1
|
Li H, Mei L, Nie X, Wu L, Lv L, Ren X, Yang J, Cao H, Wu J, Zhang Y, Hu Y, Wang W, Turck CW, Shi B, Li J, Xu L, Hu X. The Tree Shrew Model of Parkinson Disease: A Cost-Effective Alternative to Nonhuman Primate Models. J Transl Med 2024; 104:102145. [PMID: 39343009 DOI: 10.1016/j.labinv.2024.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
The surge in demand for experimental monkeys has led to a rapid increase in their costs. Consequently, there is a growing need for a cost-effective model of Parkinson disease (PD) that exhibits all core clinical and pathologic phenotypes. Evolutionarily, tree shrews (Tupaia belangeri) are closer to primates in comparison with rodents and could be an ideal species for modeling PD. To develop a tree shrew PD model, we used the 1-methyl-4-phenylpyridinium (MPP+), a metabolite derived from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, to induce lesions in dopaminergic neurons of the unilateral substantia nigra. The induced tree shrew model consistently exhibited and maintained all classic clinical manifestations of PD for a 5-month period. The symptoms included bradykinesia, rest tremor, and postural instability, and ∼50% individuals showed apomorphine-induced rotations, a classic phenotype of unilateral PD models. All these are closely resembled the ones observed in PD monkeys. Meanwhile, this model was also sensitive to L-dopa treatment in a dose-dependent manner, which suggested that the motor deficits are dopamine dependent. Immunostaining showed a significant loss of dopaminergic neurons (∼95%) in the lesioned substantia nigra, which is a crucial PD pathological marker. Moreover, a control group of nigral saline injection did not show any motor deficits and pathological changes. Cytomorphologic analysis revealed that the size of nigral dopaminergic neurons in tree shrews is much bigger than that of rodents and is close to that of macaques. The morphologic similarity may be an important structural basis for the manifestation of the highly similar phenotypes between monkey and tree shrew PD models. Collectively, in this study, we have successfully developed a PD model in a small animal species that faithfully recapitulated the classic clinical symptoms and key pathological indicators of PD monkeys, providing a novel and low-cost avenue for evaluation of PD treatments and underlying mechanisms.
Collapse
Affiliation(s)
- Hao Li
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Leyi Mei
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Institutes of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Xiupeng Nie
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Liping Wu
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Longbao Lv
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Xiaofeng Ren
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Jitong Yang
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Haonan Cao
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jing Wu
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yuhua Zhang
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingzhou Hu
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenchao Wang
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Christoph W Turck
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Max Planck Institute of Psychiatry, Munich, Germany.
| | - Bingyin Shi
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Department of Neurology, Hackensack Meridian School of Medicine, Nutley, New Jersey.
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | - Xintian Hu
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
2
|
Li J, Li N, Wei J, Feng C, Chen Y, Chen T, Ai Z, Zhu X, Ji W, Li T. Genetically engineered mesenchymal stem cells with dopamine synthesis for Parkinson's disease in animal models. NPJ Parkinsons Dis 2022; 8:175. [PMID: 36550118 PMCID: PMC9780305 DOI: 10.1038/s41531-022-00440-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Although striatal delivery of three critical genes for dopamine synthesis by viruses is a potential clinical approach for treating Parkinson's disease (PD), the approach makes it difficult to finely control dopamine secretion amounts and brings safety concerns. Here, we generate genetically engineered mesenchymal stem cells encoding three critical genes for dopamine synthesis (DOPA-MSCs). DOPA-MSCs retain their MSC identity and stable ability to secrete dopamine during passaging. Following transplantation, DOPA-MSCs reinstate striatal dopamine levels and correct motor function in PD rats. Importantly, after grafting into the caudate and putamen, DOPA-MSCs provide homotopic reconstruction of midbrain dopamine pathways by restoring striatal dopamine levels, and safely and long-term (up to 51 months) correct motor disorders and nonmotor deficits in acute and chronic PD rhesus monkey models of PD even with advanced PD symptoms. The long-term benefits and safety results support the idea that the development of dopamine-synthesized engineered cell transplantation is an important strategy for treating PD.
Collapse
Affiliation(s)
- Jun Li
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Nan Li
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Jingkuan Wei
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Chun Feng
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Yanying Chen
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Tingwei Chen
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Zongyong Ai
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Xiaoqing Zhu
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Weizhi Ji
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| | - Tianqing Li
- grid.218292.20000 0000 8571 108XState Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, 650500 Kunming, Yunnan China ,grid.218292.20000 0000 8571 108XYunnan Key Laboratory of Primate Biomedical Research, 650500 Kunming, Yunnan China
| |
Collapse
|
3
|
Darbin O, Hatanaka N, Takara S, Kaneko N, Chiken S, Naritoku D, Martino A, Nambu A. Subthalamic nucleus deep brain stimulation driven by primary motor cortex γ2 activity in parkinsonian monkeys. Sci Rep 2022; 12:6493. [PMID: 35444245 PMCID: PMC9021287 DOI: 10.1038/s41598-022-10130-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
In parkinsonism, subthalamic nucleus (STN) electrical deep brain stimulation (DBS) improves symptoms, but may be associated with side effects. Adaptive DBS (aDBS), which enables modulation of stimulation, may limit side effects, but limited information is available about clinical effectiveness and efficaciousness. We developed a brain-machine interface for aDBS, which enables modulation of stimulation parameters of STN-DBS in response to γ2 band activity (80-200 Hz) of local field potentials (LFPs) recorded from the primary motor cortex (M1), and tested its effectiveness in parkinsonian monkeys. We trained two monkeys to perform an upper limb reaching task and rendered them parkinsonian with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Bipolar intracortical recording electrodes were implanted in the M1, and a recording chamber was attached to access the STN. In aDBS, the M1 LFPs were recorded, filtered into the γ2 band, and discretized into logic pulses by a window discriminator, and the pulses were used to modulate the interval and amplitude of DBS pulses. In constant DBS (cDBS), constant stimulus intervals and amplitudes were used. Reaction and movement times during the task were measured and compared between aDBS and cDBS. The M1-γ2 activities were increased before and during movements in parkinsonian monkeys and these activities modulated the aDBS pulse interval, amplitude, and dispersion. With aDBS and cDBS, reaction and movement times were significantly decreased in comparison to DBS-OFF. The electric charge delivered was lower with aDBS than cDBS. M1-γ2 aDBS in parkinsonian monkeys resulted in clinical benefits that did not exceed those from cDBS. However, M1-γ2 aDBS achieved this magnitude of benefit for only two thirds of the charge delivered by cDBS. In conclusion, M1-γ2 aDBS is an effective therapeutic approach which requires a lower electrical charge delivery than cDBS for comparable clinical benefits.
Collapse
Affiliation(s)
- Olivier Darbin
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan. .,Department of Neurology, University South Alabama College of Medicine, 307 University Blvd, Mobile, AL, 36688, USA.
| | - Nobuhiko Hatanaka
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Sayuki Takara
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan.,Department of Physiology, Faculty of Medecine, Kindai University, Osaka-Sayama, Osaka, Japan
| | - Nobuya Kaneko
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Dean Naritoku
- Department of Neurology, University South Alabama College of Medicine, 307 University Blvd, Mobile, AL, 36688, USA
| | - Anthony Martino
- Department of Neurosurgery, University South Alabama College of Medicine, Mobile, AL, USA
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan. .,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan.
| |
Collapse
|
4
|
Li H, Su LY, Yang L, Li M, Liu Q, Li Z, Hu Y, Li H, Wu S, Wang W, Hu Y, Wang Z, Rizak JD, Huang B, Xu M, Wu J, Lv LB, Turck CW, Yin Y, Yao YG, Su B, Hu X. A cynomolgus monkey with naturally occurring Parkinson's disease. Natl Sci Rev 2021; 8:nwaa292. [PMID: 34691601 PMCID: PMC8288342 DOI: 10.1093/nsr/nwaa292] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Hao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Ling-Yan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Lixin Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Min Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Zhenhui Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Yan Hu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Hongwei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Shihao Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Wenchao Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Yingzhou Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Zhengbo Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Joshua D Rizak
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Baihui Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Jing Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Long-Bao Lv
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Christoph W Turck
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Yong Yin
- Department of Rehabilitation Medicine, the Second People's Hospital of Yunnan Province, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| |
Collapse
|
5
|
Li H, Wu S, Ma X, Li X, Cheng T, Chen Z, Wu J, Lv L, Li L, Xu L, Wang W, Hu Y, Jiang H, Yin Y, Qiu Z, Hu X. Co-editing PINK1 and DJ-1 Genes Via Adeno-Associated Virus-Delivered CRISPR/Cas9 System in Adult Monkey Brain Elicits Classical Parkinsonian Phenotype. Neurosci Bull 2021; 37:1271-1288. [PMID: 34165772 PMCID: PMC8423927 DOI: 10.1007/s12264-021-00732-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
Whether direct manipulation of Parkinson's disease (PD) risk genes in the adult monkey brain can elicit a Parkinsonian phenotype remains an unsolved issue. Here, we used an adeno-associated virus serotype 9 (AAV9)-delivered CRISPR/Cas9 system to directly co-edit PINK1 and DJ-1 genes in the substantia nigras (SNs) of two monkey groups: an old group and a middle-aged group. After the operation, the old group exhibited all the classic PD symptoms, including bradykinesia, tremor, and postural instability, accompanied by key pathological hallmarks of PD, such as severe nigral dopaminergic neuron loss (>64%) and evident α-synuclein pathology in the gene-edited SN. In contrast, the phenotype of their middle-aged counterparts, which also showed clear PD symptoms and pathological hallmarks, were less severe. In addition to the higher final total PD scores and more severe pathological changes, the old group were also more susceptible to gene editing by showing a faster process of PD progression. These results suggested that both genetic and aging factors played important roles in the development of PD in the monkeys. Taken together, this system can effectively develop a large number of genetically-edited PD monkeys in a short time (6-10 months), and thus provides a practical transgenic monkey model for future PD studies.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
| | - Shihao Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xia Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Xiao Li
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tianlin Cheng
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhifang Chen
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jing Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
| | - Longbao Lv
- National Resource Center for Non-human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, 650107, Kunming, China
| | - Ling Li
- Diagnostic Radiology Department, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
| | - Liqi Xu
- Ultrasound diagnosis Department, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
| | - Wenchao Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
| | - Yingzhou Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
| | - Haisong Jiang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yong Yin
- Department of Rehabilitation Medicine, The Second People's Hospital of Yunnan Province, Kunming, 650021, China.
| | - Zilong Qiu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200433, China.
