51
|
Fischer DL, Gombash SE, Kemp CJ, Manfredsson FP, Polinski NK, Duffy MF, Sortwell CE. Viral Vector-Based Modeling of Neurodegenerative Disorders: Parkinson's Disease. Methods Mol Biol 2016; 1382:367-82. [PMID: 26611600 DOI: 10.1007/978-1-4939-3271-9_26] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gene therapy methods are increasingly used to model Parkinson's disease (PD) in animals in an effort to test experimental therapeutics within a more relevant context to disease pathophysiology and neuropathology. We have detailed several criteria that are critical or advantageous to accurately modeling PD in a murine model or in a nonhuman primate. Using these criteria, we then evaluate approaches made to model PD using viral vectors to date, including both adeno-associated viruses and lentiviruses. Lastly, we comment on the consideration of aging as a critical factor for modeling PD.
Collapse
Affiliation(s)
- D Luke Fischer
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- MD/PhD Program, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Sara E Gombash
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Christopher J Kemp
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
| | - Fredric P Manfredsson
- Translational Science and Molecular Medicine, Michigan State University, College of Human Science, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
| | - Nicole K Polinski
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Caryl E Sortwell
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA.
| |
Collapse
|
52
|
De Deurwaerdère P, Di Giovanni G, Millan MJ. Expanding the repertoire of L-DOPA's actions: A comprehensive review of its functional neurochemistry. Prog Neurobiol 2016; 151:57-100. [PMID: 27389773 DOI: 10.1016/j.pneurobio.2016.07.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/18/2016] [Accepted: 07/03/2016] [Indexed: 01/11/2023]
Abstract
Though a multi-facetted disorder, Parkinson's disease is prototypically characterized by neurodegeneration of nigrostriatal dopaminergic neurons of the substantia nigra pars compacta, leading to a severe disruption of motor function. Accordingly, L-DOPA, the metabolic precursor of dopamine (DA), is well-established as a treatment for the motor deficits of Parkinson's disease despite long-term complications such as dyskinesia and psychiatric side-effects. Paradoxically, however, despite the traditional assumption that L-DOPA is transformed in residual striatal dopaminergic neurons into DA, the mechanism of action of L-DOPA is neither simple nor entirely clear. Herein, focussing on its influence upon extracellular DA and other neuromodulators in intact animals and experimental models of Parkinson's disease, we highlight effects other than striatal generation of DA in the functional profile of L-DOPA. While not excluding a minor role for glial cells, L-DOPA is principally transformed into DA in neurons yet, interestingly, with a more important role for serotonergic than dopaminergic projections. Moreover, in addition to the striatum, L-DOPA evokes marked increases in extracellular DA in frontal cortex, nucleus accumbens, the subthalamic nucleus and additional extra-striatal regions. In considering its functional profile, it is also important to bear in mind the marked (probably indirect) influence of L-DOPA upon cholinergic, GABAergic and glutamatergic neurons in the basal ganglia and/or cortex, while anomalous serotonergic transmission is incriminated in the emergence of L-DOPA elicited dyskinesia and psychosis. Finally, L-DOPA may exert intrinsic receptor-mediated actions independently of DA neurotransmission and can be processed into bioactive metabolites. In conclusion, L-DOPA exerts a surprisingly complex pattern of neurochemical effects of much greater scope that mere striatal transformation into DA in spared dopaminergic neurons. Their further experimental and clinical clarification should help improve both L-DOPA-based and novel strategies for controlling the motor and other symptoms of Parkinson's disease.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- CNRS (Centre National de la Recherche Scientifique), Institut des Maladies Neurodégénératives, UMR CNRS 5293, F-33000 Bordeaux, France.
| | - Giuseppe Di Giovanni
- Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK; Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta
| | - Mark J Millan
- Institut de Recherche Servier, Pole for Therapeutic Innovation in Neuropsychiatry, 78290 Croissy/Seine,Paris, France
| |
Collapse
|
53
|
Volpicelli-Daley LA, Kirik D, Stoyka LE, Standaert DG, Harms AS. How can rAAV-α-synuclein and the fibril α-synuclein models advance our understanding of Parkinson's disease? J Neurochem 2016; 139 Suppl 1:131-155. [PMID: 27018978 DOI: 10.1111/jnc.13627] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/29/2016] [Accepted: 03/16/2016] [Indexed: 12/23/2022]
Abstract
Animal models of Parkinson's disease (PD) are important for understanding the mechanisms of the disease and can contribute to developing and validating novel therapeutics. Ideally, these models should replicate the cardinal features of PD, such as progressive neurodegeneration of catecholaminergic neurons and motor defects. Many current PD models emphasize pathological forms of α-synuclein, based on findings that autosomal dominant mutations in α-synuclein and duplications/triplications of the SNCA gene cause PD. In addition, Lewy bodies and Lewy neurites, primarily composed of α-synuclein, represent the predominant pathological characteristics of PD. These inclusions have defined features, such as insolubility in non-ionic detergent, hyperphosphorylation, proteinase K sensitivity, a filamentous appearance by electron microscopy, and β-sheet structure. Furthermore, it has become clear that Lewy bodies and Lewy neurites are found throughout the peripheral and central nervous system, and could account not only for motor symptoms, but also for non-motor symptoms of the disease. The goal of this review is to describe two new α-synuclein-based models: the recombinant adeno-associated viral vector-α-synuclein model and the α-synuclein fibril model. An advantage of both models is that they do not require extensive crossbreeding of rodents transgenic for α-synuclein with other rodents transgenic for genes of interest to study the impact of such genes on PD-related pathology and phenotypes. In addition, abnormal α-synuclein can be expressed in brain regions relevant for disease. Here, we discuss the features of each model, how each model has contributed thus far to our understanding of PD, and the advantages and potential caveats of each model. This review describes two α-synuclein-based rodent models of Parkinson's disease: the rAAV-α-synuclein model and the α-synuclein fibril model. The key features of these models are described, and the extent to which they recapitulate features of PD, such as α-synuclein inclusion formation, loss of dopaminergic synapses in the striatum, motor defects, inflammation, and dopamine neuron death. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Laura A Volpicelli-Daley
- From the Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama.
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lindsay E Stoyka
- From the Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - David G Standaert
- From the Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Ashley S Harms
- From the Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
54
|
Chu Y, Morfini GA, Kordower JH. Alterations in Activity-Dependent Neuroprotective Protein in Sporadic and Experimental Parkinson’s Disease. JOURNAL OF PARKINSONS DISEASE 2016; 6:77-97. [DOI: 10.3233/jpd-160812] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yaping Chu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Gerardo A. Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jeffrey H. Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
55
|
Arcuri L, Viaro R, Bido S, Longo F, Calcagno M, Fernagut PO, Zaveri NT, Calò G, Bezard E, Morari M. Genetic and pharmacological evidence that endogenous nociceptin/orphanin FQ contributes to dopamine cell loss in Parkinson's disease. Neurobiol Dis 2016; 89:55-64. [PMID: 26804029 DOI: 10.1016/j.nbd.2016.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/09/2016] [Accepted: 01/19/2016] [Indexed: 12/30/2022] Open
Abstract
To investigate whether the endogenous neuropeptide nociceptin/orphanin FQ (N/OFQ) contributes to the death of dopamine neurons in Parkinson's disease, we undertook a genetic and a pharmacological approach using NOP receptor knockout (NOP(-/-)) mice, and the selective and potent small molecule NOP receptor antagonist (-)-cis-1-methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol (SB-612111). Stereological unbiased methods were used to estimate the total number of dopamine neurons in the substantia nigra of i) NOP(-/-) mice acutely treated with the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP), ii) naïve mice subacutely treated with MPTP, alone or in combination with SB-612111, iii) rats injected with a recombinant adeno-associated viral (AAV) vector overexpressing human mutant p.A53T α-synuclein, treated with vehicle or SB-612111. NOP(-/-) mice showed a 50% greater amount of nigral dopamine neurons spared in response to acute MPTP compared to controls, which was associated with a milder motor impairment. SB-612111, given 4 days after MPTP treatment to mimic the clinical condition, prevented the loss of nigral dopamine neurons and striatal dopaminergic terminals caused by subacute MPTP. SB-612111, administered a week after the AAV injections in a clinically-driven protocol, also increased by 50% both the number of spared nigral dopamine neurons and striatal dopamine terminals, and prevented accompanying motor deficits induced by α-synuclein. We conclude that endogenous N/OFQ contributes to dopamine neuron loss in pathogenic and etiologic models of Parkinson's disease through NOP receptor-mediated mechanisms. NOP receptor antagonists might prove effective as disease-modifying agents in Parkinson's disease, through the rescue of degenerating nigral dopamine neurons and/or the protection of the healthy ones.
