1
|
Gao V, Chlebowicz J, Gaskin K, Briano JA, Komer LE, Pineda A, Jhalani S, Ahmad S, Uwaifo E, Black LS, Haller JE, Przedborski S, Lane DA, Zhang S, Sharma M, Burré J. Synaptic vesicle-omics in mice captures signatures of aging and synucleinopathy. Nat Commun 2025; 16:4079. [PMID: 40312501 PMCID: PMC12046008 DOI: 10.1038/s41467-025-59441-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 04/23/2025] [Indexed: 05/03/2025] Open
Abstract
Neurotransmitter release occurs through exocytosis of synaptic vesicles. α-Synuclein's function and dysfunction in Parkinson's disease and other synucleinopathies is thought to be tightly linked to synaptic vesicle binding. Age is the biggest risk factor for synucleinopathy, and ~15% of synaptic vesicle proteins have been linked to central nervous system diseases. Yet, age- and disease-induced changes in synaptic vesicles remain unexplored. Via systematic analysis of synaptic vesicles at the ultrastructural, protein, and lipid levels, we reveal specific changes in synaptic vesicle populations, proteins, and lipids over age in wild-type mice and in α-synuclein knockout mice with and without expression of human α-synuclein. Strikingly, we find several previously undescribed synaptic changes in mice lacking α-synuclein, suggesting that loss of α-synuclein function contributes to synaptic dysfunction. These findings not only provide insights into synaptic vesicle biology and disease mechanisms in synucleinopathy, but also serve as a baseline for further mechanistic exploration of age- and disease-related alterations in synaptic vesicles.
Collapse
Affiliation(s)
- Virginia Gao
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Julita Chlebowicz
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Karlton Gaskin
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Juan A Briano
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Lauren E Komer
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - André Pineda
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Shrey Jhalani
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Saad Ahmad
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Eseosa Uwaifo
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Luca S Black
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jillian E Haller
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Serge Przedborski
- Departments of Neurology, Pathology & Cell Biology and Neuroscience, Columbia University, New York, NY, 10032, USA
| | - Diane A Lane
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, 14853, USA
| | - Manu Sharma
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jacqueline Burré
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
2
|
Yang N, Shi L, Xu P, Ren F, Lv S, Li C, Qi X. Identification of potential drug targets for insomnia by Mendelian randomization analysis based on plasma proteomics. Front Neurol 2024; 15:1380321. [PMID: 38725646 PMCID: PMC11079244 DOI: 10.3389/fneur.2024.1380321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Insomnia, a common clinical disorder, significantly impacts the physical and mental well-being of patients. Currently, available hypnotic medications are unsatisfactory due to adverse reactions and dependency, necessitating the identification of new drug targets for the treatment of insomnia. Methods In this study, we utilized 734 plasma proteins as genetic instruments obtained from genome-wide association studies to conduct a Mendelian randomization analysis, with insomnia as the outcome variable, to identify potential drug targets for insomnia. Additionally, we validated our results externally using other datasets. Sensitivity analyses entailed reverse Mendelian randomization analysis, Bayesian co-localization analysis, and phenotype scanning. Furthermore, we constructed a protein-protein interaction network to elucidate potential correlations between the identified proteins and existing targets. Results Mendelian randomization analysis indicated that elevated levels of TGFBI (OR = 1.01; 95% CI, 1.01-1.02) and PAM ((OR = 1.01; 95% CI, 1.01-1.02) in plasma are associated with an increased risk of insomnia, with external validation supporting these findings. Moreover, there was no evidence of reverse causality for these two proteins. Co-localization analysis confirmed that PAM (coloc.abf-PPH4 = 0.823) shared the same variant with insomnia, further substantiating its potential role as a therapeutic target. There are interactive relationships between the potential proteins and existing targets of insomnia. Conclusion Overall, our findings suggested that elevated plasma levels of TGFBI and PAM are connected with an increased risk of insomnia and might be promising therapeutic targets, particularly PAM. However, further exploration is necessary to fully understand the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liangyuan Shi
- Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital) Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Pengfei Xu
- Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital) Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Fang Ren
- Department of Laboratory, Jimo District Qingdao Hospital of Traditional Chinese Medicine, Qingdao, China
| | - Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunlin Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianghua Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Sturchio A, Rocha EM, Kauffman MA, Marsili L, Mahajan A, Saraf AA, Vizcarra JA, Guo Z, Espay AJ. Recalibrating the Why and Whom of Animal Models in Parkinson Disease: A Clinician's Perspective. Brain Sci 2024; 14:151. [PMID: 38391726 PMCID: PMC10887152 DOI: 10.3390/brainsci14020151] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 02/24/2024] Open
Abstract
Animal models have been used to gain pathophysiologic insights into Parkinson's disease (PD) and aid in the translational efforts of interventions with therapeutic potential in human clinical trials. However, no disease-modifying therapy for PD has successfully emerged from model predictions. These translational disappointments warrant a reappraisal of the types of preclinical questions asked of animal models. Besides the limitations of experimental designs, the one-size convergence and oversimplification yielded by a model cannot recapitulate the molecular diversity within and between PD patients. Here, we compare the strengths and pitfalls of different models, review the discrepancies between animal and human data on similar pathologic and molecular mechanisms, assess the potential of organoids as novel modeling tools, and evaluate the types of questions for which models can guide and misguide. We propose that animal models may be of greatest utility in the evaluation of molecular mechanisms, neural pathways, drug toxicity, and safety but can be unreliable or misleading when used to generate pathophysiologic hypotheses or predict therapeutic efficacy for compounds with potential neuroprotective effects in humans. To enhance the translational disease-modification potential, the modeling must reflect the biology not of a diseased population but of subtypes of diseased humans to distinguish What data are relevant and to Whom.
