties of α-synuclein. J Biol Chem 2018; 293:13910-13920. [PMID: 30030380 PMCID: PMC6130941 DOI: 10.1074/jbc.ra118.001862] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/15/2018] [Indexed: 11/06/2022] Open
Abstract
Increasing evidence suggests that α-synuclein (αS) aggregates in brains of individuals with Parkinson's disease and dementia with Lewy bodies can spread in a prion-like manner. Although the initial αS nuclei are pivotal in determining αS fibril polymorphs and resulting phenotypes, it is not clear how the initial fibril seeds are generated. Previous studies have shown that αS truncation might have an important role in αS aggregation. However, little is known about how this truncation influences αS's propagation properties. In the present study, we generated αS fibrils from a series of truncated human αS constructs, characterized their structures and conformational stabilities, and investigated their ability to convert the conformation of full-length αS in vitro, in cultured cells, and in WT mice. We show that both C- and N-terminal truncations of human αS induce fibril polymorphs and exhibit different cross-seeding activities. N-terminally 10- or 30-residue-truncated human αS fibrils induced more abundant αS pathologies than WT fibrils in mice, whereas other truncated fibrils induced less abundant pathologies. Biochemical analyses of these truncated fibrils revealed that N-terminal 10- or 30-residue truncations of human αS change the fibril conformation in a manner that increases their structural compatibility with WT mouse αS fibrils and reduces their stability. C-terminally 20-residue-truncated fibrils displayed enhanced seeding activity in vitro Our findings imply that truncation of αS can influence its prion-like pathogenicity, resulting in phenotypic diversity of α-synucleinopathies.
Collapse
Affiliation(s)
- Makoto Terada
- From the Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan and
- Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Genjiro Suzuki
- From the Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan and
| | - Takashi Nonaka
- From the Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan and
| | - Fuyuki Kametani
- From the Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan and
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Masato Hasegawa
- From the Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan and
| |
Collapse
|
25
|
Quinolinic Acid Amyloid-like Fibrillar Assemblies Seed α-Synuclein Aggregation. J Mol Biol 2018; 430:3847-3862. [PMID: 30098337 DOI: 10.1016/j.jmb.2018.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/17/2018] [Accepted: 08/01/2018] [Indexed: 01/08/2023]
Abstract
Quinolinic acid (QA), a downstream neurometabolite in the kynurenine pathway, the biosynthetic pathway of tryptophan, is associated with neurodegenerative diseases pathology. Mutations in genes encoding kynurenine pathway enzymes, which control the level of QA production, are linked with elevated risk of developing Parkinson's disease. Recent findings have revealed the accumulation and deposition of QA in post-mortem samples, as well as in cellular models of Alzheimer's disease and related disorders. Furthermore, intrastriatal inoculation of mice with QA results in increased levels of phosphorylated α-synuclein and neurodegenerative pathological and behavioral characteristics. However, the cellular and molecular mechanisms underlying the involvement of QA accumulation in protein aggregation and neurodegeneration remain elusive. We recently established that self-assembled ordered structures are formed by various metabolites and hypothesized that these "metabolite amyloids" may seed amyloidogenic proteins. Here we demonstrate the formation of QA amyloid-like fibrillar assemblies and seeding of α-synuclein aggregation by these nanostructures both in vitro and in cell culture. Notably, α-synuclein aggregation kinetics was accelerated by an order of magnitude. Additional amyloid-like properties of QA assemblies were demonstrated using thioflavin T assay, powder X-ray diffraction and cell apoptosis analysis. Moreover, fluorescently labeled QA assemblies were internalized by neuronal cells and co-localized with α-synuclein aggregates. In addition, we observed cell-to-cell propagation of fluorescently labeled QA assemblies in a co-culture of treated and untreated cells. Our findings suggest that excess QA levels, due to mutations in the kynurenine pathway, for example, may lead to the formation of metabolite assemblies that seed α-synuclein aggregation, resulting in neuronal toxicity and induction of Parkinson's disease.
