51
|
Daniels MJ, Nourse JB, Kim H, Sainati V, Schiavina M, Murrali MG, Pan B, Ferrie JJ, Haney CM, Moons R, Gould NS, Natalello A, Grandori R, Sobott F, Petersson EJ, Rhoades E, Pierattelli R, Felli I, Uversky VN, Caldwell KA, Caldwell GA, Krol ES, Ischiropoulos H. Cyclized NDGA modifies dynamic α-synuclein monomers preventing aggregation and toxicity. Sci Rep 2019; 9:2937. [PMID: 30814575 PMCID: PMC6393491 DOI: 10.1038/s41598-019-39480-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022] Open
Abstract
Growing evidence implicates α-synuclein aggregation as a key driver of neurodegeneration in Parkinson’s disease (PD) and other neurodegenerative disorders. Herein, the molecular and structural mechanisms of inhibiting α-synuclein aggregation by novel analogs of nordihydroguaiaretic acid (NDGA), a phenolic dibenzenediol lignan, were explored using an array of biochemical and biophysical methodologies. NDGA analogs induced modest, progressive compaction of monomeric α-synuclein, preventing aggregation into amyloid-like fibrils. This conformational remodeling preserved the dynamic adoption of α-helical conformations, which are essential for physiological membrane interactions. Oxidation-dependent NDGA cyclization was required for the interaction with monomeric α-synuclein. NDGA analog-pretreated α-synuclein did not aggregate even without NDGA-analogs in the aggregation mixture. Strikingly, NDGA-pretreated α-synuclein suppressed aggregation of naïve untreated aggregation-competent monomeric α-synuclein. Further, cyclized NDGA reduced α-synuclein-driven neurodegeneration in Caenorhabditis elegans. The cyclized NDGA analogs may serve as a platform for the development of small molecules that stabilize aggregation-resistant α-synuclein monomers without interfering with functional conformations yielding potential therapies for PD and related disorders.
Collapse
Affiliation(s)
- Malcolm J Daniels
- Pharmacology Graduate Group, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - J Brucker Nourse
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Hanna Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Valerio Sainati
- CERM and Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, 50019, Italy
| | - Marco Schiavina
- CERM and Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, 50019, Italy
| | - Maria Grazia Murrali
- CERM and Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, 50019, Italy
| | - Buyan Pan
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John J Ferrie
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Conor M Haney
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rani Moons
- Department of Chemistry, University of Antwerp, Antwerp, Belgium
| | - Neal S Gould
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, 19104, USA
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Rita Grandori
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Frank Sobott
- Biomolecular & Analytical Mass Spectrometry, Antwerp University, Antwerp, Belgium.,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom.,School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Elizabeth Rhoades
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Roberta Pierattelli
- CERM and Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, 50019, Italy
| | - Isabella Felli
- CERM and Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, 50019, Italy
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.,Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, 142292, Russian Federation
| | - Kim A Caldwell
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Edward S Krol
- College of Pharmacy & Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Harry Ischiropoulos
- Pharmacology Graduate Group, Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, 19104, USA. .,Children's Hospital of Philadelphia Research Institute and Systems Pharmacology and Translational Therapeutics, the Raymond and Ruth Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
52
|
Ganguly U, Ganguly A, Sen O, Ganguly G, Cappai R, Sahoo A, Chakrabarti S. Dopamine Cytotoxicity on SH-SY5Y Cells: Involvement of α-Synuclein and Relevance in the Neurodegeneration of Sporadic Parkinson’s Disease. Neurotox Res 2019; 35:898-907. [DOI: 10.1007/s12640-019-0001-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/29/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
|
53
|
Zhang S, Wang R, Wang G. Impact of Dopamine Oxidation on Dopaminergic Neurodegeneration. ACS Chem Neurosci 2019; 10:945-953. [PMID: 30592597 DOI: 10.1021/acschemneuro.8b00454] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. The characteristic feature of PD is the progressive degeneration of the dopaminergic (DAergic) neurons in the substantia nigra (SN). DAergic neurons in the SN accumulate black and insoluble membrane structures known as neuromelanin during aging. The oxidation of dopamine (DA) to form neuromelanin generates many o-quinones, including DA o-quinones, aminochrome, and 5,6-indolequinone. The focus of this review is to discuss the role of DA oxidation in association with PD. The oxidation of DA produces oxidative products, inducing mitochondrial dysfunction, impaired protein degradation, α-synuclein aggregation into neurotoxic oligomers, and oxidative stress, in vitro. Recent studies have demonstrated that the DA content is critical for both DJ-1 knockout and A53T α-synuclein transgenic mice to develop PD pathological features, providing evidence for DA action in PD pathogenesis in vivo. The effects of L-DOPA, as the most effective anti-PD drug, are also briefly discussed.
Collapse
Affiliation(s)
- Shun Zhang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Rui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
54
|
Ponzini E, De Palma A, Cerboni L, Natalello A, Rossi R, Moons R, Konijnenberg A, Narkiewicz J, Legname G, Sobott F, Mauri P, Santambrogio C, Grandori R. Methionine oxidation in α-synuclein inhibits its propensity for ordered secondary structure. J Biol Chem 2019; 294:5657-5665. [PMID: 30755483 DOI: 10.1074/jbc.ra118.001907] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 01/30/2019] [Indexed: 11/06/2022] Open
Abstract
α-Synuclein (AS) is an intrinsically disordered protein highly expressed in dopaminergic neurons. Its amyloid aggregates are the major component of Lewy bodies, a hallmark of Parkinson's disease (PD). AS is particularly exposed to oxidation of its methionine residues, both in vivo and in vitro Oxidative stress has been implicated in PD and oxidized α-synuclein has been shown to assemble into soluble, toxic oligomers, rather than amyloid fibrils. However, the structural effects of methionine oxidation are still poorly understood. In this work, oxidized AS was obtained by prolonged incubations with dopamine (DA) or epigallocatechin-3-gallate (EGCG), two inhibitors of AS aggregation, indicating that EGCG promotes the same final oxidation product as DA. The conformational transitions of the oxidized and non-oxidized protein were monitored by complementary biophysical techniques, including MS, ion mobility (IM), CD, and FTIR spectroscopy assays. Although the two variants displayed very similar structures under conditions that stabilize highly disordered or highly ordered states, differences emerged in the intermediate points of transitions induced by organic solvents, such as trifluoroethanol (TFE) and methanol (MeOH), indicating a lower propensity of the oxidized protein for forming either α- or β-type secondary structures. Furthermore, oxidized AS displayed restricted secondary-structure transitions in response to dehydration and slightly amplified tertiary-structure transitions induced by ligand binding. This difference in susceptibility to induced folding could explain the loss of fibrillation potential observed for oxidized AS. Finally, site-specific oxidation kinetics point out a minor delay in Met-127 modification, likely due to the effects of AS intrinsic structure.
Collapse
Affiliation(s)
- Erika Ponzini
- From the Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Antonella De Palma
- the Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Lucilla Cerboni
- From the Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Antonino Natalello
- From the Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Rossana Rossi
- the Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Rani Moons
- the Biomolecular and Analytical Mass Spectrometry, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Albert Konijnenberg
- the Biomolecular and Analytical Mass Spectrometry, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Joanna Narkiewicz
- the Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA) and ELETTRA-Sincrotrone Trieste S.C.p.A, 34136 Trieste, Italy
| | - Giuseppe Legname
- the Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA) and ELETTRA-Sincrotrone Trieste S.C.p.A, 34136 Trieste, Italy
| | - Frank Sobott
- the Biomolecular and Analytical Mass Spectrometry, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium.,the School of Molecular and Cellular Biology, University of Leeds, Leeds LS29JT, United Kingdom, and.,the Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - PierLuigi Mauri
- the Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20090 Milan, Italy
| | - Carlo Santambrogio
- From the Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy,
| | - Rita Grandori
- From the Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy,
| |
Collapse
|
55
|
Mor DE, Daniels MJ, Ischiropoulos H. The usual suspects, dopamine and alpha-synuclein, conspire to cause neurodegeneration. Mov Disord 2019; 34:167-179. [PMID: 30633814 PMCID: PMC6379109 DOI: 10.1002/mds.27607] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/15/2018] [Accepted: 12/03/2018] [Indexed: 01/07/2023] Open
Abstract
Parkinson's disease (PD) is primarily a movement disorder driven by the loss of dopamine-producing neurons in the substantia nigra (SN). Early identification of the oxidative properties of dopamine implicated it as a potential source of oxidative stress in PD, yet few studies have investigated dopamine neurotoxicity in vivo. The discovery of PD-causing mutations in α-synuclein and the presence of aggregated α-synuclein in the hallmark Lewy body pathology of PD revealed another important player. Despite extensive efforts, the precise role of α-synuclein aggregation in neurodegeneration remains unclear. We recently manipulated both dopamine levels and α-synuclein expression in aged mice and found that only the combination of these 2 factors caused progressive neurodegeneration of the SN and an associated motor deficit. Dopamine modified α-synuclein aggregation in the SN, resulting in greater abundance of α-synuclein oligomers and unique dopamine-induced oligomeric conformations. Furthermore, disruption of the dopamine-α-synuclein interaction rescued dopaminergic neurons from degeneration in transgenic Caenorhabditis elegans models. In this Perspective, we discuss these findings in the context of known α-synuclein and dopamine biology, review the evidence for α-synuclein oligomer toxicity and potential mechanisms, and discuss therapeutic implications. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Danielle E. Mor
- Lewis-Sigler Institute for Integrative Genomics and Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Malcolm J. Daniels
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Harry Ischiropoulos
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| |
Collapse
|
56
|
Oliveri V. Toward the discovery and development of effective modulators of α-synuclein amyloid aggregation. Eur J Med Chem 2019; 167:10-36. [PMID: 30743095 DOI: 10.1016/j.ejmech.2019.01.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
Abstract
A host of human diseases, including Parkinson's disease and Dementia with Lewy bodies, are suspected to be directly linked to protein aggregation. Amyloid protein aggregates and oligomeric intermediates of α-synuclein are observed in synucleinopathies and considered to be mediators of cellular toxicity. Hence, α-synuclein has seen as one of the leading and most compelling targets and is receiving a great deal of attention from researchers. Nevertheless, there is no neuroprotective approach directed toward Parkinson's disease or other synucleinopathies so far. In this review, we summarize the available data concerning inhibitors of α-synuclein aggregation and their advancing towards clinical use. The compounds are grouped according to their chemical structures, providing respective insights into their mechanism of action, pharmacology, and pharmacokinetics. Overall, shared structure-activity elements are emerging, as well as specific binding modes related to the ability of the modulators to establish hydrophobic and hydrogen bonds interactions with the protein. Some molecules with encouraging in vivo data support the possibility of translation to the clinic.
