851
|
Nordströma U, Beauvais G, Ghosh A, Pulikkaparambil Sasidharan BC, Lundblad M, Fuchs J, Joshi RL, Lipton JW, Roholt A, Medicetty S, Feinstein TN, Steiner JA, Escobar Galvis ML, Prochiantz A, Brundin P. Progressive nigrostriatal terminal dysfunction and degeneration in the engrailed1 heterozygous mouse model of Parkinson's disease. Neurobiol Dis 2014; 73:70-82. [PMID: 25281317 DOI: 10.1016/j.nbd.2014.09.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/28/2014] [Accepted: 09/21/2014] [Indexed: 01/17/2023] Open
Abstract
Current research on Parkinson's disease (PD) pathogenesis requires relevant animal models that mimic the gradual and progressive development of neuronal dysfunction and degeneration that characterizes the disease. Polymorphisms in engrailed 1 (En1), a homeobox transcription factor that is crucial for both the development and survival of mesencephalic dopaminergic neurons, are associated with sporadic PD. This suggests that En1 mutant mice might be a promising candidate PD model. Indeed, a mouse that lacks one En1 allele exhibits decreased mitochondrial complex I activity and progressive midbrain dopamine neuron degeneration in adulthood, both features associated with PD. We aimed to further characterize the disease-like phenotype of these En1(+/-) mice with a focus on early neurodegenerative changes that can be utilized to score efficacy of future disease modifying studies. We observed early terminal defects in the dopaminergic nigrostriatal pathway in En1(+/-) mice. Several weeks before a significant loss of dopaminergic neurons in the substantia nigra could be detected, we found that striatal terminals expressing high levels of dopaminergic neuron markers TH, VMAT2, and DAT were dystrophic and swollen. Using transmission electron microscopy, we identified electron dense bodies consistent with abnormal autophagic vacuoles in these terminal swellings. In line with these findings, we detected an up-regulation of the mTOR pathway, concurrent with a downregulation of the autophagic marker LC3B, in ventral midbrain and nigral dopaminergic neurons of the En1(+/-) mice. This supports the notion that autophagic protein degradation is reduced in the absence of one En1 allele. We imaged the nigrostriatal pathway using the CLARITY technique and observed many fragmented axons in the medial forebrain bundle of the En1(+/-) mice, consistent with axonal maintenance failure. Using in vivo electrochemistry, we found that nigrostriatal terminals in the dorsal striatum were severely deficient in dopamine release and reuptake. Our findings support a progressive retrograde degeneration of En1(+/-) nigrostriatal neurons, akin to what is suggested to occur in PD. We suggest that using the En1(+/-) mice as a model will provide further key insights into PD pathogenesis, and propose that axon terminal integrity and function can be utilized to estimate dopaminergic neuron health and efficacy of experimental PD therapies.
Collapse
Affiliation(s)
- Ulrika Nordströma
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC B11, 221 84 Lund, Sweden
| | - Geneviève Beauvais
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| | - Anamitra Ghosh
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| | - Baby Chakrapani Pulikkaparambil Sasidharan
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| | - Martin Lundblad
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A11, 221 84, Lund University, Sweden
| | - Julia Fuchs
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Labex Memolife, Paris, France
| | - Rajiv L Joshi
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Labex Memolife, Paris, France
| | - Jack W Lipton
- Department of Translational Science and Molecular Medicine and The Udall Center of Excellence in Parkinson's Disease Research, Michigan State University, Grand Rapids, MI, USA
| | - Andrew Roholt
- Renovo Neural, Inc. 10000 Cedar Avenue, Cleveland, OH 44106, USA
| | - Satish Medicetty
- Renovo Neural, Inc. 10000 Cedar Avenue, Cleveland, OH 44106, USA
| | - Timothy N Feinstein
- Confocal Microscopy and Quantitative Imaging Core Facility,Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| | - Jennifer A Steiner
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| | - Martha L Escobar Galvis
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| | - Alain Prochiantz
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Labex Memolife, Paris, France
| | - Patrik Brundin
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC B11, 221 84 Lund, Sweden
- Laboratory for Translational Parkinson's Disease Research, Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave, N.E., Grand Rapids, MI 49503, USA
| |
Collapse
|
852
|
Driver-Dunckley E, Adler CH, Hentz JG, Dugger BN, Shill HA, Caviness JN, Sabbagh MN, Beach TG. Olfactory dysfunction in incidental Lewy body disease and Parkinson's disease. Parkinsonism Relat Disord 2014; 20:1260-2. [PMID: 25172126 DOI: 10.1016/j.parkreldis.2014.08.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/18/2014] [Accepted: 08/11/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Olfactory dysfunction in Parkinson's disease (PD) is well-established and may represent one of the earliest signs of the disease. OBJECTIVE & METHODS The objective of this study was to evaluate the relationship of olfactory dysfunction, using the University of Pennsylvania Smell Identification Test (UPSIT), to clinical and pathological parameters of clinicopathologically diagnosed PD (n = 10), incidental Lewy body disease (ILBD) (n = 13), and identically assessed controls who lacked a neurodegenerative disease (n = 69). RESULTS Mean UPSIT scores were significantly lower in PD (16.3, p < 0.001) and ILBD (22.2, p = 0.004) compared to controls (27.7). Using an UPSIT cutoff score of <22 (the 15th percentile) the sensitivity for detecting PD was 9/10 (90%) and ILBD 6/13 (46%), while the specificity was 86% (Controls with score of <22 = 10/69). CONCLUSIONS These results add to the growing body of evidence suggesting that olfactory testing could be useful as a screening tool for identifying early, pre-motor PD.