Collapse
|
26
|
Tang BL. Unconventional Secretion and Intercellular Transfer of Mutant Huntingtin. Cells 2018; 7:59. [PMID: 29904030 PMCID: PMC6025013 DOI: 10.3390/cells7060059] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 01/17/2023] Open
Abstract
The mechanism of intercellular transmission of pathological agents in neurodegenerative diseases has received much recent attention. Huntington's disease (HD) is caused by a monogenic mutation in the gene encoding Huntingtin (HTT). Mutant HTT (mHTT) harbors a CAG repeat extension which encodes an abnormally long polyglutamine (polyQ) repeat at HTT's N-terminus. Neuronal pathology in HD is largely due to the toxic gain-of-function by mHTT and its proteolytic products, which forms both nuclear and cytoplasmic aggregates that perturb nuclear gene transcription, RNA splicing and transport as well cellular membrane dynamics. The neuropathological effects of mHTT have been conventionally thought to be cell-autonomous in nature. Recent findings have, however, indicated that mHTT could be secreted by neurons, or transmitted from one neuronal cell to another via different modes of unconventional secretion, as well as via tunneling nanotubes (TNTs). These modes of transmission allow the intercellular spread of mHTT and its aggregates, thus plausibly promoting neuropathology within proximal neuronal populations and between neurons that are connected within neural circuits. Here, the various possible modes for mHTT's neuronal cell exit and intercellular transmission are discussed.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, 117597 Singapore, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, 117456 Singapore, Singapore.
| |
Collapse
|
27
|
α-synuclein oligomers interact with ATP synthase and open the permeability transition pore in Parkinson's disease. Nat Commun 2018; 9:2293. [PMID: 29895861 PMCID: PMC5997668 DOI: 10.1038/s41467-018-04422-2] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 04/20/2018] [Indexed: 12/18/2022] Open
Abstract
Protein aggregation causes α-synuclein to switch from its physiological role to a pathological toxic gain of function. Under physiological conditions, monomeric α-synuclein improves ATP synthase efficiency. Here, we report that aggregation of monomers generates beta sheet-rich oligomers that localise to the mitochondria in close proximity to several mitochondrial proteins including ATP synthase. Oligomeric α-synuclein impairs complex I-dependent respiration. Oligomers induce selective oxidation of the ATP synthase beta subunit and mitochondrial lipid peroxidation. These oxidation events increase the probability of permeability transition pore (PTP) opening, triggering mitochondrial swelling, and ultimately cell death. Notably, inhibition of oligomer-induced oxidation prevents the pathological induction of PTP. Inducible pluripotent stem cells (iPSC)-derived neurons bearing SNCA triplication, generate α-synuclein aggregates that interact with the ATP synthase and induce PTP opening, leading to neuronal death. This study shows how the transition of α-synuclein from its monomeric to oligomeric structure alters its functional consequences in Parkinson's disease.
Collapse
|
28
|
Abstract
Parkinson's disease (PD) is characterized by intracellular inclusions of aggregated and misfolded α-Synuclein (α-Syn), and the loss of dopaminergic (DA) neurons in the brain. The resulting motor abnormalities mark the progression of PD, while non-motor symptoms can already be identified during early, prodromal stages of disease. Recent studies provide evidence that during this early prodromal phase, synaptic and axonal abnormalities occur before the degenerative loss of neuronal cell bodies. These early phenotypes can be attributed to synaptic accumulation of toxic α-Syn. Under physiological conditions, α-Syn functions in its native conformation as a soluble monomer. However, PD patient brains are characterized by intracellular inclusions of insoluble fibrils. Yet, oligomers and protofibrils of α-Syn have been identified to be the most toxic species, with their accumulation at presynaptic terminals affecting several steps of neurotransmitter release. First, high levels of α-Syn alter the size of synaptic vesicle pools and impair their trafficking. Second, α-Syn overexpression can either misregulate or redistribute proteins of the presynaptic SNARE complex. This leads to deficient tethering, docking, priming and fusion of synaptic vesicles at the active zone (AZ). Third, α-Syn inclusions are found within the presynaptic AZ, accompanied by a decrease in AZ protein levels. Furthermore, α-Syn overexpression reduces the endocytic retrieval of synaptic vesicle membranes during vesicle recycling. These presynaptic alterations mediated by accumulation of α-Syn, together impair