Collapse
Affiliation(s)
- Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, viale A. Doria 6, 95125, Catania, Italy.
| |
Collapse
|
57
|
In vitro neurotoxicity of salsolinol is attenuated by the presynaptic protein α-synuclein. Biochim Biophys Acta Gen Subj 2018; 1862:2835-2845. [DOI: 10.1016/j.bbagen.2018.08.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 01/08/2023]
|
58
|
Sun Y, Pham AN, Hare DJ, Waite TD. Kinetic Modeling of pH-Dependent Oxidation of Dopamine by Iron and Its Relevance to Parkinson's Disease. Front Neurosci 2018; 12:859. [PMID: 30534046 PMCID: PMC6275323 DOI: 10.3389/fnins.2018.00859] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/02/2018] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disease. While age is the most significant risk factor, the exact cause of this disease and the most effective approaches to mitigation remain unclear. It has long been proposed that dopamine may play a role in the pathology of Parkinson's disease in view of its ability to generate both protein-modifying quinones such as aminochrome and reactive oxygen species, especially in the presence of pathological iron accumulation in the primary site of neuron loss. Given the clinically measured acidosis of post-mortem Parkinson's disease brain tissue, the interaction between dopamine and iron was investigated over a pH range of 7.4 to 6.5 with emphasis on the accumulation of toxic quinones and generation of reactive oxygen species. Our results show that the presence of iron accelerates the formation of aminochrome with ferrous iron (Fe[II]) being more efficient in this regard than ferric iron (Fe[III]). Our results further suggest that a reduced aminochrome rearrangement rate coupled with an enhanced turnover rate of Fe[II] as a result of brain tissue acidosis could result in aminochrome accumulation within cells. Additionally, under these conditions, the enhanced redox cycling of iron in the presence of dopamine aggravates oxidative stress as a result of the production of damaging reactive species, including hydroxyl radicals.
Collapse
Affiliation(s)
- Yingying Sun
- Water Research Centre, School of Civil and Environmental Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - A Ninh Pham
- Water Research Centre, School of Civil and Environmental Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - Dominic J Hare
- Atomic Pathology Laboratory, Melbourne Dementia Research Centre at the Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, VIC, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - T David Waite
- Water Research Centre, School of Civil and Environmental Engineering, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
59
|
Nam E, Derrick JS, Lee S, Kang J, Han J, Lee SJC, Chung SW, Lim MH. Regulatory Activities of Dopamine and Its Derivatives toward Metal-Free and Metal-Induced Amyloid-β Aggregation, Oxidative Stress, and Inflammation in Alzheimer's Disease. ACS Chem Neurosci 2018; 9:2655-2666. [PMID: 29782798 DOI: 10.1021/acschemneuro.8b00122] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A catecholamine neurotransmitter, dopamine (DA), is suggested to be linked to the pathology of dementia; however, the involvement of DA and its structural analogues in the pathogenesis of Alzheimer's disease (AD), the most common form of dementia, composed of multiple pathogenic factors has not been clear. Herein, we report that DA and its rationally designed structural derivatives (1-6) based on DA's oxidative transformation are able to modulate multiple pathological elements found in AD [i.e., metal ions, metal-free amyloid-β (Aβ), metal-bound Aβ (metal-Aβ), and reactive oxygen species (ROS)], with demonstration of detailed molecular-level mechanisms. Our multidisciplinary studies validate that the protective effects of DA and its derivatives on Aβ aggregation and Aβ-mediated toxicity are induced by their oxidative transformation with concomitant ROS generation under aerobic conditions. In particular, DA and the derivatives (i.e., 3 and 4) show their noticeable anti-amyloidogenic ability toward metal-free Aβ and/or metal-Aβ, verified to occur via their oxidative transformation that facilitates Aβ oxidation. Moreover, in primary pan-microglial marker (CD11b)-positive cells, the major producers of inflammatory mediators in the brain, DA and its derivatives significantly diminish inflammation and oxidative stress triggered by lipopolysaccharides and Aβ through the reduced induction of inflammatory mediators as well as upregulated expression of heme oxygenase-1, the enzyme responsible for production of antioxidants. Collectively, we illuminate how DA and its derivatives could prevent multiple pathological features found in AD. The overall studies could advance our understanding regarding distinct roles of neurotransmitters in AD and identify key interactions for alleviation of AD pathology.
Collapse
Affiliation(s)
- Eunju Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeffrey S. Derrick
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seunghee Lee
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jiyeon Han
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Shin Jung C. Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Su Wol Chung
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
60
|
Saha S, Khan MA, Mudhara D, Deep S. Tuning the Balance between Fibrillation and Oligomerization of α-Synuclein in the Presence of Dopamine. ACS OMEGA 2018; 3:14213-14224. [PMID: 30411062 PMCID: PMC6217689 DOI: 10.1021/acsomega.8b00993] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/02/2018] [Indexed: 05/19/2023]
Abstract
The aggregates of α-synuclein bear a close connection with Parkinson's disease, which is largely characterized by the loss of the dopaminergic neurons. Dopamine promotes the formation of undesirable sodium dodecyl sulfate (SDS)-resistant oligomers of α-synuclein. In this study, we have shown that the inhibition of fibrillation by an additive may not always be the ultimate deciding factor in the context of its potential as a successful additive. Copper promotes the fibrillation of α-synuclein in buffer alone but inhibits the formation of SDS-resistant oligomers in the presence of dopamine. Glycerol, on the other hand, increases the population of such dopamine-mediated SDS-resistant oligomers. We speculate such an effect to be a manifestation of the distinct oxidation pathway of dopamine in the presence of copper.
Collapse
|
61
|
Huenchuguala S, Sjödin B, Mannervik B, Segura-Aguilar J. Novel Alpha-Synuclein Oligomers Formed with the Aminochrome-Glutathione Conjugate Are Not Neurotoxic. Neurotox Res 2018; 35:432-440. [PMID: 30343424 DOI: 10.1007/s12640-018-9969-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 01/20/2023]
Abstract
Aminochrome induces neurotoxic alpha-synuclein oligomer formation relevant to the etiology of Parkinson's disease. Oxidative stress produces aminochrome from dopamine, but conjugation with glutathione catalyzed by glutathione transferase M2-2 significantly decreases aminochrome-induced toxicity and alpha-synuclein oligomer formation. Notably, in the presence of the aminochrome-glutathione conjugate, previously unknown species of alpha-synuclein oligomers are formed. These aminochrome-glutathione oligomers of alpha-synuclein differ from formerly characterized oligomers and (i) have high molecular weight, and are stable and SDS-resistant, as determined by the Western blot method, (ii) show positive NBT-quinone-protein staining, which indicates the formation of alpha-synuclein adducts containing aminochrome. Furthermore, aminochrome-glutathione alpha-synuclein oligomers (iii) have distinctive shape and size, as determined by transmission electron microscopy, and (iv) are not toxic in U373MG cells. In conclusion, glutathione conjugated with aminochrome induces a new type of alpha-synuclein oligomers of a different size and shape, which have no demonstrable toxicity.
Collapse
Affiliation(s)
- Sandro Huenchuguala
- Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago-7, Chile.,Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras, 753, Osorno, Chile
| | - Birgitta Sjödin
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Bengt Mannervik
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago-7, Chile.
| |
Collapse
|
62
|
Iftikhar I, El-Nour KMA, Brajter-Toth A. DOPA and Adrenaline Oxidation Kinetics and Intermediates Identified by Electrospray Ionization Mass Spectrometry in Real Time. ChemElectroChem 2018. [DOI: 10.1002/celc.201701322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Imran Iftikhar
- Department of Chemistry; University of Florida; Gainesville, Florida 32611-7200 USA
| | | | - Anna Brajter-Toth
- Department of Chemistry; University of Florida; Gainesville, Florida 32611-7200 USA
| |
Collapse
|
63
|
Biosa A, Outeiro TF, Bubacco L, Bisaglia M. Diabetes Mellitus as a Risk Factor for Parkinson's Disease: a Molecular Point of View. Mol Neurobiol 2018; 55:8754-8763. [PMID: 29594935 DOI: 10.1007/s12035-018-1025-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/20/2018] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by elevated concentrations of glucose in the blood. The chronic hyperglycemic state accounts for most of the vascular complications associated to the disease and the prevalent mechanism proposed is related to the glycating chemistry mediated by methylglyoxal (MG), which accumulates in T2DM. In recent years, a higher risk of Parkinson's disease (PD) onset in people affected by T2DM has become evident, but the molecular mechanisms underlying the interplay between T2DM and PD are still unknown. The oxidative chemistry of dopamine and its reactivity towards the protein α-Synuclein (aS) has been associated to the pathogenesis of PD. Recently, aS has also been described to interact with MG. Interestingly, MG and the dopamine oxidation products share both structural similarity and chemical reactivity. The ability of MG to spread over the site of its production and react with aS could represent the rationale to explain the higher incidence of PD in T2DM-affected people and may open opportunities for the development of novel strategies to antagonize the raise of PD.