Collapse
Affiliation(s)
- Erika Driver-Dunckley
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic, Scottsdale, AZ, USA.
| | - Charles H Adler
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | | | | | - Holly A Shill
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | - John N Caviness
- Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | | | | | | |
Collapse
|
853
|
Wang Z, Guo Y, Myers KG, Heintz R, Peng YH, Maarek JMI, Holschneider DP. Exercise alters resting-state functional connectivity of motor circuits in parkinsonian rats. Neurobiol Aging 2014; 36:536-44. [PMID: 25219465 DOI: 10.1016/j.neurobiolaging.2014.08.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 07/28/2014] [Accepted: 08/12/2014] [Indexed: 01/23/2023]
Abstract
Few studies have examined changes in functional connectivity after long-term aerobic exercise. We examined the effects of 4 weeks of forced running wheel exercise on the resting-state functional connectivity (rsFC) of motor circuits of rats subjected to bilateral 6-hydroxydopamine lesion of the dorsal striatum. Our results showed substantial similarity between lesion-induced changes in rsFC in the rats and alterations in rsFC reported in Parkinson's disease subjects, including disconnection of the dorsolateral striatum. Exercise in lesioned rats resulted in: (1) normalization of many of the lesion-induced alterations in rsFC, including reintegration of the dorsolateral striatum into the motor network; (2) emergence of the ventrolateral striatum as a new broadly connected network hub; and (3) increased rsFC among the motor cortex, motor thalamus, basal ganglia, and cerebellum. Our results showed for the first time that long-term exercise training partially reversed lesion-induced alterations in rsFC of the motor circuits, and in addition enhanced functional connectivity in specific motor pathways in the parkinsonian rats, which could underlie recovery in motor functions observed in these animals.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Yumei Guo
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Kalisa G Myers
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Ryan Heintz
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Yu-Hao Peng
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Jean-Michel I Maarek
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Daniel P Holschneider
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Neurology, University of Southern California, Los Angeles, CA, USA; Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
854
|
Sánchez-Ferro Á, Rábano A, Catalán MJ, Rodríguez-Valcárcel FC, Fernández Díez S, Herreros-Rodríguez J, García-Cobos E, Álvarez-Santullano MM, López-Manzanares L, Mosqueira AJ, Vela Desojo L, López-Lozano JJ, López-Valdés E, Sánchez-Sánchez R, Molina-Arjona JA. In vivo gastric detection of α-synuclein inclusions in Parkinson's disease. Mov Disord 2014; 30:517-24. [PMID: 25113060 DOI: 10.1002/mds.25988] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/09/2013] [Accepted: 07/13/2014] [Indexed: 12/18/2022] Open
Abstract
α-Synuclein inclusions have been identified in the brain and some parts of the enteric nervous system in Parkinson's disease cases. We aimed to assess these inclusions in gastric mucosa samples from patients with symptomatic Parkinson's disease. Random biopsies were performed by gastroscopy in 28 patients with Parkinson's disease and in 29 age- and sex-matched controls. Gastroscopy was performed to start enteral levodopa (L-dopa) therapy in cases and for diagnostic purposes in controls (gastroesophageal reflux, anemia, and abdominal pain were the main indications). The clinical definition of cases and controls was made a priori. Six controls had data suggestive of "mild presymptomatic parkinsonism". Biopsy specimens were immunostained for α-synuclein. The neuropathological diagnosis was established post hoc. No differences were found in the baseline characteristics of the groups. Positive fibers for the α-synuclein protein were observed in 17 of 28 (60.7%) Parkinson's disease patients, 1 of 23 controls (4.3%), and 1 of 6 (16.7%) cases of incident "mild presymptomatic parkinsonism." Neuropathological diagnosis based on α-synuclein immunostaining showed a sensitivity of 85% (95% confidence interval [CI] 62.1-96.8), specificity of 95% (95% CI 76.2-99.9) and area under the receiver operating characteristics curve (AUC) of 0.90 (95% CI 0.80-1.00). No adverse events occurred. Detection of α-synuclein inclusions in the gastric mucosa is a useful and safe tool providing in vivo evidence of the underlying neurodegenerative peripheral involvement linked to Parkinson's disease. Further studies are warranted to determine its pathophysiological implications.
Collapse
Affiliation(s)
- Álvaro Sánchez-Ferro
- Department of Neurology, Hospital Universitario 12 de Octubre, Madrid, Spain; Department of Medicine, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
855
|
Schapira AHV, Olanow CW, Greenamyre JT, Bezard E. Slowing of neurodegeneration in Parkinson's disease and Huntington's disease: future therapeutic perspectives. Lancet 2014; 384:545-55. [PMID: 24954676 DOI: 10.1016/s0140-6736(14)61010-2] [Citation(s) in RCA: 287] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several important advances have been made in our understanding of the pathways that lead to cell dysfunction and death in Parkinson's disease and Huntington's disease. These advances have been informed by both direct analysis of the post-mortem brain and by study of the biological consequences of the genetic causes of these diseases. Some of the pathways that have been implicated so far include mitochondrial dysfunction, oxidative stress, kinase pathways, calcium dysregulation, inflammation, protein handling, and prion-like processes. Intriguingly, these pathways seem to be important in the pathogenesis of both diseases and have led to the identification of molecular targets for candidate interventions designed to slow or reverse their course. We review some recent advances that underlie putative therapies for neuroprotection in Parkinson's disease and Huntington's disease, and potential targets that might be exploited in the future. Although we will need to overcome important hurdles, especially in terms of clinical trial design, we propose several target pathways that merit further study. In Parkinson's disease, these targets include agents that might improve mitochondrial function or increase degradation of defective mitochondria, kinase inhibitors, calcium channel blockers, and approaches that interfere with the misfolding, templating, and transmission of α-synuclein. In Huntington's disease, strategies might also be directed at mitochondrial bioenergetics and turnover, the prevention of protein dysregulation, disruption of the interaction between huntingtin and p53 or huntingtin-interacting protein 1 to reduce apoptosis, and interference with expression of mutant huntingtin at both the nucleic acid and protein levels.
