1
|
Moore J, Wu T, Dhindsa J, El Fadel O, Le A, Perez A, Amoh B, Tarkunde A, Zhu KF, Avalos M, Dammer EB, Duong DM, Seyfried NT, Shulman JM, Al-Ramahi I, Botas J. Longitudinal multi-omics in alpha-synuclein Drosophila model discriminates disease- from age-associated pathologies in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:46. [PMID: 40069190 PMCID: PMC11897226 DOI: 10.1038/s41531-025-00899-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Parkinson's disease (PD) starts decades before symptoms appear, usually in the later decades of life, when age-related changes are occurring. To identify molecular changes early in the disease course and distinguish PD pathologies from aging, we generated Drosophila expressing alpha-synuclein (αSyn) in neurons and performed longitudinal bulk transcriptomics and proteomics on brains at six time points across the lifespan and compared the data to healthy control flies as well as human post-mortem brain datasets. We found that translational and energy metabolism pathways were downregulated in αSyn flies at the earliest timepoints; comparison with the aged control flies suggests that elevated αSyn accelerates changes associated with normal aging. Unexpectedly, single-cell analysis at a mid-disease stage revealed that neurons upregulate protein synthesis and nonsense-mediated decay, while glia drive their overall downregulation. Longitudinal multi-omics approaches in animal models can thus help elucidate the molecular cascades underlying neurodegeneration vs. aging and co-pathologies.
Collapse
Affiliation(s)
- Justin Moore
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Quantitative and Computational Bioscience Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Timothy Wu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Justin Dhindsa
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Omar El Fadel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Anh Le
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Alma Perez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Bismark Amoh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Akash Tarkunde
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Katy F Zhu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Matthew Avalos
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Duc M Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Joshua M Shulman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Deparment of Neurology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ismael Al-Ramahi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Quantitative and Computational Bioscience Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, 77030, USA.
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Lim D, Matute C, Cavaliere F, Verkhratsky A. Neuroglia in neurodegeneration: Alzheimer, Parkinson, and Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:9-44. [PMID: 40148060 DOI: 10.1016/b978-0-443-19102-2.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The conspicuous rise of chronic neurodegenerative diseases, including Alzheimer (AD), Parkinson (PD), and Huntington (HD) diseases, is currently without disease-modifying therapies and accompanied by an excessive rate of unsuccessful clinical trials. This reflects a profound lack of understanding of the pathogenesis of these diseases, indicating that the current paradigms guiding disease modeling and drug development are in need of reconsideration. The role of neuroglia, namely astrocytes, microglial cells, and oligodendrocytes, in the pathogenesis of neurodegenerative diseases emerged during the last decades. This chapter provides the state-of-the-art update on the changes of astrocytes, microglial cells, and oligodendrocytes in AD, PD, and HD. A growing body of evidence suggests that homeostatic and defensive functions of glial cells are compromised at different disease stages, leading to increased susceptibility of neurons to noxious stimuli, eventually resulting in their malfunction and degeneration. Investments are needed in the generation of novel preclinical models suitable for studying glial pathology, in "humanizing" research, and in-depth investigation of glial cell alterations to slow down and, possibly, halt and prevent the rise of neurodegenerative disease. Targeting glial cells opens new therapeutic avenues to treat AD, PD, and HD.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy.
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain
| | - Fabio Cavaliere
- The Basque Biomodels Platform for Human Research (BBioH), Achucarro Basque Center for Neuroscience & Fundación Biofisica Bizkaia, Leioa, Spain
| | - Alexei Verkhratsky
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
3
|
Lau K, Kotzur R, Richter F. Blood-brain barrier alterations and their impact on Parkinson's disease pathogenesis and therapy. Transl Neurodegener 2024; 13:37. [PMID: 39075566 PMCID: PMC11285262 DOI: 10.1186/s40035-024-00430-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
There is increasing evidence for blood-brain barrier (BBB) alterations in Parkinson's disease (PD), the second most common neurodegenerative disorder with rapidly rising prevalence. Altered tight junction and transporter protein levels, accumulation of α-synuclein and increase in inflammatory processes lead to extravasation of blood molecules and vessel degeneration. This could result in a self-perpetuating pathophysiology of inflammation and BBB alteration, which contribute to neurodegeneration. Toxin exposure or α-synuclein over-expression in animal models has been shown to initiate similar pathologies, providing a platform to study underlying mechanisms and therapeutic interventions. Here we provide a comprehensive review of the current knowledge on BBB alterations in PD patients and how rodent models that replicate some of these changes can be used to study disease mechanisms. Specific challenges in assessing the BBB in patients and in healthy controls are discussed. Finally, a potential role of BBB alterations in disease pathogenesis and possible implications for therapy are explored. The interference of BBB alterations with current and novel therapeutic strategies requires more attention. Brain region-specific BBB alterations could also open up novel opportunities to target specifically vulnerable neuronal subpopulations.
Collapse
Affiliation(s)
- Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Rebecca Kotzur
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
4
|
Yamanaka T, Matsui H. Modeling familial and sporadic Parkinson's disease in small fishes. Dev Growth Differ 2024; 66:4-20. [PMID: 37991125 DOI: 10.1111/dgd.12904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/26/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
The establishment of animal models for Parkinson's disease (PD) has been challenging. Nevertheless, once established, they will serve as valuable tools for elucidating the causes and pathogenesis of PD, as well as for developing new strategies for its treatment. Following the recent discovery of a series of PD causative genes in familial cases, teleost fishes, including zebrafish and medaka, have often been used to establish genetic PD models because of their ease of breeding and gene manipulation, as well as the high conservation of gene orthologs. Some of the fish lines can recapitulate PD phenotypes, which are often more pronounced than those in rodent genetic models. In addition, a new experimental teleost fish, turquoise killifish, can be used as a sporadic PD model, because it spontaneously manifests age-dependent PD phenotypes. Several PD fish models have already made significant contributions to the discovery of novel PD pathological features, such as cytosolic leakage of mitochondrial DNA and pathogenic phosphorylation in α-synuclein. Therefore, utilizing various PD fish models with distinct degenerative phenotypes will be an effective strategy for identifying emerging facets of PD pathogenesis and therapeutic modalities.
Collapse
Affiliation(s)
- Tomoyuki Yamanaka
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
5
|
Scott L, Karuppagounder SS, Neifert S, Kang BG, Wang H, Dawson VL, Dawson TM. The Absence of Parkin Does Not Promote Dopamine or Mitochondrial Dysfunction in PolgA D257A/D257A Mitochondrial Mutator Mice. J Neurosci 2022; 42:9263-9277. [PMID: 36280265 PMCID: PMC9761676 DOI: 10.1523/jneurosci.0545-22.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/10/2022] [Accepted: 10/16/2022] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). In this study, we generated a transgenic model by crossing germline Parkin-/- mice with PolgAD257A mice, an established model of premature aging and mitochondrial stress. We hypothesized that loss of Parkin-/- in PolgAD257A/D257A mice would exacerbate mitochondrial dysfunction, leading to loss of dopamine neurons and nigral-striatal specific neurobehavioral motor dysfunction. We found that aged Parkin-/-/PolgAD257A/D257A male and female mice exhibited severe behavioral deficits, nonspecific to the nigral-striatal pathway, with neither dopaminergic neurodegeneration nor reductions in striatal dopamine. We saw no difference in expression levels of nuclear-encoded subunits of mitochondrial markers and mitochondrial Complex I and IV activities, although we did observe substantial reductions in mitochondrial-encoded COX41I, indicating mitochondrial dysfunction as a result of PolgAD257A/D257A mtDNA mutations. Expression levels of mitophagy markers LC3I/LC3II remained unchanged between cohorts, suggesting no overt mitophagy defects. Expression levels of the parkin substrates, VDAC, NLRP3, and AIMP2 remained unchanged, suggesting no parkin dysfunction. In summary, we were unable to observe dopaminergic neurodegeneration with corresponding nigral-striatal neurobehavioral deficits, nor Parkin or mitochondrial dysfunction in Parkin-/-/PolgAD257A/D257A mice. These findings support a lack of synergism of Parkin loss on mitochondrial dysfunction in mouse models of mitochondrial deficits.SIGNIFICANCE STATEMENT Producing a mouse model of Parkinson's disease (PD) that is etiologically relevant, recapitulates clinical hallmarks, and exhibits reproducible results is crucial to understanding the underlying pathology and in developing disease-modifying therapies. Here, we show that Parkin-/-/PolgAD257A/D257A mice, a previously reported PD mouse model, fails to reproduce a Parkinsonian phenotype. We show that these mice do not display dopaminergic neurodegeneration nor nigral-striatal-dependent motor deficits. Furthermore, we report that Parkin loss does not synergize with mitochondrial dysfunction. Our results demonstrate that Parkin-/-/PolgAD257A/D257A mice are not a reliable model for PD and adds to a growing body of work demonstrating that Parkin loss does not synergize with mitochondrial dysfunction in mouse models of mitochondrial deficits.