Collapse
Affiliation(s)
- Ludovico Arcuri
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Riccardo Viaro
- Department of Biomedical and Specialty Surgical Sciences, Section of Human Physiology, University of Ferrara, via Fossato di Mortara 19, 44121 Ferrara, Italy
| | - Simone Bido
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Francesco Longo
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Mariangela Calcagno
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Pierre-Olivier Fernagut
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Nurulain T Zaveri
- Astraea Therapeutics, 320 Logue Avenue, Mountain View, CA 94040, USA
| | - Girolamo Calò
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy.
| |
Collapse
|
56
|
Tamburrino A, Churchill MJ, Wan OW, Colino-Sanguino Y, Ippolito R, Bergstrand S, Wolf DA, Herz NJ, Sconce MD, Björklund A, Meshul CK, Decressac M. Cyclosporin promotes neurorestoration and cell replacement therapy in pre-clinical models of Parkinson's disease. Acta Neuropathol Commun 2015; 3:84. [PMID: 26666562 PMCID: PMC4678733 DOI: 10.1186/s40478-015-0263-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/03/2015] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The early clinical trials using fetal ventral mesencephalic (VM) allografts in Parkinson's disease (PD) patients have shown efficacy (albeit not in all cases) and have paved the way for further development of cell replacement therapy strategies in PD. The preclinical work that led to these clinical trials used allografts of fetal VM tissue placed into 6-OHDA lesioned rats, while the patients received similar allografts under cover of immunosuppression in an α-synuclein disease state. Thus developing models that more faithfully replicate the clinical scenario would be a useful tool for the translation of such cell-based therapies to the clinic. RESULTS Here, we show that while providing functional recovery, transplantation of fetal dopamine neurons into the AAV-α-synuclein rat model of PD resulted in smaller-sized grafts as compared to similar grafts placed into the 6-OHDA-lesioned striatum. Additionally, we found that cyclosporin treatment was able to promote the survival of the transplanted cells in this allografted state and surprisingly also provided therapeutic benefit in sham-operated animals. We demonstrated that delayed cyclosporin treatment afforded neurorestoration in three complementary models of PD including the Thy1-α-synuclein transgenic mouse, a novel AAV-α-synuclein mouse model, and the MPTP mouse model. We then explored the mechanisms for this benefit of cyclosporin and found it was mediated by both cell-autonomous mechanisms and non-cell autonomous mechanisms. CONCLUSION This study provides compelling evidence in favor for the use of immunosuppression in all grafted PD patients receiving cell replacement therapy, regardless of the immunological mismatch between donor and host cells, and also suggests that cyclosporine treatment itself may act as a disease-modifying therapy in all PD patients.
Collapse
Affiliation(s)
- Anna Tamburrino
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Madeline J Churchill
- Veterans Hospital/Research Services/Portland and Department of Behavioral Neuroscience Oregon Health &, Science University, Portland, OR, USA
| | - Oi W Wan
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Yolanda Colino-Sanguino
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Rossana Ippolito
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Sofie Bergstrand
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Daniel A Wolf
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Niculin J Herz
- Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Houston, USA
| | - Michelle D Sconce
- Veterans Hospital/Research Services/Portland and Department of Behavioral Neuroscience Oregon Health &, Science University, Portland, OR, USA
| | - Anders Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Charles K Meshul
- Veterans Hospital/Research Services/Portland and Department of Behavioral Neuroscience Oregon Health &, Science University, Portland, OR, USA
| | - Mickael Decressac
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
| |
Collapse
|
57
|
Caudal D, Alvarsson A, Björklund A, Svenningsson P. Depressive-like phenotype induced by AAV-mediated overexpression of human α-synuclein in midbrain dopaminergic neurons. Exp Neurol 2015; 273:243-52. [DOI: 10.1016/j.expneurol.2015.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 08/18/2015] [Accepted: 09/03/2015] [Indexed: 01/10/2023]
|
58
|
Oueslati A, Lovisa B, Perrin J, Wagnières G, van den Bergh H, Tardy Y, Lashuel HA. Photobiomodulation Suppresses Alpha-Synuclein-Induced Toxicity in an AAV-Based Rat Genetic Model of Parkinson's Disease. PLoS One 2015; 10:e0140880. [PMID: 26484876 PMCID: PMC4617694 DOI: 10.1371/journal.pone.0140880] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/01/2015] [Indexed: 12/13/2022] Open
Abstract
Converging lines of evidence indicate that near-infrared light treatment, also known as photobiomodulation (PBM), may exert beneficial effects and protect against cellular toxicity and degeneration in several animal models of human pathologies, including neurodegenerative disorders. In the present study, we report that chronic PMB treatment mitigates dopaminergic loss induced by unilateral overexpression of human α-synuclein (α-syn) in the substantia nigra of an AAV-based rat genetic model of Parkinson's disease (PD). In this model, daily exposure of both sides of the rat's head to 808-nm near-infrared light for 28 consecutive days alleviated α-syn-induced motor impairment, as assessed using the cylinder test. This treatment also significantly reduced dopaminergic neuronal loss in the injected substantia nigra and preserved dopaminergic fibers in the ipsilateral striatum. These beneficial effects were sustained for at least 6 weeks after discontinuing the treatment. Together, our data point to PBM as a possible therapeutic strategy for the treatment of PD and other related synucleinopathies.
Collapse
Affiliation(s)
- Abid Oueslati
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
- Centre de Recherche du Centre Hospitalier de Québec, Axe Neuroscience et Département de Médecine Moléculaire de l'Université Laval, Québec, G1V4G2, Canada
- * E-mail: (HAL); (AO)
| | - Blaise Lovisa
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
- Medos International Sàrl, a Johnson&Johnson company, Chemin Blanc 38, CH-2400, Le Locle, Switzerland
| | - John Perrin
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Georges Wagnières
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Hubert van den Bergh
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Yanik Tardy
- Medos International Sàrl, a Johnson&Johnson company, Chemin Blanc 38, CH-2400, Le Locle, Switzerland
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, P.O. Box 5825, Doha, Qatar
- * E-mail: (HAL); (AO)
| |
Collapse
|
59
|
Comparison between α-synuclein wild-type and A53T mutation in a progressive Parkinson's disease model. Biochem Biophys Res Commun 2015; 464:988-993. [PMID: 26192120 DOI: 10.1016/j.bbrc.2015.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Vector based over-expression of α-synuclein is a newly developed method to establish animal Parkinson's disease (PD) model. In this paper, we inject the rat brain with recombinant adeno-associated virus (rAAV) to express α-synuclein wild-type and A53T mutation, and compared the degeneration of dopaminergic neurons between them. METHOD AND RESULTS The rAAV vectors were injected into the substantia nigra pars compacta (SNpc) of rat brain. In different time point, immunohistochemistry was used to detect the expression of α-synuclein. The expression level was lower in the 3rd and 6th week and increased from the 9th week. α-synuclein spread around the neurons in SNpc in the 12th week. The loss of dopaminergic neurons was increasing along the expression of α-synuclein, and damage extent was more serious in the A53T group than the WT group. In the A53T group, there were more insoluble inclusions can be detected, and the phosphorylation of α-synuclein was also higher. CONLUSION The result of comparison between the two types of α-synuclein showed that A53T mutated α-synuclein was more effective to establish PD model, and the model based A53T mutated α-synuclein was a suitable model to early-onset PD.