Collapse
Affiliation(s)
- Andrea Sturchio
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Emily M. Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Marcelo A. Kauffman
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología José María Ramos Mejía, Buenos Aires C1221ADC, Argentina;
| | - Luca Marsili
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Abhimanyu Mahajan
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Ameya A. Saraf
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| | - Joaquin A. Vizcarra
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 15213, USA;
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA;
| | - Alberto J. Espay
- James J. and Joan A. Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH 45219, USA; (A.S.); (L.M.); (A.A.S.)
| |
Collapse
|
4
|
Marín I. Emergence of the Synucleins. BIOLOGY 2023; 12:1053. [PMID: 37626939 PMCID: PMC10451939 DOI: 10.3390/biology12081053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
This study establishes the origin and evolutionary history of the synuclein genes. A combination of phylogenetic analyses of the synucleins from twenty-two model species, characterization of local synteny similarities among humans, sharks and lampreys, and statistical comparisons among lamprey and human chromosomes, provides conclusive evidence for the current diversity of synuclein genes arising from the whole-genome duplications (WGDs) that occurred in vertebrates. An ancestral synuclein gene was duplicated in a first WGD, predating the diversification of all living vertebrates. The two resulting genes are still present in agnathan vertebrates. The second WGD, specific to the gnathostome lineage, led to the emergence of the three classical synuclein genes, SNCA, SNCB and SNCG, which are present in all jawed vertebrate lineages. Additional WGDs have added new genes in both agnathans and gnathostomes, while some gene losses have occurred in particular species. The emergence of synucleins through WGDs prevented these genes from experiencing dosage effects, thus avoiding the potential detrimental effects associated with individual duplications of genes that encode proteins prone to aggregation. Additional insights into the structural and functional features of synucleins are gained through the analysis of the highly divergent synuclein proteins present in chondrichthyans and agnathans.
Collapse
Affiliation(s)
- Ignacio Marín
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), 46010 Valencia, Spain
| |
Collapse
|
5
|
Sharma M, Burré J. α-Synuclein in synaptic function and dysfunction. Trends Neurosci 2023; 46:153-166. [PMID: 36567199 PMCID: PMC9877183 DOI: 10.1016/j.tins.2022.11.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
α-Synuclein is a neuronal protein that is enriched in presynaptic terminals. Under physiological conditions, it binds to synaptic vesicle membranes and functions in neurotransmitter release, although the molecular details remain unclear, and it is controversial whether α-synuclein inhibits or facilitates neurotransmitter release. Pathologically, in synucleinopathies including Parkinson's disease (PD), α-synuclein forms aggregates that recruit monomeric α-synuclein and spread throughout the brain, which triggers neuronal dysfunction at molecular, cellular, and organ levels. Here, we present an overview of the effects of α-synuclein on SNARE-complex assembly, neurotransmitter release, and synaptic vesicle pool homeostasis, and discuss how the observed divergent effects of α-synuclein on neurotransmitter release can be reconciled. We also discuss how gain-of-function versus loss-of-function of α-synuclein may contribute to pathogenesis in synucleinopathies.
Collapse
Affiliation(s)
- Manu Sharma
- Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Jacqueline Burré
- Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Myers AJ, Brahimi A, Jenkins IJ, Koob AO. The Synucleins and the Astrocyte. BIOLOGY 2023; 12:biology12020155. [PMID: 36829434 PMCID: PMC9952504 DOI: 10.3390/biology12020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Synucleins consist of three proteins exclusively expressed in vertebrates. α-Synuclein (αS) has been identified as the main proteinaceous aggregate in Lewy bodies, a pathological hallmark of many neurodegenerative diseases. Less is understood about β-synuclein (βS) and γ-synuclein (γS), although it is known βS can interact with αS in vivo to inhibit aggregation. Likewise, both γS and βS can inhibit αS's propensity to aggregate in vitro. In the central nervous system, βS and αS, and to a lesser extent γS, are highly expressed in the neural presynaptic terminal, although they are not strictly located there, and emerging data have shown a more complex expression profile. Synapse loss and astrocyte atrophy are early aspects of degenerative diseases of the brain and correlate with disease progression. Synucleins appear to be involved in synaptic transmission, and astrocytes coordinate and organize synaptic function, with excess αS degraded by astrocytes and microglia adjacent to the synapse. βS and γS have also been observed in the astrocyte and may provide beneficial roles. The astrocytic responsibility for degradation of αS as well as emerging evidence on possible astrocytic functions of βS and γS, warrant closer inspection on astrocyte-synuclein interactions at the synapse.
Collapse
Affiliation(s)
- Abigail J. Myers
- Neuroscience Program, Health Science Research Facility, University of Vermont, 149 Beaumont Ave., Burlington, VT 05405, USA
| | - Ayat Brahimi
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
| | - Imani J. Jenkins
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
| | - Andrew O. Koob
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
- Correspondence: ; Tel.: +1-860-768-5780
| |
Collapse
|
7
|
Dias SB, de Lemos L, Sousa L, Bitoque DB, Silva GA, Seabra MC, Tenreiro S. Age-Related Changes of the Synucleins Profile in the Mouse Retina. Biomolecules 2023; 13:biom13010180. [PMID: 36671565 PMCID: PMC9855780 DOI: 10.3390/biom13010180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Alpha-synuclein (aSyn) plays a central role in Parkinson's disease (PD) and has been extensively studied in the brain. This protein is part of the synuclein family, which is also composed of beta-synuclein (bSyn) and gamma-synuclein (gSyn). In addition to its neurotoxic role, synucleins have important functions in the nervous system, modulating synaptic transmission. Synucleins are expressed in the retina, but they have been poorly characterized. However, there is evidence that they are important for visual function and that they can play a role in retinal degeneration. This study aimed to profile synucleins in the retina of naturally aged mice and to correlate their patterns with specific retinal cells. With aging, we observed a decrease in the thickness of specific retinal layers, accompanied by an increase in glial reactivity. Moreover, the aSyn levels decreased, whereas bSyn increased with aging. The colocalization of both proteins was decreased in the inner plexiform layer (IPL) of the aged retina. gSyn presented an age-related decrease at the inner nuclear layer but was not significantly changed in the ganglion cell layer. The synaptic marker synaptophysin was shown to be preferentially colocalized with aSyn in the IPL with aging. At the same time, aSyn was found to exist at the presynaptic endings of bipolar cells and was affected by aging. Overall, this study suggests that physiological aging can be responsible for changes in the retinal tissue, implicating functional alterations that could affect synuclein family function.
