1
|
Ali NH, Al-Kuraishy HM, Al-Gareeb AI, Alnaaim SA, Hetta HF, Saad HM, Batiha GES. A Mutual Nexus Between Epilepsy and α-Synuclein: A Puzzle Pathway. Mol Neurobiol 2024; 61:10198-10215. [PMID: 38703341 DOI: 10.1007/s12035-024-04204-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
Alpha-synuclein (α-Syn) is a specific neuronal protein that regulates neurotransmitter release and trafficking of synaptic vesicles. Exosome-associated α-Syn which is specific to the central nervous system (CNS) is involved in the pathogenesis of epilepsy. Therefore, this review aimed to elucidate the possible link between α-Syn and epilepsy, and how it affects the pathophysiology of epilepsy. A neurodegenerative protein such as α-Syn is implicated in the pathogenesis of epilepsy. Evidence from preclinical and clinical studies revealed that upregulation of α-Syn induces progressive neuronal dysfunctions through induction of oxidative stress, neuroinflammation, and inhibition of autophagy in a vicious cycle with subsequent development of severe epilepsy. In addition, accumulation of α-Syn in epilepsy could be secondary to the different cellular alterations including oxidative stress, neuroinflammation, reduction of brain-derived neurotrophic factor (BDNF) and progranulin (PGN), and failure of the autophagy pathway. However, the mechanism of α-Syn-induced-epileptogenesis is not well elucidated. Therefore, α-Syn could be a secondary consequence of epilepsy. Preclinical and clinical studies are warranted to confirm this causal relationship.
Collapse
Affiliation(s)
- Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Kingdom of Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, M.B.Ch.B, FRCP, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu, P.O. Box 13, Kufa, Najaf, Iraq
| | - Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
2
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 PMCID: PMC11694735 DOI: 10.1016/j.neuro.2024.06.001] [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: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
3
|
Salaramoli S, Joshaghani HR, Hosseini M, Hashemy SI. Therapeutic Effects of Selenium on Alpha-Synuclein Accumulation in Substantia Nigra Pars Compacta in a Rat Model of Parkinson's Disease: Behavioral and Biochemical Outcomes. Biol Trace Elem Res 2024; 202:1115-1125. [PMID: 37386228 DOI: 10.1007/s12011-023-03748-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 06/24/2023] [Indexed: 07/01/2023]
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder characterized by the accumulation of accumulated alpha-synuclein (α-Syn) in substantia nigra. Research has shown that selenium (Se) can protect neural cells through the actions of selenoproteins, including selenoprotein P (SelP) and selenoprotein S (SelS), which participate in endoplasmic reticulum-associated protein degradation (ERAD). In this study, we investigated the potential protective role of Se in a pre-clinical PD rat model.We aimed to evaluate the therapeutic effects of Se administration in the 6-hydroxydopamine (6-OHDA) induced unilateral rat PD model. Male Wistar rats were utilised for unilateral PD animal model which were subjected to stereotaxic surgery and injected with 20 μg 6-OHDA/5 μl 0.2% ascorbate saline. After confirming the model, the rats were intraperitoneally injected with 0.1, 0.2, and 0.3 mg/kg of sodium selenite for 7 days. We then performed behavioral tests, including apomorphine-induced rotation, hanging, and rotarod tests. Following sacrifice, we analysed the substantia nigra area of the brain and serum for protein quantification, element analysis, and gene expression analysis.Our results indicate that the administration of 0.3 mg/kg of Se improved the motor deficiency in hanging, rotarod, and apomorphine-induced rotational tests. While there was no significant improvement in the expression of α-Syn, Se increased the expression of selenoproteins. Additionally, levels of selenoproteins, Se, and α-Syn both brain and serum were re-established by the treatment, suggesting the role of Se on the α-Syn accumulation. Furthermore, Se improved PD-induced biochemical deficits by increasing the levels of SelS and SelP (p<0.005).In conclusion, our findings suggest that Se may have a protective role in PD. 0.3 mg/kg dosage of Se increased the expression of selenoproteins, reduced the accumulation of α-Syn in the brain, and improved PD-induced motor deficits. These results suggest that Se may be a potential therapeutic option for PD treatment.
Collapse
Affiliation(s)
- Sanaz Salaramoli
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Joshaghani
- Laboratory Sciences Research Centre, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioural Sciences Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Chung TD, Linville RM, Guo Z, Ye R, Jha R, Grifno GN, Searson PC. Effects of acute and chronic oxidative stress on the blood-brain barrier in 2D and 3D in vitro models. Fluids Barriers CNS 2022; 19:33. [PMID: 35551622 PMCID: PMC9097350 DOI: 10.1186/s12987-022-00327-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is a shared pathology of neurodegenerative disease and brain injuries, and is derived from perturbations to normal cell processes by aging or environmental factors such as UV exposure and air pollution. As oxidative cues are often present in systemic circulation, the blood-brain barrier (BBB) plays a key role in mediating the effect of these cues on brain dysfunction. Therefore, oxidative damage and disruption of the BBB is an emergent focus of neurodegenerative disease etiology and progression. We assessed barrier dysfunction in response to chronic and acute oxidative stress in 2D and 3D in vitro models of the BBB with human iPSC-derived brain microvascular endothelial-like cells (iBMECs). We first established doses of hydrogen peroxide to induce chronic damage (modeling aging and neurodegenerative disease) and acute damage (modeling the response to traumatic brain injury) by assessing barrier function via transendothelial electrical resistance in 2D iBMEC monolayers and permeability and monolayer integrity in 3D tissue-engineered iBMEC microvessels. Following application of these chronic and acute doses in our in vitro models, we found local, discrete structural changes were the most prevalent responses (rather than global barrier loss). Additionally, we validated unique functional changes in response to oxidative stress, including dysfunctional cell turnover dynamics and immune cell adhesion that were consistent with changes in gene expression.
Collapse
Affiliation(s)
- Tracy D Chung
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
| | - Robert Ye
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Ria Jha
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielle N Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Mechanistic Insights Expatiating the Redox-Active-Metal-Mediated Neuronal Degeneration in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23020678. [PMID: 35054862 PMCID: PMC8776156 DOI: 10.3390/ijms23020678] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a complicated and incapacitating neurodegenerative malady that emanates following the dopaminergic (DArgic) nerve cell deprivation in the substantia nigra pars compacta (SN-PC). The etiopathogenesis of PD is still abstruse. Howbeit, PD is hypothesized to be precipitated by an amalgamation of genetic mutations and exposure to environmental toxins. The aggregation of α-synucelin within the Lewy bodies (LBs), escalated oxidative stress (OS), autophagy-lysosome system impairment, ubiquitin-proteasome system (UPS) impairment, mitochondrial abnormality, programmed cell death, and neuroinflammation are regarded as imperative events that actively participate in PD pathogenesis. The central nervous system (CNS) relies heavily on redox-active metals, particularly iron (Fe) and copper (Cu), in order to modulate pivotal operations, for instance, myelin generation, synthesis of neurotransmitters, synaptic signaling, and conveyance of oxygen (O2). The duo, namely, Fe and Cu, following their inordinate exposure, are viable of permeating across the blood–brain barrier (BBB) and moving inside the brain, thereby culminating in the escalated OS (through a reactive oxygen species (ROS)-reliant pathway), α-synuclein aggregation within the LBs, and lipid peroxidation, which consequently results in the destruction of DArgic nerve cells and facilitates PD emanation. This review delineates the metabolism of Fe and Cu in the CNS, their role and disrupted balance in PD. An in-depth investigation was carried out by utilizing the existing publications obtained from prestigious medical databases employing particular keywords mentioned in the current paper. Moreover, we also focus on decoding the role of metal complexes and chelators in PD treatment. Conclusively, metal chelators hold the aptitude to elicit the scavenging of mobile/fluctuating metal ions, which in turn culminates in the suppression of ROS generation, and thereby prelude the evolution of PD.
Collapse
|
6
|
von Linstow CU, DeLano-Taylor M, Kordower JH, Brundin P. Does Developmental Variability in the Number of Midbrain Dopamine Neurons Affect Individual Risk for Sporadic Parkinson's Disease? JOURNAL OF PARKINSONS DISEASE 2021; 10:405-411. [PMID: 31958098 PMCID: PMC7242832 DOI: 10.3233/jpd-191877] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parkinson’s disease (PD) is a slowly progressing neurodegenerative disorder that is coupled to both widespread protein aggregation and to loss of substantia nigra dopamine (DA) neurons, resulting in a wide variety of motor and non-motor signs and symptoms. Recent findings suggest that the PD process is triggered several years before there is sufficient degeneration of DA neurons to cause onset of overt motor symptoms. According to this concept, the number of DA neurons present in the substantia nigra at birth could influence the time from the molecular triggering event until the clinical diagnosis with lower number of neurons at birth increasing the risk to develop the disease. Conversely, the risk for diagnosis would be reduced if the number of DA neurons is high at birth. In this commentary, we discuss the genetic and epigenetic factors that might influence the number of nigral DA neurons that each individual is born with and how these may be linked to PD risk.