Collapse
|
60
|
Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, Van den Haute C, Melki R, Baekelandt V. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature 2015; 522:340-4. [DOI: 10.1038/nature14547] [Citation(s) in RCA: 896] [Impact Index Per Article: 89.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/01/2015] [Indexed: 01/13/2023]
|
61
|
Viral vector delivery of neurotrophic factors for Parkinson's disease therapy. Expert Rev Mol Med 2015; 17:e8. [DOI: 10.1017/erm.2015.6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by the progressive loss of midbrain dopaminergic neurons, which causes motor impairments. Current treatments involve dopamine replacement to address the disease symptoms rather than its cause. Factors that promote the survival of dopaminergic neurons have been proposed as novel therapies for PD. Several dopaminergic neurotrophic factors (NTFs) have been examined for their ability to protect and/or restore degenerating dopaminergic neurons, both in animal models and in clinical trials. These include glial cell line-derived neurotrophic factor, neurturin, cerebral dopamine neurotrophic factor and growth/differentiation factor 5. Delivery of these NTFs via injection or infusion to the brain raises several practical problems. A new delivery approach for NTFs involves the use of recombinant viral vectors to enable long-term expression of these factors in brain cells. Vectors used include those based on adenoviruses, adeno-associated viruses and lentiviruses. Here we review progress to date on the potential of each of these four NTFs as novel therapeutic strategies for PD, as well as the challenges that have arisen, from pre-clinical analysis to clinical trials. We conclude by discussing recently-developed approaches to optimise the delivery of NTF-carrying viral vectors to the brain.
Collapse
|
62
|
Van der Perren A, Toelen J, Casteels C, Macchi F, Van Rompuy AS, Sarre S, Casadei N, Nuber S, Himmelreich U, Osorio Garcia MI, Michotte Y, D'Hooge R, Bormans G, Van Laere K, Gijsbers R, Van den Haute C, Debyser Z, Baekelandt V. Longitudinal follow-up and characterization of a robust rat model for Parkinson's disease based on overexpression of alpha-synuclein with adeno-associated viral vectors. Neurobiol Aging 2015; 36:1543-58. [DOI: 10.1016/j.neurobiolaging.2014.11.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/17/2014] [Accepted: 11/25/2014] [Indexed: 11/28/2022]
|
63
|
Cebrián C, Loike JD, Sulzer D. Neuroinflammation in Parkinson's disease animal models: a cell stress response or a step in neurodegeneration? Curr Top Behav Neurosci 2015; 22:237-270. [PMID: 25293443 DOI: 10.1007/7854_2014_356] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The motor symptoms of Parkinson's disease are due to the progressive degeneration of dopaminergic neurons in the substantia nigra. Multiple neuroinflammatory processes are exacerbated in Parkinson's disease, including glial-mediated reactions, increased expression of proinflammatory substances, and lymphocytic infiltration, particularly in the substantia nigra. Neuroinflammation is also implicated in the neurodegeneration and consequent behavioral symptoms of many Parkinson's disease animal models, although it is not clear whether these features emulate pathogenic steps in the genuine disorder or if some inflammatory features provide protective stress responses. Here, we compare and summarize findings on neuroinflammatory responses and effects on behavior in a wide range of toxin-based, inflammatory and genetic Parkinson's disease animal models.
Collapse
Affiliation(s)
- Carolina Cebrián
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | | | | |
Collapse
|
64
|
TNF-α regulates miRNA targeting mitochondrial complex-I and induces cell death in dopaminergic cells. Biochim Biophys Acta Mol Basis Dis 2014; 1852:451-61. [PMID: 25481834 DOI: 10.1016/j.bbadis.2014.11.019] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/22/2014] [Accepted: 11/26/2014] [Indexed: 01/06/2023]
Abstract
Parkinson's disease (PD) is a complex neurological disorder of the elderly population and majorly shows the selective loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) region of the brain. The mechanisms leading to increased cell death of DAergic neurons are not well understood. Tumor necrosis factor-alpha (TNF-α), a pro-inflammatory cytokine is elevated in blood, CSF and striatum region of the brain in PD patients. The increased level of TNF-α and its role in pathogenesis of PD are not well understood. In the current study, we investigated the role of TNF-α in the regulation of cell death and miRNA mediated mitochondrial functions using, DAergic cell line, SH-SY5Y (model of dopaminergic neuron degeneration akin to PD). The cells treated with low dose of TNF-α for prolonged period induce cell death which was rescued in the presence of zVAD.fmk, a caspase inhibitor and N-acetyl-cysteine (NAC), an antioxidant. TNF-α alters mitochondrial complex-I activity, decreases adenosine triphosphate (ATP) levels, increases reactive oxygen species levels and mitochondrial turnover through autophagy. TNF-α differentially regulates miRNA expression involved in pathogenesis of PD. Bioinformatics analysis revealed that the putative targets of altered miRNA included both pro/anti apoptotic genes and subunits of mitochondrial complex. The cells treated with TNF-α showed decreased level of nuclear encoded transcript of mitochondrial complexes, the target of miRNA. To our knowledge, the evidences in the current study demonstrated that TNF-α is a potential regulator of miRNAs which may regulate mitochondrial functions and neuronal cell death, having important implication in pathogenesis of PD.
Collapse
|
65
|
α-Synuclein-induced dopaminergic neurodegeneration in a rat model of Parkinson's disease occurs independent of ATP13A2 (PARK9). Neurobiol Dis 2014; 73:229-43. [PMID: 25461191 DOI: 10.1016/j.nbd.2014.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 10/02/2014] [Accepted: 10/12/2014] [Indexed: 11/21/2022] Open
Abstract
Mutations in the ATP13A2 (PARK9) gene cause early-onset, autosomal recessive Parkinson's disease (PD) and Kufor-Rakeb syndrome. ATP13A2 mRNA is spliced into three distinct isoforms encoding a P5-type ATPase involved in regulating heavy metal transport across vesicular membranes. Here, we demonstrate that three ATP13A2 mRNA isoforms are expressed in the normal human brain and are modestly increased in the cingulate cortex of PD cases. ATP13A2 can mediate protection toward a number of stressors in mammalian cells and can protect against α-synuclein-induced toxicity in cellular and invertebrate models of PD. Using a primary cortical neuronal model combined with lentiviral-mediated gene transfer, we demonstrate that human ATP13A2 isoforms 1 and 2 display selective neuroprotective effects toward toxicity induced by manganese and hydrogen peroxide exposure through an ATPase-independent mechanism. The familial PD mutations, F182L and G504R, abolish the neuroprotective effects of ATP13A2 consistent with a loss-of-function mechanism. We further demonstrate that the AAV-mediated overexpression of human ATP13A2 is not sufficient to attenuate dopaminergic neurodegeneration, neuropathology, and striatal dopamine and motoric deficits induced by human α-synuclein expression in a rat model of PD. Intriguingly, the delivery of an ATPase-deficient form of ATP13A2 (D513N) to the substantia nigra is sufficient to induce dopaminergic neuronal degeneration and motor deficits in rats, potentially suggesting a dominant-negative mechanism of action. Collectively, our data demonstrate a distinct lack of ATP13A2-mediated protection against α-synuclein-induced neurotoxicity in the rat nigrostriatal dopaminergic pathway, and limited neuroprotective capacity overall, and raise doubts about the potential of ATP13A2 as a therapeutic target for PD.
Collapse
|
66
|
Abnormal alpha-synuclein reduces nigral voltage-dependent anion channel 1 in sporadic and experimental Parkinson's disease. Neurobiol Dis 2014; 69:1-14. [PMID: 24825319 DOI: 10.1016/j.nbd.2014.05.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/09/2014] [Accepted: 05/02/2014] [Indexed: 01/24/2023] Open
Abstract
Both the misfolding of α-synuclein and mitochondrial dysfunction are considered two major contributors to Parkinson's disease (PD). However, the relationship between the two in normal and PD states remains unclear. Here, we report that voltage-dependent anion channel 1 (VDAC1), a major component of the outer mitochondrial membrane known to regulate mitochondrial functions, is down-regulated in response to α-synuclein accumulation and aggregation. Stereological analysis revealed that 58.33% of the neurons were VDAC1 immunoreactive in the remaining neuromelanin laden neurons in the PD group while 87.48% of the nigral neurons were VDAC1 immunoreactive in the age-matched control group. The relative levels of VDAC1 were significantly decreased in PD nigral neurons when compared to age-matched controls. In PD, this decrease was significantly greater in nigral neurons with α-synuclein inclusions. VDAC1 was observed in fibers with granular α-synuclein but not in fibers with aggregated α-synuclein. Viral vector-mediated overexpression of mutant human α-synuclein (A30P) in rats resulted in significantly decreased VDAC1 in nigral neurons and striatal fibers. These results indicate that mitochondrial function associated with VDAC1 is decreased in sporadic and experimental PD, and this decrease is associated with α-synuclein accumulation and aggregation.