Collapse
Affiliation(s)
| | - C Warren Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, 33000 Bordeaux, France
| |
Collapse
|
856
|
Rodriguez M, Morales I, Rodriguez-Sabate C, Sanchez A, Castro R, Brito JM, Sabate M. The degeneration and replacement of dopamine cells in Parkinson's disease: the role of aging. Front Neuroanat 2014; 8:80. [PMID: 25147507 PMCID: PMC4124707 DOI: 10.3389/fnana.2014.00080] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/22/2014] [Indexed: 01/06/2023] Open
Abstract
Available data show marked similarities for the degeneration of dopamine cells in Parkinson’s disease (PD) and aging. The etio-pathogenic agents involved are very similar in both cases, and include free radicals, different mitochondrial disturbances, alterations of the mitophagy and the ubiquitin-proteasome system. Proteins involved in PD such as α-synuclein, UCH-L1, PINK1 or DJ-1, are also involved in aging. The anomalous behavior of astrocytes, microglia and stem cells of the subventricular zone (SVZ) also changes similarly in aging brains and PD. Present data suggest that PD could be the expression of aging on a cell population with high vulnerability to aging. The future knowledge of mechanisms involved in aging could be critical for both understanding the etiology of PD and developing etiologic treatments to prevent the onset of this neurodegenerative illness and to control its progression.
Collapse
Affiliation(s)
- Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna La Laguna, Tenerife, Canary Islands, Spain ; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna La Laguna, Tenerife, Canary Islands, Spain ; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Clara Rodriguez-Sabate
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna La Laguna, Tenerife, Canary Islands, Spain
| | - Rafael Castro
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna La Laguna, Tenerife, Canary Islands, Spain
| | - Jose Miguel Brito
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna La Laguna, Tenerife, Canary Islands, Spain
| | - Magdalena Sabate
- Rehabilitation Service, Department of Physical Medicine and Pharmacology, Faculty of Medicine, University of La Laguna La Laguna, Tenerife, Canary Islands, Spain
| |
Collapse
|
857
|
Kraemmer J, Kovacs GG, Perju-Dumbrava L, Pirker S, Traub-Weidinger T, Pirker W. Correlation of striatal dopamine transporter imaging with post mortem substantia nigra cell counts. Mov Disord 2014; 29:1767-73. [PMID: 25048738 DOI: 10.1002/mds.25975] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/05/2014] [Accepted: 06/24/2014] [Indexed: 11/10/2022] Open
Abstract
Dopamine transporter imaging is widely used for the differential diagnosis of parkinsonism. Only limited data are available on the relationship between striatal dopamine transporter binding and dopaminergic cell loss in the substantia nigra (SN). We analyzed postmortem SN cell counts in patients who had previously undergone dopamine transporter single-photon emission computed tomography (SPECT). Pathological diagnoses included Parkinson's disease (n = 1), dementia with Lewy bodies (n = 2), multiple system atrophy (n = 1), corticobasal degeneration (n = 2), atypical parkinsonism with multiple pathological conditions (n = 1), Alzheimer's disease (n = 1), and Creutzfeldt-Jakob disease (n = 1). [(12) (3) I]β-CIT SPECT had been performed in all subjects using a standardized protocol on the same triple-head gamma camera. The density of neuromelanin-containing and tyrosine hydroxylase-positive substantia nigra neurons/mm(2) was evaluated in paraffin-embedded tissue sections by morphometric methods. Mean disease duration at the time of dopamine transporter imaging was 2.3 years, and the mean interval from imaging to death was 29.3 months (range, 4-68 months). Visual analysis of dopamine transporter images showed reduced striatal uptake in all seven patients with neurodegenerative parkinsonism, but not in Alzheimer's and Creutzfeldt-Jakob disease cases. Averaged [(right+left)/2] striatal uptake was highly correlated with averaged SN cell counts (rs = 0.98, P < 0.0005 for neuromelanin- and rs = 0.96, P < 0.0005 for tyrosine hydroxylase-positive cells). Similar strong correlations were found in separate analyses for the right and left sides. Striatal dopamine transporter binding highly correlated with postmortem SN cell counts, confirming the validity of dopamine transporter imaging as an excellent in vivo marker of nigrostriatal dopaminergic degeneration.
Collapse
Affiliation(s)
- Julia Kraemmer
- Department of Neurology, Medical University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
858
|
Abstract
Positron emission tomography (PET) is a powerful technique to quantitatively assess brain function in vivo. In Parkinson's disease (PD), PET can assist in the identification of dopamine deficiency, the characterization of dopamine and other neurotransmitter receptors and transporters, serve as a biomarker and provide insights into motor and non-motor complications of PD. PET can also shed light on mechanisms that underlie disease, such as aberrant protein deposition and neuroinflammation. Emerging developments in multimodal imaging offer the opportunity to study multiple questions concurrently and offer great promise for the future.