Collapse
Affiliation(s)
| | - Merritt DeLano-Taylor
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.,Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
7
|
α-Synuclein E46K Mutation and Involvement of Oxidative Stress in a Drosophila Model of Parkinson's Disease. PARKINSONS DISEASE 2021; 2021:6621507. [PMID: 34285796 PMCID: PMC8275411 DOI: 10.1155/2021/6621507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 06/10/2021] [Accepted: 06/23/2021] [Indexed: 01/24/2023]
Abstract
Parkinson's disease (PD) is an age-associated neurodegenerative condition in which some genetic variants are known to increase disease susceptibility on interaction with environmental factors inducing oxidative stress. Different mutations in the SNCA gene are reported as the major genetic contributors to PD. E46K mutation pathogenicity has not been investigated as intensive as other SNCA gene mutations including A30P and A53T. In this study, based on the GAL4-UAS binary genetic tool, transgenic Drosophila melanogaster flies expressing wild-type and E46K-mutated copies of the human SNCA gene were constructed. Western blotting, immunohistochemical analysis, and light and confocal microscopy of flies' brains were undertaken along with the survival rate measurement, locomotor function assay, and ethanol and paraquat (PQ) tolerance to study α-synuclein neurotoxicity. Biochemical bioassays were carried out to investigate the activity of antioxidant enzymes and alterations in levels of oxidative markers following damages induced by human α-synuclein to the neurons of the transgenic flies. Overexpression of human α-synuclein in the central nervous system of these transgenic flies led to disorganized ommatidia structures and loss of dopaminergic neurons. E46K α-synuclein caused remarkable climbing defects, reduced survivorship, higher ethanol sensitivity, and increased PQ-mediated mortality. A noticeable decline in activity of catalase and superoxide dismutase enzymes besides considerable increase in the levels of lipid peroxidation and reactive oxygen species was observed in head capsule homogenates of α-synuclein-expressing flies, which indicates obvious involvement of oxidative stress as a causal factor in SNCAE46K neurotoxicity. In all the investigations, E46K copy of the SNCA gene was found to impose more severe defects when compared to wild-type SNCA. It can be concluded that the constructed Drosophila models developed PD-like symptoms that facilitate comparative studies of molecular and cellular pathways implicated in the pathogenicity of different α-synuclein mutations.
Collapse
|
8
|
Wu M, Su H, Zhao M. The Role of α-Synuclein in Methamphetamine-Induced Neurotoxicity. Neurotox Res 2021; 39:1007-1021. [PMID: 33555547 DOI: 10.1007/s12640-021-00332-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 12/28/2022]
Abstract
Methamphetamine (METH), a highly addictive psychostimulant, is the second most widely used illicit drug. METH produces damage dopamine neurons and apoptosis via multiple inter-regulating mechanisms, including dopamine overload, hyperthermia, oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, protein degradation system dysfunction, and neuroinflammation. Increasing evidence suggests that chronic METH abuse is associated with neurodegenerative changes in the human brain and an increased risk of Parkinson's disease (PD). METH use and PD may share some common steps in causing neurotoxicity. Accumulation of α-synuclein, a presynaptic protein, is the pathological hallmark of PD. Intriguingly, α-synuclein upregulation and aggregation are also found in dopaminergic neurons in the substantia nigra in chronic METH users. This suggests α-synuclein may play a role in METH-induced neurotoxicity. The mechanism of α-synuclein cytotoxicity in PD has attracted considerable attention; however, how α-synuclein affects METH-induced neurotoxicity has not been reviewed. In this review, we summarize the relationship between METH use and PD, interdependent mechanisms that are involved in METH-induced neurotoxicity and the significance of α-synuclein upregulation in response to METH use. The identification of α-synuclein overexpression and aggregation as a contributor to METH-induced neurotoxicity may provide a novel therapeutic target for the treatment of the deleterious effect of this drug and drug addiction.
Collapse
Affiliation(s)
- Manqing Wu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang Su
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
- Shanghai Clinical Research Center for Mental Health, Shanghai, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
9
|
Seleem AA. Immunohistochemical localization of alpha-synuclein in the retina of some nocturnal and diurnal animals. Biotech Histochem 2020; 95:360-372. [PMID: 31951746 DOI: 10.1080/10520295.2019.1703218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Although alpha-synuclein has been reported to participate in neurodegenerative diseases, the actual normal biological function of alpha-synuclein remains unclear. I investigated the correlation of alpha-synuclein expression with nocturnal and diurnal activity for various species. Hematoxylin and eosin staining, periodic acid-Schiff's reaction (PAS) and immunohistochemistry of alpha-synuclein expression were performed for the retinas of diurnal, nocturnal, nocturnal with diurnal activity species. I found different intensity of alpha-synuclein expression in the retinal layers. I found alpha-synuclein expression in the outer segment of the photoreceptor layer in the diurnal studied species and absence of alpha-synuclein expression in the compartments of photoreceptor layer in the retina of nocturnal species. I found localization of alpha-synuclein in the inner and outer segments of photoreceptors of the retina of nocturnal with diurnal activity species. The retinas of diurnal animals exhibited glycogen in the paraboloid structure in the inner segment of the photoreceptor layer. The retinas of nocturnal and nocturnal with diurnal activity species were devoid of glycogen in the photoreceptor layer. I conclude that the function of alpha-synuclein is more related to diurnal than to nocturnal species.
Collapse
Affiliation(s)
- Amin A Seleem
- Amin A. Seleem, Zoology Department, Faculty of Science, Sohag University, Sohag, Egypt and Biology Department, Faculty of Science and Arts, Alula, Taibah University, Kingdom Saudi Arabia
| |
Collapse
|
10
|
Liu Y, Zhang Y, Zhu K, Chi S, Wang C, Xie A. Emerging Role of Sirtuin 2 in Parkinson's Disease. Front Aging Neurosci 2020; 11:372. [PMID: 31998119 PMCID: PMC6965030 DOI: 10.3389/fnagi.2019.00372] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/19/2019] [Indexed: 12/17/2022] Open
Abstract
Parkinson’s disease (PD), the main risk factor of which is age, is one of the most common neurodegenerative diseases, thus presenting a substantial burden on the health of affected individuals as well as an economic burden. Sirtuin 2 (SIRT2), a subtype in the family of sirtuins, belongs to class III histone deacetylases (HDACs). It is known that SIRT2 levels increase with aging, and a growing body of evidence has been accumulating, showing that the activity of SIRT2 mediates various processes involved in PD pathogenesis, including aggregation of α-synuclein (α-syn), microtubule function, oxidative stress, inflammation, and autophagy. There have been conflicting reports about the role of SIRT2 in PD, in that some studies indicate its potential to induce the death of dopaminergic (DA) neurons, and that inhibition of SIRT2 may, therefore, have protective effects in PD. Other studies suggest a protective role of SIRT2 in the context of neuronal damage. As current treatments for PD are directed at alleviating symptoms and are very limited, a comprehensive understanding of the enzymology of SIRT2 in PD may be essential for developing novel therapeutic agents for the treatment of this disease. This review article will provide an update on our knowledge of the structure, distribution, and biological characteristics of SIRT2, and highlight its role in the pathogenesis of PD.
Collapse
Affiliation(s)
- Yumei Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingying Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Konghua Zhu
- Department of Neurology, The Eighth People Hospital of Qingdao City, Qingdao, China
| | - Song Chi
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chong Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Martínez-Orozco H, Mariño L, Uceda AB, Ortega-Castro J, Vilanova B, Frau J, Adrover M. Nitration and Glycation Diminish the α-Synuclein Role in the Formation and Scavenging of Cu 2+-Catalyzed Reactive Oxygen Species. ACS Chem Neurosci 2019; 10:2919-2930. [PMID: 30973706 DOI: 10.1021/acschemneuro.9b00142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Human α-synuclein is a small monomeric protein (140 residues) essential to maintain the function of the dopaminergic neurons and the neuronal redox balance. However, it holds a dark side since it is able to clump inside the neurons forming insoluble aggregates known as Lewy bodies, which are considered the hallmark of Parkinson's disease. Sporadic mutations and nonenzymatic post-translational modifications are well-known to stimulate the formation of Lewy bodies. Yet, the effect of nonenzymatic post-translational modifications on the function of α-synuclein has been studied less intense. Therefore, here we study how nitration and glycation mediated by methylglyoxal affect the redox features of α-synuclein. Both diminish the ability of α-synuclein to chelate Cu2+, except when Nε-(carboxyethyl)lysine or Nε-(carboxymethyl)lysine (two advanced glycation end products highly prevalent in vivo) are formed. This results in a lower capacity to prevent the Cu-catalyzed ascorbic acid degradation and to delay the formation of H2O2. However, only methylglyoxal was able to abolish the ability of α-synuclein to inhibit the free radical release. Both nitration and glycation enhanced the α-synuclein availability to be damaged by O2•-, although glycation made α-synuclein less reactive toward HO•. Our data represent the first report describing how nonenzymatic post-translational modifications might affect the redox function of α-synuclein, thus contributing to a better understanding of its pathological implications.