Collapse
Affiliation(s)
- Laura Scott
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana 70130-2685
| | - Senthilkumar S Karuppagounder
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Stewart Neifert
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana 70130-2685
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Bong Gu Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Hu Wang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana 70130-2685
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana 70130-2685
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
6
|
Ahnaou A, Whim D. REM sleep behavior and olfactory dysfunction: improving the utility and translation of animal models in the search for neuroprotective therapies for Parkinson's disease. Neurosci Biobehav Rev 2022; 143:104897. [PMID: 36183864 DOI: 10.1016/j.neubiorev.2022.104897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
Parkinson's disease (PD) is a heterogeneous neurodegenerative disease that belongs to the family of synucleiopathies, varying in age, symptoms and progression. Hallmark of the disease is the accumulation of misfolded α-synuclein protein (α-Syn) in neuronal and non-neuronal brain cells. In past decades, diagnosis and treatment of PD has focused on motor deficits, which for the clinical endpoint, have contributed to the prevalence of deficits in the nigrostriatal dopaminergic system and animal models related to motor behavior to study disease. However, clinical trials have failed to translate results from animal models into effective treatments. PD as a multisystem disorder therefore requires additional assessment of motor and non-motor symptoms. Braak's staging revealed early α-Syn pathology in pontine brainstem and olfactory circuits controlling rapid eye movement sleep behavior disorder (RBD) and olfaction, respectively. Recent converging evidence from multicenter clinical studies supports that RBD is the most important risk factor for prodromal PD and the conduct of neuroprotective therapeutic trials in RBD-enriched cohorts has been recommended. Animal models of RBD and olfaction dysfunction can aid to fill the gap in translational research.
Collapse
Affiliation(s)
- A Ahnaou
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV. Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | - Drinkenburg Whim
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV. Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
7
|
Ruiz MCM, Guimarães RP, Mortari MR. Parkinson’s Disease Rodent Models: are they suitable for DBS research? J Neurosci Methods 2022; 380:109687. [DOI: 10.1016/j.jneumeth.2022.109687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/20/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022]
|
8
|
Rauschenberger L, Behnke J, Grotemeyer A, Knorr S, Volkmann J, Ip CW. Age-dependent neurodegeneration and neuroinflammation in a genetic A30P/A53T double-mutated α-synuclein mouse model of Parkinson's disease. Neurobiol Dis 2022; 171:105798. [PMID: 35750147 DOI: 10.1016/j.nbd.2022.105798] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/31/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022] Open
Abstract
The pathogenesis of Parkinson's disease (PD) is closely interwoven with the process of aging. Moreover, increasing evidence from human postmortem studies and from animal models for PD point towards inflammation as an additional factor in disease development. We here assessed the impact of aging and inflammation on dopaminergic neurodegeneration in the hm2α-SYN-39 mouse model of PD that carries the human, A30P/A53T double-mutated α-synuclein gene. At 2-3 months of age, no significant differences were observed comparing dopaminergic neuron numbers of the substantia nigra (SN) pars compacta of hm2α-SYN-39 mice with wildtype controls. At an age of 16-17 months, however, hm2α-SYN-39 mice revealed a significant loss of dopaminergic SN neurons, of dopaminergic terminals in the striatum as well as a reduction of striatal dopamine levels compared to young, 2-3 months transgenic mice and compared to 16-17 months old wildtype littermates. A significant age-related correlation of infiltrating CD4+ and CD8+ T cell numbers with dopaminergic terminal loss of the striatum was found in hm2α-SYN-39 mice, but not in wildtype controls. In the striatum of 16-17 months old wildtype mice a slightly elevated CD8+ T cell count and CD11b+ microglia cell count was observed compared to younger aged mice. Additional analyses of neuroinflammation in the nigrostriatal tract of wildtype mice did not yield any significant age-dependent changes of CD4+, CD8+ T cell and B220+ B cell numbers, respectively. In contrast, a significant age-dependent increase of CD8+ T cells, GFAP+ astrocytes as well as a pronounced increase of CD11b+ microglia numbers were observed in the SN of hm2α-SYN-39 mice pointing towards a neuroinflammatory processes in this genetic mouse model for PD. The findings in the hm2α-SYN-39 mouse model strengthen the evidence that T cell and glial cell responses are involved in the age-related neurodegeneration in PD. The slow and age-dependent progression of neurodegeneration and neuroinflammation in the hm2α-SYN-39 PD rodent model underlines its translational value and makes it suitable for studying anti-inflammatory therapies.
Collapse
Affiliation(s)
- Lisa Rauschenberger
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Jennifer Behnke
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Alexander Grotemeyer
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Susanne Knorr
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| |
Collapse
|
9
|
Morais VA, Vos M. Reduced penetrance of Parkinson's disease models. MED GENET-BERLIN 2022; 34:117-124. [PMID: 38835909 PMCID: PMC11006373 DOI: 10.1515/medgen-2022-2138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The etiology and progression of Parkinson's Disease (PD), the second most prevalent neurological disorder, have been widely investigated for several decades; however, a cure is still lacking. Despite the development of several neurotoxins and animal models to study this rather heterogeneous disease, a complete recapitulation of the neurophysiology and neuropathology of PD has not been fully achieved. One underlying cause for this could be that mutations in PD-associated genes have reduced penetrance. Therefore, the quest for novel PD models is required where a double hit approach needs to be evoked - a combination of genetic alterations and environmental factors need to be accounted for in one unique model simultaneously.
Collapse
Affiliation(s)
- Vanessa A Morais
- iMM, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028, Portugal
| | - Melissa Vos
- Institute of Neurogenetics, University of Luebeck, Ratzeburger Allee 160 building 67, 23562 Luebeck, Germany
| |
Collapse
|
10
|
Oli V, Gupta R, Kumar P. FOXO and related transcription factors binding elements in the regulation of neurodegenerative disorders. J Chem Neuroanat 2021; 116:102012. [PMID: 34400291 DOI: 10.1016/j.jchemneu.2021.102012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/16/2021] [Accepted: 08/07/2021] [Indexed: 12/16/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and others, are characterized by progressive loss of neuronal cells, which causes memory impairment and cognitive decline. Mounting evidence demonstrated the possible implications of diverse biological processes, namely oxidative stress, mitochondrial dysfunction, aberrant cell cycle re-entry, post-translational modifications, protein aggregation, impaired proteasome dysfunction, autophagy, and many others that cause neuronal cell death. The condition worsens as there is no effective treatment for such diseases due to their complex pathogenesis and mechanism. Mounting evidence demonstrated the role of regulatory transcription factors, such as NFκβ, FoxO, Myc, CREB, and others that regulate the biological processes and diminish the disease progression and pathogenesis. Studies demonstrated that forkhead box O (FoxO) transcription factors had been implicated in the regulation of aging and longevity. Further, the functions of FoxO proteins are regulated by different post-translational modifications (PTMs), namely acetylation, and ubiquitination. Various studies concluded that FoxO proteins exert both neuroprotective and neurotoxic properties depending on their regulation mechanism and activity in the brain. Thus, understanding the nature of FoxO expression and activity in the brain will help develop effective therapeutic strategies. Herein, firstly, we discuss the role of FoxO protein in cell cycle regulation and cell proliferation, followed by the regulation of FoxO proteins through acetylation and ubiquitination. We also briefly explain the activity and expression pattern of FoxO proteins in the neuronal cells and explain the mechanism through which FoxO proteins are rescued from oxidative stress-induced neurotoxicity. Later on, we present a detailed view of the implication of FoxO proteins in neurodegenerative disease and FoxO proteins as an effective therapeutic target.
Collapse
Affiliation(s)
- Vaibhav Oli
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), India.
| |
Collapse
|
11
|
Martinez Hernandez A, Silbern I, Geffers I, Tatenhorst L, Becker S, Urlaub H, Zweckstetter M, Griesinger C, Eichele G. Low-Expressing Synucleinopathy Mouse Models Based on Oligomer-Forming Mutations and C-Terminal Truncation of α-Synuclein. Front Neurosci 2021; 15:643391. [PMID: 34220415 PMCID: PMC8248494 DOI: 10.3389/fnins.2021.643391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
α-synuclein (αSyn) is the main protein component of Lewy bodies, intracellular inclusions found in the brain of Parkinson's disease (PD) patients. Neurotoxic αSyn species are broadly modified post-translationally and, in patients with genetic forms of PD, carry genetically encoded amino acid substitutions. Mutations and C-terminal truncation can increase αSyn oligomerization and fibrillization. Although several genetic mouse models based on αSyn mutations and/or truncations exist, there is still a lack of mouse models for synucleinopathies not relying on overexpression. We report here two synucleinopathy mouse models, which are based on a triple alanine to proline mutation and a C-terminal truncation of αSyn, but do not overexpress the mutant protein when compared to the endogenous mouse protein. We knocked hαSyn TP or hαSynΔ119 (h stands for "human") into the murine αSyn locus. hαSynTP is a structure-based mutant with triple alanine to proline substitutions that favors oligomers, is neurotoxic and evokes PD-like symptoms in Drosophila melanogaster. hαSynΔ119 lacks 21 amino acids at the C-terminus, favors fibrillary aggregates and occurs in PD. Knocking-in of hαSyn TP or hαSynΔ119 into the murine αSyn locus places the mutant protein under the control of the endogenous regulatory elements while simultaneously disrupting the mαSyn gene. Mass spectrometry revealed that hαSyn TP and hαSynΔ119 mice produced 12 and 10 times less mutant protein, compared to mαSyn in wild type mice. We show phenotypes in 1 and 1.5 years old hαSyn TP and hαSynΔ119 mice, despite the lower levels of hαSynTP and hαSynΔ119 expression. Direct comparison of the two mouse models revealed many commonalities but also aspects unique to each model. Commonalities included strong immunoactive state, impaired olfaction and motor coordination deficits. Neither model showed DAergic neuronal loss. Impaired climbing abilities at 1 year of age and a deviant gait pattern at 1.5 years old were specific for hαSynΔ119 mice, while a compulsive behavior was exclusively detected in hαSyn TP mice starting at 1 year of age. We conclude that even at very moderate levels of expression the two αSyn variants evoke measurable and progressive deficiencies in mutant mice. The two transgenic mouse models can thus be suitable to study αSyn-variant-based pathology in vivo and test new therapeutic approaches.