Collapse
Affiliation(s)
- Alice Biosa
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, Padova, Italy
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Max Planck Institute for Experimental Medicine, Goettingen, Germany
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Luigi Bubacco
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, Padova, Italy
| | - Marco Bisaglia
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
64
|
de Araújo FM, Ferreira RS, Souza CS, Dos Santos CC, Rodrigues TLRS, E Silva JHC, Gasparotto J, Gelain DP, El-Bachá RS, D Costa MDF, Fonseca JCM, Segura-Aguilar J, Costa SL, Silva VDA. Aminochrome decreases NGF, GDNF and induces neuroinflammation in organotypic midbrain slice cultures. Neurotoxicology 2018; 66:98-106. [PMID: 29588162 DOI: 10.1016/j.neuro.2018.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 01/19/2023]
Abstract
Recent evidence shows that aminochrome induces glial activation related to neuroinflammation. This dopamine derived molecule induces formation and stabilization of alpha-synuclein oligomers, mitochondria dysfunction, oxidative stress, dysfunction of proteasomal and lysosomal systems, endoplasmic reticulum stress and disruption of the microtubule network, but until now there has been no evidence of effects on production of cytokines and neurotrophic factors, that are mechanisms involved in neuronal loss in Parkinson's disease (PD). This study examines the potential role of aminochrome on the regulation of NGF, GDNF, TNF-α and IL-1β production and microglial activation in organotypic midbrain slice cultures from P8 - P9 Wistar rats. We demonstrated aminochrome (25 μM, for 24 h) induced reduction of GFAP expression, reduction of NGF and GDNF mRNA levels, morphological changes in Iba1+ cells, and increase of both TNF-α, IL-1β mRNA and protein levels. Moreover, aminochrome (25 μM, for 48 h) induced morphological changes in the edge of slices and reduction of TH expression. These results demonstrate neuroinflammation, as well as negative regulation of neurotrophic factors (GDNF and NGF), may be involved in aminochrome-induced neurodegeneration, and they contribute to a better understanding of PD pathogenesis.
Collapse
Affiliation(s)
- Fillipe M de Araújo
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil.
| | - Rafael S Ferreira
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Cleide S Souza
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Cleonice Creusa Dos Santos
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Tácio L R S Rodrigues
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Juliana Helena C E Silva
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Juciano Gasparotto
- Centro de estudos em Estresse oxidativo, Departamento de Bioquimica, PPG Bioquimica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de estudos em Estresse oxidativo, Departamento de Bioquimica, PPG Bioquimica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ramon S El-Bachá
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Maria de Fátima D Costa
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - José Claudio M Fonseca
- Centro de estudos em Estresse oxidativo, Departamento de Bioquimica, PPG Bioquimica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Silvia L Costa
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Victor Diogenes A Silva
- Laboratório de Neuroquímica e Biologia Celular, Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
65
|
Segura-Aguilar J, Huenchuguala S. Aminochrome Induces Irreversible Mitochondrial Dysfunction by Inducing Autophagy Dysfunction in Parkinson's Disease. Front Neurosci 2018; 12:106. [PMID: 29593482 PMCID: PMC5859232 DOI: 10.3389/fnins.2018.00106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/12/2018] [Indexed: 01/21/2023] Open
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, Faculty of Medicine, Instituto de Ciencias Biomédicas (ICBM), University of Chile, Santiago, Chile
| | - Sandro Huenchuguala
- Departamento de Ciencias Biológicas y Químicas, Facultad de Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| |
Collapse
|
66
|
Mor DE, Ischiropoulos H. The Convergence of Dopamine and α-Synuclein: Implications for Parkinson's Disease. J Exp Neurosci 2018; 12:1179069518761360. [PMID: 29559809 PMCID: PMC5846926 DOI: 10.1177/1179069518761360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 12/02/2022] Open
Abstract
In Parkinson’s disease (PD), the loss of dopamine-producing neurons in the substantia nigra (SN) leads to severe motor impairment, and pathological inclusions known as Lewy bodies contain aggregated α-synuclein protein. The relationship of α-synuclein aggregation and dopaminergic degeneration is unclear. This commentary highlights a recent study showing that the interaction of α-synuclein with dopamine may be an important mechanism underlying disease. Elevating dopamine levels in mice expressing human α-synuclein with the A53T familial PD mutation recapitulated key features of PD, including progressive neurodegeneration of the SN and decreased ambulation. The toxicity of dopamine was dependent on α-synuclein expression; hence, raising dopamine levels in nontransgenic mice did not result in neuronal injury. This interaction is likely mediated through soluble α-synuclein oligomers, which had modified conformations and were more abundant as a result of dopamine elevation in the mouse brain. Specific mutation of the dopamine interaction motif in the C-terminus of α-synuclein rescued dopamine neurons from degeneration in Caenorhabditis elegans models. Here, these findings are discussed, particularly regarding possible mechanisms of oligomer toxicity, relevance of these models to sporadic and autosomal recessive forms of PD, and implications for current PD treatment.
Collapse
Affiliation(s)
- Danielle E Mor
- Lewis-Sigler Institute for Integrative Genomics and Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Harry Ischiropoulos
- Perelman School of Medicine, University of Pennsylvania, and the Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| |
Collapse
|
67
|
Ahlemeyer B, Halupczok S, Rodenberg-Frank E, Valerius KP, Baumgart-Vogt E. Endogenous Murine Amyloid-β Peptide Assembles into Aggregates in the Aged C57BL/6J Mouse Suggesting These Animals as a Model to Study Pathogenesis of Amyloid-β Plaque Formation. J Alzheimers Dis 2018; 61:1425-1450. [DOI: 10.3233/jad-170923] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Barbara Ahlemeyer
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Sascha Halupczok
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Elke Rodenberg-Frank
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Klaus-Peter Valerius
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
68
|
Liu XL, Wang YD, Yu XM, Li DW, Li GR. Mitochondria-mediated damage to dopaminergic neurons in Parkinson's disease (Review). Int J Mol Med 2017; 41:615-623. [PMID: 29207041 DOI: 10.3892/ijmm.2017.3255] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 10/20/2017] [Indexed: 11/06/2022] Open
Abstract
Mitochondria are important organelles in virtually all eukaryotic cells, and are involved in a wide range of physiological and pathophysiological processes. Besides the generation of cellular energy in the form of adenosine triphosphate, mitochondria are also involved in calcium homeostasis, reactive oxygen species production and the activation of the intrinsic cell death pathway, thus determining cell survival and death. Mitochondrial abnormalities have been implicated in a wide range of disorders, including neurodegenerative disease such as Parkinson's disease (PD), and considered as a primary cause and central event responsible for the progressive loss of dopaminergic neurons in PD. Thus, reversion or attenuation of mitochondrial dysfunction should alleviate the severity or progression of the disease. The present review systematically summarizes the possible mechanisms associated with mitochondria‑mediated dopaminergic neuron damage in PD, in an attempt to elucidate the requirement for further studies for the development of effective PD treatments.
Collapse
Affiliation(s)
- Xiao-Liang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 132021, P.R. China
| | - Ying-Di Wang
- Department of Urinary Surgery, The Tumor Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| | - Xiu-Ming Yu
- Department of Immunology, The First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Da-Wei Li
- Department of Neurology, Affiliated Hospital of Beihua University, Jilin, Jilin 132000, P.R. China
| | - Guang-Ren Li
- Department of Neurology, The Third Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
69
|
Mor DE, Tsika E, Mazzulli JR, Gould NS, Kim H, Daniels MJ, Doshi S, Gupta P, Grossman JL, Tan VX, Kalb RG, Caldwell KA, Caldwell GA, Wolfe JH, Ischiropoulos H. Dopamine induces soluble α-synuclein oligomers and nigrostriatal degeneration. Nat Neurosci 2017; 20:1560-1568. [PMID: 28920936 PMCID: PMC5893155 DOI: 10.1038/nn.4641] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/22/2017] [Indexed: 01/27/2023]
Abstract
Parkinson's disease (PD) is defined by the loss of dopaminergic neurons in the substantia nigra and the formation of Lewy body inclusions containing aggregated α-synuclein. Efforts to explain dopamine neuron vulnerability are hindered by the lack of dopaminergic cell death in α-synuclein transgenic mice. To address this, we manipulated both dopamine levels and α-synuclein expression. Nigrally targeted expression of mutant tyrosine hydroxylase with enhanced catalytic activity increased dopamine levels without damaging neurons in non-transgenic mice. In contrast, raising dopamine levels in mice expressing human A53T mutant α-synuclein induced progressive nigrostriatal degeneration and reduced locomotion. Dopamine elevation in A53T mice increased levels of potentially toxic α-synuclein oligomers, resulting in conformationally and functionally modified species. Moreover, in genetically tractable Caenorhabditis elegans models, expression of α-synuclein mutated at the site of interaction with dopamine prevented dopamine-induced toxicity. These data suggest that a unique mechanism links two cardinal features of PD: dopaminergic cell death and α-synuclein aggregation.
Collapse
Affiliation(s)
- Danielle E. Mor
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elpida Tsika
- AC Immune SA, Ecole Polytechnique fédérale de Lausanne Innovation Park, Lausanne, Switzerland
| | - Joseph R. Mazzulli
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Neal S. Gould
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Hanna Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Malcolm J. Daniels
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shachee Doshi
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Preetika Gupta
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer L. Grossman
- State University of New York Downstate College of Medicine, Brooklyn, New York, USA
| | - Victor X. Tan
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert G. Kalb
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Kim A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Guy A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - John H. Wolfe
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harry Ischiropoulos
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
70
|
Zucca FA, Segura-Aguilar J, Ferrari E, Muñoz P, Paris I, Sulzer D, Sarna T, Casella L, Zecca L. Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson's disease. Prog Neurobiol 2017; 155:96-119. [PMID: 26455458 PMCID: PMC4826627 DOI: 10.1016/j.pneurobio.2015.09.012] [Citation(s) in RCA: 423] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 09/14/2015] [Accepted: 09/17/2015] [Indexed: 12/11/2022]
Abstract
There are several interrelated mechanisms involving iron, dopamine, and neuromelanin in neurons. Neuromelanin accumulates during aging and is the catecholamine-derived pigment of the dopamine neurons of the substantia nigra and norepinephrine neurons of the locus coeruleus, the two neuronal populations most targeted in Parkinson's disease. Many cellular redox reactions rely on iron, however an altered distribution of reactive iron is cytotoxic. In fact, increased levels of iron in the brain of Parkinson's disease patients are present. Dopamine accumulation can induce neuronal death; however, excess dopamine can be removed by converting it into a stable compound like neuromelanin, and this process rescues the cell. Interestingly, the main iron compound in dopamine and norepinephrine neurons is the neuromelanin-iron complex, since neuromelanin is an effective metal chelator. Neuromelanin serves to trap iron and provide neuronal protection from oxidative stress. This equilibrium between iron, dopamine, and neuromelanin is crucial for cell homeostasis and in some cellular circumstances can be disrupted. Indeed, when neuromelanin-containing organelles accumulate high load of toxins and iron during aging a neurodegenerative process can be triggered. In addition, neuromelanin released by degenerating neurons activates microglia and the latter cause neurons death with further release of neuromelanin, then starting a self-propelling mechanism of neuroinflammation and neurodegeneration. Considering the above issues, age-related accumulation of neuromelanin in dopamine neurons shows an interesting link between aging and neurodegeneration.