Collapse
|
67
|
Multiple system atrophy: a prototypical synucleinopathy for disease-modifying therapeutic strategies. Neurobiol Dis 2014; 67:133-9. [PMID: 24727096 DOI: 10.1016/j.nbd.2014.03.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 03/24/2014] [Accepted: 03/31/2014] [Indexed: 02/08/2023] Open
Abstract
Despite active fundamental, translational and clinical research, no therapeutic intervention has yet shown convincing effects on disease progression in Parkinson's disease (PD) patients. Indeed, several disease-modification trials failed or proved to be inconclusive due to lack of consistency between clinical rating scales and putative surrogate markers of disease progression, or confounding symptomatic effects of the tested compound. Multiple system atrophy (MSA) is a rapidly progressing orphan disorder leading to severe motor disability within a few years. Together with PD and dementia with Lewy bodies (DLB), MSA belongs to the synucleinopathies, a group of neurodegenerative disorders characterized by the abnormal accumulation of alpha-synuclein. Crucial milestones have been reached for successfully conducting clinical intervention trials in a large number of patients with MSA. In this personal view, we will review evidence, and discuss why MSA could prove the most relevant clinical model for assessing treatments that target mechanisms operating in all synucleinopathies.
Collapse
|
68
|
McFarland NR, Dimant H, Kibuuka L, Ebrahimi-Fakhari D, Desjardins CA, Danzer KM, Danzer M, Fan Z, Schwarzschild MA, Hirst W, McLean PJ. Chronic treatment with novel small molecule Hsp90 inhibitors rescues striatal dopamine levels but not α-synuclein-induced neuronal cell loss. PLoS One 2014; 9:e86048. [PMID: 24465863 PMCID: PMC3896461 DOI: 10.1371/journal.pone.0086048] [Citation(s) in RCA: 31] [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: 09/08/2013] [Accepted: 12/04/2013] [Indexed: 11/18/2022] Open
Abstract
Hsp90 inhibitors such as geldanamycin potently induce Hsp70 and reduce cytotoxicity due to α-synuclein expression, although their use has been limited due to toxicity, brain permeability, and drug design. We recently described the effects of a novel class of potent, small molecule Hsp90 inhibitors in cells overexpressing α-synuclein. Screening yielded several candidate compounds that significantly reduced α-synuclein oligomer formation and cytotoxicity associated with Hsp70 induction. In this study we examined whether chronic treatment with candidate Hsp90 inhibitors could protect against α-synuclein toxicity in a rat model of parkinsonism. Rats were injected unilaterally in the substantia nigra with AAV8 expressing human α-synuclein and then treated with drug for approximately 8 weeks by oral gavage. Chronic treatment with SNX-0723 or the more potent, SNX-9114 failed to reduce dopaminergic toxicity in the substantia nigra compared to vehicle. However, SNX-9114 significantly increased striatal dopamine content suggesting a positive neuromodulatory effect on striatal terminals. Treatment was generally well tolerated, but higher dose SNX-0723 (6–10 mg/kg) resulted in systemic toxicity, weight loss, and early death. Although still limited by potential toxicity, Hsp90 inhibitors tested herein demonstrate oral efficacy and possible beneficial effects on dopamine production in a vertebrate model of parkinsonism that warrant further study.
Collapse
Affiliation(s)
- Nikolaus R. McFarland
- Center for Translational Research in Neurodegenerative Disease, Department of Neurology, University of Florida, Gainesville, Florida, United States of America
- * E-mail: (NRM); (PJM)
| | - Hemi Dimant
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Laura Kibuuka
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Darius Ebrahimi-Fakhari
- Division of Neurology and Inherited Metabolic Diseases, Children’s Hospital, Heidelberg University Hospital, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Cody A. Desjardins
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Karin M. Danzer
- Deparment of Neurology, Universitatsklinikum Ulm, Ulm, Germany
| | - Michael Danzer
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Zhanyun Fan
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Michael A. Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Warren Hirst
- Pfizer Neuroscience Research Unit, Cambridge, Massachusetts, United States of America
| | - Pamela J. McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
- * E-mail: (NRM); (PJM)
| |
Collapse
|
69
|
Gombash SE, Manfredsson FP, Kemp CJ, Kuhn NC, Fleming SM, Egan AE, Grant LM, Ciucci MR, MacKeigan JP, Sortwell CE. Morphological and behavioral impact of AAV2/5-mediated overexpression of human wildtype alpha-synuclein in the rat nigrostriatal system. PLoS One 2013; 8:e81426. [PMID: 24312298 PMCID: PMC3842242 DOI: 10.1371/journal.pone.0081426] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/12/2013] [Indexed: 01/06/2023] Open
Abstract
The discovery of the involvement of alpha-synuclein (α-syn) in Parkinson’s disease (PD) pathogenesis has resulted in the development and use of viral vector-mediated α-syn overexpression rodent models. The goal of these series of experiments was to characterize the neurodegeneration and functional deficits resulting from injection of recombinant adeno-associated virus (rAAV) serotype 2/5-expressing human wildtype α-syn in the rat substantia nigra (SN). Rats were unilaterally injected into two sites in the SN with either rAAV2/5-expressing green fluorescent protein (GFP, 1.2 x 1013) or varying titers (2.2 x 1012, 1.0 x 1013, 5.9 x 1013, or 1.0 x 1014) of rAAV2/5-α-syn. Cohorts of rats were euthanized 4, 8, or 12 weeks following vector injection. The severity of tyrosine hydroxylase immunoreactive (THir) neuron death in the SN pars compacta (SNpc) was dependent on vector titer. An identical magnitude of nigrostriatal degeneration (60-70% SNpc THir neuron degeneration and 40-50% loss of striatal TH expression) was observed four weeks following 1.0 x 1014 titer rAAV2/5-α-syn injection and 8 weeks following 1.0 x 1013 titer rAAV2/5-α-syn injection. THir neuron degeneration was relatively uniform throughout the rostral-caudal axis of the SNpc. Despite equivalent nigrostriatal degeneration between the 1.0 x 1013 and 1.0 x 1014 rAAV2/5-α-syn groups, functional impairment in the cylinder test and the adjusting steps task was only observed in rats with the longer 8 week duration of α-syn expression. Motor impairment in the cylinder task was highly correlated to striatal TH loss. Further, 8 weeks following 5.9 x 1013 rAAV2/5-α-syn injection deficits in ultrasonic vocalizations were observed. In conclusion, our rAAV2/5-α-syn overexpression model demonstrates robust nigrostriatal α-syn overexpression, induces significant nigrostriatal degeneration that is both vector and duration dependent and under specific parameters can result in motor impairment that directly relates to the level of striatal TH denervation.
Collapse
Affiliation(s)
- Sara E. Gombash
- Graduate Program in Neuroscience, University of Cincinnati, Cincinnati, Ohio, United States of America
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Fredric P. Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Christopher J. Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Nathan C. Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Sheila M. Fleming
- Departments of Psychology and Neurology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Ann E. Egan
- Graduate Program in Neuroscience, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Laura M. Grant
- Departments of Surgery and Communications Sciences and Disorders, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Michelle R. Ciucci
- Departments of Surgery and Communications Sciences and Disorders, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Jeffrey P. MacKeigan
- Laboratory of Systems Biology, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Caryl E. Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, United States of America
- * E-mail:
| |
Collapse
|
70
|
Oliveras-Salvá M, Van der Perren A, Casadei N, Stroobants S, Nuber S, D'Hooge R, Van den Haute C, Baekelandt V. rAAV2/7 vector-mediated overexpression of alpha-synuclein in mouse substantia nigra induces protein aggregation and progressive dose-dependent neurodegeneration. Mol Neurodegener 2013; 8:44. [PMID: 24267638 PMCID: PMC4222878 DOI: 10.1186/1750-1326-8-44] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 11/18/2013] [Indexed: 12/25/2022] Open
Abstract
Background Alpha-synuclein is a key protein implicated in the pathogenesis of Parkinson's disease (PD). It is the main component of the Lewy bodies, a cardinal neuropathological feature in the disease. In addition, whole locus multiplications and point mutations in the gene coding for alpha-synuclein lead to autosomal dominant monogenic PD. Over the past decade, research on PD has impelled the development of new animal models based on alpha-synuclein. In this context, transgenic mouse lines have failed to reproduce several hallmarks of PD, especially the strong and progressive dopaminergic neurodegeneration over time that occurs in the patients. In contrast, viral vector-based models in rats and non-human primates display prominent, although highly variable, nigral dopaminergic neuron loss. However, the few studies available on viral vector-mediated overexpression of alpha-synuclein in mice report a weak neurodegenerative process and no clear Lewy body-like pathology. To address this issue, we performed a comprehensive comparative study of alpha-synuclein overexpression by means of recombinant adeno-associated viral vectors serotype 2/7 (rAAV2/7) at different doses in adult mouse substantia nigra. Results We noted a significant and dose-dependent alpha-synucleinopathy over time upon nigral viral vector-mediated alpha-synuclein overexpression. We obtained a strong, progressive and dose-dependent loss of dopaminergic neurons in the substantia nigra, reaching a maximum of 82% after 8 weeks. This effect correlated with a reduction in tyrosine hydroxylase immunoreactivity in the striatum. Moreover, behavioural analysis revealed significant motor impairments from 12 weeks after injection on. In addition, we detected the presence of alpha-synuclein-positive aggregates in the remaining surviving neurons. When comparing wild-type to mutant A53T alpha-synuclein at the same vector dose, both induced a similar degree of cell death. These data were supported by a biochemical analysis that showed a net increase in soluble and insoluble alpha-synuclein expression over time to the same extent for both alpha-synuclein variants. Conclusions In conclusion, our in vivo data provide evidence that strong and significant alpha-synuclein-induced neuropathology and progressive dopaminergic neurodegeneration can be achieved in mouse brain by means of rAAV2/7.