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- National Resource Center for Non-human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, 650107, Kunming, China.
| |
Collapse
|
6
|
Chiken S, Takada M, Nambu A. Altered Dynamic Information Flow through the Cortico-Basal Ganglia Pathways Mediates Parkinson's Disease Symptoms. Cereb Cortex 2021; 31:5363-5380. [PMID: 34268560 PMCID: PMC8568006 DOI: 10.1093/cercor/bhab164] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder caused by dopamine deficiency. To elucidate network-level changes through the cortico-basal ganglia pathways in PD, we recorded neuronal activity in PD monkeys treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. We applied electrical stimulation to the motor cortices and examined responses in the internal (GPi) and external (GPe) segments of the globus pallidus, the output and relay nuclei of the basal ganglia, respectively. In the normal state, cortical stimulation induced a triphasic response composed of early excitation, inhibition, and late excitation in the GPi and GPe. In the PD state, cortically evoked inhibition in the GPi mediated by the cortico-striato-GPi “direct” pathway was largely diminished, whereas late excitation in the GPe mediated by the cortico-striato-GPe-subthalamo (STN)-GPe pathway was elongated. l-DOPA treatment ameliorated PD signs, particularly akinesia/bradykinesia, and normalized cortically evoked responses in both the GPi and GPe. STN blockade by muscimol injection ameliorated the motor deficit and unmasked cortically evoked inhibition in the GPi. These results suggest that information flow through the direct pathway responsible for the initiation of movements is largely reduced in PD and fails to release movements, resulting in akinesia/bradykinesia. Restoration of the information flow through the direct pathway recovers execution of voluntary movements.
Collapse
Affiliation(s)
- Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI, Myodaiji, Okazaki 444-8585, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI, Myodaiji, Okazaki 444-8585, Japan
| |
Collapse
|
7
|
Pingale T, Gupta GL. Classic and evolving animal models in Parkinson's disease. Pharmacol Biochem Behav 2020; 199:173060. [PMID: 33091373 DOI: 10.1016/j.pbb.2020.173060] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and deficiency of dopamine in the striatal region. The primary objective in PD research is to understand the pathogenesis, targets, and development of therapeutic interventions to control the progress of the disease. The anatomical and physiological resemblances between humans and animals gathered the researcher's attention towards the use of animals in PD research. Due to varying age of onset, symptoms, and progression rate, PD becomes heterogeneous which demands the variety of animal models to study diverse features of the disease. Parkinson is a multifactorial disorder, selection of models become important as not a single model shows all the biochemical features of the disease. Currently, conventional pharmacological, neurotoxin-induced, genetically modified and cellular models are available for PD research, but none of them recapitulate all the biochemical characteristics of the disease. In this review, we included the updated knowledge on the main features of currently available in vivo and in vitro models as well as their strengths and weaknesses.
Collapse
Affiliation(s)
- Tanvi Pingale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| | - Girdhari Lal Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India; School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India.
| |
Collapse
|
8
|
Darbin O, Hatanaka N, Takara S, Kaneko N, Chiken S, Naritoku D, Martino A, Nambu A. Parkinsonism Differently Affects the Single Neuronal Activity in the Primary and Supplementary Motor Areas in Monkeys: An Investigation in Linear and Nonlinear Domains. Int J Neural Syst 2020; 30:2050010. [PMID: 32019380 DOI: 10.1142/s0129065720500100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The changes in neuronal firing activity in the primary motor cortex (M1) and supplementary motor area (SMA) were compared in monkeys rendered parkinsonian by treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The neuronal dynamic was characterized using mathematical tools defined in different frameworks (rate, oscillations or complex patterns). Then, and for each cortical area, multivariate and discriminate analyses were further performed on these features to identify those important to differentiate between the normal and the pathological neuronal activity. Our results show a different order in the importance of the features to discriminate the pathological state in each cortical area which suggests that the M1 and the SMA exhibit dissimilarities in their neuronal alterations induced by parkinsonism. Our findings highlight the need for multiple mathematical frameworks to best characterize the pathological neuronal activity related to parkinsonism. Future translational studies are warranted to investigate the causal relationships between cortical region-specificities, dominant pathological hallmarks and symptoms.
Collapse
Affiliation(s)
- Olivier Darbin
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Nobuhiko Hatanaka
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Sayuki Takara
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Nobuya Kaneko
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Dean Naritoku
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Anthony Martino
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
9
|
Darbin O, Hatanaka N, Takara S, Kaneko M, Chiken S, Naritoku D, Martino A, Nambu A. Local field potential dynamics in the primate cortex in relation to parkinsonism reveled by machine learning: A comparison between the primary motor cortex and the supplementary area. Neurosci Res 2020; 156:66-79. [PMID: 31991205 DOI: 10.1016/j.neures.2020.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/09/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022]
Abstract
The present study compares the cortical local field potentials (LFPs) in the primary motor cortex (M1) and the supplementary motor area (SMA) of non-human primates rendered Parkinsonian with administration of dopaminergic neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The dynamic of the LFPs was investigated under several mathematical frameworks and machine learning was used to discriminate the recordings based on these features between healthy, parkinsonian with off-medication and parkinsonian with on-medication states. The importance of each feature in the discrimination process was further investigated. The dynamic of the LFPs in M1 and SMA was affected regarding its variability (time domain analysis), oscillatory activities (frequency domain analysis) and complex patterns (non-linear domain analysis). Machine learning algorithms achieved accuracy near 0.90 for comparisons between conditions. The TreeBagger algorithm provided best accuracy. The relative importance of these features differed with the cortical location, condition and treatment. Overall, the most important features included beta oscillation, fractal dimension, gamma oscillation, entropy and asymmetry of amplitude fluctuation. The importance of features in discriminating between normal and pathological states, and on- or off-medication states depends on the pair-comparison and it is region-specific. These findings are discussed regarding the refinement of current models for movement disorders and the development of on-demand therapies.
Collapse
Affiliation(s)
- Olivier Darbin
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA.
| | - Nobuhiko Hatanaka
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Sayuki Takara
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Masaya Kaneko
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Dean Naritoku
- Department of Neurology, University South Alabama, 307 University Blvd, Mobile, AL 36688, USA
| | - Anthony Martino
- Department of Neurosurgery, University South Alabama, 307 University Blvd., Mobile, AL 36688, USA
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
10
|
Borgognon S, Cottet J, Moret V, Chatagny P, Carrara L, Fregosi M, Bloch J, Brunet JF, Rouiller EM, Badoud S. Fine Manual Dexterity Assessment After Autologous Neural Cell Ecosystem (ANCE) Transplantation in a Non-human Primate Model of Parkinson's Disease. Neurorehabil Neural Repair 2019; 33:553-567. [PMID: 31170868 DOI: 10.1177/1545968319850133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background. Autologous neural cell ecosystem (ANCE) transplantation improves motor recovery in MPTP monkeys. These motor symptoms were assessed using semi-quantitative clinical rating scales, widely used in many studies. However, limitations in terms of sensitivity, combined with relatively subjective assessment of their different items, make inter-study comparisons difficult to achieve. Objective. The aim of this study was to quantify the impact of MPTP intoxication in macaque monkeys on manual dexterity and assess whether ANCE can contribute to functional recovery. Methods. Four animals were trained to perform 2 manual dexterity tasks. After reaching a motor performance plateau, the animals were subjected to an MPTP lesion. After the occurrence of a spontaneous functional recovery plateau, all 4 animals were subjected to ANCE transplantation. Results. Two of 4 animals underwent a full spontaneous recovery before the ANCE transplantation, whereas the 2 other animals (symptomatic) presented moderate to severe Parkinson's disease (PD)-like symptoms affecting manual dexterity. The time to grasp small objects using the precision grip increased in these 2 animals. After ANCE transplantation, the 2 symptomatic animals underwent a significant functional recovery, reflected by a decrease in time to execute the different tasks, as compared with the post-lesion phase. Conclusions. Manual dexterity is affected in symptomatic MPTP monkeys. The 2 manual dexterity tasks reported here as pilot are pertinent to quantify PD symptoms and reliably assess a treatment in MPTP monkeys, such as the present ANCE transplantation, to be confirmed in a larger cohort of animals before future clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jocelyne Bloch
- 2 Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | | | | |
Collapse
|
11
|
Seo J, Lee Y, Kim BS, Park J, Yang S, Yoon HJ, Yoo J, Park HS, Hong JJ, Koo BS, Baek SH, Jeon CY, Huh JW, Kim YH, Park SJ, Won J, Ahn YJ, Kim K, Jeong KJ, Kang P, Lee DS, Lim SM, Jin YB, Lee SR. A non-human primate model for stable chronic Parkinson's disease induced by MPTP administration based on individual behavioral quantification. J Neurosci Methods 2018; 311:277-287. [PMID: 30391524 DOI: 10.1016/j.jneumeth.2018.10.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND The guidelines for applying individual adjustments to macaques according to the severity of behavioral symptoms during 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment were provided to reproduce stable chronic Parkinsonism in a recent study (Potts et al., 2014). But, since there are insufficient guidelines regarding objective severity criteria of individual symptoms for adjustments of MPTP treatment, it is difficult to develop MPTP-induced chronic non-human primate (NHP) models with stable symptoms. NEW METHOD The individual adjustments of MPTP administration based on results of automatic quantification of global activity (GA) using a video-based tracking system were applied to develop MPTP-PD model. Low-dose (0.2 mg/kg) intramuscular injection was repeated continuously until GA was lower than 8% of baseline Parkinsonian behavior scores. The positron emission tomography imaging were used to follow the longitudinal course of Parkinson's disease (PD). RESULTS Significant reductions in GA and dopamine transporter activity, along with significant increases in Parkinsonian behavior scores were found from 4 to 48 weeks following the first administration. GA was correlated with the Parkinsonian behavior score. The dopamine transporter activity was correlated with GA and the Parkinsonian behavior score. However, it was not correlated with the total dose of MPTP. Damage of dopaminergic neuronal systems in the basal ganglia was confirmed by immunohistochemistry and Western blot. COMPARISON WITH EXISTING METHOD This study reinforces previous guidelines regarding production of NHP models with stable Parkinsonian symptoms. CONCLUSIONS This novel strategy of MPTP administration based on global activity evaluations provides an important conceptual advance for the development of chronic NHP Parkinsonian models.