Collapse
Affiliation(s)
- Humberto Martínez-Orozco
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Laura Mariño
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Ana Belén Uceda
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Joaquín Ortega-Castro
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Bartolomé Vilanova
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Juan Frau
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Miquel Adrover
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| |
Collapse
|
12
|
Abstract
The past few years have resulted in an increased awareness and recognition of the prevalence and roles of intrinsically disordered proteins and protein regions (IDPs and IDRs, respectively) in synaptic vesicle trafficking and exocytosis and in overall synaptic organization. IDPs and IDRs constitute a class of proteins and protein regions that lack stable tertiary structure, but nevertheless retain biological function. Their significance in processes such as cell signaling is now well accepted, but their pervasiveness and importance in other areas of biology are not as widely appreciated. Here, we review the prevalence and functional roles of IDPs and IDRs associated with the release and recycling of synaptic vesicles at nerve terminals, as well as with the architecture of these terminals. We hope to promote awareness, especially among neuroscientists, of the importance of this class of proteins in these critical pathways and structures. The examples discussed illustrate some of the ways in which the structural flexibility conferred by intrinsic protein disorder can be functionally advantageous in the context of cellular trafficking and synaptic function.
Collapse
Affiliation(s)
- David Snead
- From the Department of Biochemistry, Weill Cornell Medicine, New York, New York 10021
| | - David Eliezer
- From the Department of Biochemistry, Weill Cornell Medicine, New York, New York 10021
| |
Collapse
|
13
|
Mesenchymal Stem Cells-derived Exosomes: A New Possible Therapeutic Strategy for Parkinson's Disease? Cells 2019; 8:cells8020118. [PMID: 30717429 PMCID: PMC6406999 DOI: 10.3390/cells8020118] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD) is the second most prevalent neurodegenerative disorder worldwide. Clinically, it is characterized by severe motor complications caused by a progressive degeneration of dopaminergic neurons (DAn) and dopamine loss. Current treatment is focused on mitigating the symptoms through administration of levodopa, rather than on preventing DAn damage. Therefore, the use and development of neuroprotective/disease-modifying strategies is an absolute need, which can lead to promising gains on PD translational research. Mesenchymal stem cells (MSCs)–derived exosomes have been proposed as a promising therapeutic tool, since it has been demonstrated that they can act as biological nanoparticles with beneficial effects in different pathological conditions, including PD. Thus, considering their potential protective action in lesioned sites, MSCs-derived exosomes might also be active modulators of the neuroregeneration processes, opening a door for their future use as therapeutical strategies in human clinical trials. Therefore, in this review, we analyze the current understanding of MSCs-derived exosomes as a new possible therapeutic strategy for PD, by providing an overview about the potential role of miRNAs in the cellular and molecular basis of PD.
Collapse
|
14
|
Iyer A, Claessens MMAE. Disruptive membrane interactions of alpha-synuclein aggregates. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1867:468-482. [PMID: 30315896 DOI: 10.1016/j.bbapap.2018.10.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/14/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022]
Abstract
Alpha synuclein (αS) is a ~14 kDa intrinsically disordered protein. Decades of research have increased our knowledge on αS yet its physiological function remains largely elusive. The conversion of monomeric αS into oligomers and amyloid fibrils is believed to play a central role of the pathology of Parkinson's disease (PD). It is becoming increasingly clear that the interactions of αS with cellular membranes are important for both αS's functional and pathogenic actions. Therefore, understanding interactions of αS with membranes seems critical to uncover functional or pathological mechanisms. This review summarizes our current knowledge of how physicochemical properties of phospholipid membranes affect the binding and aggregation of αS species and gives an overview of how post-translational modifications and point mutations in αS affect phospholipid membrane binding and protein aggregation. We discuss the disruptive effects resulting from the interaction of αS aggregate species with membranes and highlight current approaches and hypotheses that seek to understand the pathogenic and/or protective role of αS in PD.
Collapse
Affiliation(s)
- Aditya Iyer
- Membrane Enzymology Group, University of Groningen, Groningen 9747 AG, The Netherlands
| | | |
Collapse
|
15
|
Brown J, Horrocks MH. A sticky situation: Aberrant protein-protein interactions in Parkinson's disease. Semin Cell Dev Biol 2018; 99:65-77. [PMID: 29738882 DOI: 10.1016/j.semcdb.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 02/21/2018] [Accepted: 05/04/2018] [Indexed: 12/13/2022]
Abstract
The aberrant aggregation of normally soluble proteins into amyloid fibrils is the pathological hallmark of several neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Understanding this process will be key to developing both diagnostic and therapeutic approaches for neurodegenerative diseases. Recent advances in biophysical techniques, coupled with kinetic analyses have enabled a thorough description of the key molecular steps involved in protein aggregation. In this review, we discuss these advances and how they have been applied to study the ability of one such protein, α-Synuclein, to form neurotoxic oligomers.
Collapse
Affiliation(s)
- James Brown
- EMBL Australia Node in Single Molecule Science, The University of New South Wales, Sydney, NSW, 2032, Australia.
| | - Mathew H Horrocks
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
16
|
Small Molecules Attenuate the Interplay between Conformational Fluctuations, Early Oligomerization and Amyloidosis of Alpha Synuclein. Sci Rep 2018; 8:5481. [PMID: 29615762 PMCID: PMC5882917 DOI: 10.1038/s41598-018-23718-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/20/2018] [Indexed: 11/08/2022] Open
Abstract
Aggregation of alpha synuclein has strong implications in Parkinson’s disease. The heterogeneity of folding/aggregation landscape and transient nature of the early intermediates result in difficulty in developing a successful therapeutic intervention. Here we used fluorescence measurements at ensemble and single molecule resolution to study how the late and early events of alpha synuclein aggregation modulate each other. In-vitro aggregation data was complemented using measurements inside live neuroblastoma cells by employing a small molecule labeling technique. An inhibitor molecule (arginine), which delayed the late event of amyloidosis, was found to bind to the protein, shifting the early conformational fluctuations towards a compact state. In contrast, a facilitator of late aggregation (glutamate), was found to be excluded from the protein surface. The presence of glutamate was found to speed up the oligomer formation at the early stage. We found that the effects of the inhibitor and facilitator were additive and as a result they maintained a ratio at which they cancelled each other’s influence on different stages of alpha synuclein aggregation.
Collapse
|
17
|
Rey NL, Wesson DW, Brundin P. The olfactory bulb as the entry site for prion-like propagation in neurodegenerative diseases. Neurobiol Dis 2018; 109:226-248. [PMID: 28011307 PMCID: PMC5972535 DOI: 10.1016/j.nbd.2016.12.013] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
Olfactory deficits are present in numerous neurodegenerative disorders and are accompanied by pathology in related brain regions. In several of these disorders, olfactory disturbances appear early and are considered as prodromal symptoms of the disease. In addition, pathological protein aggregates affect olfactory regions prior to other regions, suggesting that the olfactory system might be particularly vulnerable to neurodegenerative diseases. Exposed to the external environment, the olfactory epithelium and olfactory bulb allow pathogen and toxin penetration into the brain, a process that has been proposed to play a role in neurodegenerative diseases. Determining whether the olfactory bulb could be a starting point of pathology and of pathology spread is crucial to understanding how neurodegenerative diseases evolve. We argue that pathological changes following environmental insults contribute to the initiation of protein aggregation in the olfactory bulb, which then triggers the spread of the pathology within the brain by a templating mechanism in a prion-like manner. We review the evidence for the early involvement of olfactory structures in neurodegenerative diseases and the relationship between neuropathology and olfactory function. We discuss the vulnerability and putative underlying mechanisms by which pathology could be initiated in the olfactory bulb, from the entry of pathogens (promoted by increased permeability of the olfactory epithelium with aging or inflammation) to the sensitivity of the olfactory system to oxidative stress and inflammation. Finally, we review changes in protein expression and neural excitability triggered by pathogenic proteins that can promote pathogenesis in the olfactory bulb and beyond.
Collapse
Affiliation(s)
- Nolwen L Rey
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| | - Daniel W Wesson
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
18
|
Sharma N, Nehru B. Curcumin affords neuroprotection and inhibits α-synuclein aggregation in lipopolysaccharide-induced Parkinson's disease model. Inflammopharmacology 2017; 26:349-360. [PMID: 29027056 DOI: 10.1007/s10787-017-0402-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 09/24/2017] [Indexed: 11/28/2022]
Abstract
Parkinson's disease (PD) pathology is characterized by the abnormal accumulation and aggregation of the pre-synaptic protein α-synuclein in the dopaminergic neurons as Lewy bodies (LBs). Curcumin, which plays a neuroprotective role in various animal models of PD, was found to directly modulate the aggregation of α-synuclein in in vitro as well as in in vivo studies. While curcumin has been shown to exhibit strong anti-oxidant and anti-inflammatory properties, there are a number of other possible mechanisms by which curcumin may alter α-synuclein aggregation which still remains obscure. Therefore, the present study was designed to understand such concealed mechanisms behind neuroprotective effects of curcumin. An animal model of PD was established by injecting lipopolysaccharide (LPS, 5 µg/5 µl PBS) into the substantia nigra (SN) of rats which was followed by curcumin administration (40 mg/kg b.wt (i.p.)) daily for a period of 21 days. Modulatory functions of curcumin were evident from the inhibition of astrocytic activation (GFAP) by immunofluorescence and NADPH oxidase complex activation by RT-PCR. Curcumin supplementation prevented the LPS-induced upregulation in the protein activity of transcription factor NFκB, proinflammatory cytokines (TNF-α, IL-1β, and IL-1α), inducible nitric oxide synthase (iNOS) as well as the regulating molecules of the intrinsic apoptotic pathway (Bax, Bcl-2, Caspase 3 and Caspase 9) by ELISA. Curcumin also resulted in significant improvement in the glutathione system (GSH, GSSG and redox ratio) and prevented iron deposition in the dopaminergic neurons as depicted from atomic absorption spectroscopy (AAS) and Prussian blue staining, respectively. Curcumin also prevented α-synuclein aggregates in the dopaminergic neurons as observed from gene as well as protein activity of α-synuclein using RT-PCR and IHC. Collectively, our results suggest that curcumin can be further pursued as a candidate drug in the molecules targeted therapy for PD and other related synucleopathies.