Collapse
Affiliation(s)
- Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Juan Segura-Aguilar
- Faculty of Medicine, Molecular and Clinical Pharmacology, ICBM, University of Chile, Santiago, Chile
| | - Emanuele Ferrari
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Patricia Muñoz
- Faculty of Medicine, Molecular and Clinical Pharmacology, ICBM, University of Chile, Santiago, Chile
| | - Irmgard Paris
- Faculty of Medicine, Molecular and Clinical Pharmacology, ICBM, University of Chile, Santiago, Chile; Department of Basic Sciences, Faculty of Sciences, Santo Tomás University, Viña del Mar, Chile
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Department of Neurology, Columbia University Medical Center, New York, NY, USA; Department of Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Tadeusz Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Luigi Casella
- Department of Chemistry, University of Pavia, Pavia, Italy
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy.
| |
Collapse
|
71
|
Duce JA, Wong BX, Durham H, Devedjian JC, Smith DP, Devos D. Post translational changes to α-synuclein control iron and dopamine trafficking; a concept for neuron vulnerability in Parkinson's disease. Mol Neurodegener 2017; 12:45. [PMID: 28592304 PMCID: PMC5463308 DOI: 10.1186/s13024-017-0186-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 06/02/2017] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease is a multifactorial neurodegenerative disorder, the aetiology of which remains elusive. The primary clinical feature of progressively impaired motor control is caused by a loss of midbrain substantia nigra dopamine neurons that have a high α-synuclein (α-syn) and iron content. α-Syn is a neuronal protein that is highly modified post-translationally and central to the Lewy body neuropathology of the disease. This review provides an overview of findings on the role post translational modifications to α-syn have in membrane binding and intracellular vesicle trafficking. Furthermore, we propose a concept in which acetylation and phosphorylation of α-syn modulate endocytic import of iron and vesicle transport of dopamine during normal physiology. Disregulated phosphorylation and oxidation of α-syn mediate iron and dopamine dependent oxidative stress through impaired cellular location and increase propensity for α-syn aggregation. The proposition highlights a connection between α-syn, iron and dopamine, three pathological components associated with disease progression in sporadic Parkinson's disease.
Collapse
Affiliation(s)
- James A Duce
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, UK. .,Oxidation Biology Unit, the Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, VIC, Australia.
| | - Bruce X Wong
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, UK.,Oxidation Biology Unit, the Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, VIC, Australia
| | - Hannah Durham
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, UK
| | | | - David P Smith
- Biomolecular Research Centre, Sheffield Hallam University, Howard Street, Sheffield, UK
| | - David Devos
- Department of Medical Pharmacology, Lille University, INSERM U1171, CHU of Lille, Lille, France
| |
Collapse
|
72
|
Muñoz PS, Segura-Aguilar J. DT-diaphorase Protects Against Autophagy Induced by Aminochrome-Dependent Alpha-Synuclein Oligomers. Neurotox Res 2017; 32:362-367. [DOI: 10.1007/s12640-017-9747-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 12/09/2022]
|
73
|
Herrera A, Muñoz P, Steinbusch HWM, Segura-Aguilar J. Are Dopamine Oxidation Metabolites Involved in the Loss of Dopaminergic Neurons in the Nigrostriatal System in Parkinson's Disease? ACS Chem Neurosci 2017; 8:702-711. [PMID: 28233992 DOI: 10.1021/acschemneuro.7b00034] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In 1967, L-dopa was introduced as part of the pharmacological therapy of Parkinson's disease (PD) and, in spite of extensive research, no additional effective drugs have been discovered to treat PD. This brings forward the question: why have no new drugs been developed? We consider that one of the problems preventing the discovery of new drugs is that we still have no information on the pathophysiology of the neurodegeneration of the neuromelanin-containing nigrostriatal dopaminergic neurons. Currently, it is widely accepted that the degeneration of dopaminergic neurons, i.e., in the substantia nigra pars compacta, involves mitochondrial dysfunction, the formation of neurotoxic oligomers of alpha-synuclein, the dysfunction of protein degradation systems, neuroinflammation, and oxidative and endoplasmic reticulum stress. However, the initial trigger of these mechanisms in the nigrostriatal system is still unknown. It has been reported that aminochrome induces the majority of these mechanisms involved in the neurodegeneration process. Aminochrome is formed within the cytoplasm of neuromelanin-containing dopaminergic neurons during the oxidation of dopamine to neuromelanin. The oxidation of dopamine to neuromelanin is a normal and harmless process, because healthy individuals have intact neuromelanin-containing dopaminergic neurons. Interestingly, aminochrome-induced neurotoxicity is prevented by two enzymes: DT-diaphorase and glutathione transferase M2-2, which explains why melanin-containing dopaminergic neurons are intact in healthy human brains.
Collapse
Affiliation(s)
- Andrea Herrera
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
- Department of Neuroscience, Faculty of
Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Patricia Muñoz
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Harry W. M. Steinbusch
- Department of Neuroscience, Faculty of
Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Juan Segura-Aguilar
- Molecular & Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
74
|
Haney CM, Cleveland CL, Wissner RF, Owei L, Robustelli J, Daniels M, Canyurt M, Rodriguez P, Ischiropoulos H, Baumgart T, Petersson EJ. Site-Specific Fluorescence Polarization for Studying the Disaggregation of α-Synuclein Fibrils by Small Molecules. Biochemistry 2017; 56:683-691. [PMID: 28045494 PMCID: PMC5520965 DOI: 10.1021/acs.biochem.6b01060] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fibrillar aggregates of the protein α-synuclein (αS) are one of the hallmarks of Parkinson's disease. Here, we show that measuring the fluorescence polarization (FP) of labels at several sites on αS allows one to monitor changes in the local dynamics of the protein after binding to micelles or vesicles, and during fibril formation. Most significantly, these site-specific FP measurements provide insight into structural remodeling of αS fibrils by small molecules and have the potential for use in moderate-throughput screens to identify small molecules that could be used to treat Parkinson's disease.
Collapse
Affiliation(s)
- Conor M. Haney
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| | - Christina L. Cleveland
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| | - Rebecca F. Wissner
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| | - Lily Owei
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| | - Jaclyn Robustelli
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| | - Malcolm Daniels
- Pharmacology Graduate Group; University of Pennsylvania; 3400 Civic Center Blvd, Philadelphia, PA 19104
| | | | | | - Harry Ischiropoulos
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Tobias Baumgart
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| | - E. James Petersson
- Department of Chemistry, University of Pennsylvania, 231 South 34 Street, Philadelphia, PA 19104
| |
Collapse
|
75
|
Structural Characteristics of α-Synuclein Oligomers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 329:79-143. [DOI: 10.1016/bs.ircmb.2016.08.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
76
|
van Diggelen F, Tepper AWJW, Apetri MM, Otzen DE. α-Synuclein Oligomers: A Study in Diversity. Isr J Chem 2016. [DOI: 10.1002/ijch.201600116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Femke van Diggelen
- Crossbeta Biosciences; Padualaan 8 3584CH Utrecht The Netherlands
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| | | | | | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO); Aarhus University; Gustav Wieds Vej 14 8000C Aarhus Denmark
| |
Collapse
|
77
|
Konijnenberg A, Ranica S, Narkiewicz J, Legname G, Grandori R, Sobott F, Natalello A. Opposite Structural Effects of Epigallocatechin-3-gallate and Dopamine Binding to α-Synuclein. Anal Chem 2016; 88:8468-75. [PMID: 27467405 DOI: 10.1021/acs.analchem.6b00731] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The intrinsically disordered and amyloidogenic protein α-synuclein (AS) has been linked to several neurodegenerative states, including Parkinson's disease. Here, nanoelectrospray-ionization mass spectrometry (nano-ESI-MS), ion mobility (IM), and native top-down electron transfer dissociation (ETD) techniques are employed to study AS interaction with small molecules known to modulate its aggregation, such as epigallocatechin-3-gallate (EGCG) and dopamine (DA). The complexes formed by the two ligands under identical conditions reveal peculiar differences. While EGCG engages AS in compact conformations, DA preferentially binds to the protein in partially extended conformations. The two ligands also have different effects on AS structure as assessed by IM, with EGCG leading to protein compaction and DA to its extension. Native top-down ETD on the protein-ligand complexes shows how the different observed modes of binding of the two ligands could be related to their known opposite effects on AS aggregation. The results also show that the protein can bind either ligand in the absence of any covalent modifications, such as oxidation.
Collapse
Affiliation(s)
- Albert Konijnenberg
- Biomolecular & Analytical Mass Spectrometry, University of Antwerp , Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Simona Ranica
- Department of Biotechnology and Biosciences, University of Milano-Bicocca , Piazza della Scienza 2, 20126 Milan, Italy
| | - Joanna Narkiewicz
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA) and ELETTRA-Sincrotrone Trieste S.C.p.A , 34136 Trieste, Italy
| | - Giuseppe Legname
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA) and ELETTRA-Sincrotrone Trieste S.C.p.A , 34136 Trieste, Italy
| | - Rita Grandori
- Department of Biotechnology and Biosciences, University of Milano-Bicocca , Piazza della Scienza 2, 20126 Milan, Italy
| | - Frank Sobott
- Biomolecular & Analytical Mass Spectrometry, University of Antwerp , Groenenborgerlaan 171, 2020 Antwerp, Belgium.,Astbury Centre for Structural Molecular Biology, University of Leeds , Leeds, LS2 9JT, U.K.,School of Molecular and Cellular Biology, University of Leeds , Leeds, LS2 9JT, U.K
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milano-Bicocca , Piazza della Scienza 2, 20126 Milan, Italy.,Consorzio Nazionale Interuniversitario per le Scienze Fisiche della Materia (CNISM), UdR of Milano-Bicocca, and Milan Center of Neuroscience (NeuroMI), 20126 Milan, Italy
| |
Collapse
|
78
|
Petta I, Lievens S, Libert C, Tavernier J, De Bosscher K. Modulation of Protein-Protein Interactions for the Development of Novel Therapeutics. Mol Ther 2016; 24:707-18. [PMID: 26675501 PMCID: PMC4886928 DOI: 10.1038/mt.2015.214] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/12/2015] [Indexed: 01/10/2023] Open
Abstract
Protein-protein interactions (PPIs) underlie most biological processes. An increasing interest to investigate the unexplored potential of PPIs in drug discovery is driven by the need to find novel therapeutic targets for a whole range of diseases with a high unmet medical need. To date, PPI inhibition with small molecules is the mechanism that has most often been explored, resulting in significant progress towards drug development. However, also PPI stabilization is gradually gaining ground. In this review, we provide a focused overview of a number of PPIs that control critical regulatory pathways and constitute targets for the design of novel therapeutics. We discuss PPI-modulating small molecules that are already pursued in clinical trials. In addition, we review a number of PPIs that are still under preclinical investigation but for which preliminary data support their use as therapeutic targets.