Collapse
Affiliation(s)
- Jincheol Seo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Bom Sahn Kim
- Department of Nuclear medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Sejung Yang
- Department of Biomedical Engineering, Yonsei University, Wonju 220-710, Republic of Korea
| | - Hai-Jeon Yoon
- Department of Nuclear medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Jang Yoo
- Department of Nuclear medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Hyun Soo Park
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jung-Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Bon-Sang Koo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Seung Ho Baek
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Sang Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Yu-Jin Ahn
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Keonwoo Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Physical Therapy, Graduate School of Inje University, Gimhae, Republic of Korea
| | - Kang Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Philyong Kang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Soo Mee Lim
- Department of Radiology, Ewha Womans University School of Medicine, Seoul, Republic of Korea.
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea.
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
12
|
Inoue KI, Miyachi S, Nishi K, Okado H, Nagai Y, Minamimoto T, Nambu A, Takada M. Recruitment of calbindin into nigral dopamine neurons protects against MPTP-Induced parkinsonism. Mov Disord 2018; 34:200-209. [PMID: 30161282 DOI: 10.1002/mds.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/06/2018] [Accepted: 06/29/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Parkinson's disease is caused by dopamine deficiency in the striatum, which is a result of loss of dopamine neurons from the substantia nigra pars compacta. There is a consensus that a subpopulation of nigral dopamine neurons that expresses the calcium-binding protein calbindin is selectively invulnerable to parkinsonian insults. The objective of the present study was to test the hypothesis that dopamine neuron degeneration might be prevented by viral vector-mediated gene delivery of calbindin into the dopamine neurons that do not normally contain it. METHODS A calbindin-expressing adenoviral vector was injected into the striatum of macaque monkeys to be conveyed to cell bodies of nigral dopamine neurons through retrograde axonal transport, or the calbindin-expressing lentiviral vector was injected into the nigra directly because of its predominant uptake from cell bodies and dendrites. The animals in which calbindin was successfully recruited into nigral dopamine neurons were administered systemically with MPTP. RESULTS In the monkeys that had received unilateral vector injections, parkinsonian motor deficits, such as muscular rigidity and akinesia/bradykinesia, appeared predominantly in the limbs corresponding to the non-calbindin-recruited hemisphere after MPTP administration. Data obtained from tyrosine hydroxylase immunostaining and PET imaging for the dopamine transporter revealed that the nigrostriatal dopamine system was preserved better on the calbindin-recruited side. Conversely, on the non-calbindin-recruited control side, many more dopamine neurons expressed α-synuclein. CONCLUSIONS The present results indicate that calbindin recruitment into nigral dopamine neurons protects against the onset of parkinsonian insults, thus providing a novel approach to PD prevention. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ken-Ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan.,Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Shigehiro Miyachi
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Cognitive Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | - Katsunori Nishi
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan
| | - Haruo Okado
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yuji Nagai
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takafumi Minamimoto
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Atsushi Nambu
- Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan.,Division of System Neurophysiology, National Institute for Physiological Sciences and Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan.,Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo, Japan
| |
Collapse
|
13
|
Huang B, Wu S, Wang Z, Ge L, Rizak JD, Wu J, Li J, Xu L, Lv L, Yin Y, Hu X, Li H. Phosphorylated α-Synuclein Accumulations and Lewy Body-like Pathology Distributed in Parkinson's Disease-Related Brain Areas of Aged Rhesus Monkeys Treated with MPTP. Neuroscience 2018; 379:302-315. [PMID: 29592843 DOI: 10.1016/j.neuroscience.2018.03.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/09/2018] [Accepted: 03/16/2018] [Indexed: 12/28/2022]
Abstract
Phosphorylation of α-synuclein at serine 129 (P-Ser 129 α-syn) is involved in the pathogenesis of Parkinson's disease (PD) and Lewy body (LB) formation. However, there is no clear evidence indicates the quantitative relation of P-Ser 129 α-syn accumulation and dopaminergic cell loss, LBs pathology and the affected brain areas in PD monkeys. Here, pathological changes in the substantia nigra (SN) and PD-related brain areas were measured in aged monkeys treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) utilizing a modeling-recovery-remodeling strategy. Compared to age-matched controls, the MPTP-treated monkeys showed significantly reduced tyrosine hydroxylase (TH)-positive neurons and increased P-Ser 129 α-syn-positive aggregations in the SN. Double-labeling Immunofluorescence found some TH-positive neurons to be co-localized with P-Ser129 α-syn in the SN, suggesting the inverse correlation between P-Ser 129 α-syn aggregations and dopaminergic cell loss in the SN may represent an interactive association related to the progression of the PD symptoms in the model. P-Ser 129 α-syn aggregations or LB-like pathology was also found in the midbrain and the neocortex, specifically in the oculomotor nucleus (CN III), temporal cortex (TC), prefrontal cortex (PFC) and in cells surrounding the third ventricle. Notably, the occipital cortex (OC) was P-Ser 129 α-syn negative. The findings of LB-like pathologies, dopaminergic cell loss and the stability of the PD symptoms in this model suggest that the modeling-recovery-remodeling strategy in aged monkeys may provide a new platform for biomedical investigations into the pathogenesis of PD and potential therapeutic development.
Collapse
Affiliation(s)
- Baihui Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Shihao Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Zhengbo Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Longjiao Ge
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Joshua D Rizak
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Jing Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Longbao Lv
- Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Yong Yin
- Department of Rehabilitation Medicine, Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, China.
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Hao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
14
|
Huot P, Sgambato-Faure V, Fox SH, McCreary AC. Serotonergic Approaches in Parkinson's Disease: Translational Perspectives, an Update. ACS Chem Neurosci 2017; 8:973-986. [PMID: 28460160 DOI: 10.1021/acschemneuro.6b00440] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) has long been seen as a disorder caused by degeneration of the dopaminergic system, leading to the classic motor manifestations of the disease. However, there is now overwhelming evidence that PD is more than a disease merely caused by dopamine depletion. It is well-known that a myriad of other neurotransmitters are affected by the disease process. One such neurotransmitter is serotonin (5-HT). 5-HT has been shown to play a role in several motor and nonmotor manifestations of PD, including tremor, cognition, depression and psychosis. 5-HT also seems to play a critical role in L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia. A breadth of preclinical studies and clinical trials have been conducted that aimed at modulating the 5-HT system in order to alleviate depression, cognitive deficits, psychosis, and dyskinesia. In this Review, we summarize recent advances in the 5-HT field in PD, but with a translational emphasis. We start by presenting a novel nonhuman primate model of PD that presents with dual dopamine and 5-HT lesions. We then present preclinical and clinical data that introduce new concepts, such as the use of biased and partial agonists, as well as molecules recently introduced to the field of PD, such as eltoprazine, pimavanserin, nelotanserin, and SYN-120, to enhance therapeutic benefit while minimizing adverse events, notably on parkinsonian disability.
Collapse
Affiliation(s)
- Philippe Huot
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 0A9, Canada
- Department
of Pharmacology, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Unité
des Troubles du Mouvement André Barbeau, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2L 4M1, Canada
- Division
of Neurology, Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Véronique Sgambato-Faure
- Institute of Cognitive
Neuroscience Marc Jeannerod, UMR 5229 CNRS, 69 675 Cedex Bron, France
- University Lyon 1, 69100 Villeurbanne, France
| | - Susan H. Fox
- Movement
Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Andrew C. McCreary
- Janssen Vaccines & Prevention B.V., Archimedesweg 4, 2333 CN Leiden, The Netherlands
| |
Collapse
|
15
|
Neuroprotective Effects of 7, 8-dihydroxyflavone on Midbrain Dopaminergic Neurons in MPP +-treated Monkeys. Sci Rep 2016; 6:34339. [PMID: 27731318 PMCID: PMC5059638 DOI: 10.1038/srep34339] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is one common neurodegenerative disease caused by a significant loss of midbrain dopaminergic neurons. Previous reports showed that 7, 8- dihydroxyflavone (7, 8-DHF) as a potent TrkB agonist can mimic BDNF and play neuroprotective roles for mouse dopaminergic neurons. Nonetheless, the safety and neuroprotective effects are unclear in monkey models of PD. Here, we find that 7, 8-DHF could be absorbed and metabolized into 7-hydroxy-8-methoxyflavone through oral administration in monkeys. The half-life time of 7, 8-DHF in monkey plasma is about 4–8 hrs. Furthermore, these monkeys maintain health state throughout the course of seven-month treatments of 7, 8-DHF (30 mg/kg/day). Importantly, 7, 8-DHF treatments can prevent the progressive degeneration of midbrain dopaminergic neurons by attenuating neurotoxic effects of MPP+ and display strong neuroprotective effects in monkeys. Our study demonstrates that this promising small molecule may be transited into a clinical useful pharmacological agent.