Collapse
Affiliation(s)
- Marusela Oliveras-Salvá
- Department of Neurosciences and Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, Laboratory for Neurobiology and Gene Therapy, Kapucijnenvoer 33, box 7001, 3000 Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Xilouri M, Brekk OR, Landeck N, Pitychoutis PM, Papasilekas T, Papadopoulou-Daifoti Z, Kirik D, Stefanis L. Boosting chaperone-mediated autophagy in vivo mitigates α-synuclein-induced neurodegeneration. Brain 2013; 136:2130-46. [PMID: 23757764 DOI: 10.1093/brain/awt131] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
α-Synuclein levels are critical to Parkinson's disease pathogenesis. Wild-type α-synuclein is degraded partly by chaperone-mediated autophagy, and aberrant α-synuclein may act as an inhibitor of the pathway. To address whether the induction of chaperone-mediated autophagy may represent a potential therapy against α-synuclein-induced neurotoxicity, we overexpressed lysosomal-associated membrane protein 2a, the rate-limiting step of chaperone-mediated autophagy, in human neuroblastoma SH-SY5Y cells, rat primary cortical neurons in vitro, and nigral dopaminergic neurons in vivo. Overexpression of the lysosomal-associated membrane protein 2a in cellular systems led to upregulation of chaperone-mediated autophagy, decreased α-synuclein turnover, and selective protection against adenoviral-mediated wild-type α-synuclein neurotoxicity. Protection was observed even when the steady-state levels of α-synuclein were unchanged, suggesting that it occurred through the attenuation of α-synuclein-mediated dysfunction of chaperone-mediated autophagy. Overexpression of the lysosomal receptor through the nigral injection of recombinant adeno-associated virus vectors effectively ameliorated α-synuclein-induced dopaminergic neurodegeneration by increasing the survival of neurons located in the substantia nigra as well as the axon terminals located in the striatum, which was associated with a reduction in total α-synuclein levels and related aberrant species. We conclude that induction of chaperone-mediated autophagy may provide a novel therapeutic strategy in Parkinson's disease and related synucleinopathies through two different mechanisms: amelioration of dysfunction of chaperone-mediated autophagy and lowering of α-synuclein levels.
Collapse
Affiliation(s)
- Maria Xilouri
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Zhou JX, Zhang HB, Huang Y, He Y, Zheng Y, Anderson JP, Gai WP, Liang ZG, Wang Y, Ren XM, Wang Q, Gong XL, Yang J, Wang X, Halliday G, Wang XM. Tenuigenin attenuates α-synuclein-induced cytotoxicity by down-regulating polo-like kinase 3. CNS Neurosci Ther 2013; 19:688-94. [PMID: 23710708 DOI: 10.1111/cns.12124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 04/18/2013] [Accepted: 04/18/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIMS Tenuigenin (Ten) is a Chinese herbal extract with antioxidative and antiinflammatory effects on toxin-induced cell models of Parkinson's disease (PD); however, its effects on α-synuclein toxicity-based PD models remain unknown. α-synuclein hyperphosphorylation is a key event in PD pathogenesis and potential target of therapeutic interventions. We tested whether Ten alleviates α-synuclein-induced cytotoxicity via reducing kinases that phosphorylate α-synuclein. METHODS SH-SY5Y cells transiently transfected with wild-type or A53T mutant α-synuclein were used to evaluate the effect of Ten on the levels of α-synuclein phosphorylation-related kinases. Cells treated with 10 μM Ten for 24 h were measured for viability (proliferation and apoptosis assays) and cellular proteins harvested and fractioned. The levels of total and phosphorylated α-synuclein and five associated kinases (polo-like kinase [PLK] 1-3, casein kinase [CK] 1-2) were evaluated by Western blotting. RESULTS Overexpression of either wild-type or A53T mutant α-synuclein decreased cell viability and increased α-synuclein phosphorylation. Ten treatment-protected cells from this α-synuclein-induced toxicity and dramatically reduced α-synuclein phosphorylation and PLK3 (but not other kinase) levels. CONCLUSION In α-synuclein cell model of PD, Ten is effective in attenuating α-synuclein-induced toxicity and α-synuclein phosphorylation probably via targeting PLK3, suggesting it could be an efficient therapeutic drug to treat α-synuclein-related neurodegeneration.
Collapse
Affiliation(s)
- Jin-Xia Zhou
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Fauss D, Motter R, Dofiles L, Rodrigues MAV, You M, Diep L, Yang Y, Seto P, Tanaka K, Baker J, Bergeron M. Development of an enzyme-linked immunosorbent assay (ELISA) to measure the level of tyrosine hydroxylase protein in brain tissue from Parkinson's disease models. J Neurosci Methods 2013; 215:245-57. [DOI: 10.1016/j.jneumeth.2013.03.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/15/2013] [Accepted: 03/18/2013] [Indexed: 01/06/2023]
|
74
|
Sanchez-Guajardo V, Barnum C, Tansey M, Romero-Ramos M. Neuroimmunological processes in Parkinson's disease and their relation to α-synuclein: microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro 2013; 5:113-39. [PMID: 23506036 PMCID: PMC3639751 DOI: 10.1042/an20120066] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 03/15/2013] [Accepted: 03/19/2013] [Indexed: 12/15/2022] Open
Abstract
The role of neuroinflammation and the adaptive immune system in PD (Parkinson's disease) has been the subject of intense investigation in recent years, both in animal models of parkinsonism and in post-mortem PD brains. However, how these processes relate to and modulate α-syn (α-synuclein) pathology and microglia activation is still poorly understood. Specifically, how the peripheral immune system interacts, regulates and/or is induced by neuroinflammatory processes taking place during PD is still undetermined. We present herein a comprehensive review of the features and impact that neuroinflamation has on neurodegeneration in different animal models of nigral cell death, how this neuroinflammation relates to microglia activation and the way microglia respond to α-syn in vivo. We also discuss a possible role for the peripheral immune system in animal models of parkinsonism, how these findings relate to the state of microglia activation observed in these animal models and how these findings compare with what has been observed in humans with PD. Together, the available data points to the need for development of dual therapeutic strategies that modulate microglia activation to change not only the way microglia interact with the peripheral immune system, but also to modulate the manner in which microglia respond to encounters with α-syn. Lastly, we discuss the immune-modulatory strategies currently under investigation in animal models of parkinsonism and the degree to which one might expect their outcomes to translate faithfully to a clinical setting.