neurotransmitter exocytosis and neuronal communication. Although α-Syn is expressed throughout the brain and enriched at presynaptic terminals, DA neurons are the most vulnerable in PD, likely because α-Syn directly regulates dopamine levels. Indeed, evidence suggests that α-Syn is a negative modulator of dopamine by inhibiting enzymes responsible for its synthesis. In addition, α-Syn is able to interact with and reduce the activity of VMAT2 and DAT. The resulting dysregulation of dopamine levels directly contributes to the formation of toxic α-Syn oligomers. Together these data suggest a vicious cycle of accumulating α-Syn and deregulated dopamine that triggers synaptic dysfunction and impaired neuronal communication, ultimately causing synaptopathy and progressive neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Jessika C Bridi
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | - Frank Hirth
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| |
Collapse
|
29
|
Leighton PLA, Allison WT. Protein Misfolding in Prion and Prion-Like Diseases: Reconsidering a Required Role for Protein Loss-of-Function. J Alzheimers Dis 2018; 54:3-29. [PMID: 27392869 DOI: 10.3233/jad-160361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Prion disease research has contributed much toward understanding other neurodegenerative diseases, including recent demonstrations that Alzheimer's disease (AD) and other neurodegenerative diseases are prion-like. Prion-like diseases involve the spread of degeneration between individuals and/or among cells or tissues via template directed misfolding, wherein misfolded protein conformers propagate disease by causing normal proteins to misfold. Here we use the premise that AD, amyotrophic lateral sclerosis, Huntington's disease, and other similar diseases are prion-like and ask: Can we apply knowledge gained from studies of these prion-like diseases to resolve debates about classical prion diseases? We focus on controversies about what role(s) protein loss-of-function might have in prion diseases because this has therapeutic implications, including for AD. We examine which loss-of-function events are recognizable in prion-like diseases by considering the normal functions of the proteins before their misfolding and aggregation. We then delineate scenarios wherein gain-of-function and/or loss-of-function would be necessary or sufficient for neurodegeneration. We consider roles of PrPC loss-of-function in prion diseases and in AD, and conclude that the conventional wisdom that prion diseases are 'toxic gain-of-function diseases' has limitations. While prion diseases certainly have required gain-of-function components, we propose that disease phenotypes are predominantly caused by deficits in the normal physiology of PrPC and its interaction partners as PrPC converts to PrPSc. In this model, gain-of-function serves mainly to spread disease, and loss-of-function directly mediates neuron dysfunction. We propose experiments and predictions to assess our conclusion. Further study on the normal physiological roles of these key proteins is warranted.
Collapse
Affiliation(s)
- Patricia L A Leighton
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - W Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Lim YJ, Lee SJ. Are exosomes the vehicle for protein aggregate propagation in neurodegenerative diseases? Acta Neuropathol Commun 2017; 5:64. [PMID: 28851422 PMCID: PMC5576311 DOI: 10.1186/s40478-017-0467-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 02/08/2023] Open
Abstract
Abnormal protein aggregation has been implicated in neurodegenerative processes in human neurological disorders, such as Alzheimer’s disease and Parkinson’s disease. Recently, studies have established a novel concept that protein aggregates are transmitted among neuronal cells. By extension, such interneuronal aggregate transmission has been hypothesized to be the underlying mechanism for the pathological and clinical disease progression. However, the precise mechanism of the interneuronal aggregate transmission remains ill-defined. Recent reports have suggested that exosomes, a specific group of extracellular vesicles that are involved in intercellular transfer of cellular macromolecules such as proteins and RNAs, could play an important role in the aggregate transmission among neurons. Here, we review various types of extracellular vesicles and critically evaluate the evidence supporting the role of exosomes in interneuronal aggregate transmission and neurodegeneration. We also discuss the competing mechanisms other than the exosome-mediated transmission. By doing so, we aim to assess the current state of knowledge on the mechanism of interneuronal aggregate transmission and suggest the future directions of research towards understanding the mechanism.