Collapse
Affiliation(s)
- Neha Sharma
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Bimla Nehru
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
19
|
Menges S, Minakaki G, Schaefer PM, Meixner H, Prots I, Schlötzer-Schrehardt U, Friedland K, Winner B, Outeiro TF, Winklhofer KF, von Arnim CAF, Xiang W, Winkler J, Klucken J. Alpha-synuclein prevents the formation of spherical mitochondria and apoptosis under oxidative stress. Sci Rep 2017; 7:42942. [PMID: 28224980 PMCID: PMC5320486 DOI: 10.1038/srep42942] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/17/2017] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress (OS), mitochondrial dysfunction, and dysregulation of alpha-synuclein (aSyn) homeostasis are key pathogenic factors in Parkinson's disease. Nevertheless, the role of aSyn in mitochondrial physiology remains elusive. Thus, we addressed the impact of aSyn specifically on mitochondrial response to OS in neural cells. We characterize a distinct type of mitochondrial fragmentation, following H2O2 or 6-OHDA-induced OS, defined by spherically-shaped and hyperpolarized mitochondria, termed "mitospheres". Mitosphere formation mechanistically depended on the fission factor Drp1, and was paralleled by reduced mitochondrial fusion. Furthermore, mitospheres were linked to a decrease in mitochondrial activity, and preceded Caspase3 activation. Even though fragmentation of dysfunctional mitochondria is considered to be a prerequisite for mitochondrial degradation, mitospheres were not degraded via Parkin-mediated mitophagy. Importantly, we provide compelling evidence that aSyn prevents mitosphere formation and reduces apoptosis under OS. In contrast, aSyn did not protect against Rotenone, which led to a different, previously described donut-shaped mitochondrial morphology. Our findings reveal a dichotomic role of aSyn in mitochondrial biology, which is linked to distinct types of stress-induced mitochondrial fragmentation. Specifically, aSyn may be part of a cellular defense mechanism preserving neural mitochondrial homeostasis in the presence of increased OS levels, while not protecting against stressors directly affecting mitochondrial function.
Collapse
Affiliation(s)
- Stefanie Menges
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Georgia Minakaki
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Holger Meixner
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Iryna Prots
- IZKF Junior Research Group III and BMBF Research Group Neuroscience, IZKF, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.,Department of Stem Cell Biology, Institute of Human Genetics, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Kristina Friedland
- Molecular and Clinical Pharmacy, Department of Chemistry and Pharmacy, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Beate Winner
- IZKF Junior Research Group III and BMBF Research Group Neuroscience, IZKF, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.,Department of Stem Cell Biology, Institute of Human Genetics, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tiago F Outeiro
- Department of Neurodegeneration and Restorative Research, University Medical Center Göttingen, 37073 Göttingen, Germany.,Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Konstanze F Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr-University Bochum, 44801 Bochum, Germany
| | | | - Wei Xiang
- Institute of Biochemistry, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jochen Klucken
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
20
|
Rcom-H'cheo-Gauthier AN, Osborne SL, Meedeniya ACB, Pountney DL. Calcium: Alpha-Synuclein Interactions in Alpha-Synucleinopathies. Front Neurosci 2016; 10:570. [PMID: 28066161 PMCID: PMC5167751 DOI: 10.3389/fnins.2016.00570] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/25/2016] [Indexed: 11/20/2022] Open
Abstract
Aggregation of the pre-synaptic protein, α-synuclein (α-syn), is the key etiological factor in Parkinson's disease (PD) and other alpha-synucleinopathies, such as multiple system atrophy (MSA) and Dementia with Lewy bodies (DLB). Various triggers for pathological α-syn aggregation have been elucidated, including post-translational modifications, oxidative stress, and binding of metal ions, such as calcium. Raised neuronal calcium levels in PD may occur due to mitochondrial dysfunction and/or may relate to calcium channel dysregulation or the reduced expression of the neuronal calcium buffering protein, calbindin-D28k. Recent results on human tissue and a mouse oxidative stress model show that neuronal calbindin-D28k expression excludes α-syn inclusion bodies. Previously, cell culture model studies have shown that transient increases of intracellular free Ca(II), such as by opening of the voltage-gated plasma calcium channels, could induce cytoplasmic aggregates of α-syn. Raised intracellular free calcium and oxidative stress also act cooperatively to promote α-syn aggregation. The association between raised neuronal calcium, α-syn aggregation, oxidative stress, and neurotoxicity is reviewed in the context of neurodegenerative α-syn disease and potential mechanism-based therapies.
Collapse
Affiliation(s)
| | | | | | - Dean L. Pountney
- Menzies Health Institute Queensland, Griffith UniversityGold Coast, QLD, Australia
| |
Collapse
|
21
|
Braak H, Del Tredici K. Potential Pathways of Abnormal Tau and α-Synuclein Dissemination in Sporadic Alzheimer's and Parkinson's Diseases. Cold Spring Harb Perspect Biol 2016; 8:a023630. [PMID: 27580631 PMCID: PMC5088528 DOI: 10.1101/cshperspect.a023630] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Experimental data indicate that transneuronal propagation of abnormal protein aggregates in neurodegenerative proteinopathies, such as sporadic Alzheimer's disease (AD) and Parkinson's disease (PD), is capable of a self-propagating process that leads to a progression of neurodegeneration and accumulation of prion-like particles. The mechanisms by which misfolded tau and α-synuclein possibly spread from one involved nerve cell to the next in the neuronal chain to induce abnormal aggregation are still unknown. Based on findings from studies of human autopsy cases, we review potential pathways and mechanisms related to axonal and transneuronal dissemination of tau (sporadic AD) and α-synuclein (sporadic PD) aggregates between anatomically interconnected regions.
Collapse
Affiliation(s)
- Heiko Braak
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section/Department of Neurology, Center for Biomedical Research, University of Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| |
Collapse
|
22
|
Alpha-synuclein aggregates are excluded from calbindin-D28k-positive neurons in dementia with Lewy bodies and a unilateral rotenone mouse model. Mol Cell Neurosci 2016; 77:65-75. [PMID: 27746320 DOI: 10.1016/j.mcn.2016.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/14/2016] [Accepted: 10/13/2016] [Indexed: 12/11/2022] Open
Abstract
α-Synuclein (α-syn) aggregates (Lewy bodies) in Dementia with Lewy Bodies (DLB) may be associated with disturbed calcium homeostasis and oxidative stress. We investigated the interplay between α-syn aggregation, expression of the calbindin-D28k (CB) neuronal calcium-buffering protein and oxidative stress, combining immunofluorescence double labelling and Western analysis, and examining DLB and normal human cases and a unilateral oxidative stress lesion model of α-syn disease (rotenone mouse). DLB cases showed a greater proportion of CB+ cells in affected brain regions compared to normal cases with Lewy bodies largely present in CB- neurons and virtually undetected in CB+ neurons. The unilateral rotenone-lesioned mouse model showed a greater proportion of CB+ cells and α-syn aggregates within the lesioned hemisphere than the control hemisphere, especially proximal to the lesion site, and α-syn inclusions occurred primarily in CB- cells and were almost completely absent in CB+ cells. Consistent with the immunofluorescence data, Western analysis showed the total CB level was 25% higher in lesioned compared to control hemisphere in aged animals that are more sensitive to lesion and 20% higher in aged compared to young mice in lesioned hemisphere, but not significantly different between young and aged in the control hemisphere. Taken together, the findings show α-syn aggregation is excluded from CB+ neurons, although the increased sensitivity of aged animals to lesion was not related to differential CB expression.