Collapse
Key Words
- lymphocytes
- m1/m2 phenotype
- microglia
- neuroinflammation
- parkinson’s disease
- α-synuclein
- 6-ohda, 6-hydroxydopamine
- ad, alzheimer’s disease
- apc, antigen-presenting cell
- α-syn, α-synuclein
- bbb, brain–blood barrier
- bcg, bacille calmette–guérin
- bm, bone marrow
- cfa, complete freund’s adjuvant
- cm, conditioned media
- cns, central nervous system
- cox, cyclooxygenase
- cr, complement receptor
- csf, cerebrospinal fluid
- da, dopamine
- eae, experimental autoimmune encephalomyelitis
- ga, galatiramer acetate
- gdnf, glial-derived neurotrophic factor
- gfp, green fluorescent protein
- hla-dr, human leucocyte antigen type dr
- ifnγ, interferon γ
- igg, immunoglobulin g
- il, interleukin
- inos, inducible nitric oxide synthase
- lamp, lysosome-associated membrane protein
- lb, lewy body
- lps, lipopolysaccharide
- mhc, major histocompatibility complex
- mptp, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- nfκb, nuclear factor κb
- nk, natural killer
- no, nitric oxide
- pd, parkinson’s disease
- pet, positron-emission tomography
- prp, prion protein
- raav, recombinant adeno-associated virus
- rns, reactive nitrogen species
- ros, reactive oxygen species
- sn, substantia nigra
- snp, single nucleotide polymorphism
- tcr, t-cell receptor
- tgfβ, tumour growth factor β
- th, tyrosine hydroxylase
- th1, t helper 1
- tlr, toll-like receptor
- tnf, tumour necrosis factor
- treg, regulatory t-cell
- vip, vasoactive intestinal peptide
- wt, wild-type
Collapse
Affiliation(s)
- Vanesa Sanchez-Guajardo
- *CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Allé 3,
Aarhus University, DK-8000 Aarhus C, Denmark
| | - Christopher J. Barnum
- †Department of Physiology, Emory University, School of Medicine, Atlanta, GA
30233, U.S.A
| | - Malú G. Tansey
- †Department of Physiology, Emory University, School of Medicine, Atlanta, GA
30233, U.S.A
| | - Marina Romero-Ramos
- *CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Allé 3,
Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
75
|
Mulcahy P, O’Doherty A, Paucard A, O’Brien T, Kirik D, Dowd E. The behavioural and neuropathological impact of intranigral AAV-α-synuclein is exacerbated by systemic infusion of the Parkinson's disease-associated pesticide, rotenone, in rats. Behav Brain Res 2013; 243:6-15. [DOI: 10.1016/j.bbr.2012.12.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/13/2012] [Accepted: 12/27/2012] [Indexed: 12/22/2022]
|
76
|
Li X, Lee J, Parsons D, Janaurajs K, Standaert DG. Evaluation of TorsinA as a target for Parkinson disease therapy in mouse models. PLoS One 2012; 7:e50063. [PMID: 23185535 PMCID: PMC3503809 DOI: 10.1371/journal.pone.0050063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 10/18/2012] [Indexed: 11/19/2022] Open
Abstract
Parkinson disease (PD) is a common and disabling disorder. No current therapy can slow or reverse disease progression. An important aspect of research in this field is target validation, a systematic approach to evaluating the likelihood that modification of a certain molecule, mechanism or biological pathway may be useful for the development of pharmacological or molecular treatments for the disease. TorsinA, a member of the AAA+ family of chaperone proteins, has been proposed as a potential target of neuroprotective therapy. TorsinA is found in Lewy bodies in human PD, and can suppress toxicity in cellular and invertebrate models of PD. Here, we evaluated the neuroprotective properties of torsinA in mouse models of PD based on intoxication with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) as well as recombinant adeno associated virus (rAAV) induced overexpression of alpha-synuclein (α-syn). Using either transgenic mice with overexpression of human torsinA (hWT mice) or mice in which torsinA expression was induced using an rAAV vector, we found no evidence for protection against acute MPTP intoxication. Similarly, genetic deletion of the endogenous mouse gene for torsinA (Dyt1) using an rAAV delivered Cre recombinase did not enhance the vulnerability of dopaminergic neurons to MPTP. Overexpression of α-syn using rAAV in the mouse substantia nigra lead to a loss of TH positive neurons six months after administration, and no difference in the degree of loss was observed between transgenic animals expressing forms of torsinA and wild type controls. Collectively, we did not observe evidence for a protective effect of torsinA in the mouse models we examined. Each of these models has limitations, and there is no single model with established predictive value with respect to the human disease. Nevertheless, these data do seem to support the view that torsinA is unlikely to be successfully translated as a target of therapy for human PD.
Collapse
Affiliation(s)
- Xinru Li
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jenny Lee
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dee Parsons
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Karen Janaurajs
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David G. Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
77
|
Lindgren HS, Lelos MJ, Dunnett SB. Do α-synuclein vector injections provide a better model of Parkinson's disease than the classic 6-hydroxydopamine model? Exp Neurol 2012; 237:36-42. [PMID: 22727767 DOI: 10.1016/j.expneurol.2012.05.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 05/25/2012] [Accepted: 05/31/2012] [Indexed: 12/21/2022]
Abstract
Improvements in modelling Parkinson's disease in rodents contribute to the advancement of scientific knowledge and open innumerable pathways for the development of new therapeutic interventions. In a recent article in this journal, Decressac and co-workers present an interesting comparison between two classic 6-hydroxydopamine (6-OHDA) models and the more recently established rodent model of Parkinson's disease induced by over-expression of α-synuclein using adeno-associated viral vectors. As expected, injections of 6-OHDA result in extensive loss of dopamine associated with pronounced motor deficits. Interestingly, over-expression of α-synuclein in the substantia nigra pars compacta also results in a considerable loss of dopamine as well as motor impairments. Both the level of dopamine loss and the motor deficits seen after α-synuclein over-expression were similar in extent to that seen after intrastriatal injections of 6-OHDA, but the temporal profile of degeneration and the development of motor deficits were progressive, more closely mimicking the clinical condition. This commentary offers further insights into the differences between these two rodent models, and asks how well they each replicate idiopathic PD. In addition, the translational relevance, reliability, and predictive value of this more recently developed AAV α-synuclein model are considered.
Collapse
Affiliation(s)
- Hanna S Lindgren
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, Wales, UK.
| | | | | |
Collapse
|
78
|
Drouin-Ouellet J, Cicchetti F. Inflammation and neurodegeneration: the story 'retolled'. Trends Pharmacol Sci 2012; 33:542-51. [PMID: 22944460 DOI: 10.1016/j.tips.2012.07.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/30/2012] [Accepted: 07/11/2012] [Indexed: 01/18/2023]
Abstract
Toll-like receptors (TLRs) play a crucial role in innate immunity by recognizing conserved motifs predominantly found in microorganisms. Increasing evidence supports a role for TLRs in sterile inflammation as observed in neurodegenerative disorders. This includes work suggesting a contribution for these receptors to the pathophysiology of Alzheimer's disease (AD), Parkinson's disease (PD), and related disorders. In this review, the potential role of TLRs in the context of protein aggregation, neuronal degeneration, and genetic risk factors is addressed. In particular, we discuss the evidence derived from experimental models of both AD and PD which suggests that activation of TLRs can have beneficial and detrimental effects on pathological features such as protein aggregation and neuronal death. A deeper understanding of these dichotomous observations could be used for therapeutic benefit.
Collapse
|
79
|
Hirsch EC, Jenner P, Przedborski S. Pathogenesis of Parkinson's disease. Mov Disord 2012; 28:24-30. [PMID: 22927094 DOI: 10.1002/mds.25032] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/03/2012] [Accepted: 04/08/2012] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease is a common adult-onset neurodegenerative disorder whose pathogenesis remains essentially unknown. Currently, it is believed that the neurodegenerative process in Parkinson's disease is a combination of both cell-autonomous and non-cell-autonomous mechanisms. Proposed cell-autonomous mechanisms include alterations in mitochondrial bioenergetics, dysregulation of calcium homeostasis, and impaired turnover of mitochondria. As for the proposed non-cell-autonomous mechanisms, they involve prion-like behavior of misfolded proteins and neuroinflammation. This suggests that cell death in Parkinson's disease is caused by a multifactorial cascade of pathogenic events and argues that effective neuroprotective therapy for Parkinson's disease may have to rely on multiple drug interventions.