Collapse
Affiliation(s)
- Ana Martinez Hernandez
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ivan Silbern
- Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.,Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Insa Geffers
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany.,Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Becker
- NMR-Based Structural Biology Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Henning Urlaub
- Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.,Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Markus Zweckstetter
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany.,NMR-Based Structural Biology Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,German Center for Neurodegenerative Diseases, DZNE, Göttingen, Germany
| | - Christian Griesinger
- Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany.,NMR-Based Structural Biology Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Gregor Eichele
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
12
|
Petiet A. Current and Emerging MR Methods and Outcome in Rodent Models of Parkinson's Disease: A Review. Front Neurosci 2021; 15:583678. [PMID: 33897339 PMCID: PMC8058186 DOI: 10.3389/fnins.2021.583678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 03/05/2021] [Indexed: 12/03/2022] Open
Abstract
Parkinson’s disease (PD) is a major neurodegenerative disease characterized by massive degeneration of the dopaminergic neurons in the substantia nigra pars compacta, α-synuclein-containing Lewy bodies, and neuroinflammation. Magnetic resonance (MR) imaging plays a crucial role in the diagnosis and monitoring of disease progression and treatment. A variety of MR methods are available to characterize neurodegeneration and other disease features such as iron accumulation and metabolic changes in animal models of PD. This review aims at giving an overview of how those physiopathological features of PD have been investigated using various MR methods in rodent models. Toxin-based and genetic-based models of PD are first described. MR methods for neurodegeneration evaluation, iron load, and metabolism alterations are then detailed, and the main findings are provided in those models. Ultimately, future directions are suggested for neuroinflammation and neuromelanin evaluations in new animal models.
Collapse
Affiliation(s)
- Alexandra Petiet
- Centre de Neuroimagerie de Recherche, Institut du Cerveau, Paris, France.,Inserm U1127, CNRS UMR 7225, Sorbonne Universités, Paris, France
| |
Collapse
|
13
|
de Bem AF, Krolow R, Farias HR, de Rezende VL, Gelain DP, Moreira JCF, Duarte JMDN, de Oliveira J. Animal Models of Metabolic Disorders in the Study of Neurodegenerative Diseases: An Overview. Front Neurosci 2021; 14:604150. [PMID: 33536868 PMCID: PMC7848140 DOI: 10.3389/fnins.2020.604150] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence of metabolic disorders, as well as of neurodegenerative diseases—mainly the sporadic forms of Alzheimer’s and Parkinson’s disease—are increasing worldwide. Notably, obesity, diabetes, and hypercholesterolemia have been indicated as early risk factors for sporadic forms of Alzheimer’s and Parkinson’s disease. These conditions share a range of molecular and cellular features, including protein aggregation, oxidative stress, neuroinflammation, and blood-brain barrier dysfunction, all of which contribute to neuronal death and cognitive impairment. Rodent models of obesity, diabetes, and hypercholesterolemia exhibit all the hallmarks of these degenerative diseases, and represent an interesting approach to the study of the phenotypic features and pathogenic mechanisms of neurodegenerative disorders. We review the main pathological aspects of Alzheimer’s and Parkinson’s disease as summarized in rodent models of obesity, diabetes, and hypercholesterolemia.
Collapse
Affiliation(s)
- Andreza Fabro de Bem
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brazilia, Brazil
| | - Rachel Krolow
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Hémelin Resende Farias
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Victória Linden de Rezende
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Miguel das Neves Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
14
|
Blosser JA, Podolsky E, Lee D. L-DOPA-Induced Dyskinesia in a Genetic Drosophila Model of Parkinson's Disease. Exp Neurobiol 2020; 29:273-284. [PMID: 32921640 PMCID: PMC7492844 DOI: 10.5607/en20028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 01/05/2023] Open
Abstract
Motor symptoms in Parkinson’s disease (PD) are directly related to the reduction of a neurotransmitter dopamine. Therefore, its precursor L-DOPA became the gold standard for PD treatment. However, chronic use of L-DOPA causes uncontrollable, involuntary movements, called L-DOPA-induced dyskinesia (LID) in the majority of PD patients. LID is complicated and very difficult to manage. Current rodent and non-human primate models have been developed to study LID mainly using neurotoxins. Therefore, it is necessary to develop a LID animal model with defects in genetic factors causing PD in order to study the relation between LID and PD genes such as α-synuclein. In this study, we first showed that a low concentration of L-DOPA (100 µM) rescues locomotion defects (i.e., speed, angular velocity, pause time) in Drosophila larvae expressing human mutant α-synuclein (A53T). This A53T larval model of PD was used to further examine dyskinetic behaviors. High concentrations of L-DOPA (5 or 10 mM) causes hyperactivity such as body bending behavior (BBB) in A53T larva, which resembles axial dyskinesia in rodents. Using ImageJ plugins and other third party software, dyskinetic BBB has been accurately and efficiently quantified. Further, we showed that a dopamine agonist pramipexole (PRX) partially rescues BBB caused by high L-DOPA. Our Drosophila genetic LID model will provide an important experimental platform to examine molecular and cellular mechanisms underlying LID, to study the role of PD causing genes in the development of LID, and to identify potential targets to slow/reverse LID pathology.
Collapse
Affiliation(s)
- Joshua A Blosser
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Eric Podolsky
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
15
|
Hanif S, Muhammad P, Chesworth R, Rehman FU, Qian RJ, Zheng M, Shi BY. Nanomedicine-based immunotherapy for central nervous system disorders. Acta Pharmacol Sin 2020; 41:936-953. [PMID: 32467570 PMCID: PMC7468531 DOI: 10.1038/s41401-020-0429-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
Abstract
Central nervous system (CNS) disorders represent a broad spectrum of brain ailments with short- and long-term disabilities, and nanomedicine-based approaches provide a new therapeutic approach to treating CNS disorders. A variety of potential drugs have been discovered to treat several neuronal disorders; however, their therapeutic success can be limited by the presence of the blood-brain barrier (BBB). Furthermore, unique immune functions within the CNS provide novel target mechanisms for the amelioration of CNS diseases. Recently, various therapeutic approaches have been applied to fight brain-related disorders, with moderate outcomes. Among the various therapeutic strategies, nanomedicine-based immunotherapeutic systems represent a new era that can deliver useful cargo with promising pharmacokinetics. These approaches exploit the molecular and cellular targeting of CNS disorders for enhanced safety, efficacy, and specificity. In this review, we focus on the efficacy of nanomedicines that utilize immunotherapy to combat CNS disorders. Furthermore, we detailed summarize nanomedicine-based pathways for CNS ailments that aim to deliver drugs across the BBB by mimicking innate immune actions. Overview of how nanomedicines can utilize multiple immunotherapy pathways to combat CNS disorders. ![]()
Collapse
|
16
|
Dirr ER, Ekhator OR, Blackwood R, Holden JG, Masliah E, Schultheis PJ, Fleming SM. Exacerbation of sensorimotor dysfunction in mice deficient in Atp13a2 and overexpressing human wildtype alpha-synuclein. Behav Brain Res 2018; 343:41-49. [PMID: 29407413 PMCID: PMC5829010 DOI: 10.1016/j.bbr.2018.01.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 12/26/2022]
Abstract
Loss of function mutations in the gene ATP13A2 are associated with Kufor-Rakeb Syndrome and Neuronal Ceroid Lipofuscinosis, the former designated as an inherited form of Parkinson's disease (PD). The function of ATP13A2 is unclear but in vitro studies indicate it is a lysosomal protein and may interact with the presynaptic protein alpha-synuclein (aSyn) and certain heavy metals. Accumulation of aSyn is a major component of lewy bodies, the pathological hallmark of PD. Atp13a2-deficient (13a2) mice develop age-dependent sensorimotor deficits, and accumulation of insoluble aSyn in the brain. To better understand the interaction between ATP13A2 and aSyn, double mutant mice with loss of Atp13a2 function combined with overexpression of human wildtype aSyn were generated. Female and male wildtype (WT), 13a2, aSyn, and 13a2-aSyn mice were tested on a battery of sensorimotor tests including adhesive removal, challenging beam traversal, spontaneous activity, gait, locomotor activity, and nest-building at 2, 4, and 6 months of age. Double mutant mice showed an earlier onset and accelerated alterations in sensorimotor function that were age, sex and test-dependent. Female 13a2-aSyn mice showed early and progressive dysfunction on the beam and in locomotor activity. In males, 13a2-aSyn mice showed more severe impairments in spontaneous activity and adhesive removal. Sex differences were also observed in aSyn and 13a2-aSyn mice on the beam, cylinder, and adhesive removal tests. In other tasks, double mutant mice displayed deficits similar to aSyn mice. These results indicate loss of Atp13a2 function exacerbates the sensorimotor phenotype in aSyn mice in an age and sex-dependent manner.