Collapse
|
23
|
Benskey MJ, Perez RG, Manfredsson FP. The contribution of alpha synuclein to neuronal survival and function - Implications for Parkinson's disease. J Neurochem 2016; 137:331-59. [PMID: 26852372 PMCID: PMC5021132 DOI: 10.1111/jnc.13570] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/29/2016] [Indexed: 02/06/2023]
Abstract
The aggregation of alpha synuclein (α-syn) is a neuropathological feature that defines a spectrum of disorders collectively termed synucleinopathies, and of these, Parkinson's disease (PD) is arguably the best characterized. Aggregated α-syn is the primary component of Lewy bodies, the defining pathological feature of PD, while mutations or multiplications in the α-syn gene result in familial PD. The high correlation between α-syn burden and PD has led to the hypothesis that α-syn aggregation produces toxicity through a gain-of-function mechanism. However, α-syn has been implicated to function in a diverse range of essential cellular processes such as the regulation of neurotransmission and response to cellular stress. As such, an alternative hypothesis with equal explanatory power is that the aggregation of α-syn results in toxicity because of a toxic loss of necessary α-syn function, following sequestration of functional forms α-syn into insoluble protein aggregates. Within this review, we will provide an overview of the literature linking α-syn to PD and the knowledge gained from current α-syn-based animal models of PD. We will then interpret these data from the viewpoint of the α-syn loss-of-function hypothesis and provide a potential mechanistic model by which loss of α-syn function could result in at least some of the neurodegeneration observed in PD. By providing an alternative perspective on the etiopathogenesis of PD and synucleinopathies, this may reveal alternative avenues of research in order to identify potential novel therapeutic targets for disease modifying strategies. The correlation between α-synuclein burden and Parkinson's disease pathology has led to the hypothesis that α-synuclein aggregation produces toxicity through a gain-of-function mechanism. However, in this review, we discuss data supporting the alternative hypothesis that the aggregation of α-synuclein results in toxicity because of loss of necessary α-synuclein function at the presynaptic terminal, following sequestration of functional forms of α-synuclein into aggregates.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Ruth G Perez
- Department of Biomedical Sciences, Center of Emphasis in Neuroscience, Paul L. Foster School of Medicine, Texas Tech University of the Health Sciences El Paso, El Paso, Texas, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA
| |
Collapse
|
24
|
Collier TJ, Redmond DE, Steece-Collier K, Lipton JW, Manfredsson FP. Is Alpha-Synuclein Loss-of-Function a Contributor to Parkinsonian Pathology? Evidence from Non-human Primates. Front Neurosci 2016; 10:12. [PMID: 26858591 PMCID: PMC4731516 DOI: 10.3389/fnins.2016.00012] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/11/2016] [Indexed: 01/20/2023] Open
Abstract
Accumulation of alpha-synuclein (α-syn) in Lewy bodies and neurites of midbrain dopamine neurons is diagnostic for Parkinson's disease (PD), leading to the proposal that PD is a toxic gain-of-function synucleinopathy. Here we discuss the alternative viewpoint that α-syn displacement from synapses by misfolding and aggregation results in a toxic loss-of-function. In support of this hypothesis we provide evidence from our pilot study demonstrating that knockdown of endogenous α-syn in dopamine neurons of non-human primates reproduces the pattern of nigrostriatal degeneration characteristic of PD.
Collapse
Affiliation(s)
- Timothy J Collier
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| | - D Eugene Redmond
- Departments of Psychiatry & Neurosurgery, Yale University School of MedicineNew Haven, CT, USA; Axion Research FoundationHamden, CT, USA
| | - Kathy Steece-Collier
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| | - Jack W Lipton
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| |
Collapse
|
25
|
Jagmag SA, Tripathi N, Shukla SD, Maiti S, Khurana S. Evaluation of Models of Parkinson's Disease. Front Neurosci 2016; 9:503. [PMID: 26834536 PMCID: PMC4718050 DOI: 10.3389/fnins.2015.00503] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/21/2015] [Indexed: 12/01/2022] Open
Abstract
Parkinson's disease is one of the most common neurodegenerative diseases. Animal models have contributed a large part to our understanding and therapeutics developed for treatment of PD. There are several more exhaustive reviews of literature that provide the initiated insights into the specific models; however a novel synthesis of the basic advantages and disadvantages of different models is much needed. Here we compare both neurotoxin based and genetic models while suggesting some novel avenues in PD modeling. We also highlight the problems faced and promises of all the mammalian models with the hope of providing a framework for comparison of various systems.
Collapse
Affiliation(s)
- Shail A Jagmag
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| | - Naveen Tripathi
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| | - Sunil D Shukla
- Department of Zoology, Government Meera Girl's College Udaipur, India
| | - Sankar Maiti
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| | - Sukant Khurana
- Department of Biology, Indian Institute of Science Education and Research Kolkata, India
| |
Collapse
|
26
|
Cardoso A, Guedes J, Cardoso A, Morais C, Cunha P, Viegas A, Costa R, Jurado A, Pedroso de Lima M. Recent Trends in Nanotechnology Toward CNS Diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 130:1-40. [DOI: 10.1016/bs.irn.2016.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
27
|
Schikowski T, Vossoughi M, Vierkötter A, Schulte T, Teichert T, Sugiri D, Fehsel K, Tzivian L, Bae IS, Ranft U, Hoffmann B, Probst-Hensch N, Herder C, Krämer U, Luckhaus C. Association of air pollution with cognitive functions and its modification by APOE gene variants in elderly women. ENVIRONMENTAL RESEARCH 2015; 142:10-6. [PMID: 26092807 DOI: 10.1016/j.envres.2015.06.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 05/05/2015] [Accepted: 06/06/2015] [Indexed: 05/07/2023]
Abstract
BACKGROUND Epidemiological studies have shown effects of long-term exposure to air pollution on cardiovascular and respiratory health. However, studies investigating the effects of air pollution on cognition and brain function are limited. We investigated if neurocognitive functions are associated with air pollution exposure and whether apolipoprotein E (ApoE) alleles modify the association of air pollution exposure with cognition. METHODS We investigated 789 women from the SALIA cohort during the 22-year follow-up examination (2008-2009). Exposure to particulate matter (PM) size fractions and nitrogen oxides (NOx) were assigned to home addresses. Traffic indicators were used to assess residential proximity to high traffic load. Level of cognitive performance was assessed using the CERAD-Plus test. Air pollution effects on cognitive functioning were estimated cross-sectionally using adjusted linear regression models. RESULTS Air pollution was negatively associated with cognitive function and cognitive performance in the subtests for semantic memory and visuo-construction. Significant associations could be observed for figure copying with an interquartile range increase of NO2 (β=-0.28 (95%CI:-0.44;-0.12)), NOx (β=-0.25 (95%CI:-0.40;-0.09)), PM10 (β=-0.14 (95%CI:-0.26;-0.02)) and PM2.5 (β=-0.19 (95%CI:-0.36;-0.02)). The association with traffic load was significant in carriers of one or two ApoE ɛ4 risk alleles. CONCLUSION In this study of elderly women, markers of air pollution were associated with cognitive impairment in the visuospatial domain. The association of traffic exposure is significant in participants carrying the ApoE ε4 risk allele.
Collapse
Affiliation(s)
- Tamara Schikowski
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany; Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Switzerland.
| | - Mohammad Vossoughi
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Andrea Vierkötter
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Thomas Schulte
- Department of Psychiatry and Psychotherapy, Heinrich Heine University Düsseldorf, Germany
| | - Tom Teichert
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Centre for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), partner site Düsseldorf, Germany
| | - Dorothee Sugiri
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Karin Fehsel
- Department of Psychiatry and Psychotherapy, Heinrich Heine University Düsseldorf, Germany
| | - Lilian Tzivian
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Il-seok Bae
- Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Ulrich Ranft
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Barbara Hoffmann
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany; Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Nicole Probst-Hensch
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Switzerland
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Centre for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), partner site Düsseldorf, Germany
| | - Ursula Krämer
- IUF- Leibniz Research Institute for Environmental Medicine (IUF), Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | - Christian Luckhaus
- Department of Psychiatry and Psychotherapy, Heinrich Heine University Düsseldorf, Germany
| |
Collapse
|
28
|
Abstract
Neurodegenerative diseases (NDs) collectively afflict more than 40 million people worldwide. The majority of these diseases lack therapies to slow or stop progression due in large part to the challenge of disentangling the simultaneous presentation of broad, multifaceted pathophysiologic changes. Present technologies and computational capabilities suggest an optimistic future for deconvolving these changes to identify novel mechanisms driving ND onset and progression. In particular, integration of highly multi-dimensional omic analytical techniques (e.g., microarray, mass spectrometry) with computational systems biology approaches provides a systematic methodology to elucidate new mechanisms driving NDs. In this review, we begin by summarizing the complex pathophysiology of NDs associated with protein aggregation, emphasizing the shared complex dysregulation found in all of these diseases, and discuss available experimental ND models. Next, we provide an overview of technological and computational techniques used in systems biology that are applicable to studying NDs. We conclude by reviewing prior studies that have applied these approaches to NDs and comment on the necessity of combining analysis from both human tissues and model systems to identify driving mechanisms. We envision that the integration of computational approaches with multiple omic analyses of human tissues, and mouse and in vitro models, will enable the discovery of new therapeutic strategies for these devastating diseases.