Collapse
|
16
|
Su LY, Li H, Lv L, Feng YM, Li GD, Luo R, Zhou HJ, Lei XG, Ma L, Li JL, Xu L, Hu XT, Yao YG. Melatonin attenuates MPTP-induced neurotoxicity via preventing CDK5-mediated autophagy and SNCA/α-synuclein aggregation. Autophagy 2016; 11:1745-59. [PMID: 26292069 DOI: 10.1080/15548627.2015.1082020] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Autophagy is involved in the pathogenesis of neurodegenerative diseases including Parkinson disease (PD). However, little is known about the regulation of autophagy in neurodegenerative process. In this study, we characterized aberrant activation of autophagy induced by neurotoxin 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) and demonstrated that melatonin has a protective effect on neurotoxicity. We found an excessive activation of autophagy in monkey brain tissues and C6 cells, induced by MPTP, which is mediated by CDK5 (cyclin-dependent kinase 5). MPTP treatment significantly reduced total dendritic length and dendritic complexity of cultured primary cortical neurons and melatonin could reverse this effect. Decreased TH (tyrosine hydroxylase)-positive cells and dendrites of dopaminergic neurons in the substantia nigra pars compacta (SNc) were observed in MPTP-treated monkeys and mice. Along with decreased TH protein level, we observed an upregulation of CDK5 and enhanced autophagic activity in the striatum of mice with MPTP injection. These changes could be salvaged by melatonin treatment or knockdown of CDK5. Importantly, melatonin or knockdown of CDK5 reduced MPTP-induced SNCA/α-synuclein aggregation in mice, which is widely thought to trigger the pathogenesis of PD. Finally, melatonin or knockdown of CDK5 counteracted the PD phenotype in mice induced by MPTP. Our findings uncover a potent role of CDK5-mediated autophagy in the pathogenesis of PD, and suggest that control of autophagic pathways may provide an important clue for exploring potential target for novel therapeutics of PD.
Collapse
Affiliation(s)
- Ling-Yan Su
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Hao Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Li Lv
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Yue-Mei Feng
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - Guo-Dong Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China.,d School of Life Science; Anhui University ; Hefei, Anhui , China
| | - Rongcan Luo
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - He-Jiang Zhou
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - Xiao-Guang Lei
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - Liang Ma
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Jia-Li Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,c Kunming Primate Research Center of the Chinese Academy of Sciences; Kunming Institute of Zoology; Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Lin Xu
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,e CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences ; Shanghai , China
| | - Xin-Tian Hu
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,c Kunming Primate Research Center of the Chinese Academy of Sciences; Kunming Institute of Zoology; Chinese Academy of Sciences ; Kunming, Yunnan , China.,e CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences ; Shanghai , China
| | - Yong-Gang Yao
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China.,c Kunming Primate Research Center of the Chinese Academy of Sciences; Kunming Institute of Zoology; Chinese Academy of Sciences ; Kunming, Yunnan , China.,e CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences ; Shanghai , China
| |
Collapse
|
17
|
Grow DA, McCarrey JR, Navara CS. Advantages of nonhuman primates as preclinical models for evaluating stem cell-based therapies for Parkinson's disease. Stem Cell Res 2016; 17:352-366. [PMID: 27622596 DOI: 10.1016/j.scr.2016.08.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 01/29/2023] Open
Abstract
The derivation of dopaminergic neurons from induced pluripotent stem cells brings new hope for a patient-specific, stem cell-based replacement therapy to treat Parkinson's disease (PD) and related neurodegenerative diseases; and this novel cell-based approach has already proven effective in animal models. However, there are several aspects of this procedure that have yet to be optimized to the extent required for translation to an optimal cell-based transplantation protocol in humans. These challenges include pinpointing the optimal graft location, appropriately scaling up the graft volume, and minimizing the risk of chronic immune rejection, among others. To advance this procedure to the clinic, it is imperative that a model that accurately and fully recapitulates characteristics most pertinent to a cell-based transplantation to the human brain is used to optimize key technical aspects of the procedure. Nonhuman primates mimic humans in multiple ways including similarities in genomics, neuroanatomy, neurophysiology, immunogenetics, and age-related changes in immune function. These characteristics are critical to the establishment of a relevant model in which to conduct preclinical studies to optimize the efficacy and safety of cell-based therapeutic approaches to the treatment of PD. Here we review previous studies in rodent models, and emphasize additional advantages afforded by nonhuman primate models in general, and the baboon model in particular, for preclinical optimization of cell-based therapeutic approaches to the treatment of PD and other neurodegenerative diseases. We outline current unresolved challenges to the successful application of stem cell therapies in humans and propose that the baboon model in particular affords a number of traits that render it most useful for preclinical studies designed to overcome these challenges.
Collapse
Affiliation(s)
- Douglas A Grow
- Department of Biology, University of Texas at San Antonio, San Antonio Cellular Therapeutics Institute, PriStem, United States
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio Cellular Therapeutics Institute, PriStem, United States
| | - Christopher S Navara
- Department of Biology, University of Texas at San Antonio, San Antonio Cellular Therapeutics Institute, PriStem, United States.
| |
Collapse
|
18
|
Combining Diffusion Tensor Imaging and Susceptibility Weighted Imaging on the Substantia Nigra of 1-Methyl-4-Phenyl-1, 2, 3, 6-Tetrahydropyridine (MPTP)-induced Rhesus Monkey Model of Parkinson's Disease. W INDIAN MED J 2016; 64:480-486. [PMID: 27400227 DOI: 10.7727/wimj.2016.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/08/2016] [Indexed: 12/18/2022]
Abstract
Objective The aim of this study was to evaluate whether combining diffusion tensor magnetic resonance imaging (DTI) and susceptibility weighted imaging (SWI) techniques would provide a sensitive method for differentiating between 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced rhesus monkey model of Parkinson's disease (PD) and wild-type controls. Subjects and Methods Seventeen rhesus monkeys were divided into two groups. A series of intramuscular injections of either saline (control group, n = 8) or MPTP (0.2 mg/kg body weight; PD group, n = 9) were given to the monkeys, twice a week. Then, SWI and DTI scans were obtained from the monkeys with Siemens Magnetom Verio 3.0T superconductive MRI system. Region of interest analysis was performed on substantia nigra pars compacta (SNc) and substantia nigra pars reticulata (SNr). In addition, immunohistochemical staining of tyrosine hydroxylase was applied to assess degeneration of SN dopaminergic neurons. Results Monkeys in the PD group displayed mild to moderate motor symptoms assessed using Kurlan's scale. With SWI scans, decreased width of SNc but increased width of SNr was found in PD group monkeys compared to controls. Calculation of the ratios of widths of SNc and SNr to the anterior and posterior mesencephalic diameter also reflected narrower SNc but wider SNr than controls. Decreased SWI signal intensity of SNc and SNr suggested iron deposition in both subregions of SN. The DTI scans showed lower fractional anisotropy (FA) values in SNc of the PD group monkeys, while no change of FA values in SNr was detected. Immunohistochemical test displayed generalized loss of dopaminergic neurons in SN of PD group monkeys. Conclusion Combining the use of DTI and SWI can provide a sensitive method for differentiating between MPTP-induced rhesus monkey model of PD and wild-type controls. This effective imaging modality might provide additional information for characteristic identification of PD at early stages, thus enhancing the ability to make early diagnosis, and monitor progression of the natural history and treatment effects.
Collapse
|
19
|
Asakawa T, Fang H, Sugiyama K, Nozaki T, Hong Z, Yang Y, Hua F, Ding G, Chao D, Fenoy AJ, Villarreal SJ, Onoe H, Suzuki K, Mori N, Namba H, Xia Y. Animal behavioral assessments in current research of Parkinson's disease. Neurosci Biobehav Rev 2016; 65:63-94. [PMID: 27026638 DOI: 10.1016/j.neubiorev.2016.03.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/22/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder, is traditionally classified as a movement disorder. Patients typically suffer from many motor dysfunctions. Presently, clinicians and scientists recognize that many non-motor symptoms are associated with PD. There is an increasing interest in both motor and non-motor symptoms in clinical studies on PD patients and laboratory research on animal models that imitate the pathophysiologic features and symptoms of PD patients. Therefore, appropriate behavioral assessments are extremely crucial for correctly understanding the mechanisms of PD and accurately evaluating the efficacy and safety of novel therapies. This article systematically reviews the behavioral assessments, for both motor and non-motor symptoms, in various animal models involved in current PD research. We addressed the strengths and weaknesses of these behavioral tests and their appropriate applications. Moreover, we discussed potential mechanisms behind these behavioral tests and cautioned readers against potential experimental bias. Since most of the behavioral assessments currently used for non-motor symptoms are not particularly designed for animals with PD, it is of the utmost importance to greatly improve experimental design and evaluation in PD research with animal models. Indeed, it is essential to develop specific assessments for non-motor symptoms in PD animals based on their characteristics. We concluded with a prospective view for behavioral assessments with real-time assessment with mobile internet and wearable device in future PD research.