Collapse
Affiliation(s)
- Ioanna Petta
- Receptor Research Laboratories, Cytokine Receptor Lab (CRL), VIB Department of Medical Protein Research, Ghent, Belgium
- Department of Biochemistry, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sam Lievens
- Receptor Research Laboratories, Cytokine Receptor Lab (CRL), VIB Department of Medical Protein Research, Ghent, Belgium
- Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Claude Libert
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jan Tavernier
- Receptor Research Laboratories, Cytokine Receptor Lab (CRL), VIB Department of Medical Protein Research, Ghent, Belgium
- Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Cytokine Receptor Lab (CRL), VIB Department of Medical Protein Research, Ghent, Belgium
- Department of Biochemistry, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Lab (NRL), VIB Department of Medical Protein Research, Ghent, Belgium
| |
Collapse
|
79
|
Impact of Plant-Derived Flavonoids on Neurodegenerative Diseases. Neurotox Res 2016; 30:41-52. [PMID: 26951456 DOI: 10.1007/s12640-016-9600-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/24/2015] [Accepted: 01/21/2016] [Indexed: 12/27/2022]
Abstract
Neurodegenerative disorders have a common characteristic that is the involvement of different cell types, typically the reactivity of astrocytes and microglia, characterizing gliosis, which in turn contributes to the neuronal dysfunction and or death. Flavonoids are secondary metabolites of plant origin widely investigated at present and represent one of the most important and diversified among natural products phenolic groups. Several biological activities are attributed to this class of polyphenols, such as antitumor activity, antioxidant, antiviral, and anti-inflammatory, among others, which give significant pharmacological importance. Our group have observed that flavonoids derived from Brazilian plants Dimorphandra mollis Bent., Croton betulaster Müll. Arg., e Poincianella pyramidalis Tul., botanical synonymous Caesalpinia pyramidalis Tul. also elicit a broad spectrum of responses in astrocytes and neurons in culture as activation of astrocytes and microglia, astrocyte associated protection of neuronal progenitor cells, neuronal differentiation and neuritogenesis. It was observed the flavonoids also induced neuronal differentiation of mouse embryonic stem cells and human pluripotent stem cells. Moreover, with the objective of seeking preclinical pharmacological evidence of these molecules, in order to assess its future use in the treatment of neurodegenerative disorders, we have evaluated the effects of flavonoids in preclinical in vitro models of neuroinflammation associated with Parkinson's disease and glutamate toxicity associated with ischemia. In particular, our efforts have been directed to identify mechanisms involved in the changes in viability, morphology, and glial cell function induced by flavonoids in cultures of glial cells and neuronal cells alone or in interactions and clarify the relation with their neuroprotective and morphogetic effects.
Collapse
|
80
|
Li DW, Li GR, Zhang BL, Feng JJ, Zhao H. Damage to dopaminergic neurons is mediated by proliferating cell nuclear antigen through the p53 pathway under conditions of oxidative stress in a cell model of Parkinson's disease. Int J Mol Med 2015; 37:429-35. [PMID: 26677001 DOI: 10.3892/ijmm.2015.2430] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/30/2015] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress is widely considered as a central event in the pathogenesis of Parkinson's disease (PD). The mechanisms underlying the oxidative damage-mediated loss of dopaminergic neurons in PD are not yet fully understood. Accumulating evidence has indicated that oxidative DNA damage plays a crucial role in programmed neuronal cell death, and is considered to be at least partly responsible for the degeneration of dopaminergic neurons in PD. This process involves a number of signaling cascades and molecular proteins. Proliferating cell nuclear antigen (PCNA) is a pleiotropic protein affecting a wide range of vital cellular processes, including chromatin remodelling, DNA repair and cell cycle control, by interacting with a number of enzymes and regulatory proteins. In the present study, the exposure of PC12 cells to 1-methyl-4-phenylpyridinium (MPP+) led to the loss of cell viability and decreased the expression levels of PCNA in a dose- and time-dependent manner, indicating that this protein may be involved in the neurotoxic actions of MPP+ in dopaminergic neuronal cells. In addition, a significant upregulation in p53 expression was also observed in this cellular model of PD. p53 is an upstream inducer of PCNA and it has been recognized as a key contributor responsible for dopaminergic neuronal cell death in mouse models of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. This indicates that MPP+-induced oxidative damage is mediated by the downregulation of PCNA through the p53 pathway in a cellular model of PD. Thus, our results may provide some novel insight into the molecular mechanisms responsible for the development of PD and provide new possible therapeutic targets for the treatment of PD.
Collapse
Affiliation(s)
- Da-Wei Li
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guang-Ren Li
- Department of Neurology, The Third Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Bei-Lin Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing-Jing Feng
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hua Zhao
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
81
|
Luna E, Luk KC. Bent out of shape: α-Synuclein misfolding and the convergence of pathogenic pathways in Parkinson's disease. FEBS Lett 2015; 589:3749-59. [PMID: 26505673 DOI: 10.1016/j.febslet.2015.10.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/15/2015] [Accepted: 10/18/2015] [Indexed: 02/03/2023]
Abstract
Protein inclusions made up primarily of misfolded α-synuclein (α-Syn) are the hallmark of a set of disorders known as synucleinopathies, most notably Parkinson's disease (PD). It is becoming increasingly appreciated that α-Syn misfolding can spread to anatomically connected regions in a prion-like manner. The protein aggregates that ensue are correlated with neurodegeneration in the various yet select neuronal populations that are affected. Recent advances have begun to shed light on the spreading and toxicity mechanisms that may be occurring in PD. Several key emerging themes are arising from this work suggesting that α-Syn mediated neurodegeneration is due to a combination of relative α-Syn expression level, connectivity to affected brain regions, and intrinsic vulnerability to pathology.
Collapse
Affiliation(s)
- Esteban Luna
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, United States.
| |
Collapse
|
82
|
Janowska MK, Baum J. The loss of inhibitory C-terminal conformations in disease associated P123H β-synuclein. Protein Sci 2015; 25:286-94. [PMID: 26332674 DOI: 10.1002/pro.2798] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/27/2015] [Indexed: 11/06/2022]
Abstract
β-synuclein (βS) is a homologue of α-synuclein (αS), the major protein component of Lewy bodies in patients with Parkinson's disease. In contrast to αS, βS does not form fibrils, mitigates αS toxicity in vivo and inhibits αS fibril formation in vitro. Previously a missense mutation of βS, P123H, was identified in patients with Dementia with Lewy Body disease. The single P123H mutation at the C-terminus of βS is able to convert βS from a nontoxic to a toxic protein that is also able to accelerate formation of inclusions when it is in the presence of αS in vivo. To elucidate the molecular mechanisms of these processes, we compare the conformational properties of the monomer forms of αS, βS and P123H-βS, and the effects on fibril formation of coincubation of αS with βS, and with P123H-βS. NMR residual dipolar couplings and secondary structure propensities show that the P123H mutation of βS renders it more flexible C-terminal to the mutation site and more αS-like. In vitro Thioflavin T fluorescence experiments show that P123H-βS accelerates αS fibril formation upon coincubation, as opposed to wild type βS that acts as an inhibitor of αS aggregation. When P123H-βS becomes more αS-like it is unable to perform the protective function of βS, which suggests that the extended polyproline II motif of βS in the C-terminus is critical to its nontoxic nature and to inhibition of αS upon coincubation. These studies may provide a basis for understanding which regions to target for therapeutic intervention in Parkinson's disease.
Collapse
Affiliation(s)
- Maria K Janowska
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854
| |
Collapse
|
83
|
Khalife M, Morshedi D, Aliakbari F, Tayaranian Marvian A, Mohammad Beigi H, Azimzadeh Jamalkandi S, Pan-Montojo F. Alpha-Synuclein Fibrils Interact with Dopamine Reducing its Cytotoxicity on PC12 Cells. Protein J 2015; 34:291-303. [DOI: 10.1007/s10930-015-9625-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
84
|
Hayden EY, Kaur P, Williams TL, Matsui H, Yeh SR, Rousseau DL. Heme Stabilization of α-Synuclein Oligomers during Amyloid Fibril Formation. Biochemistry 2015; 54:4599-610. [PMID: 26161848 DOI: 10.1021/acs.biochem.5b00280] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
α-Synuclein (αSyn), which forms amyloid fibrils, is linked to the neuronal pathology of Parkinson's disease, as it is the major fibrillar component of Lewy bodies, the inclusions that are characteristic of the disease. Oligomeric structures, common to many neurodegenerative disease-related proteins, may in fact be the primary toxic species, while the amyloid fibrils exist either as a less toxic dead-end species or even as a beneficial mechanism for clearing damaged proteins. To alter the progression of the aggregation and gain insights into the prefibrillar structures, we determined the effect of heme on αSyn oligomerization by several different techniques, including native (nondenaturing) polyacrylamide gel electrophoresis, thioflavin T fluorescence, transmission electron microscopy, atomic force microscopy, circular dichroism, and membrane permeation using a calcein release assay. During aggregation, heme is able to bind the αSyn in a specific fashion, stabilizing distinct oligomeric conformations and promoting the formation of αSyn into annular structures, thereby delaying and/or inhibiting the fibrillation process. These results indicate that heme may play a regulatory role in the progression of Parkinson's disease; in addition, they provide insights into how the aggregation process may be altered, which may be applicable to the understanding of many neurodegenerative diseases.