Collapse
|
31
|
Abstract
Clearing misfolded proteins from the cytoplasm is essential to maintain cellular homeostasis. Now, a parallel clearance system is described that uses the deubiquitylase USP19 to enable secretion of misfolded cytoplasmic proteins when conventional proteasomal degradation is compromised. Misfolding-associated protein secretion (MAPS) has important implications for protein quality control and prion-like transmission.
Collapse
|
32
|
Hinz FI, Geschwind DH. Molecular Genetics of Neurodegenerative Dementias. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a023705. [PMID: 27940516 DOI: 10.1101/cshperspect.a023705] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative dementias are clinically heterogeneous, progressive diseases with frequently overlapping symptoms, such as cognitive impairments and behavior and movement deficits. Although a majority of cases appear to be sporadic, there is a large genetic component that has yet to be fully explained. Here, we review the recent genetic and genomic findings pertaining to Alzheimer's disease, frontotemporal dementia, Lewy body dementia, and prion dementia. In this review, we describe causal and susceptibility genes identified for these dementias and discuss recent research pertaining to the molecular function of these genes. Of particular interest, there is a large overlap in clinical phenotypes, genes, and/or aggregating protein products involved in these diseases, as well as frequent comorbid presentation, indicating that these dementias may represent a continuum of syndromes rather than individual diseases.
Collapse
Affiliation(s)
- Flora I Hinz
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095.,Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California 90024
| |
Collapse
|
33
|
Bickle L, Hopwood JJ, Karageorgos L. Analysis of sheep α-synuclein provides a molecular strategy for the reduction of fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:261-273. [DOI: 10.1016/j.bbapap.2016.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/16/2016] [Accepted: 12/16/2016] [Indexed: 12/15/2022]
|
34
|
Abstract
Transmissible spongiform encephalopathies (TSEs) are caused by an infectious agent that is thought to consist of only misfolded and aggregated prion protein (PrP). Unlike conventional micro-organisms, the agent spreads and propagates by binding to and converting normal host PrP into the abnormal conformer, increasing the infectious titre. Synthetic prions, composed of refolded fibrillar forms of recombinant PrP (rec-PrP) have been generated to address whether PrP aggregates alone are indeed infectious prions. In several reports, the development of TSE disease has been described following inoculation and passage of rec-PrP fibrils in transgenic mice and hamsters. However in studies described here we show that inoculation of rec-PrP fibrils does not always cause clinical TSE disease or increased infectious titre, but can seed the formation of PrP amyloid plaques in PrP-P101L knock-in transgenic mice (101LL). These data are reminiscent of the "prion-like" spread of misfolded protein in other models of neurodegenerative disease following inoculation of transgenic mice with pre-formed amyloid seeds. Protein misfolding, even when the protein is PrP, does not inevitably lead to the development of an infectious TSE disease. It is possible that most in vivo and in vitro produced misfolded PrP is not infectious and that only a specific subpopulation is associated with infectivity and neurotoxicity.
Collapse
Affiliation(s)
- Rona M. Barron
- Neurobiology Division, The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush, UK,Correspondence to: Rona M. Barron; The Roslin Institute, Easter Bush, Midlothian, UK EH25 9RG;
| |
Collapse
|
35
|
Dahmene M, Bérard M, Oueslati A. Dissecting the Molecular Pathway Involved in PLK2 Kinase-mediated α-Synuclein-selective Autophagic Degradation. J Biol Chem 2017; 292:3919-3928. [PMID: 28154193 DOI: 10.1074/jbc.m116.759373] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 01/27/2017] [Indexed: 12/19/2022] Open
Abstract
Increasing lines of evidence support the causal link between α-synuclein (α-syn) accumulation in the brain and Parkinson's disease (PD) pathogenesis. Therefore, lowering α-syn protein levels may represent a viable therapeutic strategy for the treatment of PD and related disorders. We recently described a novel selective α-syn degradation pathway, catalyzed by the activity of the Polo-like kinase 2 (PLK2), capable of reducing α-syn protein expression and suppressing its toxicity in vivo However, the exact molecular mechanisms underlying this degradation route remain elusive. In the present study we report that among PLK family members, PLK3 is also able to catalyze α-syn phosphorylation and degradation in living cells. Using pharmacological and genetic approaches, we confirmed the implication of the macroautophagy on PLK2-mediated α-syn turnover, and our observations suggest a concomitant co-degradation of these two proteins. Moreover, we showed that the N-terminal region of α-syn is important for PLK2-mediated α-syn phosphorylation and degradation and is implicated in the physical interaction between the two proteins. We also demonstrated that PLK2 polyubiquitination is important for PLK2·α-syn protein complex degradation, and we hypothesize that this post-translational modification may act as a signal for the selective recognition by the macroautophagy machinery. Finally, we observed that the PD-linked mutation E46K enhances PLK2-mediated α-syn degradation, suggesting that this mutated form is a bona fide substrate of this degradation pathway. In conclusion, our study provides a detailed description of the new degradation route of α-syn and offers new opportunities for the development of therapeutic strategies aiming to reduce α-syn protein accumulation and toxicity.