Collapse
Affiliation(s)
- Tetsuya Asakawa
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan; Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan.
| | - Huan Fang
- Department of Pharmacy, Jinshan Hospital of Fudan University, Shanghai, China
| | - Kenji Sugiyama
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Takao Nozaki
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Zhen Hong
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yilin Yang
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Fei Hua
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Guanghong Ding
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Dongman Chao
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Albert J Fenoy
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Sebastian J Villarreal
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Hirotaka Onoe
- Functional Probe Research Laboratory, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Katsuaki Suzuki
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Norio Mori
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Ying Xia
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA.
| |
Collapse
|
20
|
Yan T, Rizak JD, Wang J, Yang S, Ma Y, Hu X. Severe dopaminergic neuron loss in rhesus monkey brain impairs morphine-induced conditioned place preference. Front Behav Neurosci 2015; 9:273. [PMID: 26528155 PMCID: PMC4600774 DOI: 10.3389/fnbeh.2015.00273] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 09/28/2015] [Indexed: 11/15/2022] Open
Abstract
It is well known that dopamine (DA) is critical for reward, but the precise role of DA in reward remains uncertain. The aim of this study was to determine what percentage of dopaminergic neurons in the primate brain is required for the expression of conditioned reward by measuring the performance of DA-deficient rhesus monkeys in a morphine-induced conditioned place preference (CPP) paradigm. Animals with mild Parkinsonian symptoms successfully developed and retained a morphine preference that was equivalent to control monkeys. However, these monkeys could not maintain the preference as well as controls when they retained severe Parkinsonian symptoms. On the other hand, monkeys initially in a severe Parkinsonian state developed a preference for morphine, but this preference was weaker than that of the controls. Histological results showed that the loss of dopaminergic neurons in monkeys that had severe Parkinsonian symptoms was about 80% in comparison to the control monkeys. All these data suggest that a severely impaired DA system alters rewarding-seeking behavior in non-human primates.
Collapse
Affiliation(s)
- Ting Yan
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China ; University of Chinese Academy of Sciences Beijing, China
| | - Joshua Dominic Rizak
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China ; University of Chinese Academy of Sciences Beijing, China
| | - Jianhong Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China
| | - Shangchuan Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China
| | - Yuanye Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China ; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China ; Yunnan Key Laboratory of Primate Biomedical Research Kunming, Yunnan, China
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China ; Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences Kunming, Yunnan, China ; Yunnan Key Laboratory of Primate Biomedical Research Kunming, Yunnan, China
| |
Collapse
|
21
|
Li H, Lei X, Yan T, Li H, Huang B, Li L, Xu L, Liu L, Chen N, Lü L, Ma Y, Xu L, Li J, Wang Z, Zhang B, Hu X. The temporary and accumulated effects of transcranial direct current stimulation for the treatment of advanced Parkinson's disease monkeys. Sci Rep 2015. [PMID: 26220760 PMCID: PMC4518219 DOI: 10.1038/srep12178] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) is a useful noninvasive technique of cortical brain stimulation for the treatment of neurological disorders. Clinical research has demonstrated tDCS with anodal stimulation of primary motor cortex (M1) in Parkinson’s disease (PD) patients significantly improved their motor function. However, few studies have been focused on the optimization of parameters which contributed significantly to the treatment effects of tDCS and exploration of the underline neuronal mechanisms. Here, we used different stimulation parameters of anodal tDCS on M1 for the treatment of aged advanced PD monkeys induced with 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) administration, and then analyzed the temporary and accumulated effects of tDCS treatment. The results indicated anodal tDCS on M1 very significantly improved motor ability temporarily; importantly, the treatment effects of anodal tDCS on M1 were quantitatively correlated to the accumulated stimulation instead of the stimuli intensity or duration respectively. In addition, c-fos staining showed tDCS treatment effects activated the neurons both in M1 and substantia nigra (SN). Therefore, we propose that long time and continue anodal tDCS on M1 is a better strategy to improve the motor symptoms of PD than individual manipulation of stimuli intensity or duration.
Collapse
Affiliation(s)
- Hao Li
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China [2] University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoguang Lei
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Ting Yan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Hongwei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Baihui Huang
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China [2] University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ling Li
- Medical imaging department, Kunming general hospital of PLA, Kunming, Yunnan, 650032, China
| | - Liqi Xu
- Medical imaging department, Kunming general hospital of PLA, Kunming, Yunnan, 650032, China
| | - Li Liu
- Medical imaging department, Kunming general hospital of PLA, Kunming, Yunnan, 650032, China
| | - Nanhui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Longbao Lü
- Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Yuanye Ma
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China [2] Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Lin Xu
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China [2] CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Zhengbo Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Xintian Hu
- 1] Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences &Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China [2] CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai, 200031, China [3] Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| |
Collapse
|
22
|
Lei X, Li H, Huang B, Rizak J, Li L, Xu L, Liu L, Wu J, Lü L, Wang Z, Hu Y, Le W, Deng X, Li J, Yao Y, Xu L, Hu X, Zhang B. 1-Methyl-4-phenylpyridinium stereotactic infusion completely and specifically ablated the nigrostriatal dopaminergic pathway in rhesus macaque. PLoS One 2015; 10:e0127953. [PMID: 26010745 PMCID: PMC4444358 DOI: 10.1371/journal.pone.0127953] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 04/22/2015] [Indexed: 01/09/2023] Open
Abstract
Introduction Complete and specific ablation of a single dopaminergic (DA) pathway is a critical step to distinguish the roles of DA pathways in vivo. However, this kind of technique has not been reported in non-human primates. This study aimed to establish a lesioning method with a complete and specific ablation. Method A carefully designed infusion route based on a MRI stereotactic technique was developed to deliver the highly selective dopaminergic toxin 1-methyl-4-phenylpyridinium (MPP+) unilaterally into multiple sites of compact part of substantia nigra (SNc) and striatum in monkeys. The nigrostriatal DA pathway was selected because lesioning of this pathway may induce symptoms that are suitable for evaluation. The pathological, behavioral, neuropharmacological, and clinical laboratorial data were collected to evaluate the lesioning effects. Result Pathological examination revealed a complete ablation of tyrosine hydroxylase positive (TH+) neurons in the SNc, while preserving intact TH+ neurons in the ventral tegmental area (VTA) nearby. TH+ projections in the striatum were also unilaterally lost. The monkeys displayed stable (>28 weeks) rotations and symptoms which were expected with loss of DA neurons in the SNc, with rest tremor being an exception. No item implied the presence of a severe side effect caused by the operation or the intracerebral MPP+ infusion. The results suggested that rest tremor may not directly rely on the nigrostriatal pathway. Conclusion Taken together, in addition to providing a specific nigrostriatal DA lesioned model, this method, combined with brain stimulation or other techniques, can be applied as a powerful tool for the complete lesion of any desired DA pathway in order to study its specific functions in the brain.
Collapse
Affiliation(s)
- Xiaoguang Lei
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Baihui Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Joshua Rizak
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ling Li
- Medical Imaging Department, Kunming General Hospital of PLA, Kunming, Yunnan, China
| | - Liqi Xu
- Medical Imaging Department, Kunming General Hospital of PLA, Kunming, Yunnan, China
| | - Li Liu
- Medical Imaging Department, Kunming General Hospital of PLA, Kunming, Yunnan, China
| | - Jing Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Longbao Lü
- Kunming Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhengbo Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yingzhou Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Weidong Le
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xingli Deng
- Neurosurgery Department, 1st Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yonggang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai, China
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (XH); (BZ)
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail: (XH); (BZ)
| |
Collapse
|
23
|
A quantitative approach to developing Parkinsonian monkeys (Macaca fascicularis) with intracerebroventricular 1-methyl-4-phenylpyridinium injections. J Neurosci Methods 2015; 251:99-107. [PMID: 26003862 DOI: 10.1016/j.jneumeth.2015.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Non-human primate Parkinson's disease (PD) models are essential for PD research. The most extensively used PD monkey models are induced with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, the modeling processes of developing PD monkeys cannot be quantitatively controlled with MPTP. Therefore, a new approach to quantitatively develop chronic PD monkey models will help to advance the goals of "reduction, replacement and refinement" in animal experiments. NEW METHOD A novel chronic PD monkey models was reported using the intracerebroventricular administration of 1-methyl-4-phenylpyridinium (MPP(+)) in Cynomolgus monkeys (Macaca fascicularis). RESULTS This approach successfully produced stable and consistent PD monkeys with typical motor symptoms and pathological changes. More importantly, a sigmoidal relationship (Y=8.15801e(-0.245/x); R=0.73) was discovered between PD score (Y) and cumulative dose of MPP(+) (X). This relationship was then used to develop two additional PD monkeys under a specific time schedule (4 weeks), with planned PD scores (7) by controlling the dose and frequency of the MPP(+) administration as an independent validation of the formula. COMPARISON WITH EXISTING METHOD(S) We developed Parkinsonian monkeys within controlled time frames by regulating the accumulated dose of MPP(+) intracerebroventricular administered, while limiting side effects often witnessed in models developed with the peripheral administration of MPTP, makes this model highly suitable for treatment development. CONCLUSIONS This novel approach provides an edge in evaluating the mechanisms of PD pathology associated with environmental toxins and novel treatment approaches as the formula developed provides a "map" to control and predict the modeling processes.