Collapse
Affiliation(s)
- Emily R Dirr
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | - Osunde R Ekhator
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | - Rachel Blackwood
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | - John G Holden
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | | | | | - Sheila M Fleming
- Department of Pharmaceutical Sciences and Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, USA.
| |
Collapse
|
17
|
Yue P, Gao L, Wang X, Ding X, Teng J. Ultrasound‐triggered effects of the microbubbles coupled to GDNF‐ and Nurr1‐loaded PEGylated liposomes in a rat model of Parkinson's disease. J Cell Biochem 2018; 119:4581-4591. [DOI: 10.1002/jcb.26608] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/07/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Peijian Yue
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Lin Gao
- Department of Neurological Intensive Care UnitThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xuejing Wang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xuebing Ding
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Junfang Teng
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
18
|
Resveratrol and pinostilbene confer neuroprotection against aging-related deficits through an ERK1/2-dependent mechanism. J Nutr Biochem 2017; 54:77-86. [PMID: 29268122 DOI: 10.1016/j.jnutbio.2017.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 09/18/2017] [Accepted: 10/24/2017] [Indexed: 01/02/2023]
Abstract
Age-related declines in motor function may be due, in part, to an increase in oxidative stress in the aging brain leading to dopamine (DA) neuronal cell death. In this study, we examined the neuroprotective effects of natural antioxidants resveratrol and pinostilbene against age-related DAergic cell death and motor dysfunction using SH-SY5Y neuroblastoma cells and young, middle-aged, and old male C57BL/6 mice. Resveratrol and pinostilbene protected SH-SY5Y cells from a DA-induced decrease in cell viability. Dietary supplementation with resveratrol and pinostilbene inhibited the decline of motor function observed with age. While DA and its metabolites (DOPAC and HVA), dopamine transporter, and tyrosine hydroxylase levels remain unchanged during aging or treatment, resveratrol and pinostilbene increased ERK1/2 activation in vitro and in vivo in an age-dependent manner. Inhibition of ERK1/2 in SH-SY5Y cells decreased the protective effects of both compounds. These data suggest that resveratrol and pinostilbene alleviate age-related motor decline via the promotion of DA neuronal survival and activation of the ERK1/2 pathways.
Collapse
|
19
|
Dall'Ara E, Boudiffa M, Taylor C, Schug D, Fiegle E, Kennerley AJ, Damianou C, Tozer GM, Kiessling F, Müller R. Longitudinal imaging of the ageing mouse. Mech Ageing Dev 2016; 160:93-116. [PMID: 27530773 DOI: 10.1016/j.mad.2016.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/30/2016] [Accepted: 08/04/2016] [Indexed: 12/13/2022]
Abstract
Several non-invasive imaging techniques are used to investigate the effect of pathologies and treatments over time in mouse models. Each preclinical in vivo technique provides longitudinal and quantitative measurements of changes in tissues and organs, which are fundamental for the evaluation of alterations in phenotype due to pathologies, interventions and treatments. However, it is still unclear how these imaging modalities can be used to study ageing with mice models. Almost all age related pathologies in mice such as osteoporosis, arthritis, diabetes, cancer, thrombi, dementia, to name a few, can be imaged in vivo by at least one longitudinal imaging modality. These measurements are the basis for quantification of treatment effects in the development phase of a novel treatment prior to its clinical testing. Furthermore, the non-invasive nature of such investigations allows the assessment of different tissue and organ phenotypes in the same animal and over time, providing the opportunity to study the dysfunction of multiple tissues associated with the ageing process. This review paper aims to provide an overview of the applications of the most commonly used in vivo imaging modalities used in mouse studies: micro-computed-tomography, preclinical magnetic-resonance-imaging, preclinical positron-emission-tomography, preclinical single photon emission computed tomography, ultrasound, intravital microscopy, and whole body optical imaging.
Collapse
Affiliation(s)
- E Dall'Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK.
| | - M Boudiffa
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK.
| | - C Taylor
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| | - D Schug
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany.
| | - E Fiegle
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany.
| | - A J Kennerley
- Biological Services Unit, University of Sheffield, Sheffield, UK.
| | - C Damianou
- Department of Electrical Engineering, Cyprus University of Technology, Limassol, Cyprus.
| | - G M Tozer
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK.
| | - F Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany.
| | - R Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
20
|
Benskey MJ, Perez RG, Manfredsson FP. The contribution of alpha synuclein to neuronal survival and function - Implications for Parkinson's disease. J Neurochem 2016; 137:331-59. [PMID: 26852372 PMCID: PMC5021132 DOI: 10.1111/jnc.13570] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/29/2016] [Indexed: 02/06/2023]
Abstract
The aggregation of alpha synuclein (α-syn) is a neuropathological feature that defines a spectrum of disorders collectively termed synucleinopathies, and of these, Parkinson's disease (PD) is arguably the best characterized. Aggregated α-syn is the primary component of Lewy bodies, the defining pathological feature of PD, while mutations or multiplications in the α-syn gene result in familial PD. The high correlation between α-syn burden and PD has led to the hypothesis that α-syn aggregation produces toxicity through a gain-of-function mechanism. However, α-syn has been implicated to function in a diverse range of essential cellular processes such as the regulation of neurotransmission and response to cellular stress. As such, an alternative hypothesis with equal explanatory power is that the aggregation of α-syn results in toxicity because of a toxic loss of necessary α-syn function, following sequestration of functional forms α-syn into insoluble protein aggregates. Within this review, we will provide an overview of the literature linking α-syn to PD and the knowledge gained from current α-syn-based animal models of PD. We will then interpret these data from the viewpoint of the α-syn loss-of-function hypothesis and provide a potential mechanistic model by which loss of α-syn function could result in at least some of the neurodegeneration observed in PD. By providing an alternative perspective on the etiopathogenesis of PD and synucleinopathies, this may reveal alternative avenues of research in order to identify potential novel therapeutic targets for disease modifying strategies. The correlation between α-synuclein burden and Parkinson's disease pathology has led to the hypothesis that α-synuclein aggregation produces toxicity through a gain-of-function mechanism. However, in this review, we discuss data supporting the alternative hypothesis that the aggregation of α-synuclein results in toxicity because of loss of necessary α-synuclein function at the presynaptic terminal, following sequestration of functional forms of α-synuclein into aggregates.
Collapse
Affiliation(s)
- Matthew J Benskey
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Ruth G Perez
- Department of Biomedical Sciences, Center of Emphasis in Neuroscience, Paul L. Foster School of Medicine, Texas Tech University of the Health Sciences El Paso, El Paso, Texas, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA
| |
Collapse
|
21
|
Flores-Cuadrado A, Ubeda-Bañon I, Saiz-Sanchez D, de la Rosa-Prieto C, Martinez-Marcos A. Hippocampal α-synuclein and interneurons in Parkinson's disease: Data from human and mouse models. Mov Disord 2016; 31:979-88. [DOI: 10.1002/mds.26586] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/20/2016] [Accepted: 01/24/2016] [Indexed: 01/15/2023] Open
Affiliation(s)
- Alicia Flores-Cuadrado
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Ciudad Real Spain
| | - Isabel Ubeda-Bañon
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Ciudad Real Spain
| | - Daniel Saiz-Sanchez
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Ciudad Real Spain
| | - Carlos de la Rosa-Prieto
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Ciudad Real Spain
| | - Alino Martinez-Marcos
- Laboratorio de Neuroplasticidad y Neurodegeneración, Facultad de Medicina de Ciudad Real, Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Ciudad Real Spain
| |
Collapse
|
22
|
Van der Perren A, Casteels C, Van Laere K, Gijsbers R, Van den Haute C, Baekelandt V. Development of an Alpha-synuclein Based Rat Model for Parkinson's Disease via Stereotactic Injection of a Recombinant Adeno-associated Viral Vector. J Vis Exp 2016:53670. [PMID: 26967677 PMCID: PMC4828209 DOI: 10.3791/53670] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In order to study the molecular pathways of Parkinson's disease (PD) and to develop novel therapeutic strategies, scientific investigators rely on animal models. The identification of PD-associated genes has led to the development of genetic PD models. Most transgenic α-SYN mouse models develop gradual α-SYN pathology but fail to display clear dopaminergic cell loss and dopamine-dependent behavioral deficits. This hurdle was overcome by direct targeting of the substantia nigra with viral vectors overexpressing PD-associated genes. Local gene delivery using viral vectors provides an attractive way to express transgenes in the central nervous system. Specific brain regions can be targeted (e.g. the substantia nigra), expression can be induced in the adult setting and high expression levels can be achieved. Further, different vector systems based on various viruses can be used. The protocol outlines all crucial steps to perform a viral vector injection in the substantia nigra of the rat to develop a viral vector-based alpha-synuclein animal model for Parkinson's disease.