Collapse
Affiliation(s)
- Levi B Wood
- Cancer Research Institute, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
29
|
The proposed mechanisms of radio frequency waves (RFWs) on nervous system functions impairment. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s00580-015-2096-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
30
|
Jahromi SR, Haddadi M, Shivanandappa T, Ramesh SR. Attenuation of neuromotor deficits by natural antioxidants of Decalepis hamiltonii in transgenic Drosophila model of Parkinson's disease. Neuroscience 2015; 293:136-50. [PMID: 25754960 DOI: 10.1016/j.neuroscience.2015.02.048] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 02/20/2015] [Accepted: 02/25/2015] [Indexed: 12/21/2022]
Abstract
Oxidative stress is believed to be a major factor for the onset of Parkinson's disease (PD). In this study, we have investigated oxidative status in transgenic Drosophila model of PD. Our results revealed elevated levels of reactive oxygen species (ROS) and lipid peroxidation (LPO) in A30P and A53T α-synuclein PD model flies compared to control. We have demonstrated for the first time the ameliorating potential of natural antioxidants characterized from the roots of Dh in A30P and A53T α-synuclein PD model flies. Feeding of transgenic flies with aqueous Dh root extract for 21 days significantly improved their climbing ability and circadian rhythm of locomotor activity which was associated with reduction in levels of ROS and LPO and enhancement in the activities of catalase (CAT) and superoxide dismutase (SOD). Dh protected against paraquat (PQ) sensitivity in α-synuclein transgenic flies and delayed the onset of PD-like symptoms which appears to be mediated by suppression of oxidative stress.
Collapse
Affiliation(s)
- S R Jahromi
- Department of Studies in Zoology, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - M Haddadi
- Department of Biology, Faculty of Science, University of Zabol, Zabol, Iran
| | - T Shivanandappa
- Department of Studies in Zoology, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - S R Ramesh
- Department of Studies in Zoology, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India.
| |
Collapse
|
31
|
Snead D, Eliezer D. Alpha-synuclein function and dysfunction on cellular membranes. Exp Neurobiol 2014; 23:292-313. [PMID: 25548530 PMCID: PMC4276801 DOI: 10.5607/en.2014.23.4.292] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/15/2014] [Accepted: 11/16/2014] [Indexed: 11/19/2022] Open
Abstract
Alpha-synuclein is a small neuronal protein that is closely associated with the etiology of Parkinson's disease. Mutations in and alterations in expression levels of alpha-synuclein cause autosomal dominant early onset heredity forms of Parkinson's disease, and sporadic Parkinson's disease is defined in part by the presence of Lewy bodies and Lewy neurites that are composed primarily of alpha-synuclein deposited in an aggregated amyloid fibril state. The normal function of alpha-synuclein is poorly understood, and the precise mechanisms by which it leads to toxicity and cell death are also unclear. Although alpha-synuclein is a highly soluble, cytoplasmic protein, it binds to a variety of cellular membranes of different properties and compositions. These interactions are considered critical for at least some normal functions of alpha-synuclein, and may well play critical roles in both the aggregation of the protein and its mechanisms of toxicity. Here we review the known features of alpha-synuclein membrane interactions in the context of both the putative functions of the protein and of its pathological roles in disease.
Collapse
Affiliation(s)
- David Snead
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - David Eliezer
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
32
|
Siddique YH, Faisal M, Naz F, Jyoti S, Rahul. Role of Ocimum sanctum leaf extract on dietary supplementation in the transgenic Drosophila model of Parkinson's disease. Chin J Nat Med 2014; 12:777-81. [PMID: 25443371 DOI: 10.1016/s1875-5364(14)60118-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Indexed: 11/25/2022]
Abstract
AIM To evaluate the effect of Ocimum sanctum leaf extract on the dietary supplementation in the transgenic Drosophila model of Parkinson's disease. METHOD The effect of Ocimum sanctum leaf extract was studied on the transgenic Drosophila model of flies expressing normal human alpha synuclein (h-αs) in the neurons. O. sanctum extract at final concentrations of 0.042 8 × 10(-4), 0.87 × 10(-4), and 1.85 × 10(-4) g·mL(-1) of diet were established and the flies were allowed to feed for 21 days. The climbing assay and lipid peroxidation were taken as parameters for the study. RESULTS The supplementation of O. sanctum extract showed a dose-dependent significant delay in the loss of climbing ability and reduction in oxidative stress in the brain of PD model flies. CONCLUSION The results of the present study showed that the O. sanctum extract is potent in reducing the PD symptoms in transgenic Drosophila model.
Collapse
Affiliation(s)
- Yasir Hasan Siddique
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
| | - Mohammad Faisal
- Forest Entomology Division, Forest Research Institute, Dehradun 248006, UK, India
| | - Falaq Naz
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Smita Jyoti
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Rahul
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
33
|
Abstract
Many proteins associated with neurodegenerative diseases have poorly defined or unknown functions. α-Synuclein is one such protein which is associated with a range of diseases including Parkinson's disease. Now accepted as a metal-binding protein, α-synuclein's function could possibly be defined in relation to the binding of cofactors. It has been suggested recently that α-synuclein is able to reduce iron using copper as its catalytic centre. The consequence of this is that possibly the function of α-synuclein can now be defined. The evidence for this and the consequences for Parkinson's disease are discussed in the present review.
Collapse
|
34
|
Rcom-H'cheo-Gauthier A, Goodwin J, Pountney DL. Interactions between calcium and alpha-synuclein in neurodegeneration. Biomolecules 2014; 4:795-811. [PMID: 25256602 PMCID: PMC4192672 DOI: 10.3390/biom4030795] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/25/2014] [Accepted: 07/14/2014] [Indexed: 12/01/2022] Open
Abstract
In Parkinson’s disease and some atypical Parkinson’s syndromes, aggregation of the α-synuclein protein (α-syn) has been linked to neurodegeneration. Many triggers for pathological α-syn aggregation have been identified, including port-translational modifications, oxidative stress and raised metal ions, such as Ca2+. Recently, it has been found using cell culture models that transient increases of intracellular Ca2+ induce cytoplasmic α-syn aggregates. Ca2+-dependent α-syn aggregation could be blocked by the Ca2+ buffering agent, BAPTA-AM, or by the Ca2+ channel blocker, Trimethadione. Furthermore, a greater proportion of cells positive for aggregates occurred when both raised Ca2+ and oxidative stress were combined, indicating that Ca2+ and oxidative stress cooperatively promote α-syn aggregation. Current on-going work using a unilateral mouse lesion model of Parkinson’s disease shows a greater proportion of calbindin-positive neurons survive the lesion, with intracellular α-syn aggregates almost exclusively occurring in calbindin-negative neurons. These and other recent findings are reviewed in the context of neurodegenerative pathologies and suggest an association between raised Ca2+, α-syn aggregation and neurotoxicity.
Collapse
Affiliation(s)
- Alex Rcom-H'cheo-Gauthier
- Griffith Health Institute, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia.
| | - Jacob Goodwin
- Griffith Health Institute, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia.
| | - Dean L Pountney
- Griffith Health Institute, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia.
| |
Collapse
|
35
|
Kanaan NM, Manfredsson FP. Loss of functional alpha-synuclein: a toxic event in Parkinson's disease? JOURNAL OF PARKINSONS DISEASE 2014; 2:249-67. [PMID: 23938255 PMCID: PMC4736738 DOI: 10.3233/jpd-012138] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The discovery that alpha-synuclein (α-syn) is the primary component of the neuropathological hallmarks of Parkinson's disease (PD) and the identification of α-syn mutations in numerous inherited forms of PD has positioned α-syn at the top of the list of important factors in the pathogenesis of PD. Based on the pathological accumulation of α-syn in the brains of patients, the field is currently focused on therapeutic strategies that aim to reduce or eliminate α-syn. However, recent evidence suggests α-syn is a critical protein in neuron (i.e. dopamine neurons) survival and that maintaining a certain level of biologically functional α-syn is an important consideration in targeting α-syn for therapies. Despite the widespread interest in α-syn, the normal biological functions remain elusive, but a large body of work is focused on addressing this issue. In this review, we will discuss the current evidence related to α-syn function, α-syn folding and aggregation, and α-syn's role in disease. Finally, we will propose a relatively novel hypothesis on the pathogenesis of PD that hinges upon the premises that functional α-syn is critical to cell survival and that a reduction in biologically functional α-syn, whether through aggregation or reduced expression, may lead to the neurodegeneration in PD.
Collapse
Affiliation(s)
- Nicholas M Kanaan
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | | |
Collapse
|
36
|
Akbari A, Jelodar G, Nazifi S. Vitamin C protects rat cerebellum and encephalon from oxidative stress following exposure to radiofrequency wave generated by a BTS antenna model. Toxicol Mech Methods 2014; 24:347-52. [PMID: 24730455 DOI: 10.3109/15376516.2014.910852] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Fujita KA, Ostaszewski M, Matsuoka Y, Ghosh S, Glaab E, Trefois C, Crespo I, Perumal TM, Jurkowski W, Antony PMA, Diederich N, Buttini M, Kodama A, Satagopam VP, Eifes S, del Sol A, Schneider R, Kitano H, Balling R. Integrating pathways of Parkinson's disease in a molecular interaction map. Mol Neurobiol 2014; 49:88-102. [PMID: 23832570 PMCID: PMC4153395 DOI: 10.1007/s12035-013-8489-4] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/13/2013] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a major neurodegenerative chronic disease, most likely caused by a complex interplay of genetic and environmental factors. Information on various aspects of PD pathogenesis is rapidly increasing and needs to be efficiently organized, so that the resulting data is available for exploration and analysis. Here we introduce a computationally tractable, comprehensive molecular interaction map of PD. This map integrates pathways implicated in PD pathogenesis such as synaptic and mitochondrial dysfunction, impaired protein degradation, alpha-synuclein pathobiology and neuroinflammation. We also present bioinformatics tools for the analysis, enrichment and annotation of the map, allowing the research community to open new avenues in PD research. The PD map is accessible at http://minerva.uni.lu/pd_map .