Collapse
|
24
|
Acute morphine treatments alleviate tremor in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated monkeys. PLoS One 2014; 9:e88404. [PMID: 24520383 PMCID: PMC3919785 DOI: 10.1371/journal.pone.0088404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/05/2014] [Indexed: 11/24/2022] Open
Abstract
Parkinson’s disease (PD) is a chronic and progressive neurodegenerative disorder associated with decreased striatal dopamine levels. Morphine has been found to elevate dopamine levels, which indicates a potential therapeutic effect in PD treatment that has not been investigated previously. To evaluate this hypothesis, an investigation of the acute effects of morphine on PD symptoms was carried out in male rhesus PD monkeys that had been induced with MPTP. All MPTP induced monkeys displayed progressive and irreversible PD motor symptoms. The behavioral response of these animals to morphine and L-Dopa were quantified with the Kurlan scale. It was found that L-Dopa alleviated bradykinesia, but did not significantly improve tremor. In contrast, acute morphine alleviated tremor significantly. These results suggested that, compared to L-Dopa, morphine has different therapeutic effects in PD therapy and may act through different biological mechanisms to alleviate PD symptoms.
Collapse
|
25
|
Motor behavior correlates with striatal [18F]-DOPA uptake in MPTP-lesioned primates. Neurochem Int 2013; 62:349-53. [DOI: 10.1016/j.neuint.2013.01.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 01/14/2013] [Accepted: 01/19/2013] [Indexed: 11/21/2022]
|
26
|
Neurotoxin-based models of Parkinson's disease. Neuroscience 2012; 211:51-76. [DOI: 10.1016/j.neuroscience.2011.10.057] [Citation(s) in RCA: 360] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 10/27/2011] [Accepted: 10/28/2011] [Indexed: 12/21/2022]
|
27
|
van Bregt DR, Thomas TC, Hinzman JM, Cao T, Liu M, Bing G, Gerhardt GA, Pauly JR, Lifshitz J. Substantia nigra vulnerability after a single moderate diffuse brain injury in the rat. Exp Neurol 2012; 234:8-19. [PMID: 22178300 PMCID: PMC3294202 DOI: 10.1016/j.expneurol.2011.12.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 11/28/2011] [Accepted: 12/01/2011] [Indexed: 11/25/2022]
Abstract
Dementia and parkinsonism are late-onset symptoms associated with repetitive head injury, as documented in multiple contact-sport athletes. Clinical symptomatology is the likely phenotype of chronic degeneration and circuit disruption in the substantia nigra (SN). To investigate the initiating neuropathology, we hypothesize that a single diffuse brain injury is sufficient to initiate SN neuropathology including neuronal loss, vascular disruption and microglial activation, contributing to neurodegeneration and altered dopamine regulation. Adult, male Sprague-Dawley rats were subjected to sham or moderate midline fluid percussion brain injury. Stereological estimates indicated a significant 44% loss of the estimated total neuron number in the SN at 28-days post-injury, without atrophy of neuronal nuclear volumes, including 25% loss of tyrosine hydroxylase positive neurons by 28-days post-injury. Multi-focal vascular compromise occurred 1-2 days post-injury, with ensuing microglial activation (significant 40% increase at 4-days). Neurodegeneration (silver-stain technique) encompassed on average 21% of the SN by 7-days post-injury and increased to 29% by 28-days compared to sham (1%). Whole tissue SN, but not striatum, dopamine metabolism was altered at 28-days post-injury, without appreciable gene or protein changes in dopamine synthesis or regulation elements. Together, single moderate diffuse brain injury resulted in SN neurovascular pathology potentially associated with neuroinflammation or dopamine dysregulation. Compensatory mechanisms may preserve dopamine signaling acutely, but subsequent SN damage with aging or additional injury may expose clinical symptomatology of motor ataxias and dementia.
Collapse
Affiliation(s)
- Daniel R. van Bregt
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Theresa Currier Thomas
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jason M. Hinzman
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Morris K. Udall Parkinson's Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, USA
- Center for Microelectrode Technology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Tuoxin Cao
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Mei Liu
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Guoying Bing
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Greg A. Gerhardt
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Morris K. Udall Parkinson's Disease Research Center of Excellence, University of Kentucky College of Medicine, Lexington, KY, USA
- Center for Microelectrode Technology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - James R. Pauly
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Pharmaceutical Sciences University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Jonathan Lifshitz
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Physical Medicine & Rehabilitation, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
28
|
Tachibana Y, Iwamuro H, Kita H, Takada M, Nambu A. Subthalamo-pallidal interactions underlying parkinsonian neuronal oscillations in the primate basal ganglia. Eur J Neurosci 2011; 34:1470-84. [DOI: 10.1111/j.1460-9568.2011.07865.x] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
29
|
Redmond DE. Behavioral Assessment in the African Green Monkey After MPTP Administration. NEUROMETHODS 2011. [DOI: 10.1007/978-1-61779-298-4_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
30
|
The effects of reversible inactivation of the subthalamo-pallidal pathway on the behaviour of naive and hemiparkinsonian monkeys. J Clin Neurosci 2010; 4:218-27. [PMID: 18638958 DOI: 10.1016/s0967-5868(97)90076-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/1996] [Accepted: 06/28/1996] [Indexed: 11/23/2022]
Abstract
This study was designed to further investigate the role of the subthalamic nucleus (STN) and globus pallidus internus (GPi) in the pathophysiology of Parkinson's disease. The prevailing theory about the pathophysiology of Parkinson's disease (PD) predicts that there is overactivity of the subthalamo-pallidal pathway. In order to inactivate that pathway, naive and hemiparkinsonian monkeys were locally administered either muscimol (to reversibly inactivate the contralateral STN) or kynurenic acid (to reduce glutamatergic activity in the contralateral GPi). Three naive and 2 hemiparkinsonian monkeys were studied. Intra-carotid MPTP was administered to produce 2 hemiparkinsonian monkeys. Injection sites of muscimol and kynurenic acid in the brain were confirmed electrophysiologically and histologically. Injections of muscimol into the STN in naive and hemiparkinsonian monkeys caused reversible contralateral dystonia, but did not alleviate Parkinsonism. Only one kynurenic acid injection into GPi partially alleviated Parkinsonism. On the basis of the results in this study, aspects of the currently accepted hypothesis of the pathophysiology of PD cannot be confirmed. However, this study reports that the STN has an important role in the production of dystonia. This experimental model of dystonia will prove suitable for further study of both the mechanisms causing dystonia as well as for possible therapeutic approaches to its treatment.
Collapse
|
31
|
Vitale A, Manciocco A, Alleva E. The 3R principle and the use of non-human primates in the study of neurodegenerative diseases: the case of Parkinson's disease. Neurosci Biobehav Rev 2008; 33:33-47. [PMID: 18773919 DOI: 10.1016/j.neubiorev.2008.08.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 08/11/2008] [Accepted: 08/11/2008] [Indexed: 12/28/2022]
Abstract
The aim of this paper is to offer an ethical perspective on the use of non-human primates in neurobiological studies, using the Parkinson's disease (PD) as an important case study. We refer, as theoretical framework, to the 3R principle, originally proposed by Russell and Burch [Russell, W.M.S., Burch, R.L., 1959. The Principles of Humane Experimental Technique. Universities Federation for Animal Welfare Wheathampstead, England (reprinted in 1992)]. Then, the use of non-human primates in the study of PD will be discussed in relation to the concepts of Replacement, Reduction, and Refinement. Replacement and Reduction result to be the more problematic concept to be applied, whereas Refinement offers relatively more opportunities of improvement. However, although in some cases the 3R principle shows its applicative limits, its value, as conceptual and inspirational tool remains extremely valuable. It suggests to the researchers a series of questions, both theoretical and methodological, which can have the results of improving the quality of life on the experimental models, the quality of the scientific data, and the public perception from the non-scientist community.
Collapse
Affiliation(s)
- Augusto Vitale
- Section of Behavioural Neuroscience, Dipartimento di Biologia Cellulare e Neuroscienze, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy.
| | | | | |
Collapse
|
32
|
Xin T, Ai Y, Gerhardt G, Gash D, Zhang Z. Globus pallidus plays a critical role in neurotrophic factor induced functional improvements in hemiparkinsonian monkeys. Biochem Biophys Res Commun 2008; 370:434-9. [PMID: 18381061 DOI: 10.1016/j.bbrc.2008.03.108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Accepted: 03/20/2008] [Indexed: 11/18/2022]
Abstract
This study was designed to test the hypothesis that the integrity of the globus pallidus (GP) is critical for neurotrophic factor, such as glial-derived neurotrophic factor (GDNF), induced functional changes in rhesus macaques with MPTP-induced parkinsonism, because our previous studies demonstrated that the GP was one of the most affected areas as assessed by the levels of dopamine (DA) and its metabolites. A group of eight hemiparkinsonian monkeys with pallidal lesions, which positively responsed to intraventricular (ICV) injections of GDNF prior to the lesions, and a group of eight hemiparkinsonian monkeys without pallidal lesions, were treated with GDNF after a long washout period after the initial ICV infusions of GDNF. Significant behavioral improvements were only seen in the monkeys without pallidal lesions that received GDNF. Monkeys with pallidal lesions failed to exhibit any behavioral improvement even though they had elevated nigral DA levels. The results suggest that the GP is critical for neurotrophic factor induced functional changes in PD monkeys.