Collapse
Affiliation(s)
- Anke Van der Perren
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven
| | - Cindy Casteels
- Division of Nuclear Medicine, Leuven University Hospital - KU Leuven
| | - Koen Van Laere
- Division of Nuclear Medicine, Leuven University Hospital - KU Leuven
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven; Leuven Viral Vector Core, KU Leuven
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven; Leuven Viral Vector Core, KU Leuven
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven;
| |
Collapse
|
23
|
Finkelstein DI, Hare DJ, Billings JL, Sedjahtera A, Nurjono M, Arthofer E, George S, Culvenor JG, Bush AI, Adlard PA. Clioquinol Improves Cognitive, Motor Function, and Microanatomy of the Alpha-Synuclein hA53T Transgenic Mice. ACS Chem Neurosci 2016; 7:119-29. [PMID: 26481462 DOI: 10.1021/acschemneuro.5b00253] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The abnormal accumulation of alpha-synuclein (α-syn) has been linked to a number of neurodegenerative disorders, the most noteworthy of which is Parkinson's disease. Alpha-synuclein itself is not toxic and fulfills various physiological roles in the central nervous system. However, specific types of aggregates have been shown to be toxic, and metals have been linked to the assembly of these toxic aggregates. In this paper, we have characterized a transgenic mouse that overexpresses the A53T mutation of human α-syn, specifically assessing cognition, motor performance, and subtle anatomical markers that have all been observed in synucleinopathies in humans. We hypothesized that treatment with the moderate-affinity metal chelator, clioquinol (CQ), would reduce the interaction between metals and α-syn to subsequently improve the phenotype of the A53T animal model. We showed that CQ prevents an iron-synuclein interaction, the formation of urea-soluble α-syn aggregates, α-syn-related substantia nigra pars compacta cell loss, reduction in dendritic spine density of hippocampal and caudate putamen medium spiny neurons, and the decline in motor and cognitive function. In conclusion, our data suggests that CQ is capable of mitigating the pathological metal/α-syn interactions, suggesting that the modulation of metal ions warrants further study as a therapeutic approach for the synucleinopathies.
Collapse
Affiliation(s)
- David I. Finkelstein
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Dominic J. Hare
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
- Elemental
Bio-imaging Facility, University of Technology Sydney, Broadway, New South Wales 2007, Australia
- Senator
Frank R. Lautenberg Environmental Science Laboratory, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jessica L. Billings
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Amelia Sedjahtera
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Milawaty Nurjono
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Elisa Arthofer
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
- Department
of Physiology and Pharmacology, Karolinska Institut, Stockholm SE-171 77, Sweden
| | - Sonia George
- School
of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Janetta G. Culvenor
- School
of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ashley I. Bush
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Paul A. Adlard
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
24
|
Neurobehavioral Anomalies in the Pitx3/ak Murine Model of Parkinson’s Disease and MPTP. Behav Genet 2015; 46:228-41. [DOI: 10.1007/s10519-015-9753-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/23/2015] [Indexed: 01/11/2023]
|
25
|
Khairnar A, Latta P, Drazanova E, Ruda-Kucerova J, Szabó N, Arab A, Hutter-Paier B, Havas D, Windisch M, Sulcova A, Starcuk Z, Rektorova I. Diffusion Kurtosis Imaging Detects Microstructural Alterations in Brain of α-Synuclein Overexpressing Transgenic Mouse Model of Parkinson’s Disease: A Pilot Study. Neurotox Res 2015; 28:281-9. [DOI: 10.1007/s12640-015-9537-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 06/29/2015] [Indexed: 12/12/2022]
|
26
|
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]
|
27
|
The transfection of BDNF to dopamine neurons potentiates the effect of dopamine D3 receptor agonist recovering the striatal innervation, dendritic spines and motor behavior in an aged rat model of Parkinson's disease. PLoS One 2015; 10:e0117391. [PMID: 25693197 PMCID: PMC4332861 DOI: 10.1371/journal.pone.0117391] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/21/2014] [Indexed: 01/28/2023] Open
Abstract
The progressive degeneration of the dopamine neurons of the pars compacta of substantia nigra and the consequent loss of the dopamine innervation of the striatum leads to the impairment of motor behavior in Parkinson's disease. Accordingly, an efficient therapy of the disease should protect and regenerate the dopamine neurons of the substantia nigra and the dopamine innervation of the striatum. Nigral neurons express Brain Derived Neurotropic Factor (BDNF) and dopamine D3 receptors, both of which protect the dopamine neurons. The chronic activation of dopamine D3 receptors by their agonists, in addition, restores, in part, the dopamine innervation of the striatum. Here we explored whether the over-expression of BDNF by dopamine neurons potentiates the effect of the activation of D3 receptors restoring nigrostriatal innervation. Twelve-month old Wistar rats were unilaterally injected with 6-hydroxydopamine into the striatum. Five months later, rats were treated with the D3 agonist 7-hydroxy-N,N-di-n-propy1-2-aminotetralin (7-OH-DPAT) administered i.p. during 4½ months via osmotic pumps and the BDNF gene transfection into nigral cells using the neurotensin-polyplex nanovector (a non-viral transfection) that selectively transfect the dopamine neurons via the high-affinity neurotensin receptor expressed by these neurons. Two months after the withdrawal of 7-OH-DPAT when rats were aged (24 months old), immunohistochemistry assays were made. The over-expression of BDNF in rats receiving the D3 agonist normalized gait and motor coordination; in addition, it eliminated the muscle rigidity produced by the loss of dopamine. The recovery of motor behavior was associated with the recovery of the nigral neurons, the dopamine innervation of the striatum and of the number of dendritic spines of the striatal neurons. Thus, the over-expression of BDNF in dopamine neurons associated with the chronic activation of the D3 receptors appears to be a promising strategy for restoring dopamine neurons in Parkinson's disease.
Collapse
|
28
|
Batelli S, Invernizzi RW, Negro A, Calcagno E, Rodilossi S, Forloni G, Albani D. The Parkinson's Disease-Related Protein DJ-1 Protects Dopaminergic Neurons in vivo and Cultured Cells from Alpha-Synuclein and 6-Hydroxydopamine Toxicity. NEURODEGENER DIS 2014; 15:13-23. [DOI: 10.1159/000367993] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 09/01/2014] [Indexed: 11/19/2022] Open
|
29
|
Flores-Cuadrado A, Ubeda-Bañon I, Saiz-Sanchez D, de la Rosa-Prieto C, Martinez-Marcos A. α-Synuclein staging in the amygdala of a Parkinson's disease model: cell types involved. Eur J Neurosci 2014; 41:137-46. [DOI: 10.1111/ejn.12763] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 09/17/2014] [Accepted: 09/25/2014] [Indexed: 01/15/2023]
Affiliation(s)
- Alicia Flores-Cuadrado
- Laboratorio de Neuroplasticidad y Neurodegeneración; Facultad de Medicina de Ciudad Real; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Avda Moledores S/N 13005 Ciudad Real Spain
| | - Isabel Ubeda-Bañon
- Laboratorio de Neuroplasticidad y Neurodegeneración; Facultad de Medicina de Ciudad Real; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Avda Moledores S/N 13005 Ciudad Real Spain
| | - Daniel Saiz-Sanchez
- Laboratorio de Neuroplasticidad y Neurodegeneración; Facultad de Medicina de Ciudad Real; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Avda Moledores S/N 13005 Ciudad Real Spain
| | - Carlos de la Rosa-Prieto
- Laboratorio de Neuroplasticidad y Neurodegeneración; Facultad de Medicina de Ciudad Real; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Avda Moledores S/N 13005 Ciudad Real Spain
| | - Alino Martinez-Marcos
- Laboratorio de Neuroplasticidad y Neurodegeneración; Facultad de Medicina de Ciudad Real; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha; Avda Moledores S/N 13005 Ciudad Real Spain
| |
Collapse
|
30
|
Delenclos M, Carrascal L, Jensen K, Romero-Ramos M. Immunolocalization of human alpha-synuclein in the Thy1-aSyn ("Line 61") transgenic mouse line. Neuroscience 2014; 277:647-64. [PMID: 25090921 DOI: 10.1016/j.neuroscience.2014.07.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 12/15/2022]
Abstract
Alpha-synuclein (a-syn) is the major component of the intracytoplasmic inclusions known as Lewy bodies (LB), which constitute the hallmark of Parkinson's disease (PD). Mice overexpressing human a-syn under the Thy-1 promoter (ASO) show slow neurodegeneration and some behavioral deficits similar to those seen in human PD patients. Here, we describe a whole-brain distribution of human a-syn in adult ASO mice. We find that the human a-syn is ubiquitously distributed in the brain including the cerebellar cortex, but the intensity and sub-cellular localization of the staining differed in the various regions of the central nervous system. Among particular CNS areas with human a-syn immunoreactivity, we describe staining patterns in the olfactory bulb, cortex, hippocampus, thalamic region, brainstem nuclei and cerebellar cortex. This immunohistochemical study provides an anatomical map of the human a-syn distribution in ASO mice. Our data show that human a-syn, although not present at levels that were detectable by immunostaining in dopaminergic neurons of substantia nigra or noradrenergic neurons of locus coeruleus, was highly expressed in other PD relevant regions of the brain in different neuronal subtypes. These data will help to relate a-syn expression to the phenotypic manifestations observed in this widely used mouse line.