Collapse
Affiliation(s)
- A Jon Stoessl
- Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health. Vancouver, Canada.
| |
Collapse
|
859
|
Dijkstra AA, Voorn P, Berendse HW, Groenewegen HJ, Rozemuller AJM, van de Berg WDJ. Stage-dependent nigral neuronal loss in incidental Lewy body and Parkinson's disease. Mov Disord 2014; 29:1244-51. [PMID: 24996051 DOI: 10.1002/mds.25952] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 05/19/2014] [Accepted: 06/02/2014] [Indexed: 11/07/2022] Open
Abstract
To gain a better understanding of the significance of α-synuclein pathological conditions during disease progression in Parkinson's disease, we investigated whether 1) nigral neuronal loss in incidental Lewy body disease and Parkinson's disease donors is associated with the local burden α-synuclein pathological conditions during progression of pathological conditions; 2) the burden and distribution of α-synuclein pathological conditions are related to clinical measures of disease progression. Post-mortem tissue and medical records of 24 Parkinson's disease patients, 20 incidental Lewy body disease donors, and 12 age-matched controls were obtained from the Netherlands Brain Bank for morphometric analysis. We observed a 20% decrease in nigral neuronal cell density in incidental Lewy body disease compared with controls. Nigral neuronal loss (12%) was already observed before the appearance α-synuclein aggregates. The progression from Braak α-synuclein stage 3 to 4 was associated with a significant decline in neuronal cell density (46%). Nigral neuronal loss increased with later Braak α-synuclein stages but did not vary across consecutive Braak α-synuclein stages. We observed a negative correlation between neuronal density and local α-synuclein burden in the substantia nigra of Parkinson's disease patients (ρ = -0.54), but no relationship with Hoehn & Yahr stage or disease duration. In conclusion, our findings cast doubt on the pathogenic role of α-synuclein aggregates in elderly, but do suggest that the severity of neurodegeneration and local burden of α-synuclein pathological conditions are closely coupled during disease progression in Parkinson's disease.
Collapse
Affiliation(s)
- Anke A Dijkstra
- Department of Anatomy and Neurosciences, section of Functional Neuroanatomy, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
860
|
Cutaneous neuropathy in Parkinson's disease: a window into brain pathology. Acta Neuropathol 2014; 128:99-109. [PMID: 24788821 PMCID: PMC4059960 DOI: 10.1007/s00401-014-1284-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/01/2014] [Accepted: 04/05/2014] [Indexed: 01/29/2023]
Abstract
The deposition of alpha-synuclein in the brain, the neuropathological hallmark of Parkinson’s disease (PD), follows a distinct anatomical and temporal sequence. This study aimed to characterize alpha-synuclein deposition in cutaneous nerves from patients with PD. We further strived to explore whether peripheral nerve involvement is intrinsic to PD and reflective of known features of brain pathology, which could render it a useful tool for pathogenetic studies and pre-mortem histological diagnosis of PD. We obtained skin biopsies from the distal and proximal leg, back and finger of 31 PD patients and 35 controls and quantified the colocalization of phosphorylated alpha-synuclein in somatosensory and autonomic nerve fibers and the pattern of loss of different subtypes of dermal fibers. Deposits of phosphorylated alpha-synuclein were identified in 16/31 PD patients but in 0/35 controls (p < 0.0001). Quantification of nerve fibers revealed two types of peripheral neurodegeneration in PD: (1) a length-dependent reduction of intraepidermal small nerve fibers (p < 0.05) and (2) a severe non-length-dependent reduction of substance P-immunoreactive intraepidermal nerve fibers (p < 0.0001). The latter coincided with a more pronounced proximal manifestation of alpha-synuclein pathology in the skin. The histological changes did not correlate with markers of levodopa toxicity such as vitamin B12 deficiency. Our findings suggest that loss of peripheral nerve fibers is an intrinsic feature of PD and that peripheral nerve changes may reflect the two types of central alpha-synuclein-related PD pathology, namely neuronal death and axonal degeneration. Detection of phosphorylated alpha-synuclein in dermal nerve fibers might be a useful diagnostic test for PD with high specificity but low sensitivity.
Collapse
|
861
|
Geldenhuys WJ, Abdelmagid SM, Gallegos PJ, Safadi FF. Parkinson’s disease biomarker: a patent evaluation of WO2013153386. Expert Opin Ther Pat 2014; 24:947-51. [DOI: 10.1517/13543776.2014.931375] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
862
|
Kordower JH. The prion hypothesis of Parkinson's disease: This hot topic just got hotter. Mov Disord 2014; 29:988. [DOI: 10.1002/mds.25918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 04/21/2014] [Accepted: 04/27/2014] [Indexed: 11/07/2022] Open
|
863
|
Kotagal V, Albin RL, Müller MLTM, Koeppe RA, Studenski S, Frey KA, Bohnen NI. Advanced age, cardiovascular risk burden, and timed up and go test performance in Parkinson disease. J Gerontol A Biol Sci Med Sci 2014; 69:1569-75. [PMID: 24864306 DOI: 10.1093/gerona/glu070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Cardiovascular comorbidities are a known risk factor for impaired mobility in elderly individuals. Motor impairments in Parkinson disease are conventionally ascribed to nigrostriatal dopaminergic denervation although progressive gait and balance impairments become more common with aging and often show limited response to dopaminergic replacement therapies. METHODS We explored the association between elevated cardiovascular risk factors and performance on the Timed Up and Go test in cross-sectional of Parkinson disease subjects (n = 83). Cardiovascular risk factor status was estimated using the Framingham General Cardiovascular Disease risk-scoring algorithm in order to dichotomize the cohort into those with and without elevated modifiable cardiovascular risk compared with normative scores for age and gender. All subjects underwent clinical and neuroimaging evaluations including a 3-m Timed Up and Go test, [(11)C]dihydrotetrabenazine positron emission tomography imaging to estimate nigrostriatal dopamine terminal loss, and an magnetic resonance imaging assessment of leukoaraiosis. A similar analysis was performed in 49 healthy controls. RESULTS After adjusting for disease duration, leukoaraiosis, and nigrostriatal dopaminergic denervation, Parkinson disease subjects with elevated Framingham risk scores (n = 61) displayed slower Timed Up and Go test performance (β = 1.86, t = 2.41, p = .018) compared with subjects with normal range Framingham risk scores (n = 22). When age ≥65 was added to the model in a post hoc analysis, the strength of effect seen with older age (β = 1.51, t = 2.44, p = .017) was similar to that of elevated Framingham risk scoring (β = 1.87, t = 2.51, p = .014). In a multivariable regression model studying the healthy control population, advanced age (t = 2.15, p = .037) was a significant predictor of Timed Up and Go speed though striatal [(11)C]dihydrotetrabenazine (t = -1.30, p = .19) and elevated Framingham risk scores (t = 1.32, p = .19) were not. CONCLUSIONS Modifiable cardiovascular risk factors and older age may independently exacerbate balance-related disability in Parkinson disease and may exert additive or synergistic pathological effects. The pathophysiology of these impairments cannot be explained completely by nigrostriatal dopaminergic denervation or leukoaraiosis burden and may relate to systemic factors seen with accelerated aging.