Collapse
Affiliation(s)
- Etienne C Hirsch
- Université Pierre et Marie Curie-Paris 06, Centre de Recherche de l'Institut du Cerveau et de la Moëlle Épinière, Hôpital de la Salpêtrière, Paris, France
| | | | | |
Collapse
|
80
|
Chu Y, Morfini GA, Langhamer LB, He Y, Brady ST, Kordower JH. Alterations in axonal transport motor proteins in sporadic and experimental Parkinson's disease. ACTA ACUST UNITED AC 2012; 135:2058-73. [PMID: 22719003 DOI: 10.1093/brain/aws133] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The progressive loss of the nigrostriatal pathway is a distinguishing feature of Parkinson's disease. As terminal field loss seems to precede cell body loss, we tested whether alterations of axonal transport motor proteins would be early features in Parkinson's disease. There was a decline in axonal transport motor proteins in sporadic Parkinson's disease that preceded other well-known nigral cell-related pathology such as phenotypic downregulation of dopamine. Reductions in conventional kinesin levels precede the alterations in dopaminergic phenotypic markers (tyrosine hydroxylase) in the early stages of Parkinson's disease. This reduction was significantly greater in nigral neurons containing α-synuclein inclusions. Unlike conventional kinesin, reductions in the levels of the cytoplasmic dynein light chain Tctex type 3 subunit were only observed at late Parkinson's disease stages. Reductions in levels of conventional kinesin and cytoplasmic dynein subunits were recapitulated in a rat genetic Parkinson's disease model based on over-expression of human mutant α-synuclein (A30P). Together, our data suggest that α-synuclein aggregation is a key feature associated with reductions of axonal transport motor proteins in Parkinson's disease and support the hypothesis that dopaminergic neurodegeneration following a 'dying-back' pattern involving axonal transport disruption.
Collapse
Affiliation(s)
- Yaping Chu
- Department of Neurological Sciences, Rush University Medical Centre, 1735 West Harrison Street, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
81
|
Brooks SP, Dunnett SB. Cognitive deficits in animal models of basal ganglia disorders. Brain Res Bull 2012; 92:29-40. [PMID: 22588013 DOI: 10.1016/j.brainresbull.2012.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 03/01/2012] [Accepted: 04/20/2012] [Indexed: 10/28/2022]
Abstract
The two most common neurological disorders of the basal ganglia are Parkinson's disease (PD) and Huntington's disease (HD). The most overt symptoms of these diseases are motoric, reflecting the loss of the striatal medium spiny neurons in HD and ascending substantia nigra dopaminergic cells in PD. However, both disease processes induce insidious psychiatric and cognitive syndromes that can manifest well in advance of the onset of motor deficits. These early deficits provide an opportunity for prophylactic therapeutic intervention in order to retard disease progression from the earliest possible point. In order to exploit this opportunity, animal models of HD and PD are being probed for the specific cognitive deficits represented in the disease states. At the neuronal level, these deficits are typically, but not exclusively, mediated by disruption of parallel corticostriatal loops that integrate motor information with sensory and higher order, "executive" cognitive functions. Dysfunction in these systems can be probed with sensitive behavioural tests that selectively probe these cognitive functions in mouse models with focal lesions of striatal or cortical regions, or of specific neurotransmitter systems. Typically these tests were designed and validated in rats. With the advent of genetically modified mouse models of disease, validated tests provide an opportunity to screen mouse models of disease for early onset cognitive deficits. This review seeks to draw together the literature on cognitive deficits in HD and PD, to determine the extent to which these deficits are represented in the current animal models of disease, and to evaluate the viability of selecting cognitive deficits as potential therapeutic targets. This article is part of a Special Issue entitled 'Animal Models'.
Collapse
Affiliation(s)
- Simon P Brooks
- Brain Repair Group, School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, Wales, UK.
| | | |
Collapse
|
82
|
Hallett PJ, McLean JR, Kartunen A, Langston JW, Isacson O. α-Synuclein overexpressing transgenic mice show internal organ pathology and autonomic deficits. Neurobiol Dis 2012; 47:258-67. [PMID: 22549133 DOI: 10.1016/j.nbd.2012.04.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/06/2012] [Accepted: 04/11/2012] [Indexed: 12/22/2022] Open
Abstract
While studying transgenic mice that overexpress human wildtype alpha-synuclein (Thy1-ASO, ASO) for typical brain alpha-synucleinopathy and central nervous system neuropathology, we observed progressive functional changes in the gastrointestinal and other peripheral organs. A more systematic study revealed that the gastrointestinal tract in ASO mice showed severe distension and blockage of the large intestine by 9-12 months of age. Functional assessments demonstrated a reduction in fecal water content and fecal pellet output, and increased whole gut transit time, in ASO mice compared to wildtype littermates, indicative of constipation, a symptom commonly reported by Parkinson's disease (PD) patients. Food intake was increased and body weight was decreased in 12 month old ASO mice, suggestive of metabolic abnormalities. Post-mortem histological analyses showed that human alpha-synuclein protein was robustly expressed in axonal fibers and in occasional cell bodies of the enteric nervous system, and in the heart of ASO mice. Accumulation of proteinase-K insoluble alpha-synuclein, reminiscent of neurodegenerative processes in PD was also observed. The functional and pathological changes we document here in ASO mice could relate to the autonomic deficits also seen in idiopathic and alpha-synuclein-mediated genetic forms of PD. These experimental data provide a foundation for therapeutic modeling of autonomic changes in PD and related alpha-synucleinopathies.
Collapse
Affiliation(s)
- Penelope J Hallett
- Center for Neuroregeneration Research, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA.
| | | | | | | | | |
Collapse
|
83
|
Lastres-Becker I, Ulusoy A, Innamorato NG, Sahin G, Rábano A, Kirik D, Cuadrado A. α-Synuclein expression and Nrf2 deficiency cooperate to aggravate protein aggregation, neuronal death and inflammation in early-stage Parkinson's disease. Hum Mol Genet 2012; 21:3173-92. [DOI: 10.1093/hmg/dds143] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
84
|
Barkholt P, Sanchez-Guajardo V, Kirik D, Romero-Ramos M. Long-term polarization of microglia upon α-synuclein overexpression in nonhuman primates. Neuroscience 2012; 208:85-96. [DOI: 10.1016/j.neuroscience.2012.02.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 01/26/2012] [Accepted: 02/02/2012] [Indexed: 12/11/2022]
|
85
|
Comparison of the behavioural and histological characteristics of the 6-OHDA and α-synuclein rat models of Parkinson's disease. Exp Neurol 2012; 235:306-15. [PMID: 22394547 DOI: 10.1016/j.expneurol.2012.02.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 02/12/2012] [Accepted: 02/20/2012] [Indexed: 01/19/2023]
Abstract
Development of relevant models of Parkinson's disease (PD) is essential for a better understanding of the pathological processes underlying the human disease and for the evaluation of promising targets for therapeutic intervention. To date, most pre-clinical studies have been performed in the well-established rodent and non-human primate models using injection of 6-hydroxydopamine (6-OHDA) or 1-methyl-4-phenyl-1,2,3,6-tetrahydroxypyridine (MPTP). Overexpression of the disease-causing protein α-synuclein (α-syn), using adeno-associated viral (AAV) vectors, has provided a novel model that recapitulates many features of the human disease. In the present study we compared the AAV-α-syn rat model with models where the nigro-striatal pathway is lesioned by injection of 6-OHDA in the striatum (partial lesion) or the medial forebrain bundle (full lesion). Examination of the behavioural changes over time revealed a different progression and magnitude of the motor impairment. Interestingly, dopamine (DA) neuron loss is prominent in both the toxin and the AAV-α-syn models. However, α-syn overexpressing animals were seen to exhibit less cell and terminal loss for an equivalent level of motor abnormalities. Prominent and persistent axonal pathology is only observed in the α-syn rat model. We suggest that, while neuronal and terminal loss mainly accounts for the behavioural impairment in the toxin-based model, similar motor deficits result from the combination of cell death and dysfunction of the remaining nigro-striatal neurons in the AAV-α-syn model. While the two models have been developed to mimic DA neuron deficiency, they differ in their temporal and neuropathological characteristics, and replicate different aspects of the pathophysiology of the human disease. This study suggests that the AAV-α-syn model replicates the human pathology more closely than either of the other two 6-OHDA lesion models.
Collapse
|
86
|
Lelos MJ, Dowd E, Dunnett SB. Nigral grafts in animal models of Parkinson's disease. Is recovery beyond motor function possible? PROGRESS IN BRAIN RESEARCH 2012. [PMID: 23195417 DOI: 10.1016/b978-0-444-59575-1.00006-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) has long been considered predominantly to be a "movement disorder," and it is only relatively recently that nonmotor symptoms of PD have been recognized to be a major concern to patients. Consequently, there has been surprisingly little investigation into the feasibility of utilizing cell replacement therapies to ameliorate any of the nonmotor dysfunctions of PD. In this chapter, we identify nonmotor impairments associated predominately with dopaminergic dysmodulation, evaluate the few emerging studies that have identified a role for dopamine and nigral transplantation in nonmotor performance, and consider a number of outstanding questions and considerations dominating the field of nigral transplantation today. Preliminary results obtained from rodent models of PD, despite being limited in number, give clear indications of graft effects on striatal processing beyond the simple activation of motor output and promise a major, exciting, and fruitful new avenue of research for the next decade. We can now consider the prospect of rewriting the opportunities for treating patients, with new stem cell sources to be complemented by new targets for therapeutic benefit.