Collapse
Affiliation(s)
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Integrated Biobank of Luxembourg, Luxembourg City, Luxembourg
| | | | - Samik Ghosh
- The Systems Biology Institute, Minato-ku, Tokyo, Japan
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Christophe Trefois
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Isaac Crespo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Thanneer M. Perumal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Wiktor Jurkowski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Paul M. A. Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Nico Diederich
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Department of Neuroscience, Centre Hospitalier Luxembourg, Luxembourg City, Luxembourg
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Akihiko Kodama
- Faculty of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Venkata P. Satagopam
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Serge Eifes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
- Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Hiroaki Kitano
- The Systems Biology Institute, Minato-ku, Tokyo, Japan
- Sony Computer Science Laboratories, Shinagawa-ku, Tokyo, Japan
- Division of Systems Biology, Cancer Institute, Tokyo, Japan
- Open Biology Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa Japan
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
38
|
Deleersnijder A, Gerard M, Debyser Z, Baekelandt V. The remarkable conformational plasticity of alpha-synuclein: blessing or curse? Trends Mol Med 2013; 19:368-77. [DOI: 10.1016/j.molmed.2013.04.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 04/03/2013] [Accepted: 04/03/2013] [Indexed: 12/21/2022]
|
39
|
One single method to produce native and Tat-fused recombinant human α-synuclein in Escherichia coli. BMC Biotechnol 2013; 13:32. [PMID: 23557146 PMCID: PMC3621789 DOI: 10.1186/1472-6750-13-32] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 03/25/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human α-synuclein is a small-sized, natively unfolded protein that in fibrillar form is the primary component of Lewy bodies, the pathological hallmark of Parkinson's disease. Experimental evidence suggests that α-synuclein aggregation is the key event that triggers neurotoxicity although additional findings have proposed a protective role of α-synuclein against oxidative stress. One way to address the mechanism of this protective action is to evaluate α-synuclein-mediated protection by delivering this protein inside cells using a chimeric protein fused with the Tat-transduction domain of HIV Tat, named TAT-α-synuclein. RESULTS A reliable protocol was designed to efficiently express and purify two different forms of human α-synuclein. The synthetic cDNAs encoding for the native α-synuclein and the fusion protein with the transduction domain of Tat protein from HIV were overexpressed in a BL21(DE3) E. coli strain as His-tagged proteins. The recombinant proteins largely localized (≥ 85%) to the periplasmic space. By using a quick purification protocol, based on recovery of periplasmic space content and metal-chelating chromatography, the recombinant α-synuclein protein forms could be purified in a single step to ≥ 95% purity. Both α-synuclein recombinant proteins form fibrils and the TAT-α-synuclein is also cytotoxic in the micromolar concentration range. CONCLUSIONS To further characterize the molecular mechanisms of α-synuclein neurotoxicity both in vitro and in vivo and to evaluate the relevance of extracellular α-synuclein for the pathogenesis and progression of Parkinson's disease, a suitable method to produce different high-quality forms of this pathological protein is required. Our optimized expression and purification procedure offers an easier and faster means of producing different forms (i.e., both the native and the TAT-fusion form) of soluble recombinant α-synuclein than previously described procedures.
Collapse
|
40
|
Abstract
A number of neurodegenerative diseases principally affect humans as they age and are characterized by the loss of specific groups of neurons in different brain regions. Although these disorders are generally sporadic, it is now clear that many of them have a substantial genetic component. As genes are the raw material with which evolution works, we might benefit from understanding these genes in an evolutionary framework. Here, I will discuss how we can understand whether evolution has shaped genes involved in neurodegeneration and the implications for practical issues, such as our choice of model systems for studying these diseases, and more theoretical concerns, such as the level of selection against these phenotypes.
Collapse
Affiliation(s)
- Mark R Cookson
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892-3707, USA.
| |
Collapse
|
41
|
Effect of α-synuclein on amyloid β-induced toxicity: relevance to Lewy body variant of Alzheimer disease. Neurochem Res 2013; 38:797-806. [PMID: 23389658 DOI: 10.1007/s11064-013-0982-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 12/30/2012] [Accepted: 01/25/2013] [Indexed: 10/27/2022]
Abstract
Alzheimer's disease, the most prevalent age-related neurodegenerative disease, is characterized by the presence of extracellular senile plaques composed of amyloid-beta (Aβ) peptide and intracellular neurofibrillary tangles. More than 50 % of Alzheimer's disease (AD) patients also exhibit abundant accumulation of α-synuclein (α-Syn)-positive Lewy bodies. This Lewy body variant of AD (LBV-AD) is associated with accelerated cognitive dysfunction and progresses more rapidly than pure AD. In addition, it has been suggested that Aβ and α-Syn can directly interact. In this study we investigated the effect of α-Syn on Aβ-induced toxicity in cortical neurons. In order to mimic the intracellular accumulation of α-Syn observed in the brain of LBV-AD patients, we used valproic acid (VPA) to increase its endogenous expression levels. The release of α-Syn from damaged presynaptic terminals that occurs during the course of the disease was simulated by challenging cells with recombinant α-Syn. Our results showed that either VPA-induced α-Syn upregulation or addition of recombinant α-Syn protect primary cortical neurons from soluble Aβ1-42 decreasing the caspase-3-mediated cell death. It was also found that neuroprotection against Aβ-induced toxicity mediated by α-Syn overexpression involves the PI3K/Akt cell survival pathway. Furthermore, recombinant α-Syn was shown to directly interact with Aβ1-42 and to decrease the levels of Aβ1-42 oligomers, which might explain its neuroprotective effect. In conclusion, we demonstrate that either endogenous or exogenous α-Syn can be neuroprotective against Aβ-induced cell death, suggesting a cell defence mechanism during the initial stages of the mixed pathology.
Collapse
|
42
|
Mackey VR, Muthian G, Smith M, King J, Charlton CG. Prenatal exposure to methanol as a dopamine system sensitization model in C57BL/6J mice. Life Sci 2012; 91:921-7. [PMID: 23000099 DOI: 10.1016/j.lfs.2012.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 08/24/2012] [Accepted: 09/07/2012] [Indexed: 12/14/2022]
Abstract
AIMS In this study, the effects of prenatal exposure to methanol (MeOH) on the nigrostriatal dopamine (NSDA) system were examined to determine if the interaction could sensitize this system, and serve as an underpinning for Parkinson's disease (PD) like changes that occur later in life. Methanol was studied because its toxicity resembles the symptoms of PD and the symptoms are relieved by L-dopa meaning that MeOH targets the NSDA system. Since fermentation and wood combustion are major sources for MeOH, the incidence of human encounters with MeOH is high. As a superior solvent and the precursor for formaldehyde, MeOH has a powerful and sometimes, irreversible impact on chemical processes, such as cross-linking proteins and nucleic acids. It may cause subthreshold changes that sensitizes the NSDA system to PD, that occur during aging. MAIN METHODS To study the prenatal effects of MeOH, pregnant C57BL/6J mice were administered 40 mg/kg MeOH by oral gavage during gestation days 8-12, twice daily. Twelve weeks after birth, behavior impairments were recorded. The striatum was dissected for the determination of tyrosine hydroxylase (TH), L-aromatic amino acid decarboxylase (LAAD), α-synuclein and levels of dopamine (DA) and its metabolites. KEY FINDINGS MeOH reduced striatal TH and LAAD protein by 47% and 57% respectively and DA by 32%. SIGNIFICANCE The results mean that in utero exposure to toxins similar to MeOH could sensitize the striatal system to changes that cause PD. This study may help identify strategies to block this type of in utero toxicity.
Collapse
Affiliation(s)
- Veronica R Mackey
- Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | | | | | | | | |
Collapse
|
43
|
α-Synuclein Protects Neurons from Apoptosis Downstream of Free-Radical Production Through Modulation of the MAPK Signalling Pathway. Neurotox Res 2012; 23:358-69. [DOI: 10.1007/s12640-012-9352-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/28/2012] [Accepted: 08/11/2012] [Indexed: 01/08/2023]
|
44
|
Prasad K, Beach TG, Hedreen J, Richfield EK. Critical role of truncated α-synuclein and aggregates in Parkinson's disease and incidental Lewy body disease. Brain Pathol 2012; 22:811-25. [PMID: 22452578 DOI: 10.1111/j.1750-3639.2012.00597.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The role of Lewy bodies, Lewy neurites and α-synuclein (αSYN) in the pathophysiology and diagnosis of Parkinson's disease (PD) is unclear. We used postmortem human tissue, a panel of antibodies (Abs) and confocal microscopy to examine the three-dimensional neurochemical anatomy of the nigrostriatal system. Abs were specific to truncated (tαSYN), phosphorylated and full-length αSYN. The findings demonstrate the critical role of tαSYN in initiating aggregation, a role for other forms of αSYN in aggregate expansion, a reason for the wide variety of proteins present in different aggregates, an explanation for the laminar appearance of aggregates described historically using different methods, the existence of proximal greater than distal aggregation in the vulnerable nigrostriatal pathway, the independent transport of different forms of αSYN as cargo along axons and a possible sequence for the formation of Lewy bodies. Findings differed between incidental Lewy body disease and PD only quantitatively. These findings have implications for understanding the pathogenesis and treatment of PD.