Collapse
Affiliation(s)
- Vikas Kotagal
- Department of Neurology, University of Michigan, Ann Arbor.
| | - Roger L Albin
- Department of Neurology, University of Michigan, Ann Arbor. Neurology Service and GRECC, VAAAHS, Ann Arbor, Michigan
| | | | | | - Stephanie Studenski
- Department of Internal Medicine, Division of Geriatrics, University of Pittsburgh, Pennsylvania
| | - Kirk A Frey
- Department of Neurology, University of Michigan, Ann Arbor. Department of Radiology, University of Michigan, Ann Arbor
| | | |
Collapse
|
864
|
Tian L, Karimi M, Brown CA, Loftin SK, Perlmutter JS. In vivo measures of nigrostriatal neuronal response to unilateral MPTP treatment. Brain Res 2014; 1571:49-60. [PMID: 24845719 DOI: 10.1016/j.brainres.2014.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 01/01/2023]
Abstract
A single unilateral intracarotid infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) into non-human primates causes injury to the nigrostriatal pathway including nigral cell bodies, axons and striatal terminal fields. In this model, motor parkinsonism correlates well with the loss of nigral dopaminergic cell bodies but only correlates with in vitro measures of nigrostriatal terminal fields when nigral cell loss does not exceed 50%. The goals of this study are to determine the relationship of motor parkinsonism with the degree of injury to nigrostriatal axons, as reflected by in vitro fiber length density measures, and compare in vivo with in vitro measures of striatal terminal fields. We determined axon integrity by measuring fiber length density with tyrosine hydroxylase (TH) immunohistology and dopamine transporter (DAT) density with DAT immunohistology. We then calculated the terminal arbor size and compared these measures with previously published data of quantified in vivo positron emission tomography (PET) measures of presynaptic dopaminergic neurons, autoradiographic measures of DAT and vesicular monoamine transporter type 2 (VMAT2), striatal dopamine, nigral cell counts, and parkinsonian motor ratings in the same animals. Our data demonstrate that in vivo and in vitro measures of striatal terminal fields correlate with each other regardless of the method of measurement. PET-based in vivo striatal measures accurately reflect in vitro measures of DAT and VMAT2. Terminal arbor size and other terminal field measures correlate with nigral TH immunoreactive (TH-ir) cell counts only when nigral TH-ir cell loss does not exceed 50%. Fiber length density was the only striatal measure that linearly correlated with motor ratings (Spearman: r=-0.81, p<0.001, n=16).
Collapse
Affiliation(s)
- LinLin Tian
- Neurology, Washington University, St. Louis, MO 63110, USA
| | | | - Chris A Brown
- Neurology, Washington University, St. Louis, MO 63110, USA
| | - Susan K Loftin
- Neurology, Washington University, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Neurology, Washington University, St. Louis, MO 63110, USA; Radiology, Washington University, St. Louis, MO 63110, USA; Neurobiology, Washington University, St. Louis, MO 63110, USA; Occupational Therapy, Washington University, St. Louis, MO 63110, USA; Physical Therapy, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
865
|
Prodoehl J, Burciu RG, Vaillancourt DE. Resting State Functional Magnetic Resonance Imaging in Parkinson’s Disease. Curr Neurol Neurosci Rep 2014; 14:448. [DOI: 10.1007/s11910-014-0448-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
866
|
Collier TJ. Rebuilding the nigrostriatal dopamine pathway: 30 years and counting. Exp Neurol 2014; 256:21-4. [PMID: 24681002 DOI: 10.1016/j.expneurol.2014.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/05/2014] [Accepted: 03/11/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Timothy J Collier
- Michigan State University, Translational Science & Molecular Medicine, Udall Center of Excellence in Parkinson's Disease Research, Edwin A. Brophy Endowed Chair in Central Nervous System Disorders, Grand Rapids, MI 49503.