Collapse
Affiliation(s)
- Sarah Batista Dias
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Luísa de Lemos
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Luís Sousa
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Diogo B. Bitoque
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Gabriela Araújo Silva
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Miguel C. Seabra
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Sandra Tenreiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
8
|
Patterson JR, Hirst WD, Howe JW, Russell CP, Cole-Strauss A, Kemp CJ, Duffy MF, Lamp J, Umstead A, Kubik M, Stoll AC, Vega IE, Steece-Collier K, Chen Y, Campbell AC, Nezich CL, Glajch KE, Sortwell CE. Beta2-adrenoreceptor agonist clenbuterol produces transient decreases in alpha-synuclein mRNA but no long-term reduction in protein. NPJ Parkinsons Dis 2022; 8:61. [PMID: 35610264 PMCID: PMC9130326 DOI: 10.1038/s41531-022-00322-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/08/2022] [Indexed: 11/18/2022] Open
Abstract
β2-adrenoreceptor (β2AR) agonists have been associated with a decreased risk of developing Parkinson's disease (PD) and are hypothesized to decrease expression of both alpha-synuclein mRNA (Snca) and protein (α-syn). Effects of β2AR agonist clenbuterol on the levels of Snca mRNA and α-syn protein were evaluated in vivo (rats and mice) and in rat primary cortical neurons by two independent laboratories. A modest decrease in Snca mRNA in the substantia nigra was observed after a single acute dose of clenbuterol in rats, however, this decrease was not maintained after multiple doses. In contrast, α-syn protein levels remained unchanged in both single and multiple dosing paradigms. Furthermore, clenbuterol did not decrease Snca in cultured rat primary cortical neurons, or decrease Snca or α-syn in mice. Additionally, compared to the single-dose paradigm, repeat dosing resulted in substantially lower levels of clenbuterol in plasma and brain tissue in rodents. Based on our observations of a transient decrease in Snca and no effect on α-syn protein in this preclinical study, these data support the conclusion that clenbuterol is not likely a viable disease-modifying strategy for PD.
Collapse
Affiliation(s)
- Joseph R Patterson
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA.
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | - Jacob W Howe
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Christopher P Russell
- Cell and Molecular Biology Department, Grand Valley State University, Allendale, MI, USA
| | - Allyson Cole-Strauss
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Christopher J Kemp
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Jared Lamp
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Andrew Umstead
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Michael Kubik
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Anna C Stoll
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Irving E Vega
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Yi Chen
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | - Anne C Campbell
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | | | - Kelly E Glajch
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Mercy Health Hauenstein Neuroscience Medical Center, Grand Rapids, MI, USA
| |
Collapse
|
9
|
Carnazza KE, Komer LE, Xie YX, Pineda A, Briano JA, Gao V, Na Y, Ramlall T, Buchman VL, Eliezer D, Sharma M, Burré J. Synaptic vesicle binding of α-synuclein is modulated by β- and γ-synucleins. Cell Rep 2022; 39:110675. [PMID: 35417693 PMCID: PMC9116446 DOI: 10.1016/j.celrep.2022.110675] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 01/23/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
α-synuclein, β-synuclein, and γ-synuclein are abundantly expressed proteins in the vertebrate nervous system. α-synuclein functions in neurotransmitter release by binding to and clustering synaptic vesicles and chaperoning SNARE-complex assembly. Pathologically, aggregates originating from soluble pools of α-synuclein are deposited into Lewy bodies in Parkinson's disease and related synucleinopathies. The functions of β-synuclein and γ-synuclein in presynaptic terminals remain poorly studied. Using in vitro liposome binding studies, circular dichroism spectroscopy, immunoprecipitation, and fluorescence resonance energy transfer (FRET) experiments on isolated synaptic vesicles in combination with subcellular fractionation of brains from synuclein mouse models, we show that β-synuclein and γ-synuclein have a reduced affinity toward synaptic vesicles compared with α-synuclein, and that heteromerization of β-synuclein or γ-synuclein with α-synuclein results in reduced synaptic vesicle binding of α-synuclein in a concentration-dependent manner. Our data suggest that β-synuclein and γ-synuclein are modulators of synaptic vesicle binding of α-synuclein and thereby reduce α-synuclein's physiological activity at the neuronal synapse.
Collapse
Affiliation(s)
- Kathryn E Carnazza
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lauren E Komer
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ying Xue Xie
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - André Pineda
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Juan Antonio Briano
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Virginia Gao
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yoonmi Na
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Trudy Ramlall
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Vladimir L Buchman
- School of Biosciences, Cardiff University, Cardiff CF103AX, UK; Belgorod State National Research University, 85 Pobedy Street, Belgorod, Belgorod 308015, Russian Federation
| | - David Eliezer
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jacqueline Burré
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
10
|
Dudok B, Klein PM, Hwaun E, Lee BR, Yao Z, Fong O, Bowler JC, Terada S, Sparks FT, Szabo GG, Farrell JS, Berg J, Daigle TL, Tasic B, Dimidschstein J, Fishell G, Losonczy A, Zeng H, Soltesz I. Alternating sources of perisomatic inhibition during behavior. Neuron 2021; 109:997-1012.e9. [PMID: 33529646 PMCID: PMC7979482 DOI: 10.1016/j.neuron.2021.01.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/02/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Interneurons expressing cholecystokinin (CCK) and parvalbumin (PV) constitute two key GABAergic controllers of hippocampal pyramidal cell output. Although the temporally precise and millisecond-scale inhibitory regulation of neuronal ensembles delivered by PV interneurons is well established, the in vivo recruitment patterns of CCK-expressing basket cell (BC) populations has remained unknown. We show in the CA1 of the mouse hippocampus that the activity of CCK BCs inversely scales with both PV and pyramidal cell activity at the behaviorally relevant timescales of seconds. Intervention experiments indicated that the inverse coupling of CCK and PV GABAergic systems arises through a mechanism involving powerful inhibitory control of CCK BCs by PV cells. The tightly coupled complementarity of two key microcircuit regulatory modules demonstrates a novel form of brain-state-specific segregation of inhibition during spontaneous behavior.
Collapse
Affiliation(s)
- Barna Dudok
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Ernie Hwaun
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Olivia Fong
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - John C Bowler
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Satoshi Terada
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Fraser T Sparks
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Gergely G Szabo
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jim Berg
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gord Fishell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
11
|
γ-Synuclein Induces Human Cortical Astrocyte Proliferation and Subsequent BDNF Expression and Release. Neuroscience 2019; 410:41-54. [PMID: 31078687 DOI: 10.1016/j.neuroscience.2019.04.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 01/03/2023]
Abstract
γ-Synuclein (γ-syn) is expressed by astrocytes in the human nervous system, and increased extracellularly in the brain and cerebrospinal fluid of individuals diagnosed with Alzheimer's disease. Upregulation of γ-syn also coincides with proliferation of glioblastomas and other cancers. In order to better understand regulation and function of extracellular γ-syn, primary human cortical astrocytes were treated with γ-syn conditioned media at various physiological concentrations (50, 100, 150 nM) after cell synchronization. Additionally, extracellular brain-derived neurotrophic factor (BDNF), a neuroprotective growth factor released by astrocytes that has been shown to be decreased extracellularly in neurodegenerative disease, was observed in response to γ-syn treatment. Analysis of 5-bromodeoxyuridine (BrdU) and propidium iodide through flow cytometry 24 h after release from synchronization revealed an increase in G2/M phase of the cell cycle with 100 nM γ-syn during initial cell division, an effect that was reversed at 48 h. However, increased extracellular BDNF was observed at 48 h with 100 nM and 150 nM γ-syn treatment with no difference between controls at 24 h. Further analysis of cell cycle markers with immunocytochemistry of BrdU and Ki67 after treatment with 100 nM γ-syn confirmed increased initial cell proliferation and decreased non-proliferating cells. Western blot analysis demonstrated increased γ-syn levels after 100 nM treatment at 24 and 48 h, and increased pro-BDNF, mature BDNF and cell viability at 48 h. The results demonstrate that γ-syn internalization by human cortical astrocytes causes upregulation of the cell cycle, followed by subsequent BDNF expression and release.