Collapse
Affiliation(s)
- Kavita Prasad
- Department of Pathology and Lab Medicine, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
45
|
Bellucci A, Zaltieri M, Navarria L, Grigoletto J, Missale C, Spano P. From α-synuclein to synaptic dysfunctions: new insights into the pathophysiology of Parkinson's disease. Brain Res 2012; 1476:183-202. [PMID: 22560500 DOI: 10.1016/j.brainres.2012.04.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/30/2012] [Accepted: 04/09/2012] [Indexed: 10/28/2022]
Abstract
Alpha-synuclein is a natively unfolded protein playing a key role in the regulation of several neuronal synaptic functions in physiological and pathological conditions. Many studies, over the past years, have shown that it is actively involved in PD pathophysiology. Alpha-synuclein is integrated in a complex network of neuronal processes through the interaction with cytosolic and synaptic proteins. Hence, it is not the sole α-synuclein pathology but its effects on diverse protein partners and specific cellular pathways in the membrane and/or cytosolic districts such as endoplasmic reticulum/Golgi, axonal and synaptic compartments of dopaminergic neurons, that may cause the onset of neuronal cell dysfunction and degeneration which are among the key pathological features of the PD brain. Here we summarize a series of experimental data supporting that α-synuclein aggregation may induce dysfunction and degeneration of synapses via these multiple mechanisms. Taken together, these data add new insights into the complex mechanisms underlying synaptic derangement in PD and other α-synucleinopathies. This article is part of a Special Issue entitled: Brain Integration.
Collapse
Affiliation(s)
- Arianna Bellucci
- Division of Pharmacology, Department of Biomedical Sciences and Biotechnologies and National Institute of Neuroscience, University of Brescia, Brescia, Italy.
| | | | | | | | | | | |
Collapse
|
46
|
Increased expression of α-synuclein by SNCA duplication is associated with resistance to toxic stimuli. J Mol Neurosci 2012; 47:249-55. [PMID: 22392151 DOI: 10.1007/s12031-012-9732-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 02/21/2012] [Indexed: 01/16/2023]
Abstract
Duplication of alpha-synuclein gene (SNCA) is a recognized cause of Parkinson's disease (PD). However, the penetrance in families with SNCA duplication is as low as 30%, indicating that factors other than the SNCA gene dosage have an important role in neuronal death. In this study, using lymphoblastoid cell lines (LCLs) derived from a parkinsonian kindred with SNCA duplication, we examined whether there is difference in (1) the level of SNCA mRNA and protein expression and cell viability and (2) the vulnerability to various insults relevant to PD between a patient, asymptomatic carrier, and unaffected control. Expression of SNCA mRNA and protein increased in the LCLs from subjects with SNCA gene duplication, irrespective of the disease status. In the absence of treatment, LCLs from the patient and carrier showed decreased viability compared with the LCL from the control. The LCL from the patient also showed decreased viability compared to the carrier. When susceptibility to various insults including lactacystin, dexamethasone, 3-methyladenine, H(2)O(2), and rotenone was examined, surprisingly, the LCL from the patient was more resistant than the LCL from the control to all agents except for lactacystin. This study shows that both intrinsic and extrinsic factors and their interaction have important roles in cell death and in the development of PD and further indicates that the relationship between cell death and the level of alpha-synuclein may be more complicated than previously thought.
Collapse
|
47
|
Abstract
AbstractGenetic, neuropathological and biochemical evidence implicates α-synuclein, a 140 amino acid presynaptic neuronal protein, in the pathogenesis of Parkinson’s disease and other neurodegenerative disorders. The aggregated protein inclusions mainly containing aberrant α-synuclein are widely accepted as morphological hallmarks of α-synucleinopathies, but their composition and location vary between disorders along with neuronal networks affected. α-Synuclein exists physiologically in both soluble and membran-bound states, in unstructured and α-helical conformations, respectively, while posttranslational modifications due to proteostatic deficits are involved in β-pleated aggregation resulting in formation of typical inclusions. The physiological function of α-synuclein and its role linked to neurodegeneration, however, are incompletely understood. Soluble oligomeric, not fully fibrillar α-synuclein is thought to be neurotoxic, main targets might be the synapse, axons and glia. The effects of aberrant α-synuclein include alterations of calcium homeostasis, mitochondrial dysfunction, oxidative and nitric injuries, cytoskeletal effects, and neuroinflammation. Proteasomal dysfunction might be a common mechanism in the pathogenesis of neuronal degeneration in α-synucleinopathies. However, how α-synuclein induces neurodegeneration remains elusive as its physiological function. Genome wide association studies demonstrated the important role for genetic variants of the SNCA gene encoding α-synuclein in the etiology of Parkinson’s disease, possibly through effects on oxidation, mitochondria, autophagy, and lysosomal function. The neuropathology of synucleinopathies and the role of α-synuclein as a potential biomarker are briefly summarized. Although animal models provided new insights into the pathogenesis of Parkinson disease and multiple system atrophy, most of them do not adequately reproduce the cardinal features of these disorders. Emerging evidence, in addition to synergistic interactions of α-synuclein with various pathogenic proteins, suggests that prionlike induction and seeding of α-synuclein could lead to the spread of the pathology and disease progression. Intervention in the early aggregation pathway, aberrant cellular effects, or secretion of α-synuclein might be targets for neuroprotection and disease-modifying therapy.
Collapse
|
48
|
Proteomics in Parkinson's disease: An unbiased approach towards peripheral biomarkers and new therapies. J Biotechnol 2011; 156:325-37. [DOI: 10.1016/j.jbiotec.2011.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 06/24/2011] [Accepted: 08/08/2011] [Indexed: 12/27/2022]
|
49
|
Choong CJ, Say YH. Neuroprotection of α-synuclein under acute and chronic rotenone and maneb treatment is abolished by its familial Parkinson's disease mutations A30P, A53T and E46K. Neurotoxicology 2011; 32:857-63. [PMID: 21658409 DOI: 10.1016/j.neuro.2011.05.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 03/31/2011] [Accepted: 05/23/2011] [Indexed: 11/30/2022]
Abstract
α-Synuclein (α-Syn) plays a crucial role in the pathophysiology of Parkinson's disease (PD). α-Syn has been extensively studied in many neuronal cell-based PD models but has yielded mixed results. The objective of this study was to re-evaluate the dual cytotoxic/protective roles of α-Syn in dopaminergic SH-SY5Y cells. Stable SH-SY5Y cells overexpressing wild type or familial α-Syn mutants (A30P, E46K and A53T) were subjected to acute and chronic rotenone and maneb treatment. Compared with untransfected SH-SY5Y cells, wild type α-Syn attenuated rotenone and maneb-induced cell death along with an attenuation of toxin-induced mitochondrial membrane potential changes and Reactive Oxygen Species level, whereas the mutant α-Syn constructs exacerbated environmental toxins-induced cytotoxicity. After chronic treatment, wild type α-Syn but not the mutant variants was found to rescue cells from subsequent acute hydrogen peroxide insult. These results suggest that the fundamental property of wild type α-Syn may be protective, and such property may be lost by its familial PD mutations.
Collapse
Affiliation(s)
- Chi-Jing Choong
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Perak Campus, 31900 Kampar, Perak, Malaysia
| | | |
Collapse
|
50
|
Chua CEL, Tang BL. Rabs, SNAREs and α-synuclein--membrane trafficking defects in synucleinopathies. ACTA ACUST UNITED AC 2011; 67:268-81. [PMID: 21439320 DOI: 10.1016/j.brainresrev.2011.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 03/15/2011] [Accepted: 03/16/2011] [Indexed: 12/22/2022]
Abstract
Neuronal dysfunctions and neurodegeneration are often associated with defects in membrane transport. Synucleinopathies are a diverse group of neurodegenerative disorders that share a common pathological feature--insoluble aggregates composed largely of the protein α-synuclein in certain populations of neurons and glia. The actual physiological function of the brain-enriched α-synuclein is still not particularly clear. What is obvious is that when the protein is present in pathologically high amounts, or in mutant forms with enhanced membrane association and oligomerization, it causes neuronal demise with manifestations of impaired neuronal traffic, heightened oxidative stress, mitochondrial degeneration and defects in lipid metabolism. α-synuclein's direct association with the activities of key components of the eukaryotic membrane traffic machinery, namely Rabs and the soluble N-ethylmaleimide sensitive factor (NSF) attachment protein receptors (SNAREs), has highlighted a key role for membrane transport defects in α-synuclein-mediated pathology. Here, we summarize and discuss recent findings in this regard, and their implications in the molecular aspects of synucleinopathy.
Collapse
Affiliation(s)
- Christelle En Lin Chua
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| | | |
Collapse
|