| |
Collapse
|
867
|
Olanow CW. Parkinson disease: Gene therapy for Parkinson disease--a hope, or a dream? Nat Rev Neurol 2014; 10:186-7. [PMID: 24662536 DOI: 10.1038/nrneurol.2014.45] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- C Warren Olanow
- Department of Neurology, Mount Sinai School of Medicine, 1 Gustave Levy Place, Annenberg 20-82, New York, NY 10029, USA
| |
Collapse
|
868
|
Recasens A, Dehay B, Bové J, Carballo-Carbajal I, Dovero S, Pérez-Villalba A, Fernagut PO, Blesa J, Parent A, Perier C, Fariñas I, Obeso JA, Bezard E, Vila M. Lewy body extracts from Parkinson disease brains trigger α-synuclein pathology and neurodegeneration in mice and monkeys. Ann Neurol 2014; 75:351-62. [PMID: 24243558 DOI: 10.1002/ana.24066] [Citation(s) in RCA: 483] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/30/2013] [Accepted: 11/12/2013] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Mounting evidence suggests that α-synuclein, a major protein component of Lewy bodies (LB), may be responsible for initiating and spreading the pathological process in Parkinson disease (PD). Supporting this concept, intracerebral inoculation of synthetic recombinant α-synuclein fibrils can trigger α-synuclein pathology in mice. However, it remains uncertain whether the pathogenic effects of recombinant synthetic α-synuclein may apply to PD-linked pathological α-synuclein and occur in species closer to humans. METHODS Nigral LB-enriched fractions containing pathological α-synuclein were purified from postmortem PD brains by sucrose gradient fractionation and subsequently inoculated into the substantia nigra or striatum of wild-type mice and macaque monkeys. Control animals received non-LB fractions containing soluble α-synuclein derived from the same nigral PD tissue. RESULTS In both mice and monkeys, intranigral or intrastriatal inoculations of PD-derived LB extracts resulted in progressive nigrostriatal neurodegeneration starting at striatal dopaminergic terminals. No neurodegeneration was observed in animals receiving non-LB fractions from the same patients. In LB-injected animals, exogenous human α-synuclein was quickly internalized within host neurons and triggered the pathological conversion of endogenous α-synuclein. At the onset of LB-induced degeneration, host pathological α-synuclein diffusely accumulated within nigral neurons and anatomically interconnected regions, both anterogradely and retrogradely. LB-induced pathogenic effects required both human α-synuclein present in LB extracts and host expression of α-synuclein. INTERPRETATION α-Synuclein species contained in PD-derived LB are pathogenic and have the capacity to initiate a PD-like pathological process, including intracellular and presynaptic accumulations of pathological α-synuclein in different brain areas and slowly progressive axon-initiated dopaminergic nigrostriatal neurodegeneration.
Collapse
Affiliation(s)
- Ariadna Recasens
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute-Center for Networked Biomedical Research on Neurodegenerative Diseases, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
869
|
|
870
|
Tereshchenko J, Maddalena A, Bähr M, Kügler S. Pharmacologically controlled, discontinuous GDNF gene therapy restores motor function in a rat model of Parkinson's disease. Neurobiol Dis 2014; 65:35-42. [PMID: 24440408 DOI: 10.1016/j.nbd.2014.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/13/2013] [Accepted: 01/08/2014] [Indexed: 11/18/2022] Open
Abstract
Neurotrophic factors have raised hopes to be able to cure symptoms and to prevent progressive neurodegeneration in devastating neurological diseases. Gene therapy by means of viral vectors can overcome the hurdle of targeted delivery, but its current configuration is irreversible and thus much less controllable than that of classical pharmacotherapies. We thus aimed at developing a strategy allowing for both curative and controllable neurotrophic factor expression. Therefore, the short-term, intermittent and reversible expression of a neutrophic factor was evaluated for therapeutic efficacy in a slowly progressive animal model of Parkinson's disease (PD). We demonstrate that short-term induced expression of glial cell line derived neurotrophic factor (GDNF) is sufficient to provide i) substantial protection of nigral dopaminergic neurons from degeneration and ii) restoration of dopamine supply and motor behaviour in the partial striatal 6-OHDA model PD. These neurorestorative effects of GDNF lasted several weeks beyond the time of its expression. Later on, therapeutic efficacy ceased, but was restored by a second short induction of GDNF expression, demonstrating that monthly application of the inducing drug mifepristone was sufficient to maintain neuroprotective and neurorestorative GDNF levels. These findings suggest that forthcoming gene therapies for PD or other neurodegenerative disorders can be designed in a way that low frequency application of an approved drug can provide controllable and therapeutically efficient levels of GDNF or other neurotrophic factors. Neurotrophic factor expression can be withdrawn in case of off-target effects or sufficient clinical benefit, a feature that may eventually increase the acceptance of gene therapy for less advanced patients, which may profit better from such approaches.
Collapse
Affiliation(s)
- Julia Tereshchenko
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Andrea Maddalena
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Mathias Bähr
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Sebastian Kügler
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany.
| |
Collapse
|
871
|
|
872
|
Francardo V, Cenci MA. Investigating the molecular mechanisms of L-DOPA-induced dyskinesia in the mouse. Parkinsonism Relat Disord 2014; 20 Suppl 1:S20-2. [DOI: 10.1016/s1353-8020(13)70008-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
873
|
Abstract
Urinary dopamine fluctuations in the competitive inhibition state were first documented in 2009. At that time, it was noted that progressively higher daily dosing values of L-tyrosine decreased the magnitude of these fluctuations. While extensive statistical analysis has been performed by the authors since 2004, it was not until 2012 that a plausible explanation was formulated. In the process, correlations with L-tyrosine administration and the on/off effect of Parkinson's disease were defined. This paper documents the current knowledge with regard to the management of retrograde phase 1 dopamine fluctuations and investigates the hypothesis that they are caused by a melanin steal phenomenon.
Collapse
Affiliation(s)
- Marty Hinz
- Clinical Research, NeuroResearch Clinics, Inc., Cape Coral, FL, USA
| | - Alvin Stein
- Stein Orthopedic Associates, Plantation, FL, USA
| | - Ted Cole
- Cole Center for Healing, Cincinnati, OH, USA
| |
Collapse
|
874
|
Abstract
Parkinson's disease (PD) is, after Alzheimer's disease, the second most common neurodegenerative disorder with an approximate prevalence of 0.5-1% among persons 65-69 years of age, rising to 1-3% among persons 80 years of age and older. Pathologically, PD is characterized by the loss of neurons in the substantia nigra pars compacta (SNpc), and by the presence of eosinophilic protein deposits (Lewy bodies) in this region, in other aminergic nuclei and in cortical and limbic structures. Moreover, it has now been shown that pathology also involves the peripheral nervous system. Braak and colleagues suggested a thread of pathology starting from the vagal nerve to progress to the brainstem, and eventually to limbic and neocortical brain regions. This progression of pathology may account for the clinical evolution of PD toward a composite symptomatology. However, this hypothesis has been criticized by others. In this chapter, we review the clinical features of PD (motor and nonmotor) and their pathological correlates.