Collapse
|
12
|
Benskey MJ, Sellnow RC, Sandoval IM, Sortwell CE, Lipton JW, Manfredsson FP. Silencing Alpha Synuclein in Mature Nigral Neurons Results in Rapid Neuroinflammation and Subsequent Toxicity. Front Mol Neurosci 2018; 11:36. [PMID: 29497361 PMCID: PMC5819572 DOI: 10.3389/fnmol.2018.00036] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/26/2018] [Indexed: 12/19/2022] Open
Abstract
Human studies and preclinical models of Parkinson’s disease implicate the involvement of both the innate and adaptive immune systems in disease progression. Further, pro-inflammatory markers are highly enriched near neurons containing pathological forms of alpha synuclein (α-syn), and α-syn overexpression recapitulates neuroinflammatory changes in models of Parkinson’s disease. These data suggest that α-syn may initiate a pathological inflammatory response, however the mechanism by which α-syn initiates neuroinflammation is poorly understood. Silencing endogenous α-syn results in a similar pattern of nigral degeneration observed following α-syn overexpression. Here we aimed to test the hypothesis that loss of α-syn function within nigrostriatal neurons results in neuronal dysfunction, which subsequently stimulates neuroinflammation. Adeno-associated virus (AAV) expressing an short hairpin RNA (shRNA) targeting endogenous α-syn was unilaterally injected into the substantia nigra pars compacta (SNc) of adult rats, after which nigrostriatal pathology and indices of neuroinflammation were examined at 7, 10, 14 and 21 days post-surgery. Removing endogenous α-syn from nigrostriatal neurons resulted in a rapid up-regulation of the major histocompatibility complex class 1 (MHC-1) within transduced nigral neurons. Nigral MHC-1 expression occurred prior to any overt cell death and coincided with the recruitment of reactive microglia and T-cells to affected neurons. Following the induction of neuroinflammation, α-syn knockdown resulted in a 50% loss of nigrostriatal neurons in the SNc and a corresponding loss of nigrostriatal terminals and dopamine (DA) concentrations within the striatum. Expression of a control shRNA did not elicit any pathological changes. Silencing α-syn within glutamatergic neurons of the cerebellum did not elicit inflammation or cell death, suggesting that toxicity initiated by α-syn silencing is specific to DA neurons. These data provide evidence that loss of α-syn function within nigrostriatal neurons initiates a neuronal-mediated neuroinflammatory cascade, involving both the innate and adaptive immune systems, which ultimately results in the death of affected neurons.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Rhyomi C Sellnow
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Ivette M Sandoval
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| | - Jack W Lipton
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
13
|
Pathak D, Berthet A, Bendor JT, Yu K, Sellnow RC, Orr AL, Nguyen MK, Edwards RH, Manfredsson FP, Nakamura K. Loss of α-Synuclein Does Not Affect Mitochondrial Bioenergetics in Rodent Neurons. eNeuro 2017; 4:ENEURO.0216-16.2017. [PMID: 28462393 PMCID: PMC5409983 DOI: 10.1523/eneuro.0216-16.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 04/02/2017] [Accepted: 04/06/2017] [Indexed: 12/28/2022] Open
Abstract
Increased α-synuclein (αsyn) and mitochondrial dysfunction play central roles in the pathogenesis of Parkinson's disease (PD), and lowering αsyn is under intensive investigation as a therapeutic strategy for PD. Increased αsyn levels disrupt mitochondria and impair respiration, while reduced αsyn protects against mitochondrial toxins, suggesting that interactions between αsyn and mitochondria influences the pathologic and physiologic functions of αsyn. However, we do not know if αsyn affects normal mitochondrial function or if lowering αsyn levels impacts bioenergetic function, especially at the nerve terminal where αsyn is enriched. To determine if αsyn is required for normal mitochondrial function in neurons, we comprehensively evaluated how lowering αsyn affects mitochondrial function. We found that αsyn knockout (KO) does not affect the respiration of cultured hippocampal neurons or cortical and dopaminergic synaptosomes, and that neither loss of αsyn nor all three (α, β and γ) syn isoforms decreased mitochondria-derived ATP levels at the synapse. Similarly, neither αsyn KO nor knockdown altered the capacity of synaptic mitochondria to meet the energy requirements of synaptic vesicle cycling or influenced the localization of mitochondria to dopamine (DA) synapses in vivo. Finally, αsyn KO did not affect overall energy metabolism in mice assessed with a Comprehensive Lab Animal Monitoring System. These studies suggest either that αsyn has little or no significant physiological effect on mitochondrial bioenergetic function, or that any such functions are fully compensated for when lost. These results implicate that αsyn levels can be reduced in neurons without impairing (or improving) mitochondrial bioenergetics or distribution.