Collapse
Affiliation(s)
- Mariah J Lelos
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, Wales, UK.
| | | | | |
Collapse
|
87
|
Monti B, Mercatelli D, Contestabile A. Valproic acid neuroprotection in 6-OHDA lesioned rat, a model for parkinson's disease. ACTA ACUST UNITED AC 2012. [DOI: 10.7243/2050-0874-1-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
88
|
Decressac M, Mattsson B, Lundblad M, Weikop P, Björklund A. Progressive neurodegenerative and behavioural changes induced by AAV-mediated overexpression of α-synuclein in midbrain dopamine neurons. Neurobiol Dis 2011; 45:939-53. [PMID: 22182688 DOI: 10.1016/j.nbd.2011.12.013] [Citation(s) in RCA: 205] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/11/2011] [Accepted: 12/04/2011] [Indexed: 12/22/2022] Open
Abstract
Parkinson's disease (PD) is characterised by the progressive loss of nigral dopamine neurons and the presence of synucleinopathy. Overexpression of α-synuclein in vivo using viral vectors has opened interesting possibilities to model PD-like pathology in rodents. However, the attempts made so far have failed to show a consistent behavioural phenotype and pronounced dopamine neurodegeneration. Using a more efficient adeno-associated viral (AAV) vector construct, which includes a WPRE enhancer element and uses the neuron-specific synapsin-1 promoter to drive the expression of human wild-type α-synuclein, we have now been able to achieve increased levels of α-synuclein in the transduced midbrain dopamine neurons sufficient to induce profound deficits in motor function, accompanied by reduced expression of proteins involved in dopamine neurotransmission and a time-dependent loss of nigral dopamine neurons, that develop progressively over 2-4 months after vector injection. As in human PD, nigral cell loss was preceded by degenerative changes in striatal axons and terminals, and the appearance of α-synuclein positive inclusions in dystrophic axons and dendrites, supporting the idea that α-synuclein-induced pathology hits the axons and terminals first and later progresses to involve also the cell bodies. The time-course of changes seen in the AAV-α-synuclein treated animals defines distinct stages of disease progression that matches the pre-symptomatic, early symptomatic, and advanced stages seen in PD patients. This model provides new interesting possibilities for studies of stage-specific pathologic mechanisms and identification of targets for disease-modifying therapeutic interventions linked to early or late stages of the disease.
Collapse
Affiliation(s)
- M Decressac
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, BMC A11, Lund 22184, Sweden.
| | | | | | | | | |
Collapse
|
89
|
Abstract
Advances in imaging have made it possible to detect functional and, increasingly, structural changes in Parkinson's disease. Although imaging is not yet routinely used for diagnosis, such an application is becoming increasingly feasible. Of potentially greater interest, however, is the use of imaging as a biomarker to detect premotor disease and disease progression. Imaging also provides insights into complications of Parkinson's disease and its long-term treatment, and the role of dopamine in the normal brain. Furthermore, these techniques can be applied to animal models, to help validate these models and allow their use in the study of potential disease-modifying therapies.
Collapse
Affiliation(s)
- A Jon Stoessl
- Pacific Parkinson's Research Centre, University of British Columbia, Vancouver, BC, Canada.
| | | | | | | |
Collapse
|
90
|
Enhanced gene delivery to the neonatal retina through systemic administration of tyrosine-mutated AAV9. Gene Ther 2011; 19:176-81. [PMID: 22011645 DOI: 10.1038/gt.2011.163] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Delivery of therapeutic genes to a large region of the retina with minimal damage from intraocular surgery is a central goal of treatment for retinal degenerations. Recent studies have shown that AAV9 can reach the central nervous system (CNS) and retina when administered systemically to neonates, which is a promising strategy for some retinal diseases. We investigated whether the retinal transduction efficiency of systemically delivered AAV9 could be improved by mutating capsid surface tyrosines, previously shown to increase the infectivity of several AAV vectors. Specifically, we evaluated retinal transduction following neonatal intravascular administration of AAV9 vectors containing tyrosine to phenylalanine mutations at two highly conserved sites. Our results show that a novel, double tyrosine mutant of AAV9 significantly enhanced gene delivery to the CNS and retina, and that gene expression can be restricted to rod photoreceptor cells by incorporating a rhodopsin promoter. This approach provides a new methodology for the development of retinal gene therapies or creation of animal models of neurodegenerative disease.
Collapse
|
91
|
Zalachoras I, Evers MM, van Roon-Mom WMC, Aartsma-Rus AM, Meijer OC. Antisense-mediated RNA targeting: versatile and expedient genetic manipulation in the brain. Front Mol Neurosci 2011; 4:10. [PMID: 21811437 PMCID: PMC3142880 DOI: 10.3389/fnmol.2011.00010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 07/08/2011] [Indexed: 12/28/2022] Open
Abstract
A limiting factor in brain research still is the difficulty to evaluate in vivo the role of the increasing number of proteins implicated in neuronal processes. We discuss here the potential of antisense-mediated RNA targeting approaches. We mainly focus on those that manipulate splicing (exon skipping and exon inclusion), but will also briefly discuss mRNA targeting. Classic knockdown of expression by mRNA targeting is only one possible application of antisense oligonucleotides (AON) in the control of gene function. Exon skipping and inclusion are based on the interference of AONs with splicing of pre-mRNAs. These are powerful, specific and particularly versatile techniques, which can be used to circumvent pathogenic mutations, shift splice variant expression, knock down proteins, or to create molecular models using in-frame deletions. Pre-mRNA targeting is currently used both as a research tool, e.g., in models for motor neuron disease, and in clinical trials for Duchenne muscular dystrophy and amyotrophic lateral sclerosis. AONs are particularly promising in relation to brain research, as the modified AONs are taken up extremely fast in neurons and glial cells with a long residence, and without the need for viral vectors or other delivery tools, once inside the blood brain barrier. In this review we cover (1). The principles of antisense-mediated techniques, chemistry, and efficacy. (2) The pros and cons of AON approaches in the brain compared to other techniques of interfering with gene function, such as transgenesis and short hairpin RNAs, in terms of specificity of the manipulation, spatial, and temporal control over gene expression, toxicity, and delivery issues. (3) The potential applications for Neuroscience. We conclude that there is good evidence from animal studies that the central nervous system can be successfully targeted, but the potential of the diverse AON-based approaches appears to be under-recognized.
Collapse
Affiliation(s)
- Ioannis Zalachoras
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research Leiden, Netherlands
| | | | | | | | | |
Collapse
|
92
|
Decressac M, Ulusoy A, Mattsson B, Georgievska B, Romero-Ramos M, Kirik D, Bjorklund A. GDNF fails to exert neuroprotection in a rat -synuclein model of Parkinson's disease. Brain 2011; 134:2302-11. [DOI: 10.1093/brain/awr149] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
93
|
Development of advanced therapies based on viral vector-mediated overexpression of therapeutic molecules and knockdown of disease-related genes for Parkinson’s disease. Ther Deliv 2011; 2:37-50. [DOI: 10.4155/tde.10.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The last decade witnessed the translation of several gene-based therapeutic approaches from experimental studies to early clinical trials. Studies targeting the treatment of Parkinson’s disease (PD) were among the forefront of trials in the CNS. In this article, we overview three major strategies for the treatment of PD: the enzyme-replacement strategies are based on well-defined principles of functional restoration and are well suited for treatment of patients with advanced disease who would typically experience complications due to side effects of pharmacotherapy. Neurotrophic factor delivery, on the other hand, aims to delay the disability and eventually modifiy disease progression. Finally, we present an outlook to a completely new way of interfering with the disease process, which is taking advantage of recently discovered RNAi mechanisms in cells. Gene therapy is now becoming a reality in the clinics and developments in the next decade will help uncover the true potential of this approach for not only the treatment of PD patients, but also many other neurological disorders.
Collapse
|