Collapse
|
875
|
Bartus RT, Weinberg MS, Samulski RJ. Parkinson's disease gene therapy: success by design meets failure by efficacy. Mol Ther 2013; 22:487-497. [PMID: 24356252 PMCID: PMC3944322 DOI: 10.1038/mt.2013.281] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/05/2013] [Indexed: 12/13/2022] Open
Abstract
Over the past decade, nine gene therapy clinical trials for Parkinson's disease (PD) have been initiated and completed. Starting with considerable optimism at the initiation of each trial, none of the programs has yet borne sufficiently robust clinical efficacy or found a clear path toward regulatory approval. Despite the immediately disappointing nature of the efficacy outcomes in these trials, the clinical data garnered from the individual studies nonetheless represent tangible and significant progress for the gene therapy field. Collectively, the clinical trials demonstrate that we have overcome the major safety hurdles previously suppressing central nervous system (CNS) gene therapy, for none produced any evidence of untoward risk or harm after administration of various vector-delivery systems. More importantly, these studies also demonstrated controlled, highly persistent generation of biologically active proteins targeted to structures deep in the human brain. Therefore, a renewed, focused emphasis must be placed on advancing clinical efficacy by improving clinical trial design, patient selection and outcome measures, developing more predictive animal models to support clinical testing, carefully performing retrospective analyses, and most importantly moving forward—beyond our past limits.
Collapse
Affiliation(s)
- Raymond T Bartus
- Ceregene, Inc., San Diego, California, USA; RTBioconsultants, Inc., San Diego, California, USA.
| | - Marc S Weinberg
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
876
|
Abstract
There are a growing number of roles that midbrain dopamine (DA) neurons assume, such as, reward, aversion, alerting and vigor. Here I propose a theory that may be able to explain why the suggested functions of DA came about. It has been suggested that largely parallel cortico-basal ganglia-thalamo-cortico loops exist to control different aspects of behavior. I propose that (1) the midbrain DA system is organized in a similar manner, with different groups of DA neurons corresponding to these parallel neural pathways (NPs). The DA system can be viewed as the "manager" of these parallel NPs in that it recruits and activates only the task-relevant NPs when they are needed. It is likely that the functions of those NPs that have been consistently activated by the corresponding DA groups are facilitated. I also propose that (2) there are two levels of DA roles: the How and What roles. The How role is encoded in tonic and phasic DA neuron firing patterns and gives a directive to its target NP: how vigorously its function needs to be carried out. The tonic DA firing is to provide the needed level of DA in the target NPs to support their expected behavioral and mental functions; it is only when a sudden unexpected boost or suppression of activity is required by the relevant target NP that DA neurons in the corresponding NP act in a phasic manner. The What role is the implementational aspect of the role of DA in the target NP, such as binding to D1 receptors to boost working memory. This What aspect of DA explains why DA seems to assume different functions depending on the region of the brain in which it is involved. In terms of the role of the lateral habenula (LHb), the LHb is expected to suppress maladaptive behaviors and mental processes by controlling the DA system. The demand-based smart management by the DA system may have given animals an edge in evolution with adaptive behaviors and a better survival rate in resource-scarce situations.
Collapse
Affiliation(s)
- Simon Hong
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology Cambridge, MA, USA
| |
Collapse
|
877
|
Moloney TC, Hyland R, O'Toole D, Paucard A, Kirik D, O'Doherty A, Gorman AM, Dowd E. Heat shock protein 70 reduces α-synuclein-induced predegenerative neuronal dystrophy in the α-synuclein viral gene transfer rat model of Parkinson's disease. CNS Neurosci Ther 2013; 20:50-8. [PMID: 24279716 DOI: 10.1111/cns.12200] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 10/02/2013] [Accepted: 10/06/2013] [Indexed: 01/20/2023] Open
Abstract
AIMS It has become increasingly evident that the nigrostriatal degeneration associated with Parkinson's disease initiates at the level of the axonal terminals in the putamen, and this nigrostriatal terminal dystrophy is either caused or exacerbated by the presence of α-synuclein immunopositive neuronal inclusions. Therefore, strategies aimed at reducing α-synuclein-induced early neuronal dystrophy may slow or halt the progression to overt nigrostriatal neurodegeneration. Thus, this study sought to determine if adeno-associated virus (AAV) mediated overexpression of two molecular chaperone heat shock proteins, namely Hsp27 or Hsp70, in the AAV-α-synuclein viral gene transfer rat model of Parkinson's disease could prevent α-synuclein-induced early neuronal pathology. METHODS Male Sprague-Dawley rats were intranigrally coinjected with pathogenic (AAV-α-synuclein) and putative therapeutic (AAV-Hsp27 or AAV-Hsp70) viral vectors and were sacrificed 18 weeks postviral injection. RESULTS Intranigral injection of AAV-α-synuclein resulted in significant α-synuclein accumulation in the substantia nigra and striatal terminals which led to significant dystrophy of nigrostriatal dopaminergic neurons without overt nigrostriatal neurodegeneration. Coinjection of AAV-Hsp70, but not AAV-Hsp27, significantly reduced AAV-α-synuclein-induced neuronal dystrophy. CONCLUSIONS These data confirm that overexpression of Hsp70 holds significant potential as a disease-modulating therapeutic approach for Parkinson's disease, with protective effects against early-onset α-synuclein-induced pathology demonstrated in the AAV-α-synuclein model.