Collapse
Affiliation(s)
- Divya Pathak
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Amandine Berthet
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Jacob T. Bendor
- Department of Neurology and Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, San Francisco, California 94158
| | - Katharine Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Rhyomi C. Sellnow
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503
- Mercy Health Hauenstein Neuroscience Center, Grand Rapids, MI 49503
| | - Adam L. Orr
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Mai K Nguyen
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Robert H. Edwards
- Department of Neurology and Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, San Francisco, California 94158
| | - Fredric P. Manfredsson
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503
- Mercy Health Hauenstein Neuroscience Center, Grand Rapids, MI 49503
| | - Ken Nakamura
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
- Department of Neurology and Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
14
|
Benskey MJ, Perez RG, Manfredsson FP. The contribution of alpha synuclein to neuronal survival and function - Implications for Parkinson's disease. J Neurochem 2016; 137:331-59. [PMID: 26852372 PMCID: PMC5021132 DOI: 10.1111/jnc.13570] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/29/2016] [Indexed: 02/06/2023]
Abstract
The aggregation of alpha synuclein (α-syn) is a neuropathological feature that defines a spectrum of disorders collectively termed synucleinopathies, and of these, Parkinson's disease (PD) is arguably the best characterized. Aggregated α-syn is the primary component of Lewy bodies, the defining pathological feature of PD, while mutations or multiplications in the α-syn gene result in familial PD. The high correlation between α-syn burden and PD has led to the hypothesis that α-syn aggregation produces toxicity through a gain-of-function mechanism. However, α-syn has been implicated to function in a diverse range of essential cellular processes such as the regulation of neurotransmission and response to cellular stress. As such, an alternative hypothesis with equal explanatory power is that the aggregation of α-syn results in toxicity because of a toxic loss of necessary α-syn function, following sequestration of functional forms α-syn into insoluble protein aggregates. Within this review, we will provide an overview of the literature linking α-syn to PD and the knowledge gained from current α-syn-based animal models of PD. We will then interpret these data from the viewpoint of the α-syn loss-of-function hypothesis and provide a potential mechanistic model by which loss of α-syn function could result in at least some of the neurodegeneration observed in PD. By providing an alternative perspective on the etiopathogenesis of PD and synucleinopathies, this may reveal alternative avenues of research in order to identify potential novel therapeutic targets for disease modifying strategies. The correlation between α-synuclein burden and Parkinson's disease pathology has led to the hypothesis that α-synuclein aggregation produces toxicity through a gain-of-function mechanism. However, in this review, we discuss data supporting the alternative hypothesis that the aggregation of α-synuclein results in toxicity because of loss of necessary α-synuclein function at the presynaptic terminal, following sequestration of functional forms of α-synuclein into aggregates.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Ruth G Perez
- Department of Biomedical Sciences, Center of Emphasis in Neuroscience, Paul L. Foster School of Medicine, Texas Tech University of the Health Sciences El Paso, El Paso, Texas, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA
| |
Collapse
|
15
|
Collier TJ, Redmond DE, Steece-Collier K, Lipton JW, Manfredsson FP. Is Alpha-Synuclein Loss-of-Function a Contributor to Parkinsonian Pathology? Evidence from Non-human Primates. Front Neurosci 2016; 10:12. [PMID: 26858591 PMCID: PMC4731516 DOI: 10.3389/fnins.2016.00012] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/11/2016] [Indexed: 01/20/2023] Open
Abstract
Accumulation of alpha-synuclein (α-syn) in Lewy bodies and neurites of midbrain dopamine neurons is diagnostic for Parkinson's disease (PD), leading to the proposal that PD is a toxic gain-of-function synucleinopathy. Here we discuss the alternative viewpoint that α-syn displacement from synapses by misfolding and aggregation results in a toxic loss-of-function. In support of this hypothesis we provide evidence from our pilot study demonstrating that knockdown of endogenous α-syn in dopamine neurons of non-human primates reproduces the pattern of nigrostriatal degeneration characteristic of PD.
Collapse
Affiliation(s)
- Timothy J Collier
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| | - D Eugene Redmond
- Departments of Psychiatry & Neurosurgery, Yale University School of MedicineNew Haven, CT, USA; Axion Research FoundationHamden, CT, USA
| | - Kathy Steece-Collier
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| | - Jack W Lipton
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Science & Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA; Hauenstein Neuroscience Center, Mercy Health Saint Mary'sGrand Rapids, MI, USA
| |
Collapse
|
16
|
Benskey MJ, Kuhn NC, Galligan JJ, Garcia J, Boye SE, Hauswirth WW, Mueller C, Boye SL, Manfredsson FP. Targeted gene delivery to the enteric nervous system using AAV: a comparison across serotypes and capsid mutants. Mol Ther 2015; 23:488-500. [PMID: 25592336 PMCID: PMC4351472 DOI: 10.1038/mt.2015.7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/30/2014] [Indexed: 12/11/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) vectors are one of the most widely used gene transfer systems in research and clinical trials. AAV can transduce a wide range of biological tissues, however to date, there has been no investigation on targeted AAV transduction of the enteric nervous system (ENS). Here, we examined the efficiency, tropism, spread, and immunogenicity of AAV transduction in the ENS. Rats received direct injections of various AAV serotypes expressing green fluorescent protein (GFP) into the descending colon. AAV serotypes tested included; AAV 1, 2, 5, 6, 8, or 9 and the AAV2 and AAV8 capsid mutants, AAV2-Y444F, AAV2-tripleY-F, AAV2-tripleY-F+T-V, AAV8-Y733F, and AAV8-doubeY-F+T-V. Transduction, as determined by GFP-positive cells, occurred in neurons and enteric glia within the myenteric and submucosal plexuses of the ENS. AAV6 and AAV9 showed the highest levels of transduction within the ENS. Transduction efficiency scaled with titer and time, was translated to the murine ENS, and produced no vector-related immune response. A single injection of AAV into the colon covered an area of ~47 mm(2). AAV9 primarily transduced neurons, while AAV6 transduced enteric glia and neurons. This is the first report on targeted AAV transduction of neurons and glia in the ENS.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - James J Galligan
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Joanna Garcia
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Christian Mueller
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Neuroscience Program, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
17
|
Kanaan NM, Manfredsson FP. Loss of functional alpha-synuclein: a toxic event in Parkinson's disease? JOURNAL OF PARKINSONS DISEASE 2014; 2:249-67. [PMID: 23938255 PMCID: PMC4736738 DOI: 10.3233/jpd-012138] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The discovery that alpha-synuclein (α-syn) is the primary component of the neuropathological hallmarks of Parkinson's disease (PD) and the identification of α-syn mutations in numerous inherited forms of PD has positioned α-syn at the top of the list of important factors in the pathogenesis of PD. Based on the pathological accumulation of α-syn in the brains of patients, the field is currently focused on therapeutic strategies that aim to reduce or eliminate α-syn. However, recent evidence suggests α-syn is a critical protein in neuron (i.e. dopamine neurons) survival and that maintaining a certain level of biologically functional α-syn is an important consideration in targeting α-syn for therapies. Despite the widespread interest in α-syn, the normal biological functions remain elusive, but a large body of work is focused on addressing this issue. In this review, we will discuss the current evidence related to α-syn function, α-syn folding and aggregation, and α-syn's role in disease. Finally, we will propose a relatively novel hypothesis on the pathogenesis of PD that hinges upon the premises that functional α-syn is critical to cell survival and that a reduction in biologically functional α-syn, whether through aggregation or reduced expression, may lead to the neurodegeneration in PD.