Collapse
Affiliation(s)
- Eric Y Hayden
- †Department of Physiology and Biophysics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York 10461, United States
| | - Prerna Kaur
- ‡Department of Chemistry, Hunter College and Graduate Center, The City University of New York, New York, New York 10021, United States
| | - Thomas L Williams
- §Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Hiroshi Matsui
- ‡Department of Chemistry, Hunter College and Graduate Center, The City University of New York, New York, New York 10021, United States
| | - Syun-Ru Yeh
- †Department of Physiology and Biophysics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York 10461, United States
| | - Denis L Rousseau
- †Department of Physiology and Biophysics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York 10461, United States
| |
Collapse
|
85
|
Abstract
SIGNIFICANCE A constant accumulation of oxidized proteins takes place during aging. Oxidation of proteins leads to a partial unfolding and, therefore, to aggregation. Protein aggregates impair the activity of cellular proteolytic systems (proteasomes, lysosomes), resulting in further accumulation of oxidized proteins. In addition, the accumulation of highly crosslinked protein aggregates leads to further oxidant formation, damage to macromolecules, and, finally, to apoptotic cell death. Furthermore, protein oxidation seems to play a role in the development of various age-related diseases, for example, neurodegenerative diseases. RECENT ADVANCES The highly oxidized lipofuscin accumulates during aging. Lipofuscin formation might cause impaired lysosomal and proteasomal degradation, metal ion accumulation, increased reactive oxygen species formation, and apoptosis. CRITICAL ISSUES It is still unclear to which extent protein oxidation is involved in the progression of aging and in the development of some age-related diseases. FUTURE DIRECTIONS An extensive knowledge of the effects of protein oxidation on the aging process and its contribution to the development of age-related diseases could enable further strategies to reduce age-related impairments. Strategies aimed at lowering aggregate formation might be a straightforward intervention to reduce age-related malfunctions of organs.
Collapse
Affiliation(s)
- Sandra Reeg
- German Institute of Human Nutrition , Nuthetal, Germany
| | - Tilman Grune
- German Institute of Human Nutrition , Nuthetal, Germany
| |
Collapse
|
86
|
Pavlin M, Repič M, Vianello R, Mavri J. The Chemistry of Neurodegeneration: Kinetic Data and Their Implications. Mol Neurobiol 2015; 53:3400-3415. [PMID: 26081152 DOI: 10.1007/s12035-015-9284-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/03/2015] [Indexed: 01/01/2023]
Abstract
We collected experimental kinetic rate constants for chemical processes responsible for the development and progress of neurodegeneration, focused on the enzymatic and non-enzymatic degradation of amine neurotransmitters and their reactive and neurotoxic metabolites. A gross scheme of neurodegeneration on the molecular level is based on two pathways. Firstly, reactive species oxidise heavy atom ions, which enhances the interaction with alpha-synuclein, thus promoting its folding to the beta form and giving rise to insoluble amyloid plaques. The latter prevents the function of vesicular transport leading to gradual neuronal death. In the second pathway, radical species, OH(·) in particular, react with the methylene groups of the apolar part of the lipid bilayer of either the cell or mitochondrial wall, resulting in membrane leakage followed by dyshomeostasis, loss of resting potential and neuron death. Unlike all other central neural system (CNS)-relevant biogenic amines, dopamine and noradrenaline are capable of a non-enzymatic auto-oxidative reaction, which produces hydrogen peroxide. This reaction is not limited to the mitochondrial membrane where scavenging enzymes, such as catalase, are located. On the other hand, dopamine and its metabolites, such as dopamine-o-quinone, dopaminechrome, 5,6-dihydroxyindole and indo-5,6-quinone, also interact directly with alpha-synuclein and reversibly inhibit plaque formation. We consider the role of the heavy metal ions, selected scavengers and scavenging enzymes, and discuss the relevance of certain foods and food supplements, including curcumin, garlic, N-acetyl cysteine, caffeine and red wine, as well as the long-term administration of non-steroid anti-inflammatory drugs and occasional tobacco smoking, that could all act toward preventing neurodegeneration. The current analysis can be employed in developing strategies for the prevention and treatment of neurodegeneration, and, hopefully, aid in the building of an overall kinetic molecular model of neurodegeneration itself.
Collapse
Affiliation(s)
- Matic Pavlin
- Computational Biophysics, German Research School for Simulation Sciences, Joint Venture of RWTH Aachen University and Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.,Computational Biomedicine, Institute for Advanced Simulations (IAS-5/INM-9), 52425, Jülich, Germany
| | - Matej Repič
- Laboratory of Computational Chemistry and Biochemistry, Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Robert Vianello
- Quantum Organic Chemistry Group, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000, Zagreb, Croatia.
| | - Janez Mavri
- National Institute of Chemistry, Hajdrihova 19, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
87
|
Barrett PJ, Timothy Greenamyre J. Post-translational modification of α-synuclein in Parkinson's disease. Brain Res 2015; 1628:247-253. [PMID: 26080075 DOI: 10.1016/j.brainres.2015.06.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and the most prevalent degenerative movement disorder. It is estimated that the prevalence of such age-related neurodegenerative diseases will double in the next 25 years. While the etiology of Parkinson's disease is not entirely clear, a common link between both inherited and sporadic forms of disease is the protein α-synuclein. In PD brains, α-synuclein is typically found in large, insoluble protein aggregates referred to as Lewy bodies and Lewy neurites. The exact role of α-synuclein is still unknown, but it has been shown to undergo a variety of post-translational modifications, which impact α-synuclein aggregation and oligomer formation in different ways. This review highlights key post-translational modifications and the impact they have on α-synuclein aggregation and toxicity, elucidating potential mechanisms for PD pathogenesis and targets for future therapeutics. This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
Affiliation(s)
- Paul J Barrett
- Department of Neurology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - J Timothy Greenamyre
- Department of Neurology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
88
|
Stabilization of Alpha-Synuclein Oligomers In Vitro by the Neurotransmitters, Dopamine and Norepinephrine: The Effect of Oxidized Catecholamines. Neurochem Res 2015; 40:1341-9. [DOI: 10.1007/s11064-015-1597-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/20/2015] [Accepted: 04/30/2015] [Indexed: 10/23/2022]
|
89
|
The N-terminal residues 43 to 60 form the interface for dopamine mediated α-synuclein dimerisation. PLoS One 2015; 10:e0116497. [PMID: 25679387 PMCID: PMC4332483 DOI: 10.1371/journal.pone.0116497] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/10/2014] [Indexed: 12/17/2022] Open
Abstract
α-synuclein (α-syn) is a major component of the intracellular inclusions called Lewy bodies, which are a key pathological feature in the brains of Parkinson's disease patients. The neurotransmitter dopamine (DA) inhibits the fibrillisation of α-syn into amyloid, and promotes α-syn aggregation into SDS-stable soluble oligomers. While this inhibition of amyloid formation requires the oxidation of both DA and the methionines in α-syn, the molecular basis for these processes is still unclear. This study sought to define the protein sequences required for the generation of oligomers. We tested N- (α-syn residues 43-140) and C-terminally (1-95) truncated α-syn, and found that similar to full-length protein both truncated species formed soluble DA:α-syn oligomers, albeit 1-95 had a different profile. Using nuclear magnetic resonance (NMR), and the N-terminally truncated α-syn 43-140 protein, we analysed the structural characteristics of the DA:α-syn 43-140 dimer and α-syn 43-140 monomer and found the dimerisation interface encompassed residues 43 to 60. Narrowing the interface to this small region will help define the mechanism by which DA mediates the formation of SDS-stable soluble DA:α-syn oligomers.
Collapse
|
90
|
Andreasen M, Lorenzen N, Otzen D. Interactions between misfolded protein oligomers and membranes: A central topic in neurodegenerative diseases? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1897-907. [PMID: 25666871 DOI: 10.1016/j.bbamem.2015.01.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 11/19/2022]
Abstract
The deposition of amyloid material has been associated with many different diseases. Although these diseases are very diverse the amyloid material share many common features such as cross-β-sheet structure of the backbone of the proteins deposited. Another common feature of the aggregation process for a wide variety of proteins is the presence of prefibrillar oligomers. These oligomers are linked to the cytotoxicity occurring during the aggregation of proteins. These prefibrillar oligomers interact extensively with lipid membranes and in some cases leads to destabilization of lipid membranes. This interaction is however highly dependent on the nature of both the oligomer and the lipids. Anionic lipids are often required for interaction with the lipid membrane while increased exposure of hydrophobic patches from highly dynamic protein oligomers are structural determinants of cytotoxicity of the oligomers. To explore the oligomer lipid interaction in detail the interaction between oligomers of α-synuclein and the 4th fasciclin-1 domain of TGFBIp with lipid membranes will be examined here. For both proteins the dynamic species are the ones causing membrane destabilization and the membrane interaction is primarily seen when the lipid membranes contain anionic lipids. Hence the dynamic nature of oligomers with exposed hydrophobic patches alongside the presence of anionic lipids could be essential for the cytotoxicity observed for prefibrillar oligomers in general. This article is part of a Special Issue entitled: Lipid-protein interactions.
Collapse
Affiliation(s)
- Maria Andreasen
- Department of Chemistry, Cambridge University, Lensfield Road, Cambridge CB2 1EW, UK; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK 8000 Aarhus C, Denmark
| | - Nikolai Lorenzen
- Department of Protein Biophysics and Formulation, Biopharmaceuticals Research Unit, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Daniel Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK 8000 Aarhus C, Denmark.
| |
Collapse
|
91
|
Segura-Aguilar J, Kostrzewa RM. Neurotoxin mechanisms and processes relevant to Parkinson's disease: an update. Neurotox Res 2015; 27:328-54. [PMID: 25631236 DOI: 10.1007/s12640-015-9519-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 12/14/2022]
Abstract
The molecular mechanism responsible for degenerative process in the nigrostriatal dopaminergic system in Parkinson's disease (PD) remains unknown. One major advance in this field has been the discovery of several genes associated to familial PD, including alpha synuclein, parkin, LRRK2, etc., thereby providing important insight toward basic research approaches. There is an consensus in neurodegenerative research that mitochon dria dysfunction, protein degradation dysfunction, aggregation of alpha synuclein to neurotoxic oligomers, oxidative and endoplasmic reticulum stress, and neuroinflammation are involved in degeneration of the neuromelanin-containing dopaminergic neurons that are lost in the disease. An update of the mechanisms relating to neurotoxins that are used to produce preclinical models of Parkinson´s disease is presented. 6-Hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and rotenone have been the most wisely used neurotoxins to delve into mechanisms involved in the loss of dopaminergic neurons containing neuromelanin. Neurotoxins generated from dopamine oxidation during neuromelanin formation are likewise reviewed, as this pathway replicates neurotoxin-induced cellular oxidative stress, inactivation of key proteins related to mitochondria and protein degradation dysfunction, and formation of neurotoxic aggregates of alpha synuclein. This survey of neurotoxin modeling-highlighting newer technologies and implicating a variety of processes and pathways related to mechanisms attending PD-is focused on research studies from 2012 to 2014.