Collapse
Affiliation(s)
- Teresa C Moloney
- Department of Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland; National Centre for Biomedical Engineering Science (NCBES), National University of Ireland, Galway, Ireland; NCBES Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
878
|
Daviaud N, Garbayo E, Lautram N, Franconi F, Lemaire L, Perez-Pinzon M, Montero-Menei CN. Modeling nigrostriatal degeneration in organotypic cultures, a new ex vivo model of Parkinson's disease. Neuroscience 2013; 256:10-22. [PMID: 24161279 DOI: 10.1016/j.neuroscience.2013.10.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/30/2013] [Accepted: 10/10/2013] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disorder afflicting 2% of the population older than 65 years worldwide. Recently, brain organotypic slices have been used to model neurodegenerative disorders, including PD. They conserve brain three-dimensional architecture, synaptic connectivity and its microenvironment. This model has allowed researchers a simple and rapid method to observe cellular interactions and mechanisms. In the present study, we developed an organotypic PD model from rat brains that includes all the areas involved in the nigrostriatal pathway in a single slice preparation, without using neurotoxins to induce the dopaminergic lesion. The mechanical transection of the nigrostriatal pathway obtained during slice preparation induced PD-like histopathology. Progressive nigrostriatal degeneration was monitored combining innovative approaches, such as diffusion tensor magnetic resonance imaging (DT-RMI) to follow fiber degeneration and mass spectrometry to quantify striatal dopamine content, together with bright-field and fluorescence microscopy imaging. A substantia nigra dopaminergic cell number decrease was observed by immunohistochemistry against rat tyrosine hydroxylase (TH) reaching 80% after 2 days in culture associated with a 30% decrease of striatal TH-positive fiber density, a 15% loss of striatal dopamine content quantified by mass spectrometry and a 70% reduction of nigrostriatal fiber fractional anisotropy quantified by DT-RMI. In addition, a significant decline of medium spiny neuron density was observed from days 7 to 16. These sagittal organotypic slices could be used to study the early stage of PD, namely dopaminergic degeneration, and the late stage of the pathology with dopaminergic and GABAergic neuron loss. This novel model might improve the understanding of PD and may represent a promising tool to refine the evaluation of new therapeutic approaches.
Collapse
Affiliation(s)
- N Daviaud
- LUNAM University, Angers University, France; INSERM UMR S_1066, Angers University, France
| | - E Garbayo
- LUNAM University, Angers University, France; INSERM UMR S_1066, Angers University, France; Pharmacy and Pharmaceutical Technology Department, University of Navarra, Pamplona, Spain
| | - N Lautram
- LUNAM University, Angers University, France; INSERM UMR S_1066, Angers University, France
| | - F Franconi
- CIFAB-PRIMEX, LUNAM University, Angers University, France
| | - L Lemaire
- LUNAM University, Angers University, France; INSERM UMR S_1066, Angers University, France
| | - M Perez-Pinzon
- University of Miami, Miller School of Medicine, Miami, FL, USA
| | - C N Montero-Menei
- LUNAM University, Angers University, France; INSERM UMR S_1066, Angers University, France.
| |
Collapse
|
879
|
Martinez RC, Hamani C, de Carvalho MC, de Oliveira AR, Alho E, Navarro J, dos Santos Ghilardi MG, Bor-Seng-Shu E, Heinsen H, Otoch JP, Brandão ML, Barbosa ER, Teixeira MJ, Fonoff ET. Intraoperative dopamine release during globus pallidus internus stimulation in Parkinson's disease. Mov Disord 2013; 28:2027-32. [DOI: 10.1002/mds.25691] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 08/15/2013] [Accepted: 08/22/2013] [Indexed: 12/24/2022] Open
Affiliation(s)
- Raquel C.R. Martinez
- Laboratory of Neuromodulation and Experimental Pain; Hospital Sirio-Libanes São Paulo Brazil
- University of São Paulo and Institute of Neuroscience and Behavior (INeC); Campus USP Ribeirao Preto Brazil
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
- Department of Surgery, Discipline of Surgical Technique; School of Medicine, University of São Paulo; LIM 26 HC-FMUSP São Paulo Brazil
| | - Clement Hamani
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
| | | | - Amanda Ribeiro de Oliveira
- University of São Paulo and Institute of Neuroscience and Behavior (INeC); Campus USP Ribeirao Preto Brazil
| | - Eduardo Alho
- Labor für Morphologische Hirnforschung (Laboratory for Morphological Brain Research); Julius-Maximilians Universität Würzburg; Würzburg Germany
| | - Jessie Navarro
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
| | - Maria Gabriela dos Santos Ghilardi
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
| | - Edson Bor-Seng-Shu
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
| | - Helmut Heinsen
- Labor für Morphologische Hirnforschung (Laboratory for Morphological Brain Research); Julius-Maximilians Universität Würzburg; Würzburg Germany
| | - José Pinhata Otoch
- Department of Surgery, Discipline of Surgical Technique; School of Medicine, University of São Paulo; LIM 26 HC-FMUSP São Paulo Brazil
| | - Marcus Lira Brandão
- University of São Paulo and Institute of Neuroscience and Behavior (INeC); Campus USP Ribeirao Preto Brazil
| | - Egberto Reis Barbosa
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
| | - Manoel Jacobsen Teixeira
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
- Department of Surgery, Discipline of Surgical Technique; School of Medicine, University of São Paulo; LIM 26 HC-FMUSP São Paulo Brazil
| | - Erich Talamoni Fonoff
- Department of Neurology, Division of Functional Neurosurgery; Institute of Psychiatry; University of São Paulo School of Medicine; São Paulo Brazil
- Department of Surgery, Discipline of Surgical Technique; School of Medicine, University of São Paulo; LIM 26 HC-FMUSP São Paulo Brazil
| |
Collapse
|