Collapse
Affiliation(s)
- Nicholas M Kanaan
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | | |
Collapse
|
18
|
Eschbach J, Danzer KM. α-Synuclein in Parkinson's disease: pathogenic function and translation into animal models. NEURODEGENER DIS 2013; 14:1-17. [PMID: 24080741 DOI: 10.1159/000354615] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 07/22/2013] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease is a common neurodegenerative disease characterised by the loss of dopaminergic neurons in the substantia nigra pars compacta and the formation of α-synuclein aggregates found in Lewy bodies throughout the brain. Several α-synuclein transgenic mouse models have been generated, as well as viral-mediated overexpression of wild-type and mutated α-synuclein to mimic the disease and to delineate the pathogenic pathway of α-synuclein-mediated toxicity and neurodegeneration. In this review, we will recapitulate what we have learned about the function of α-synuclein and α-synuclein-mediated toxicity through studies of transgenic animal models, inducible animal models and viral-based models.
Collapse
|
19
|
Oaks AW, Frankfurt M, Finkelstein DI, Sidhu A. Age-dependent effects of A53T alpha-synuclein on behavior and dopaminergic function. PLoS One 2013; 8:e60378. [PMID: 23560093 PMCID: PMC3613356 DOI: 10.1371/journal.pone.0060378] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 02/25/2013] [Indexed: 11/30/2022] Open
Abstract
Expression of A53T mutant human alpha-synuclein under the mouse prion promoter is among the most successful transgenic models of Parkinson's disease. Accumulation of A53T alpha-synuclein causes adult mice to develop severe motor impairment resulting in early death at 8–12 months of age. In younger, pre-symptomatic animals, altered motor activity and anxiety-like behaviors have also been reported. These behavioral changes, which precede severe neuropathology, may stem from non-pathological functions of alpha-synuclein, including modulation of monoamine neurotransmission. Our analysis over the adult life-span of motor activity, anxiety-like, and depressive-like behaviors identifies perturbations both before and after the onset of disease. Young A53T mice had increased distribution of the dopamine transporter (DAT) to the membrane that was associated with increased striatal re-uptake function. DAT function decreased with aging, and was associated with neurochemical alterations that included increased expression of beta-synuclein and gamma synuclein. Prior to normalization of dopamine uptake, transient activation of Tau kinases and hyperphosphorylation of Tau in the striatum were also observed. Aged A53T mice had reduced neuron counts in the substantia nigra pars compacta, yet striatal medium spiny neuron dendritic spine density was largely maintained. These findings highlight the involvement of the synuclein family of proteins and phosphorylation of Tau in the response to dopaminergic dysfunction of the nigrostriatal pathway.
Collapse
Affiliation(s)
- Adam W. Oaks
- Laboratory of Molecular Neurochemistry, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Maya Frankfurt
- Department of Science Education, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, United States of America
| | - David I. Finkelstein
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Anita Sidhu
- Laboratory of Molecular Neurochemistry, Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
20
|
Peña-Oliver Y, Buchman VL, Dalley JW, Robbins TW, Schumann G, Ripley TL, King SL, Stephens DN. Deletion of alpha-synuclein decreases impulsivity in mice. GENES, BRAIN, AND BEHAVIOR 2012; 11:137-46. [PMID: 22142176 PMCID: PMC3380554 DOI: 10.1111/j.1601-183x.2011.00758.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 10/05/2011] [Accepted: 11/03/2011] [Indexed: 12/31/2022]
Abstract
The presynaptic protein alpha-synuclein, associated with Parkinson's Disease (PD), plays a role in dopaminergic neurotransmission and is implicated in impulse control disorders (ICDs) such as drug addiction. In this study we investigated a potential causal relationship between alpha-synuclein and impulsivity, by evaluating differences in motor impulsivity in the 5-choice serial reaction time task (5-CSRTT) in strains of mice that differ in the expression of the alpha-synuclein gene. C57BL/6JOlaHsd mice differ from their C57BL/6J ancestors in possessing a chromosomal deletion resulting in the loss of two genes, snca, encoding alpha-synuclein, and mmrn1, encoding multimerin-1. C57BL/6J mice displayed higher impulsivity (more premature responding) than C57BL/6JOlaHsd mice when the pre-stimulus waiting interval was increased in the 5-CSRTT. In order to ensure that the reduced impulsivity was indeed related to snca, and not adjacent gene deletion, wild type (WT) and mice with targeted deletion of alpha-synuclein (KO) were tested in the 5-CSRTT. Similarly, WT mice were more impulsive than mice with targeted deletion of alpha-synuclein. Interrogation of our ongoing analysis of impulsivity in BXD recombinant inbred mouse lines revealed an association of impulsive responding with levels of alpha-synuclein expression in hippocampus. Expression of beta- and gamma-synuclein, members of the synuclein family that may substitute for alpha-synuclein following its deletion, revealed no differential compensations among the mouse strains. These findings suggest that alpha-synuclein may contribute to impulsivity and potentially, to ICDs which arise in some PD patients treated with dopaminergic medication.
Collapse
Affiliation(s)
- Y Peña-Oliver
- School of Psychology, University of SussexFalmer, Brighton BN1 9QG, UK
| | - V L Buchman
- School of Biosciences, Cardiff UniversityMuseum Avenue, Cardiff CF10 3AX, UK
| | - J W Dalley
- Behavioural and Clinical Neuroscience Institute and Department of Experimental Psychology, University of CambridgeDowning Street, Cambridge CB2 3EB, UK
- Department of Psychiatry, Addenbrooke's Hospital, University of CambridgeHill's Road, Cambridge CB2 2QQ, UK
| | - T W Robbins
- Behavioural and Clinical Neuroscience Institute and Department of Experimental Psychology, University of CambridgeDowning Street, Cambridge CB2 3EB, UK
| | - G Schumann
- Institute of Psychiatry, Kings CollegeDenmark Hill, London, SE5 8AF
| | - T L Ripley
- School of Psychology, University of SussexFalmer, Brighton BN1 9QG, UK
| | - S L King
- School of Psychology, University of SussexFalmer, Brighton BN1 9QG, UK
| | - D N Stephens
- School of Psychology, University of SussexFalmer, Brighton BN1 9QG, UK
| |
Collapse
|
21
|
Peña-Oliver Y, Buchman VL, Stephens DN. Lack of involvement of alpha-synuclein in unconditioned anxiety in mice. Behav Brain Res 2010; 209:234-40. [PMID: 20138921 PMCID: PMC3132456 DOI: 10.1016/j.bbr.2010.01.049] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 01/22/2010] [Accepted: 01/29/2010] [Indexed: 10/19/2022]
Abstract
Alpha-synuclein is implicated in the pathology of Parkinson disease (PD) and is involved in synaptic function, particularly in presynaptic events in dopamine (DA) synapses. Recently, a role for alpha-synuclein in reward and addiction, especially in alcoholism, has been reported. Since PD and alcohol dependence present a strong comorbidity with anxiety disorders, a role for alpha-synuclein in anxiety has been proposed. The aim of the present investigation was to study the involvement of alpha-synuclein in anxiety by testing alpha-synuclein knock out and wild type mice in three different emotionality tests: the open field, the elevated plus maze and the light-dark box. Alpha-synuclein knock out mice and wild type controls displayed consistently similar emotionality profiles in all the tests, suggesting a lack of involvement of alpha-synuclein in unconditioned anxiety in mice.