Collapse
Affiliation(s)
- Tao Xin
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong, University School of Medicine, Jinan, Shandong 250021, PR China
| | | | | | | | | |
Collapse
|
33
|
Berger TW, Gerhardt G, Liker MA, Soussou W. The Impact of Neurotechnology on Rehabilitation. IEEE Rev Biomed Eng 2008; 1:157-97. [PMID: 22274903 DOI: 10.1109/rbme.2008.2008687] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Theodore W Berger
- Department of Biomedical Engineering, Center for Neural Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | |
Collapse
|
34
|
Lipina SJ, Colombo JA. Premorbid exercising in specific cognitive tasks prevents impairment of performance in parkinsonian monkeys. Brain Res 2007; 1134:180-6. [PMID: 17196188 DOI: 10.1016/j.brainres.2006.11.066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 11/13/2006] [Accepted: 11/21/2006] [Indexed: 11/29/2022]
Abstract
Adult Cebus apella monkeys were exposed to either one, two or four series of cognitive tasks that place a demand on working memory and inhibitory control (Spatial Delayed Response and Object Retrieval Detour), before administration of the neurotoxin 1-methyl-1-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Following MPTP treatment, monkeys receiving more than one series successfully reached criteria at delays similar to those attained during the pre-MPTP stage for the Spatial Delayed Response task and avoided increased perseveration in the Object Retrieval Detour task. Results provide evidence that protection towards a neurotoxin in specific cognitive performances can be increased by repeated exposure to task-specific cognitive demands and that motor and cognitive impairment following MPTP treatment can be effectively dissociated in primates.
Collapse
Affiliation(s)
- Sebastián J Lipina
- Unidad de Neurobiología Aplicada (UNA) (CEMIC), Av. Galván 4102 C1431FWO, Buenos Aires, Argentina
| | | |
Collapse
|
35
|
|
36
|
Abstract
Nonhuman primate (NHP) models of Parkinson's disease (PD) play an essential role in the understanding of PD pathophysiology and the assessment of PD therapies. NHP research enabled the identification of environmental risk factors for the development of PD. Electrophysiological studies in NHP models of PD identified the neural circuit responsible for PD motor symptoms, and this knowledge led to the development of subthalamic surgical ablation and deep brain stimulation. Similar to human PD patients, parkinsonian monkeys are responsive to dopamine replacement therapies and present complications associated with their long-term use, a similarity that facilitated the assessment of new symptomatic treatments, such as dopaminergic agonists. New generations of compounds and novel therapies that use directed intracerebral delivery of drugs, cells, and viral vectors benefit from preclinical evaluation in NHP models of PD. There are several NHP models of PD, each with characteristics that make it suitable for the study of different aspects of the disease or potential new therapies. Investigators who use the models and peer scientists who evaluate their use need information about the strengths and limitations of the different PD models and their methods of evaluation. This article provides a critical review of available PD monkey models, their utilization, and how they compare to emerging views of PD as a multietiologic, multisystemic disease. The various models are particularly useful for representing different aspects of PD at selected time points. This conceptualization provides clues for the development of new NHP models and facilitates the clinical translation of findings. As ever, successful application of any model depends on matching the model to the scientific question to be answered. Adequate experimental designs, with multiple outcome measures of clinical relevance and an appropriate number of animals, are essential to minimize the limitations of models and increase their predictive clinical validity.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, WI 53715, USA.
| |
Collapse
|
37
|
Kaneda K, Tachibana Y, Imanishi M, Kita H, Shigemoto R, Nambu A, Takada M. Down-regulation of metabotropic glutamate receptor 1alpha in globus pallidus and substantia nigra of parkinsonian monkeys. Eur J Neurosci 2006; 22:3241-54. [PMID: 16367790 DOI: 10.1111/j.1460-9568.2005.04488.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Enhanced glutamatergic neurotransmission via the subthalamopallidal or subthalamonigral projection seems crucial for developing parkinsonian motor signs. In the present study, the possible changes in the expression of metabotropic glutamate receptors (mGluRs) were examined in the basal ganglia of a primate model for Parkinson's disease. When the patterns of immunohistochemical localization of mGluRs in monkeys administered systemically with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) were analysed in comparison with normal controls, we found that expression of mGluR1alpha, but not of other subtypes, was significantly reduced in the internal and external segments of the globus pallidus and the substantia nigra pars reticulata. To elucidate the functional role of mGluR1 in the control of pallidal neuron activity, extracellular unit recordings combined with intrapallidal microinjections of mGluR1-related agents were then performed in normal and parkinsonian monkeys. In normal awake conditions, the spontaneous firing rates of neurons in the pallidal complex were increased by DHPG, a selective agonist of group I mGluRs, whereas they were decreased by AIDA, a selective antagonist of group I mGluRs, or LY367385, a selective antagonist of mGluR1. These electrophysiological data strongly indicate that the excitatory mechanism of pallidal neurons by glutamate is mediated at least partly through mGluR1. The effects of the mGluR1-related agents on neuronal firing in the internal pallidal segment became rather obscure after MPTP treatment. Our results suggest that the specific down-regulation of pallidal and nigral mGluR1alpha in the parkinsonian state may exert a compensatory action to reverse the overactivity of the subthalamic nucleus-derived glutamatergic input that is generated in the disease.
Collapse
Affiliation(s)
- K Kaneda
- Department of System Neuroscience, Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Research, Fuchu, Tokyo 183-8526, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Togasaki DM, Hsu A, Samant M, Farzan B, DeLanney LE, Langston JW, Di Monte DA, Quik M. The Webcam system: a simple, automated, computer-based video system for quantitative measurement of movement in nonhuman primates. J Neurosci Methods 2005; 145:159-66. [PMID: 15922034 DOI: 10.1016/j.jneumeth.2004.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Revised: 11/01/2004] [Accepted: 12/22/2004] [Indexed: 11/26/2022]
Abstract
Investigations using models of neurologic disease frequently involve quantifying animal motor activity. We developed a simple method for measuring motor activity using a computer-based video system (the Webcam system) consisting of an inexpensive video camera connected to a personal computer running customized software. Images of the animals are captured at half-second intervals and movement is quantified as the number of pixel changes between consecutive images. The Webcam system allows measurement of motor activity of the animals in their home cages, without devices affixed to their bodies. Webcam quantification of movement was validated by correlation with measures simultaneously obtained by two other methods: measurement of locomotion by interruption of infrared beams; and measurement of general motor activity using portable accelerometers. In untreated squirrel monkeys, correlations of Webcam and locomotor activity exceeded 0.79, and correlations with general activity counts exceeded 0.65. Webcam activity decreased after the monkeys were rendered parkinsonian by treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), but the correlations with the other measures of motor activity were maintained. Webcam activity also correlated with clinical ratings of parkinsonism. These results indicate that the Webcam system is reliable under both untreated and experimental conditions and is an excellent method for quantifying motor activity in animals.
Collapse
Affiliation(s)
- Daniel M Togasaki
- The Parkinson's Institute, 1170 Morse Avenue, Sunnyvale, CA 94089, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Papa SM, Auberson YP, Greenamyre JT. Prolongation of levodopa responses by glycineB antagonists in parkinsonian primates. Ann Neurol 2005; 56:723-7. [PMID: 15470750 PMCID: PMC3325144 DOI: 10.1002/ana.20279] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To examine the antiparkinsonian effects of blocking glycineB receptors, we designed a pilot study testing the potent and selective antagonist, PAMQX, in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated primates. PAMQX had no intrinsic effects but markedly potentiated the antiparkinsonian action of levodopa. In a dose-dependent fashion, coadministration of the glycineB antagonist with levodopa extended the response duration by nearly 60%. It is noteworthy that PAMQX, within a considerable dose range, did not cause ataxia or other side effects. These data indicate that blocking N-methyl-D-aspartate receptors selectively to manipulate dopaminergic-mediated motor responses may be produced effectively by glycineB antagonists.
Collapse
Affiliation(s)
- Stella M Papa
- Department of Neurology, Emory University, Atlanta, Georgia 30322, USA.
| | | | | |
Collapse
|
40
|
Dunnett SB. Chapter V Motor function(s) of the nigrostriatal dopamine system: Studies of lesions and behavior. ACTA ACUST UNITED AC 2005. [DOI: 10.1016/s0924-8196(05)80009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
41
|
Emborg ME. Evaluation of animal models of Parkinson's disease for neuroprotective strategies. J Neurosci Methods 2004; 139:121-43. [PMID: 15488225 DOI: 10.1016/j.jneumeth.2004.08.004] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2004] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of dopaminergic nigral neurons and striatal dopamine. Despite the advances of modern therapy to treat the symptoms of PD, most of the patients will eventually experience debilitating disability. The need for neuroprotective strategies that will slow or stop the progression of the disease is clear. The progress in the understanding of the cause and pathogenesis of PD is providing clues for the development of disease-modifying strategies. In that regard, animal models of PD and non-human primate models in particular, are essential for the preclinical evaluation and testing of candidate therapies. However, the diversity of models and different outcome measures used by investigators make it challenging to compare results between neuroprotective agents. In this review we will discuss methods for the selection, development and assessment of animal models of PD, the role of non-human primates and the concept of "multiple models/multiple endpoints" to predict the success in the clinic of neuroprotective strategies.