Collapse
Affiliation(s)
- Manel Dahmene
- From the CHU de Quebec Research Center, Axe Neuroscience and Department of Molecular Medicine, Laval University, Quebec, QC G1V4G2, Canada
| | - Morgan Bérard
- From the CHU de Quebec Research Center, Axe Neuroscience and Department of Molecular Medicine, Laval University, Quebec, QC G1V4G2, Canada
| | - Abid Oueslati
- From the CHU de Quebec Research Center, Axe Neuroscience and Department of Molecular Medicine, Laval University, Quebec, QC G1V4G2, Canada
| |
Collapse
|
36
|
Oueslati A. Implication of Alpha-Synuclein Phosphorylation at S129 in Synucleinopathies: What Have We Learned in the Last Decade? JOURNAL OF PARKINSONS DISEASE 2017; 6:39-51. [PMID: 27003784 PMCID: PMC4927808 DOI: 10.3233/jpd-160779] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abnormal accumulation of proteinaceous intraneuronal inclusions called Lewy bodies (LBs) is the neurpathological hallmark of Parkinson’s disease (PD) and related synucleinopathies. These inclusions are mainly constituted of a presynaptic protein, α-synuclein (α-syn). Over the past decade, growing amounts of studies reported an aberrant accumulation of phosphorylated α-syn at the residue S129 (pS129) in the brain of patients suffering from PD, as well as in transgenic animal models of synucleinopathies. Whereas only a small fraction of α-syn (<4%) is phosphorylated in healthy brains, a dramatic accumulation of pS129 (>90%) has been observed within LBs, suggesting that this post-translational modification may play an important role in the regulation of α-syn aggregation, LBs formation and neuronal degeneration. However, whether phosphorylation at S129 suppresses or enhances α-syn aggregation and toxicity in vivo remains a subject of active debate. The answer to this question has important implications for understanding the role of phosphorylation in the pathogenesis of synucleinopathies and determining if targeting kinases or phosphatases could be a viable therapeutic strategy for the treatment of these devastating neurological disorders. In the present review, we explore recent findings from in vitro, cell-based assays and in vivo studies describing the potential implications of pS129 in the regulation of α-syn physiological functions, as well as its implication in synucleinopathies pathogenesis and diagnosis.