Collapse
Affiliation(s)
- Yolanda Peña-Oliver
- Department of Psychology, University of Sussex, Falmer, Brighton BN1 9QG, UK
| | | | | |
Collapse
|
22
|
Soehn AS, Pham TT, Schaeferhoff K, Floss T, Weisenhorn DMV, Wurst W, Bonin M, Riess O. Periphilin is strongly expressed in the murine nervous system and is indispensable for murine development. Genesis 2010; 47:697-707. [PMID: 19621438 DOI: 10.1002/dvg.20553] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Periphilin is involved in multiple processes in vivo. To explore its physiological role from an organismic perspective, we generated mice with a gene trap insertion in the periphilin-1 gene. Based on beta-gal reporter activity, a widespread periphilin expression was evident, especially in the developing somites and limbs, the embryonic nervous system, and the adult brain. In accordance with this broad expression, homozygous deficiency of periphilin was lethal in early embryogenesis. Mice with a heterozygous deficiency did not show any abnormalities of brain morphology and function, neither histologically nor regarding the transcriptome. Interestingly, the reduction of the periphilin-1 gene dosage was compensated by an increased expression of the remaining wild-type allele in the brain. These results point to an indispensable function of periphilin during murine development and an important role in the nervous system, reflected by a strong and tightly regulated expression in the murine brain.
Collapse
Affiliation(s)
- Anne S Soehn
- Department of Medical Genetics, University of Tuebingen, Tuebingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Jüch M, Smalla KH, Kähne T, Lubec G, Tischmeyer W, Gundelfinger ED, Engelmann M. Congenital lack of nNOS impairs long-term social recognition memory and alters the olfactory bulb proteome. Neurobiol Learn Mem 2009; 92:469-84. [DOI: 10.1016/j.nlm.2009.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 05/19/2009] [Accepted: 06/10/2009] [Indexed: 12/21/2022]
|
24
|
Sen S, West AB. The therapeutic potential of LRRK2 and alpha-synuclein in Parkinson's disease. Antioxid Redox Signal 2009; 11:2167-87. [PMID: 19271991 PMCID: PMC2787962 DOI: 10.1089/ars.2009.2430] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Revised: 02/25/2009] [Accepted: 03/07/2009] [Indexed: 12/29/2022]
Abstract
Current treatments for Parkinson's disease fail to modify disease progression, and the underlying pathogenic mechanisms remain elusive. The identification of specific targets responsible for disease will aid in the development of relevant model systems and the discovery of neuroprotective and neurorestorative therapies. Two promising protein candidates, alpha-synuclein and LRRK2, offer unique insight into the molecular basis of disease and the potential to intervene in pathogenesis. Although multiple lines of evidence support alpha-synuclein and LRRK2 as robust targets for therapy, the connection between protein function and neurodegeneration is unclear. Technology capable of mitigating alpha-synuclein and LRRK2 disease-associated function will ultimately be required before the true value of these proteins as therapeutic targets can be discerned.
Collapse
Affiliation(s)
- Saurabh Sen
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama School of Medicine, Birmingham, Alabama 35294, USA
| | | |
Collapse
|
25
|
Surgucheva I, Shestopalov VI, Surguchov A. Effect of gamma-synuclein silencing on apoptotic pathways in retinal ganglion cells. J Biol Chem 2008; 283:36377-85. [PMID: 18936092 PMCID: PMC2606004 DOI: 10.1074/jbc.m806660200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 10/15/2008] [Indexed: 11/06/2022] Open
Abstract
gamma-Synuclein (Syn G) is highly expressed in retinal ganglion cells and the loss of these cells in glaucoma is associated with significant reduction of the intracellular Syn G level. However, a causative relationship between these two events has not been established. Here we show that the knockdown of Syn G results in a decreased viability of the immortalized retinal ganglion cells (RGC-5). The Syn G silencing reduces phosphorylation of serine 112 (Ser112) in Bad protein, a member of the Bcl-2 family that plays a critical role in apoptotic cell death signaling. Our gene expression analysis data suggests that changes in Bad phosphorylation status may be caused by a coordinated shift in activities of kinases controlling Bad phosphorylation and phosphatases catalyzing its dephosphorylation. Moreover, increased phosphorylation of Bad-sequestering protein 14-3-3 detected in these cells is also pro-apoptotic. These results suggest that the homeostatic level of Syn G in RGC-5 cells is required for transcriptional regulation of protein kinases and phosphatases, controlling phosphorylation of Bad and 14-3-3. Lowering Syn G causes Bad dephosphorylation, dissociation from phosphorylated 14-3-3, and translocation to mitochondria where it initiates apoptotic death cascade.
Collapse
Affiliation(s)
- Irina Surgucheva
- Laboratory of Retinal Biology, Veterans Affairs Medical Center, Kansas City, Missouri 64128, USA
| | | | | |
Collapse
|
26
|
Häbig K, Walter M, Stappert H, Riess O, Bonin M. Microarray expression analysis of human dopaminergic neuroblastoma cells after RNA interference of SNCA--a key player in the pathogenesis of Parkinson's disease. Brain Res 2008; 1256:19-33. [PMID: 19135032 DOI: 10.1016/j.brainres.2008.12.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 10/01/2008] [Accepted: 12/03/2008] [Indexed: 11/26/2022]
Abstract
The pre-synaptic protein alpha-synuclein is a key player in the pathogenesis of Parkinson's disease (PD). Misfolded alpha-synuclein protofibrils accumulate and serve as seed structures that cause numerous proteins in the cytoplasm of neuronal cells to aggregate into so-called Lewy bodies. Furthermore, both missense mutations and multiplications of the SNCA gene lead to autosomal dominant forms of familial PD. However, the exact biological role of alpha-synuclein in normal brains remains elusive. To gain more insight into the normal function of this protein, we evaluated changes in whole genome expression in dopaminergic neuroblastoma cells (SH-SY5Y) caused by reductions of 90% in alpha-synuclein RNA levels and of 59% in alpha-synuclein protein levels as a result of RNA interference. The expression of 361 genes was altered at least+/-1.5-fold by the RNA interference, with 82 up-regulated and 279 down-regulated. The differentially expressed gene products are involved in the regulation of transcription, cell cycle, protein degradation, apoptosis, neurogenesis, and lipid metabolism. To examine the influence of SNCA down-regulation by RNAi on apoptosis, we performed cell death assays using different stress triggers. The changes observed in the expression profile of dopaminergic neuronal cells following reduction of SNCA expression warrant studies to investigate the role of signaling cascades in familial and idiopathic PD.