Collapse
Affiliation(s)
- Juan Segura-Aguilar
- Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Independencia 1027, Casilla, 70000, Santiago 7, Chile,
| | | |
Collapse
|
92
|
Muñoz P, Cardenas S, Huenchuguala S, Briceño A, Couve E, Paris I, Segura-Aguilar J. DT-Diaphorase Prevents Aminochrome-Induced Alpha-Synuclein Oligomer Formation and Neurotoxicity. Toxicol Sci 2015; 145:37-47. [PMID: 25634539 DOI: 10.1093/toxsci/kfv016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
It was reported that aminochrome induces the formation of alpha synuclein (SNCA) oligomers during dopamine oxidation. We found that DT-diaphorase (NQO1) prevents the formation of SNCA oligomers in the presence of aminochrome determined by Western blot, transmission electron microscopy, circular dichroism, and thioflavin T fluorescence, suggesting a protective role of NQO1 by preventing the formation of SNCA oligomers in dopaminergic neurons. In order to test NQO1 protective role in SNCA neurotoxicity in cellular model, we overexpressed SNCA in both RCSN-3 cells (wild-type) and RCSN-3Nq7 cells, which have constitutive expression of a siRNA against NQO1. The expression of SNCA in RCSN-3SNCA and RCSN-3Nq7SNCA cells increased 4.2- and 4.4-fold, respectively. The overexpression of SNCA in RCSN-3Nq7SNCA cells induces a significant increase in cell death of 2.8- and 3.2-fold when they were incubated with 50 and 70 µM aminochrome, respectively. The cell death was found to be of apoptotic character determined by annexin/propidium iodide technique with flow cytometry and DNA laddering. A Western blot demonstrated that SNCA in RCSN-3SNCA is only found in monomer form both in the presence of 20 µM aminochrome or cell culture medium contrasting with RCSN-3Nq7SNCA cells where the majority SNCA is found as oligomer. The antioligomer compound scyllo-inositol induced a significant decrease in aminochrome-induced cell death in RCSN-3Nq7SNCA cells in comparison to cells incubated in the absence of scyllo-inositol. Our results suggest that NQO1 seems to play an important role in the prevention of aminochrome-induced SNCA oligomer formation and SNCA oligomers neurotoxicity in dopaminergic neurons.
Collapse
Affiliation(s)
- Patricia Muñoz
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| | - Sergio Cardenas
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| | - Sandro Huenchuguala
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| | - Andrea Briceño
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| | - Eduardo Couve
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| | - Irmgard Paris
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| | - Juan Segura-Aguilar
- *Department of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile; Faculty of Engineering, Central University of Chile; Department of Biology, University of Valparaiso, Chile; and Department Basic Sciences, University of Santo Tomas, Viña del Mar, Chile
| |
Collapse
|
93
|
Giráldez-Pérez RM, Antolín-Vallespín M, Muñoz MD, Sánchez-Capelo A. Models of α-synuclein aggregation in Parkinson's disease. Acta Neuropathol Commun 2014; 2:176. [PMID: 25497491 PMCID: PMC4272812 DOI: 10.1186/s40478-014-0176-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is not only characterized by motor disturbances but also, by cognitive, sensory, psychiatric and autonomic dysfunction. It has been proposed that some of these symptoms might be related to the widespread pathology of α-synuclein (α-syn) aggregation in different nuclei of the central and peripheral nervous system. However, the pathogenic formation of α-syn aggregates in different brain areas of PD patients is poorly understood. Most experimental models of PD are valuable to assess specific aspects of its pathogenesis, such as toxin-induced dopaminergic neurodegeneration. However, new models are required that reflect the widespread and progressive formation of α-syn aggregates in different brain areas. Such α-syn aggregation is induced in only a few animal models, for example perikaryon inclusions are found in rats administered rotenone, aggregates with a neuritic morphology develop in mice overexpressing either mutated or wild-type α-syn, and in Smad3 deficient mice, aggregates form extensively in the perikaryon and neurites of specific brain nuclei. In this review we focus on α-syn aggregation in the human disorder, its genetics and the availability of experimental models. Indeed, evidences show that dopamine (DA) metabolism may be related to α-syn and its conformational plasticity, suggesting an interesting link between the two pathological hallmarks of PD: dopaminergic neurodegeneration and Lewy body (LB) formation.
Collapse
Affiliation(s)
- Rosa María Giráldez-Pérez
- />CIBERNED - Ser. Neurobiología – Investigación, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
- />Departamento Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Mónica Antolín-Vallespín
- />CIBERNED - Ser. Neurobiología – Investigación, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
| | - María Dolores Muñoz
- />Unidad de Neurología Experimental, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
| | - Amelia Sánchez-Capelo
- />CIBERNED - Ser. Neurobiología – Investigación, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
| |
Collapse
|
94
|
Cuevas C, Huenchuguala S, Muñoz P, Villa M, Paris I, Mannervik B, Segura-Aguilar J. Glutathione transferase-M2-2 secreted from glioblastoma cell protects SH-SY5Y cells from aminochrome neurotoxicity. Neurotox Res 2014; 27:217-28. [PMID: 25403520 DOI: 10.1007/s12640-014-9500-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 12/14/2022]
Abstract
U373MG cells are able to take up aminochrome that induces glutathione transferase M2-2 (GSTM2) expression in a concentration-dependent manner where 100 µM aminochrome increases GSTM2 expression by 2.1-fold (P < 0.001) at 3 h. The uptake of (3)H-aminochrome into U373MG cells was significantly reduced in the presence of 2 µM nomifensine (P < 0.001) 100 µM imipramine (P < 0.001) and 50 mM dopamine (P < 0.001). Interestingly, U373MG cells excrete GSTM2 into the conditioned medium and the excretion was significantly increased (2.7-fold; P < 0.001) when the cells were pretreated with 50 µM aminochrome for 3 h. The U373MG-conditioned medium containing GSTM2 protects SH-SY5Y cells incubated with 10 µM aminochrome. The significant protection provided by U373MG-conditioned medium in SH-SY5Y cells incubated with aminochrome was dependent on GSTM2 internalization into SH-SY5Y cells as evidenced by (i) uptake of (14)C-GSTM2 released from U373MG cells into SH-SY5Y cells, a process inhibited by anti-GSTM2 antiserum; (ii) lack of protection of U373MG-conditioned medium in the presence of anti-GSTM2 antiserum on SH-SY5Y cells treated with aminochrome; and (iii) lack of protection of conditioned medium from U373MGsiGST6 that expresses an siRNA directed against GSTM2 on SH-SY5Y cells treated with aminochrome. In conclusion, our results demonstrated that U373MG cells protect SH-SY5Y cells against aminochrome neurotoxicity by releasing GSTM2 into the conditioned medium and subsequent internalization of GSTM2 into SH-SY5Y cells. These results suggest a new mechanism of protection of dopaminergic neurons mediated by astrocytes by releasing GSTM2 into the intersynaptic space and subsequent internalization into dopaminergic neuron in order to protect these cells against aminochrome neurotoxicity.
Collapse
Affiliation(s)
- Carlos Cuevas
- Molecular and Clinical Pharmacology ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
95
|
Gauhar A, Shaykhalishahi H, Gremer L, Mirecka EA, Hoyer W. Impact of subunit linkages in an engineered homodimeric binding protein to α-synuclein. Protein Eng Des Sel 2014; 27:473-9. [PMID: 25332193 DOI: 10.1093/protein/gzu047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aggregation of the protein α-synuclein (α-syn) has been implicated in Parkinson's disease and other neurodegenerative disorders, collectively referred to as synucleinopathies. The β-wrapin AS69 is a small engineered binding protein to α-syn that stabilizes a β-hairpin conformation of monomeric α-syn and inhibits α-syn aggregation at substoichiometric concentrations. AS69 is a homodimer whose subunits are linked via a disulfide bridge between their single cysteine residues, Cys-28. Here we show that expression of a functional dimer as a single polypeptide chain is achievable by head-to-tail linkage of AS69 subunits. Choice of a suitable linker is essential for construction of head-to-tail dimers that exhibit undiminished α-syn affinity compared with the solely disulfide-linked dimer. We characterize AS69-GS3, a head-to-tail dimer with a glycine-serine-rich linker, under oxidized and reduced conditions in order to evaluate the impact of the Cys28-disulfide bond on structure, stability and α-syn binding. Formation of the disulfide bond causes compaction of AS69-GS3, increases its thermostability, and is a prerequisite for high-affinity binding to α-syn. Comparison of AS69-GS3 and AS69 demonstrates that head-to-tail linkage promotes α-syn binding by affording accelerated disulfide bond formation.