Collapse
Affiliation(s)
- Abid Oueslati
- Correspondence to: Abid Oueslati, Centre de Recherche du CHU de Québec-Université Laval, Axe Neuroscience et Départe-ment de Médecine Moléculaire de l’Université Laval, Québec G1V4G2, Canada. Tel.: +1 4185254444/Ext 49119; Fax: +1 4186542125; E-mail:
| |
Collapse
|
37
|
Wang X, Ma M, Teng J, Zhang J, Zhou S, Zhang Y, Wu E, Ding X. Chronic exposure to cerebrospinal fluid of multiple system atrophy in neuroblastoma and glioblastoma cells induces cytotoxicity via ER stress and autophagy activation. Oncotarget 2016; 6:13278-94. [PMID: 25965819 PMCID: PMC4537014 DOI: 10.18632/oncotarget.3748] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/08/2015] [Indexed: 11/25/2022] Open
Abstract
Oncogenesis and neurodegeneration share many common pathogenic pathways, involved in endoplastic reticulum (ER) stress, autophagy, DNA repair, and oxidative stress. However, mechanisms of cross-talking between oncogenesis and neurodegeneration are still unknown. Characterized by abnormal accumulation of α-synuclein (α-syn) aggregates in central nervous system (CNS), multiple system atrophy (MSA) is classified as α-synucleinopathy. Rapidly emerging evidence suggests that ‘prion-like propagation’ of α-syn aggregates in the regional spread of CNS leads to the progression of α-synucleinopathy. Whether cerebrospinal fluid (CSF) has deteriorating effects on neurogenic tumor cells and is involved in progression of α-synucleinopathy has not been explored. Here, we first show the cytotoxic effects of MSA-CSF on the neuroblastoma and glioblastoma cells and its underlying mechanism in vitro. Remarkably, MSA-CSF induced cytotoxicity via activating ER stress-associated apoptosis and autophagy in both SH-SY5Y and U251 cells. The result from in vivo systematic neuropathological analysis reveals that abnormally activated ER stress and autophagy were confined to substantia nigra and cerebellum in mouse CNS following MSA-CSF treatment. Specifically, dopamine neurons in substantia nigra and Purkinje cells in cerebellum cortex were degenerated in MSA-CSF-injected mice. Altogether, these findings demonstrate that MSA-CSF exerts cytotoxicities on nervous system neoplasms and accelerates the progression of synucleinopathies.
Collapse
Affiliation(s)
- Xuejing Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingming Ma
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junfang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuang Zhou
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Ying Zhang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Xuebing Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|
38
|
Mollenhauer B, Parnetti L, Rektorova I, Kramberger MG, Pikkarainen M, Schulz-Schaeffer WJ, Aarsland D, Svenningsson P, Farotti L, Verbeek MM, Schlossmacher MG. Biological confounders for the values of cerebrospinal fluid proteins in Parkinson's disease and related disorders. J Neurochem 2016; 139 Suppl 1:290-317. [DOI: 10.1111/jnc.13390] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 09/11/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik; Kassel Germany
- University Medical Center (Department of Neuropathology); Georg-August University Goettingen; Goettingen Germany
| | - Lucilla Parnetti
- Centro Disturbi della Memoria- Unità Valutativa Alzheimer; Clinica Neurologica; Università di Perugia; Perugia Italy
| | - Irena Rektorova
- Applied Neuroscience Group; CEITEC MU; Masaryk University; Brno Czech Republic
| | - Milica G. Kramberger
- Department of Neurology; University Medical Center Ljubljana; Ljubljana Slovenia
- Division for Neurogeriatrics; Department of NVS; Karolinska Institutet; Center for Alzheimer Research; Stockholm Sweden
- Centre for Age-Related Medicine; Stavanger University Hospital; Stavanger Norway
| | - Maria Pikkarainen
- Institute of Clinical Medicine / Neurology; University of Eastern Finland; Kuopio Finland
| | - Walter J. Schulz-Schaeffer
- University Medical Center (Department of Neuropathology); Georg-August University Goettingen; Goettingen Germany
| | - Dag Aarsland
- Division for Neurogeriatrics; Department of NVS; Karolinska Institutet; Center for Alzheimer Research; Stockholm Sweden
- Centre for Age-Related Medicine; Stavanger University Hospital; Stavanger Norway
| | - Per Svenningsson
- Department for Clinical Neuroscience; Karolinska Institute; Stockholm Sweden
| | - Lucia Farotti
- Centro Disturbi della Memoria- Unità Valutativa Alzheimer; Clinica Neurologica; Università di Perugia; Perugia Italy
| | - Marcel M. Verbeek
- Department of Neurology; Department of Laboratory Medicine; Donders Institute for Brain, Cognition and Behaviour; Radboud University Medical Centre; Nijmegen The Netherlands
| | - Michael G. Schlossmacher
- Program in Neuroscience and Division of Neurology; The Ottawa Hospital; University of Ottawa Brain & Mind Research Institute; Ottawa Ontario Canada
| |
Collapse
|
39
|
Brundin P, Atkin G, Lamberts JT. Basic science breaks through: New therapeutic advances in Parkinson's disease. Mov Disord 2015; 30:1521-7. [PMID: 26177603 DOI: 10.1002/mds.26332] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/13/2015] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is typically associated with progressive motor dysfunction, although PD patients also exhibit a variety of non-motor symptoms. The neuropathological hallmark of PD is intraneuronal inclusions containing primarily α-Synuclein (α-Syn), and several lines of evidence point to α-Syn as a key contributor to disease progression. Thus, basic research in the field of PD is largely focused on understanding the pathogenic properties of α-Syn. Over the past 2 y, these studies helped to identify several novel therapeutic strategies that have the potential to slow PD progression; such strategies include sequestration of extracellular α-Syn through immunotherapy, reduction of α-Syn multimerization or intracellular toxicity, and attenuation of the neuroinflammatory response. This review describes these and other putative therapeutic strategies, together with the basic science research that led to their identification. The current breadth of novel targets for the treatment of PD warrants cautious optimism in the fight against this devastating disease.