Collapse
Affiliation(s)
- M Delenclos
- Synaptic Physiology Laboratory, Department of Biomedicine, Ole Worms Alle 4, Aarhus University, 8000 C, Denmark; CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Alle 3, Aarhus University, Aarhus 8000 C, Denmark
| | - L Carrascal
- Synaptic Physiology Laboratory, Department of Biomedicine, Ole Worms Alle 4, Aarhus University, 8000 C, Denmark
| | - K Jensen
- Synaptic Physiology Laboratory, Department of Biomedicine, Ole Worms Alle 4, Aarhus University, 8000 C, Denmark; The Lundbeck Foundation Research Center MIND, Department of Biomedicine, Aarhus University, Aarhus 8000 C, Denmark
| | - M Romero-Ramos
- CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Alle 3, Aarhus University, Aarhus 8000 C, Denmark; AU IDEAS Center NEURODIN, Aarhus University, Aarhus 8000 C, Denmark.
| |
Collapse
|
31
|
Stayte S, Vissel B. Advances in non-dopaminergic treatments for Parkinson's disease. Front Neurosci 2014; 8:113. [PMID: 24904259 PMCID: PMC4033125 DOI: 10.3389/fnins.2014.00113] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 04/30/2014] [Indexed: 01/05/2023] Open
Abstract
Since the 1960's treatments for Parkinson's disease (PD) have traditionally been directed to restore or replace dopamine, with L-Dopa being the gold standard. However, chronic L-Dopa use is associated with debilitating dyskinesias, limiting its effectiveness. This has resulted in extensive efforts to develop new therapies that work in ways other than restoring or replacing dopamine. Here we describe newly emerging non-dopaminergic therapeutic strategies for PD, including drugs targeting adenosine, glutamate, adrenergic, and serotonin receptors, as well as GLP-1 agonists, calcium channel blockers, iron chelators, anti-inflammatories, neurotrophic factors, and gene therapies. We provide a detailed account of their success in animal models and their translation to human clinical trials. We then consider how advances in understanding the mechanisms of PD, genetics, the possibility that PD may consist of multiple disease states, understanding of the etiology of PD in non-dopaminergic regions as well as advances in clinical trial design will be essential for ongoing advances. We conclude that despite the challenges ahead, patients have much cause for optimism that novel therapeutics that offer better disease management and/or which slow disease progression are inevitable.
Collapse
Affiliation(s)
- Sandy Stayte
- Neuroscience Department, Neurodegenerative Disorders Laboratory, Garvan Institute of Medical Research, Sydney NSW, Australia ; Faculty of Medicine, University of New South Wales, Sydney NSW, Australia
| | - Bryce Vissel
- Neuroscience Department, Neurodegenerative Disorders Laboratory, Garvan Institute of Medical Research, Sydney NSW, Australia ; Faculty of Medicine, University of New South Wales, Sydney NSW, Australia
| |
Collapse
|
32
|
Abstract
In order to study the molecular pathways of Parkinson's disease (PD) and to develop novel therapeutic strategies, scientific investigators rely on animal models. The identification of PD-associated genes has led to the development of genetic PD models as an alternative to toxin-based models. Viral vector-mediated loco-regional gene delivery provides an attractive way to express transgenes in the central nervous system. Several vector systems based on various viruses have been developed. In this chapter, we give an overview of the different viral vector systems used for targeting the CNS. Further, we describe the different viral vector-based PD models currently available based on overexpression strategies for autosomal dominant genes such as α-synuclein and LRRK2, and knockout or knockdown strategies for autosomal recessive genes, such as parkin, DJ-1, and PINK1. Models based on overexpression of α-synuclein are the most prevalent and extensively studied, and therefore the main focus of this chapter. Many efforts have been made to increase the expression levels of α-synuclein in the dopaminergic neurons. The best α-synuclein models currently available have been developed from a combined approach using newer AAV serotypes and optimized vector constructs, production, and purification methods. These third-generation α-synuclein models show improved face and predictive validity, and therefore offer the possibility to reliably test novel therapeutics.
Collapse
|
33
|
Hennis MR, Seamans KW, Marvin MA, Casey BH, Goldberg MS. Behavioral and neurotransmitter abnormalities in mice deficient for Parkin, DJ-1 and superoxide dismutase. PLoS One 2013; 8:e84894. [PMID: 24386432 PMCID: PMC3873453 DOI: 10.1371/journal.pone.0084894] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/27/2013] [Indexed: 01/10/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease characterized by loss of neurons in the substantia nigra that project to the striatum and release dopamine. The cause of PD remains uncertain, however, evidence implicates mitochondrial dysfunction and oxidative stress. Although most cases of PD are sporadic, 5-10% of cases are caused by inherited mutations. Loss-of-function mutations in Parkin and DJ-1 were the first to be linked to recessively inherited Parkinsonism. Surprisingly, mice bearing similar loss-of-function mutations in Parkin and DJ-1 do not show age-dependent loss of nigral dopaminergic neurons or depletion of dopamine in the striatum. Although the normal cellular functions of Parkin and DJ-1 are not fully understood, we hypothesized that loss-of-function mutations in Parkin and DJ-1 render cells more sensitive to mitochondrial dysfunction and oxidative stress. To test this hypothesis, we crossed mice deficient for Parkin and DJ-1 with mice deficient for the mitochondrial antioxidant protein Mn-superoxide dismutase (SOD2) or the cytosolic antioxidant protein Cu-Zn-superoxide dismutase (SOD1). Aged Parkin-/-DJ-1-/- and Mn-superoxide dismutase triple deficient mice have enhanced performance on the rotorod behavior test. Cu/Zn-superoxide dismutase triple deficient mice have elevated levels of dopamine in the striatum in the absence of nigral cell loss. Our studies demonstrate that on a Parkin/DJ-1 null background, mice that are also deficient for major antioxidant proteins do not have progressive loss of dopaminergic neurons but have behavioral and striatal dopamine abnormalities.
Collapse
Affiliation(s)
- Meghan R. Hennis
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Katherine W. Seamans
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Marian A. Marvin
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Bradford H. Casey
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Matthew S. Goldberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
34
|
Surprising behavioral and neurochemical enhancements in mice with combined mutations linked to Parkinson's disease. Neurobiol Dis 2013; 62:113-23. [PMID: 24075852 DOI: 10.1016/j.nbd.2013.09.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 08/23/2013] [Accepted: 09/17/2013] [Indexed: 11/23/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder behind Alzheimer's disease. There are currently no therapies proven to halt or slow the progressive neuronal cell loss in PD. A better understanding of the molecular and cellular causes of PD is needed to develop disease-modifying therapies. PD is an age-dependent disease that causes the progressive death of dopamine-producing neurons in the brain. Loss of substantia nigra dopaminergic neurons results in locomotor symptoms such as slowness of movement, tremor, rigidity and postural instability. Abnormalities in other neurotransmitters, such as serotonin, may also be involved in both the motor and non-motor symptoms of PD. Most cases of PD are sporadic but many families show a Mendelian pattern of inherited Parkinsonism and causative mutations have been identified in genes such as Parkin, DJ-1, PINK1, alpha-synuclein and leucine rich repeat kinase 2 (LRRK2). Although the definitive causes of idiopathic PD remain uncertain, the activity of the antioxidant enzyme glutathione peroxidase 1 (Gpx1) is reduced in PD brains and has been shown to be a key determinant of vulnerability to dopaminergic neuron loss in PD animal models. Furthermore, Gpx1 activity decreases with age in human substantia nigra but not rodent substantia nigra. Therefore, we crossed mice deficient for both Parkin and DJ-1 with mice deficient for Gpx1 to test the hypothesis that loss-of-function mutations in Parkin and DJ-1 cause PD by increasing vulnerability to Gpx1 deficiency. Surprisingly, mice lacking Parkin, DJ-1 and Gpx1 have increased striatal dopamine levels in the absence of nigral cell loss compared to wild type, Gpx1(-/-), and Parkin(-/-)DJ-1(-/-) mutant mice. Additionally, Parkin(-/-)DJ-1(-/-) mice exhibit improved rotarod performance and have increased serotonin in the striatum and hippocampus. Stereological analysis indicated that the increased serotonin levels were not due to increased serotonergic projections. The results of our behavioral, neurochemical and immunohistochemical analyses reveal that PD-linked mutations in Parkin and DJ-1 cause dysregulation of neurotransmitter systems beyond the nigrostriatal dopaminergic circuit and that loss-of-function mutations in Parkin and DJ-1 lead to adaptive changes in dopamine and serotonin especially in the context of Gpx1 deficiency.