Collapse
Affiliation(s)
- Aziz Gauhar
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Hamed Shaykhalishahi
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Lothar Gremer
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany Institute of Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Ewa A Mirecka
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Wolfgang Hoyer
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany Institute of Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| |
Collapse
|
96
|
Xiong R, Siegel D, Ross D. Quinone-induced protein handling changes: implications for major protein handling systems in quinone-mediated toxicity. Toxicol Appl Pharmacol 2014; 280:285-95. [PMID: 25151970 DOI: 10.1016/j.taap.2014.08.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/11/2014] [Accepted: 08/13/2014] [Indexed: 11/24/2022]
Abstract
Para-quinones such as 1,4-Benzoquinone (BQ) and menadione (MD) and ortho-quinones including the oxidation products of catecholamines, are derived from xenobiotics as well as endogenous molecules. The effects of quinones on major protein handling systems in cells; the 20/26S proteasome, the ER stress response, autophagy, chaperone proteins and aggresome formation, have not been investigated in a systematic manner. Both BQ and aminochrome (AC) inhibited proteasomal activity and activated the ER stress response and autophagy in rat dopaminergic N27 cells. AC also induced aggresome formation while MD had little effect on any protein handling systems in N27 cells. The effect of NQO1 on quinone induced protein handling changes and toxicity was examined using N27 cells stably transfected with NQO1 to generate an isogenic NQO1-overexpressing line. NQO1 protected against BQ-induced apoptosis but led to a potentiation of AC- and MD-induced apoptosis. Modulation of quinone-induced apoptosis in N27 and NQO1-overexpressing cells correlated only with changes in the ER stress response and not with changes in other protein handling systems. These data suggested that NQO1 modulated the ER stress response to potentiate toxicity of AC and MD, but protected against BQ toxicity. We further demonstrated that NQO1 mediated reduction to unstable hydroquinones and subsequent redox cycling was important for the activation of the ER stress response and toxicity for both AC and MD. In summary, our data demonstrate that quinone-specific changes in protein handling are evident in N27 cells and the induction of the ER stress response is associated with quinone-mediated toxicity.
Collapse
Affiliation(s)
- Rui Xiong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Ccxampus, Aurora, CO 80045, USA
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Ccxampus, Aurora, CO 80045, USA
| | - David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Ccxampus, Aurora, CO 80045, USA.
| |
Collapse
|
97
|
Uversky VN, Davé V, Iakoucheva LM, Malaney P, Metallo SJ, Pathak RR, Joerger AC. Pathological unfoldomics of uncontrolled chaos: intrinsically disordered proteins and human diseases. Chem Rev 2014; 114:6844-79. [PMID: 24830552 PMCID: PMC4100540 DOI: 10.1021/cr400713r] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute University of South Florida, Tampa, Florida 33612, United States
- Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Vrushank Davé
- Department of Pathology and Cell Biology , Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States
| | - Lilia M. Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093, United States
| | - Prerna Malaney
- Department of Pathology and Cell Biology , Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Steven J. Metallo
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Ravi Ramesh Pathak
- Department of Pathology and Cell Biology , Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Andreas C. Joerger
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
98
|
Corvaglia S, Sanavio B, Hong Enriquez RP, Sorce B, Bosco A, Scaini D, Sabella S, Pompa PP, Scoles G, Casalis L. Atomic force microscopy based nanoassay: a new method to study α-Synuclein-dopamine bioaffinity interactions. Sci Rep 2014; 4:5366. [PMID: 24947141 PMCID: PMC4064358 DOI: 10.1038/srep05366] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 05/28/2014] [Indexed: 12/25/2022] Open
Abstract
Intrinsically Disordered Proteins (IDPs) are characterized by the lack of well-defined 3-D structure and show high conformational plasticity. For this reason, they are a strong challenge for the traditional characterization of structure, supramolecular assembly and biorecognition phenomena. We show here how the fine tuning of protein orientation on a surface turns useful in the reliable testing of biorecognition interactions of IDPs, in particular α-Synuclein. We exploited atomic force microscopy (AFM) for the selective, nanoscale confinement of α-Synuclein on gold to study the early stages of α-Synuclein aggregation and the effect of small molecules, like dopamine, on the aggregation process. Capitalizing on the high sensitivity of AFM topographic height measurements we determined, for the first time in the literature, the dissociation constant of dopamine-α-Synuclein adducts.
Collapse
Affiliation(s)
- Stefania Corvaglia
- NanoInnovation Laboratory, Elettra Sincrotrone Trieste S.C.p.A., S.S.14 Km 163.5, 34149 Basovizza, Trieste, Italy
- Life Science Department, University of Trieste, via Giorgieri 1, I-34127 Trieste, Italy
| | - Barbara Sanavio
- NanoInnovation Laboratory, Elettra Sincrotrone Trieste S.C.p.A., S.S.14 Km 163.5, 34149 Basovizza, Trieste, Italy
- Department of Biological and Medical Science, University of Udine, Ospedale della Misericordia, Piazzale Santa Maria della Misericordia, 15 -33100 Udine, Italy
- Current address: Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, IFOM-IEO-campus, via Adamello, 16, 20139 Milan, Italy
| | - Rolando P. Hong Enriquez
- Department of Biological and Medical Science, University of Udine, Ospedale della Misericordia, Piazzale Santa Maria della Misericordia, 15 -33100 Udine, Italy
| | - Barbara Sorce
- Center for Bio-Molecular Nanotechnologies@UniLe, Istituto Italiano di Tecnologia, Via Barsanti-73010 Arnesano, Lecce, Italy
- Current address: ETH Zürich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Alessandro Bosco
- NanoInnovation Laboratory, Elettra Sincrotrone Trieste S.C.p.A., S.S.14 Km 163.5, 34149 Basovizza, Trieste, Italy
| | - Denis Scaini
- NanoInnovation Laboratory, Elettra Sincrotrone Trieste S.C.p.A., S.S.14 Km 163.5, 34149 Basovizza, Trieste, Italy
- Life Science Department, University of Trieste, via Giorgieri 1, I-34127 Trieste, Italy
| | - Stefania Sabella
- Center for Bio-Molecular Nanotechnologies@UniLe, Istituto Italiano di Tecnologia, Via Barsanti-73010 Arnesano, Lecce, Italy
| | - Pier Paolo Pompa
- Center for Bio-Molecular Nanotechnologies@UniLe, Istituto Italiano di Tecnologia, Via Barsanti-73010 Arnesano, Lecce, Italy
| | - Giacinto Scoles
- NanoInnovation Laboratory, Elettra Sincrotrone Trieste S.C.p.A., S.S.14 Km 163.5, 34149 Basovizza, Trieste, Italy
- Department of Biological and Medical Science, University of Udine, Ospedale della Misericordia, Piazzale Santa Maria della Misericordia, 15 -33100 Udine, Italy
| | - Loredana Casalis
- NanoInnovation Laboratory, Elettra Sincrotrone Trieste S.C.p.A., S.S.14 Km 163.5, 34149 Basovizza, Trieste, Italy
| |
Collapse
|
99
|
Lorenzen N, Nielsen SB, Yoshimura Y, Vad BS, Andersen CB, Betzer C, Kaspersen JD, Christiansen G, Pedersen JS, Jensen PH, Mulder FAA, Otzen DE. How epigallocatechin gallate can inhibit α-synuclein oligomer toxicity in vitro. J Biol Chem 2014; 289:21299-310. [PMID: 24907278 DOI: 10.1074/jbc.m114.554667] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Oligomeric species of various proteins are linked to the pathogenesis of different neurodegenerative disorders. Consequently, there is intense focus on the discovery of novel inhibitors, e.g. small molecules and antibodies, to inhibit the formation and block the toxicity of oligomers. In Parkinson disease, the protein α-synuclein (αSN) forms cytotoxic oligomers. The flavonoid epigallocatechin gallate (EGCG) has previously been shown to redirect the aggregation of αSN monomers and remodel αSN amyloid fibrils into disordered oligomers. Here, we dissect EGCG's mechanism of action. EGCG inhibits the ability of preformed oligomers to permeabilize vesicles and induce cytotoxicity in a rat brain cell line. However, EGCG does not affect oligomer size distribution or secondary structure. Rather, EGCG immobilizes the C-terminal region and moderately reduces the degree of binding of oligomers to membranes. We interpret our data to mean that the oligomer acts by destabilizing the membrane rather than by direct pore formation. This suggests that reduction (but not complete abolition) of the membrane affinity of the oligomer is sufficient to prevent cytotoxicity.
Collapse
Affiliation(s)
- Nikolai Lorenzen
- From the Interdisciplinary Nanoscience Center, Center for Insoluble Protein Structures
| | - Søren B Nielsen
- From the Interdisciplinary Nanoscience Center, Center for Insoluble Protein Structures
| | - Yuichi Yoshimura
- From the Interdisciplinary Nanoscience Center, Center for Insoluble Protein Structures, Departments of Chemistry
| | - Brian S Vad
- From the Interdisciplinary Nanoscience Center, Center for Insoluble Protein Structures
| | | | | | - Jørn D Kaspersen
- From the Interdisciplinary Nanoscience Center, Departments of Chemistry
| | - Gunna Christiansen
- Biomedicine-Medical Immunology, Aarhus University, 8000 Aarhus C, Denmark
| | - Jan S Pedersen
- From the Interdisciplinary Nanoscience Center, Departments of Chemistry
| | | | - Frans A A Mulder
- From the Interdisciplinary Nanoscience Center, Center for Insoluble Protein Structures, Departments of Chemistry
| | - Daniel E Otzen
- From the Interdisciplinary Nanoscience Center, Center for Insoluble Protein Structures,
| |
Collapse
|
100
|
MHC-I expression renders catecholaminergic neurons susceptible to T-cell-mediated degeneration. Nat Commun 2014; 5:3633. [PMID: 24736453 PMCID: PMC4024461 DOI: 10.1038/ncomms4633] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/12/2014] [Indexed: 12/13/2022] Open
Abstract
Subsets of rodent neurons are reported to express major histocompatibilty complex class I (MHC-I), but such expression has not been reported in normal adult human neurons. Here we provide evidence from immunolabel, RNA expression, and mass spectrometry analysis of postmortem samples that human catecholaminergic substantia nigra and locus coeruleus neurons express MHC-I, and that this molecule is inducible in human stem cell derived dopamine (DA) neurons. Catecholamine murine cultured neurons are more responsive to induction of MHC-I by gamma-interferon than other neuronal populations. Neuronal MHC-I is also induced by factors released from microglia activated by neuromelanin or alpha-synuclein, or high cytosolic DA and/or oxidative stress. DA neurons internalize foreign ovalbumin and display antigen derived from this protein by MHC-I, which triggers DA neuronal death in the presence of appropriate cytotoxic T-cells. Thus, neuronal MHC-I can trigger antigenic response, and catecholamine neurons may be particularly susceptible to T cell-mediated cytotoxic attack.
Collapse
|