Collapse
Affiliation(s)
- Karina Häbig
- Department of Medical Genetics, Institute of Human Genetics, University of Tübingen, Germany
| | | | | | | | | |
Collapse
|
27
|
Al-Wandi A, Ninkina N, Millership S, Williamson SJM, Jones PA, Buchman VL. Absence of alpha-synuclein affects dopamine metabolism and synaptic markers in the striatum of aging mice. Neurobiol Aging 2008; 31:796-804. [PMID: 19097673 PMCID: PMC3146702 DOI: 10.1016/j.neurobiolaging.2008.11.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 10/19/2008] [Accepted: 11/08/2008] [Indexed: 12/22/2022]
Abstract
Despite numerous evidences for neurotoxicity of overexpressed α-synuclein, a protective function was suggested for endogenous α-synuclein and other members of the synuclein family. This protective role is most important for and evident in presynaptic terminals, where synucleins are normally accumulated. However, mice lacking synucleins display no adverse phenotype. In particular, no significant changes in striatal dopamine metabolism and only subtle deficit of dopaminergic neurons in the substantia nigra were found in juvenile or adult mice. To assess whether aging and synuclein deficiency may have additive detrimental effect on the nigrostriatal system, we studied dopaminergic neurons of the substantia nigra and their striatal synapses in 24–26-month-old α-synuclein and γ-synuclein null mutant mice. Significant ∼36% reduction of the striatal dopamine was found in aging α-synuclein, but not γ-synuclein null mutant mice when compared to age-matching wild type mice. This was accompanied by the reduction of TH-positive fibers in the striatum and decrease of striatal levels of TH and DAT. However, no progressive loss of TH-positive neurons was revealed in the substantia nigra of synuclein-deficient aging animals. Our results are consistent with a hypothesis that α-synuclein is important for normal function and integrity of synapses, and suggest that in the aging nervous system dysfunction of this protein could become a predisposition factor for the development of nigrostriatal pathology.
Collapse
Affiliation(s)
- Abdelmojib Al-Wandi
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3US, United Kingdom
| | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Hoepken HH, Gispert S, Azizov M, Klinkenberg M, Ricciardi F, Kurz A, Morales-Gordo B, Bonin M, Riess O, Gasser T, Kögel D, Steinmetz H, Auburger G. Parkinson patient fibroblasts show increased alpha-synuclein expression. Exp Neurol 2008; 212:307-13. [PMID: 18511044 DOI: 10.1016/j.expneurol.2008.04.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 03/19/2008] [Accepted: 04/01/2008] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative movement disorder of advanced age with largely unknown etiology, but well documented tissue damage from oxidative stress. Increased alpha-synuclein (SNCA) expression is known to cause a rare form of PD, early-onset autosomal dominant PARK4. We have previously shown that loss-of-function mutations of the mitochondrial kinase PINK1 which cause the early-onset recessive PARK6 variant result in oxidative damage in patient fibroblasts. We now investigated the molecular chain of events from mitochondrial dysfunction to cell death which is largely unknown. Primary skin fibroblast cultures from patients were analysed for gene expression anomalies. In G309D-PINK1 patient fibroblasts, mainly genes regulated by oxidative stress, as well as genes encoding synaptic proteins such as SNCA showed altered expression. The induction of SNCA was also observed in control fibroblasts with knock-down of PINK1. The induction of SNCA expression was found to constitute a specific disease biomarker in sporadic PD patient fibroblasts. To understand the mechanism of this induction, we exposed control fibroblasts to oxidative, proteasomal and endoplasmic reticulum stress and were able to trigger the SNCA expression upregulation. Our data indicate that loss-of-function of PINK1 leads to enhanced alpha-synuclein expression and altered cell-cell contact. Alpha-synuclein induction might represent a common event for different variants of PD as well as a PD-specific trigger of neurodegeneration. We propose that the expression changes described might potentially serve as biomarkers that allow objective PD patient diagnosis in an accessible, peripheral tissue.
Collapse
|
30
|
Senior SL, Ninkina N, Deacon R, Bannerman D, Buchman VL, Cragg SJ, Wade-Martins R. Increased striatal dopamine release and hyperdopaminergic-like behaviour in mice lacking both alpha-synuclein and gamma-synuclein. Eur J Neurosci 2008; 27:947-57. [PMID: 18333965 PMCID: PMC3145106 DOI: 10.1111/j.1460-9568.2008.06055.x] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alpha-synuclein is intimately involved in the pathogenesis of Parkinson's disease, and has been implicated in the regulation of synthesis, release and reuptake of dopamine (DA). However, mice lacking members of the synuclein family have been reported to display no overt behavioural phenotype. This may be a result of compensatory upregulation of other synucleins during development. Here we report on behaviour and DA synapse function of alpha-synuclein null, gamma-synuclein null, and alpha-gamma-synuclein double-null knockout mice. Double-null mice were hyperactive in a novel environment and alternated at a lower rate in a T-maze spontaneous alternation task, a phenotype reminiscent of mice expressing reduced levels of the DA transporter. To investigate a possible hyperdopaminergic phenotype in alpha-gamma-synuclein double-null mice, we used fast-scan cyclic voltammetry at carbon-fibre microelectrodes to assess DA release and reuptake in striatal slices from wild-type, alpha-null, gamma-null and double-null mice in real time. Double-null mice were found to have a twofold increase in the extracellular concentration of DA detected after discrete electrical stimuli in the striatum. By measuring the rate of reuptake of DA and tissue DA content in these animals, we showed that the observed increase in size of striatal DA transients was not attributable to a decrease in reuptake of DA via the DA transporter, and can not be attributed to an increase in tissue DA levels in the striatum. Rather, we propose that loss of both alpha- and gamma-synuclein causes an increase in release probability from dopaminergic synapses.
Collapse
Affiliation(s)
- Steven L Senior
- Wellcome Trust Centre For Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | | | | | | | | | | | | |
Collapse
|