Collapse
|
35
|
Heuer A, Vinh NN, Dunnett SB. Behavioural recovery on simple and complex tasks by means of cell replacement therapy in unilateral 6-hydroxydopamine-lesioned mice. Eur J Neurosci 2013; 37:1691-704. [DOI: 10.1111/ejn.12150] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 01/07/2013] [Accepted: 01/09/2013] [Indexed: 01/16/2023]
Affiliation(s)
- Andreas Heuer
- Brain Repair Group; School of Bioscience; Cardiff University; Cardiff; UK
| | - Ngoc-Nga Vinh
- Brain Repair Group; School of Bioscience; Cardiff University; Cardiff; UK
| | - Stephen B. Dunnett
- Brain Repair Group; School of Bioscience; Cardiff University; Cardiff; UK
| |
Collapse
|
36
|
Arnold A, Naaldijk YM, Fabian C, Wirth H, Binder H, Nikkhah G, Armstrong L, Stolzing A. Reprogramming of Human Huntington Fibroblasts Using mRNA. ACTA ACUST UNITED AC 2012. [DOI: 10.5402/2012/124878] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The derivation of
induced pluripotent stem cells (iPS) from human
cell sources using transduction based on viral
vectors has been reported by several
laboratories. Viral vector-induced integration
is a potential cause of genetic modification. We
have derived iPS cells from human foreskin,
adult Huntington fibroblasts, and adult skin
fibroblasts of healthy donors using a nonviral
and nonintegrating procedure based on mRNA
transfer. In vitro transcribed mRNA for 5
factors, oct-4, nanog, klf-4, c-myc, sox-2 as
well as for one new factor, hTERT, was used to
induce pluripotency. Reprogramming was analyzed
by qPCR analysis of pluripotency gene
expression, differentiation, gene expression
array, and teratoma assays. iPS cells were shown
to express pluripotency markers and were able to
differentiate towards ecto-, endo-, and
mesodermal lineages. This method may represent a
safer technology for reprogramming and
derivation of iPS cells. Cells produced by this
method can more easily be transferred into the
clinical setting.
Collapse
Affiliation(s)
- Antje Arnold
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraβe 1, 04103 Leipzig, Germany
| | - Yahaira M. Naaldijk
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraβe 1, 04103 Leipzig, Germany
| | - Claire Fabian
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Henry Wirth
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, Härtelstraβe 16-18, 04107 Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, Härtelstraβe 16-18, 04107 Leipzig, Germany
| | - Guido Nikkhah
- Department Stereotactic and Functional Neurosurgery, University Hospital of Freiburg, Breisacher Straβe 67, 79106 Freiburg, Germany
| | - Lyle Armstrong
- Institute of Human Genetics, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Alexandra Stolzing
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraβe 1, 04103 Leipzig, Germany
| |
Collapse
|
37
|
Moon J, Lee HS, Kang JM, Park J, Leung A, Hong S, Chung S, Kim KS. Stem cell grafting improves both motor and cognitive impairments in a genetic model of Parkinson's disease, the aphakia (ak) mouse. Cell Transplant 2012; 22:1263-79. [PMID: 23031199 DOI: 10.3727/096368912x657242] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stem cell-based cell replacement of lost midbrain dopamine (mDA) neurons is a potential therapy for Parkinson's disease (PD). Toward this goal, it is critical to optimize various aspects of cell transplantation and to assess functional recovery through behavioral tests in validated animal model(s) of PD. At present, cell transplantation studies are being done almost exclusively in neurotoxin-based animal models, because few genetic models of PD exhibit robust mDA neuronal loss. Here we used a genetic model of PD, the aphakia mouse, which demonstrates selective degeneration of mDA neurons in the substantia nigra. We systematically investigated the functional effects of transplanting embryonic stem cell-derived cells at different stages of in vitro differentiation: embryoid body (EB), neural progenitor (NP), and neuronal differentiated (ND) stages. We found that transplantation of NP cells yielded the best outcomes for both survival and behavioral improvement, while transplantation of EB and ND cells resulted in high teratoma-like tumor formation and poor survival, respectively. In behavioral paradigms specific to basal ganglia, the NP cells group prominently improved motor behavioral defects 1 and 2 months posttransplantation. Furthermore, we found that NP cell transplantation also improved cognitive impairments of aphakia mice, as examined by the passive avoidance task. Importantly, these graft-induced functional improvements well correlated with survival of tyrosine hydroxylase-positive DA neurons. Taken together, we propose that the aphakia mouse can serve as a novel and useful platform for cell transplantation studies to assess both neurological and cognitive improvements and that NP stage cells represent an optimal stage for transplantation.
Collapse
Affiliation(s)
- Jisook Moon
- Molecular Neurobiology Laboratory, McLean Hospital/Harvard Medical School, Belmont, MA 02178, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Noorian AR, Rha J, Annerino DM, Bernhard D, Taylor GM, Greene JG. Alpha-synuclein transgenic mice display age-related slowing of gastrointestinal motility associated with transgene expression in the vagal system. Neurobiol Dis 2012; 48:9-19. [DOI: 10.1016/j.nbd.2012.06.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/27/2012] [Accepted: 06/07/2012] [Indexed: 12/29/2022] Open
|
39
|
Vermeer LMM, Florang VR, Doorn JA. Catechol and aldehyde moieties of 3,4-dihydroxyphenylacetaldehyde contribute to tyrosine hydroxylase inhibition and neurotoxicity. Brain Res 2012; 1474:100-9. [PMID: 22877852 DOI: 10.1016/j.brainres.2012.07.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 07/18/2012] [Accepted: 07/24/2012] [Indexed: 01/09/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder which leads to the selective loss of dopaminergic neurons. This causes a decrease in the important neurotransmitter dopamine (DA), which is essential for coordinated movement. Previous studies have implicated the monoamine oxidase metabolite of DA, 3,4-dihydroxphenylacetaldehyde (DOPAL), in the pathogenesis of PD and have shown it to be a reactive intermediate capable of protein modification. DOPAL also has demonstrated the ability to cause mitochondrial dysfunction and lead to significant inhibition of the rate-limiting enzyme in DA synthesis, tyrosine hydroxylase (TH). The current study was undertaken to investigate four analogs of DOPAL, including a novel nitrile analog, to determine how the structure of DOPAL is related to its toxicity and inhibition of TH. Both mitochondrial function and inhibition of TH in cell lysate were investigated. Furthermore, a novel whole cell assay was designed to determine the consequence to enzyme action when DOPAL levels were elevated. The results presented here demonstrate that changes to DOPAL structure lead to a decrease in toxicity and inhibition of enzyme activity as compared to the parent compound. Furthermore, the production of superoxide anion but not hydrogen peroxide increased in the presence of elevated DOPAL. These results reveal the toxicity of DOPAL and demonstrate that both the catechol and aldehyde are required to potently inhibit TH activity.
Collapse
Affiliation(s)
- Lydia M M Vermeer
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, The University of Iowa, 115 South Grand Ave., Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
40
|
de Almeida AM, Bendixen E. Pig proteomics: A review of a species in the crossroad between biomedical and food sciences. J Proteomics 2012; 75:4296-314. [DOI: 10.1016/j.jprot.2012.04.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 04/04/2012] [Accepted: 04/08/2012] [Indexed: 11/29/2022]
|
41
|
Overexpression of parkin ameliorates dopaminergic neurodegeneration induced by 1- methyl-4-phenyl-1,2,3,6-tetrahydropyridine in mice. PLoS One 2012; 7:e39953. [PMID: 22792139 PMCID: PMC3390003 DOI: 10.1371/journal.pone.0039953] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 05/29/2012] [Indexed: 01/23/2023] Open
Abstract
Mutations in the parkin gene are currently thought to be the most common cause of recessive familial Parkinsonism. Parkin functions as an E3 ligase to regulate protein turnover, and its function in mitochondrial quality control has been reported recently. Overexpression of parkin has been found to prevent neuronal degeneration under various conditions both in vivo and in vitro. Here, we generated a transgenic mouse model in which expression of wild type parkin was driven by neuron-specific enolase (NSE) promoter. We reported that both young and old parkin transgenic mice exhibited less reduction of striatal TH protein and number of TH positive neurons in the substantia nigra induced by 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP), compared to wild type littermates. MPTP-induced mitochondrial impairment in the substantia nigra was improved in young parkin transgenic mice. Decreased striatal α-synuclein was demonstrated in old parkin transgenic mice. These results provide reliable evidence from the transgenic mouse model for parkin that overexpression of parkin may attenuate dopaminergic neurodegeneration induced by MPTP through protection of mitochondria and reduction of α-synuclein in the nigrostriatal pathway.
Collapse
|
42
|
The emerging role of proteolysis in mitochondrial quality control and the etiology of Parkinson's disease. PARKINSONS DISEASE 2012; 2012:382175. [PMID: 22666630 PMCID: PMC3359724 DOI: 10.1155/2012/382175] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/19/2012] [Indexed: 12/15/2022]
Abstract
Mitochondria are highly dynamic organelles that are important for many diverse cellular processes, such as energy metabolism, calcium buffering, and apoptosis. Mitochondrial biology and dysfunction have recently been linked to different types of cancers and neurodegenerative diseases, most notably Parkinson's disease. Thus, a better understanding of the quality control systems that maintain a healthy mitochondrial network can facilitate the development of effective treatments for these diseases. In this perspective, we will discuss recent advances on two mitochondrial quality control pathways: the UPS and mitophagy, highlight how new players may be contributing to regulate these pathways. We believe the proteases involved will be key and novel regulators of mitochondrial quality control, and this knowledge will provide insights into future studies aimed to combat neurodegenerative diseases.