Collapse
Affiliation(s)
- Patrik Brundin
- Laboratory of Translational Parkinson's Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Graham Atkin
- Laboratory of Translational Parkinson's Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jennifer T Lamberts
- Laboratory of Translational Parkinson's Disease Research, Van Andel Research Institute, Grand Rapids, Michigan, USA.,College of Pharmacy, Ferris State University, Big Rapids, Michigan, USA
| |
Collapse
|
40
|
Distinct higher-order α-synuclein oligomers induce intracellular aggregation. Biochem J 2015; 468:485-93. [DOI: 10.1042/bj20150159] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022]
Abstract
The cell-to-cell transmission of pathology in Parkinson's disease has been linked to soluble amyloid oligomers. Ion mobility spectrometry (IMS)–MS has been used to show that these soluble oligomers have a compact ring-like conformation.
Collapse
|
41
|
Xu S, Chan P. Interaction between Neuromelanin and Alpha-Synuclein in Parkinson's Disease. Biomolecules 2015; 5:1122-42. [PMID: 26057626 PMCID: PMC4496713 DOI: 10.3390/biom5021122] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 04/29/2015] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is a very common neurodegenerative disorder characterized by the accumulation of α-synuclein (α-syn) into Lewy body (LB) inclusions and the loss of neuronmelanin (NM) containing dopamine (DA) neurons in the substantia nigra (SN). Pathological α-syn and NM are two prominent hallmarks in this selective and progressive neurodegenerative disease. Pathological α-syn can induce dopaminergic neuron death by various mechanisms, such as inducing oxidative stress and inhibiting protein degradation systems. Therefore, to explore the factors that trigger α-syn to convert from a non-toxic protein to toxic one is a pivotal question to clarify the mechanisms of PD pathogenesis. Many triggers for pathological α-syn aggregation have been identified, including missense mutations in the α-syn gene, higher concentration, and posttranslational modifications of α-Syn. Recently, the role of NM in inducing α-syn expression and aggregation has been suggested as a mechanism for this pigment to modulate neuronal vulnerability in PD. NM may be responsible for PD and age-associated increase and aggregation in α-syn. Here, we reviewed our previous study and other recent findings in the area of interaction between NM and α-syn.
Collapse
Affiliation(s)
- Shengli Xu
- Beijing Institute of Geriatrics, Xuanwu Hospital of Capital University of Medical Sciences, No.45 changchun St., Xicheng District, Beijing 100053, China.
- Parkinson's disease Center of Beijing Institute for Brain Disorders, Beijing 100053, China.
| | - Piu Chan
- Beijing Institute of Geriatrics, Xuanwu Hospital of Capital University of Medical Sciences, No.45 changchun St., Xicheng District, Beijing 100053, China.
- Parkinson's disease Center of Beijing Institute for Brain Disorders, Beijing 100053, China.
| |
Collapse
|
42
|
Propagation of dysbindin-1B aggregates: Exosome-mediated transmission of neurotoxic deposits. Neuroscience 2015; 291:301-16. [DOI: 10.1016/j.neuroscience.2015.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 11/21/2022]
|