Collapse
|
43
|
The neurobehavior ontology: an ontology for annotation and integration of behavior and behavioral phenotypes. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012. [PMID: 23195121 DOI: 10.1016/b978-0-12-388408-4.00004-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In recent years, considerable advances have been made toward our understanding of the genetic architecture of behavior and the physical, mental, and environmental influences that underpin behavioral processes. The provision of a method for recording behavior-related phenomena is necessary to enable integrative and comparative analyses of data and knowledge about behavior. The neurobehavior ontology facilitates the systematic representation of behavior and behavioral phenotypes, thereby improving the unification and integration behavioral data in neuroscience research.
Collapse
|
44
|
Glajch KE, Fleming SM, Surmeier DJ, Osten P. Sensorimotor assessment of the unilateral 6-hydroxydopamine mouse model of Parkinson's disease. Behav Brain Res 2011; 230:309-16. [PMID: 22178078 DOI: 10.1016/j.bbr.2011.12.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/30/2011] [Accepted: 12/04/2011] [Indexed: 12/30/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by marked impairments in motor function caused by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNc). Animal models of PD have traditionally been based on toxins, such as 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), that selectively lesion dopaminergic neurons. Motor impairments from 6-OHDA lesions of SNc neurons are well characterized in rats, but much less work has been done in mice. In this study, we compare the effectiveness of a series of drug-free behavioral tests in assessing sensorimotor impairments in the unilateral 6-OHDA mouse model, including six tests used for the first time in this PD mouse model (the automated treadmill "DigiGait" test, the challenging beam test, the adhesive removal test, the pole test, the adjusting steps test, and the test of spontaneous activity) and two tests used previously in 6-OHDA-lesioned mice (the limb-use asymmetry "cylinder" test and the manual gait test). We demonstrate that the limb-use asymmetry, challenging beam, pole, adjusting steps, and spontaneous activity tests are all highly robust assays for detecting sensorimotor impairments in the 6-OHDA mouse model. We also discuss the use of the behavioral tests for specific experimental objectives, such as simple screening for well-lesioned mice in studies of PD cellular pathophysiology or comprehensive behavioral analysis in preclinical therapeutic testing using a battery of sensorimotor tests.
Collapse
Affiliation(s)
- Kelly E Glajch
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60610, USA.
| | | | | | | |
Collapse
|
45
|
Beeler JA. Preservation of function in Parkinson's disease: what's learning got to do with it? Brain Res 2011; 1423:96-113. [PMID: 22000081 DOI: 10.1016/j.brainres.2011.09.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 08/06/2011] [Accepted: 09/20/2011] [Indexed: 01/16/2023]
Abstract
Dopamine denervation gives rise to abnormal corticostriatal plasticity; however, its role in the symptoms and progression of Parkinson's disease (PD) has not been articulated or incorporated into current clinical models. The 'integrative selective gain' framework proposed here integrates dopaminergic mechanisms known to modulate basal ganglia throughput into a single conceptual framework: (1) synaptic weights, the neural instantiation of accumulated experience and skill modulated by dopamine-dependent plasticity and (2) system gain, the operating parameters of the basal ganglia, modulated by dopamine's on-line effects on cell excitability, glutamatergic transmission and the balance between facilitatory and inhibitory pathways. Within this framework and based on recent work, a hypothesis is presented that prior synaptic weights and established skills can facilitate motor performance and preserve function despite diminished dopamine; however, dopamine denervation induces aberrant corticostriatal plasticity that degrades established synaptic weights and replaces them with inappropriate, inhibitory learning that inverts the function of the basal ganglia resulting in 'anti-optimization' of motor performance. Consequently, mitigating aberrant corticostriatal plasticity represents an important therapeutic objective, as reflected in the long-duration response to levodopa, reinterpreted here as the correction of aberrant learning. It is proposed that viewing aberrant corticostriatal plasticity and learning as a provisional endophenotype of PD would facilitate investigation of this hypothesis.
Collapse
Affiliation(s)
- Jeff A Beeler
- Department of Neurobiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
46
|
Ubeda-Bañon I, Saiz-Sanchez D, de la Rosa-Prieto C, Martinez-Marcos A. α-Synuclein in the olfactory system of a mouse model of Parkinson’s disease: correlation with olfactory projections. Brain Struct Funct 2011; 217:447-58. [DOI: 10.1007/s00429-011-0347-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 08/30/2011] [Indexed: 10/17/2022]
|
47
|
Efficient and stable transduction of dopaminergic neurons in rat substantia nigra by rAAV 2/1, 2/2, 2/5, 2/6.2, 2/7, 2/8 and 2/9. Gene Ther 2011; 18:517-27. [PMID: 21326331 DOI: 10.1038/gt.2010.179] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dysfunction of the nigrostriatal system is the major cause of Parkinson's disease (PD). This brain region is therefore an important target for gene delivery aiming at disease modeling and gene therapy. Recombinant adeno-associated viral (rAAV) vectors have been developed as efficient vehicles for gene transfer into the central nervous system. Recently, several serotypes have been described, with varying tropism for brain transduction. In light of the further development of a viral vector-mediated rat model for PD, we performed a comprehensive comparison of the transduction and tropism for dopaminergic neurons (DNs) in the adult Wistar rat substantia nigra (SN) of seven rAAV vector serotypes (rAAV 2/1, 2/2, 2/5, 2/6.2, 2/7, 2/8 and 2/9). All vectors were normalized by titer and volume, and stereotactically injected into the SN. Gene expression was assessed non-invasively and quantitatively in vivo by bioluminescence imaging at 2 and 5 weeks after injection, and was found to be stable over time. Immunohistochemistry at 6 weeks following injection revealed the most widespread enhanced green fluorescence protein expression and the highest number of positive nigral cells using rAAV 2/7, 2/9 and 2/1. The area transduced by rAAV 2/8 was smaller, but nevertheless almost equal numbers of nigral cells were targeted. Detailed confocal analysis revealed that serotype 2/7, 2/9, 2/1 and 2/8 transduced at least 70% of the DNs. In conclusion, these results show that various rAAV serotypes efficiently transduce nigral DNs, but significant differences in transgene expression pattern and level were observed.
Collapse
|
48
|
Inden M, Kitamura Y, Abe M, Tamaki A, Takata K, Taniguchi T. Parkinsonian Rotenone Mouse Model: Reevaluation of Long-Term Administration of Rotenone in C57BL/6 Mice. Biol Pharm Bull 2011; 34:92-6. [DOI: 10.1248/bpb.34.92] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Mari Abe
- Department of Neurobiology, Kyoto Pharmaceutical University
| | - Aya Tamaki
- Department of Neurobiology, Kyoto Pharmaceutical University
| | | | | |
Collapse
|
49
|
Panneton WM, Kumar VB, Gan Q, Burke WJ, Galvin JE. The neurotoxicity of DOPAL: behavioral and stereological evidence for its role in Parkinson disease pathogenesis. PLoS One 2010; 5:e15251. [PMID: 21179455 PMCID: PMC3001493 DOI: 10.1371/journal.pone.0015251] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 11/05/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The etiology of Parkinson disease (PD) has yet to be fully elucidated. We examined the consequences of injections of 3,4-dihydroxyphenylacetaldehyde (DOPAL), a toxic metabolite of dopamine, into the substantia nigra of rats on motor behavior and neuronal survival. METHODS/PRINCIPAL FINDINGS A total of 800 nl/rat of DOPAL (1 µg/200 nl) was injected stereotaxically into the substantia nigra over three sites while control animals received similar injections of phosphate buffered saline. Rotational behavior of these rats was analyzed, optical density of striatal tyrosine hydroxylase was calculated, and unbiased stereological counts of the substantia nigra were made. The rats showed significant rotational asymmetry ipsilateral to the lesion, supporting disruption of dopaminergic nigrostriatal projections. Such disruption was verified since the density of striatal tyrosine hydroxylase decreased significantly (p<0.001) on the side ipsilateral to the DOPAL injections when compared to the non-injected side. Stereological counts of neurons stained for Nissl in pars compacta of the substantia nigra significantly decreased (p<0.001) from control values, while counts of those in pars reticulata were unchanged after DOPAL injections. Counts of neurons immunostained for tyrosine hydroxylase also showed a significant (p=0.032) loss of dopaminergic neurons. In spite of significant loss of dopaminergic neurons, DOPAL injections did not induce significant glial reaction in the substantia nigra. CONCLUSIONS The present study provides the first in vivo quantification of substantia nigra pars compacta neuronal loss after injection of the endogenous toxin DOPAL. The results demonstrate that injections of DOPAL selectively kills SN DA neurons, suggests loss of striatal DA terminals, spares non-dopaminergic neurons of the pars reticulata, and triggers a behavioral phenotype (rotational asymmetry) consistent with other PD animal models. This study supports the "catecholaldehyde hypothesis" as an important link for the etiology of sporadic PD.
Collapse
Affiliation(s)
- W Michael Panneton
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America.
| | | | | | | | | |
Collapse
|
50
|
Planken A, Porokuokka L, Hänninen AL, Tuominen R, Andressoo JO. Medium-throughput computer aided micro-island method to assay embryonic dopaminergic neuron cultures in vitro. J Neurosci Methods 2010; 194:122-31. [DOI: 10.1016/j.jneumeth.2010.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/13/2010] [Accepted: 10/07/2010] [Indexed: 01/13/2023]
|