Collapse
Affiliation(s)
- Marina E Emborg
- National Primate Research Center and Department of Anatomy, University of Wisconsin, 1223 Capitol Court, Madison, WI 53715, USA.
| |
Collapse
|
42
|
Andringa G, Eshuis S, Perentes E, Maguire RP, Roth D, Ibrahim M, Leenders KL, Cools AR. TCH346 prevents motor symptoms and loss of striatal FDOPA uptake in bilaterally MPTP-treated primates. Neurobiol Dis 2004; 14:205-17. [PMID: 14572443 DOI: 10.1016/s0969-9961(03)00125-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The neuroprotective efficacy of the propargylamine TCH346 was studied in the primate model of Parkinson's disease, the bilaterally MPTP-treated monkey. Male rhesus monkeys received 2.5 mg MPTP into the left carotid artery and, 8 weeks later, 1.25 mg MPTP into the right carotid artery. Starting 2 h after the second MPTP infusion, either 0.014 mg/kg TCH346 or its solvent was subcutaneously injected twice per day for 14 days. The first MPTP treatment induced mild Parkinson symptoms, reduced right limb movements, and reduced FDOPA uptake in the left striatum. The second MPTP treatment made Parkinson symptoms worse, reduced left limb movements, and reduced FDOPA uptake in the right striatum of solvent-treated monkeys. In contrast, the second MPTP treatment did not further worsen motor symptoms and did not decrease FDOPA uptake in the right striatum of TCH346-treated monkeys. Although the effects of the second MPTP treatment were largely prevented, the effects of the first MPTP treatment were not reversed by TCH346. Immunohistochemical examination confirmed the dramatic loss of dopamine cells in vehicle-treated monkeys and the preservation of these neurons in the right brain side of the TCH346-treated animals. In conclusion, systemic administration of TCH346 prevented motor symptoms and nigrostriatal degeneration induced by MPTP in primates.
Collapse
Affiliation(s)
- Gerda Andringa
- Department of Psychoneuropharmacology, University of Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Grondin R, Zhang Z, Ai Y, Gash DM, Gerhardt GA. Intracranial delivery of proteins and peptides as a therapy for neurodegenerative diseases. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 61:101-23. [PMID: 14674610 DOI: 10.1007/978-3-0348-8049-7_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Parkinson's disease is characterized by a progressive degeneration of the substantia nigra pars compacta dopamine neurons that innervate the striatum. Unlike current treatments for PD, GDNF administration could potentially slow or halt the continued degeneration of nigral dopaminergic neurons. GDNF does not cross the blood-brain barrier and needs to be administered directly into the brain. Due to the progressive nature of PD, sustained delivery of trophic factors may be necessary for optimal, long-term neuronal effects. Novel methods for sustained delivery of GDNF into the nigrostriatal pathway are currently being studied in non-human primates, including computer-controlled infusion pumps. Using this approach, we have demonstrated that chronic infusions of nominally 7.5 or 22.5 microg/day GDNF into the lateral ventricle, the putamen or the substantia nigra, using programmable pumps, promotes restoration of the nigrostriatal dopaminergic system and significantly improves motor functions in MPTP-lesioned rhesus monkeys with neural deficits modeling the terminal stages of PD and in aged rhesus monkeys modeling the early stages of PD. Based on the promising studies of the chronic effects of GDNF in non-human primate models of PD, a study was recently conducted in England on five advanced PD patients. Chronic GDNF infusion into the dorsal putamen, via programmable pumps, resulted in improved motor function in all patients and limited side effects were observed. However, while the data from this intraparenchymal clinical trial in humans look encouraging, extensive blinded efficacy trials will need to be conducted before it can be determined if chronic treatment with GDNF or other trophic molecules will prove useful in treating patients with PD.
Collapse
Affiliation(s)
- Richard Grondin
- Department of Anatomy & Neurobiology and the Morris K. Udall Parkinson's Disease Research Center of Excellence, 305 Davis-Mills Bldg, University of Kentucky Medical Center, Lexington, KY 40536-0098, USA.
| | | | | | | | | |
Collapse
|
45
|
Matsumura M, Kojima J. The role of the pedunculopontine tegmental nucleus in experimental parkinsonism in primates. Stereotact Funct Neurosurg 2002; 77:108-15. [PMID: 12378066 DOI: 10.1159/000064614] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To clarify the role of the pedunculopontine tegmental nucleus (PPN) in motor behavior, we have conducted a series of experiments in primates. In the first part, PPN was damaged locally with kainic acid, which resulted in mild hemiparkinsonism in the contralateral limbs. In the second part, muscimol (a GABA agonist) was locally injected into the PPN area in monkeys who had been trained to perform a lever-pull movement with an arm, resulting in a slowness of movement and a delay of the movement onset. In the third part, a dopaminergic neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was systemically injected in monkeys with prior PPN lesioning. These monkeys developed no, or if any, very mild parkinsonism. PPN lesioning was supposed to have protected the nigral neurons from the MPTP- toxicity. The PPN facilitates the motor system through its nigral projection. The decreased activity of the PPN may underlie the pathophysiology of parkinsonism.
Collapse
Affiliation(s)
- M Matsumura
- Chuo Gunma Neurosurgery Hospital, Nakaomachi, Takasaki, Japan.
| | | |
Collapse
|
46
|
Grondin R, Zhang Z, Yi A, Cass WA, Maswood N, Andersen AH, Elsberry DD, Klein MC, Gerhardt GA, Gash DM. Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys. Brain 2002; 125:2191-201. [PMID: 12244077 DOI: 10.1093/brain/awf234] [Citation(s) in RCA: 211] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The powerful trophic effects that glial cell line-derived neurotrophic factor (GDNF) exerts on midbrain dopamine neurones suggest its use in treating Parkinson's disease. However, some important questions remain about the possible therapeutic applications of GDNF. Here we demonstrate that the chronic infusion of 5 or 15 micro g/day GDNF into the lateral ventricle or the striatum, using programmable pumps, promotes restoration of the nigrostriatal dopaminergic system and significantly improves motor functions in rhesus monkeys with neural deficits modelling the terminal stages of Parkinson's disease. The functional improvements were associated with pronounced upregulation and regeneration of nigral dopamine neurones and their processes innervating the striatum. When compared with vehicle recipients, these functional improvements were associated with (i) >30% bilateral increase in nigral dopamine neurone cell size; (ii) >20% bilateral increase in the number of nigral cells expressing the dopamine marker tyrosine hydroxylase; (iii) >70 and >50% bilateral increase in dopamine metabolite levels in the striatum and the pallidum, respectively; (iv) 233 and 155% increase in dopamine levels in the periventricular striatal region and the globus pallidus, respectively, on the lesioned side; and (v) a five-fold increase in tyrosine hydroxylase-positive fibre density in the periventricular striatal region on the lesioned side. In addition, chronic GDNF treatment did not induce the side-effects generally associated with chronic administration of levodopa, the most widely used treatment for Parkinson's disease. Thus, the results suggest that the prolonged and controlled delivery of GDNF into the brain could be used to intervene in long-term neurodegenerative disease processes like Parkinson's disease. Additional studies are required to determine the potential differences between chronic, intraventricular and intraputamenal (or intranigral) delivery of GDNF to maximize the efficacy of infusion treatments.
Collapse
Affiliation(s)
- Richard Grondin
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Rm. 305 Davis Mills Building, 800 Rose Street, Lexington, KY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Huang X, Lawler CP, Lewis MM, Nichols DE, Mailman RB. D1 dopamine receptors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2002; 48:65-139. [PMID: 11526741 DOI: 10.1016/s0074-7742(01)48014-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- X Huang
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
48
|
Zhang Z, Andersen A, Grondin R, Barber T, Avison R, Gerhardt G, Gash D. Pharmacological MRI mapping of age-associated changes in basal ganglia circuitry of awake rhesus monkeys. Neuroimage 2001; 14:1159-67. [PMID: 11697947 DOI: 10.1006/nimg.2001.0902] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
While the pathophysiological changes induced by the loss of dopamine innervation in the basal ganglia by Parkinson's disease (PD) are well studied, little is known about functional changes in the neural circuitry of this area during normal aging. Here we report the first survey of age-associated changes in the basal ganglia of behaviorally characterized, awake rhesus monkeys, using pharmacological MRI to map responses to dopaminergic stimulation. Apomorphine, a mixed D(1)/D(2) dopamine receptor agonist, evoked little change in the substantia nigra (SN) of aged animals while significantly reducing activation in young adult monkeys. Compared to young animals, both apomorphine and d-amphetamine (which increases synaptic dopamine levels) significantly increased activation of the aged rhesus globus pallidus externa (GPe). In addition, the aged animals showed decreased activity in the putamen in response to d-amphetamine administration. Although the responses in the SN and putamen of the aged monkeys differed from those in animal models of PD, the apomorphine-evoked activation of their GPe corresponded with apomorphine-induced increases in neuronal activity seen in Parkinson's patients and animal models. Given the major role of the GPe in regulating motor behavior, the altered responses in the aged GPe may contribute significantly to the motor slowing and movement dysfunctions characterizing advanced age.
Collapse
Affiliation(s)
- Z Zhang
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Lipina SJ, Colombo JA. Dissociated functional recovery in parkinsonian monkeys following transplantation of astroglial cells. Brain Res 2001; 911:176-80. [PMID: 11511388 DOI: 10.1016/s0006-8993(01)02682-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bilateral astroglial transplantation into the neostriatum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys resulted in significant performance improvement in a spatial delayed response task, but failed to modify perseveration in an object retrieval detour task, or to improve motor clinical rating. Results suggest that brain circuits subserving various motor and cognitive performances can be functionally dissociated, and that remaining resources for the reorganization of neural circuits involved in spatial working memory performance in parkinsonian monkeys, appear to be responsive to striatal transplantation of subcultured, fetal striatal astroglial cells.
Collapse
Affiliation(s)
- S J Lipina
- Unidad de Neurobiología Aplicada (UNA) (CEMIC-CONICET), Av. Galván 4102, 1431 Buenos Aires, Argentina
| | | |
Collapse
|
50
|
Andringa G, Cools AR. The neuroprotective effects of CGP 3466B in the best in vivo model of Parkinson's disease, the bilaterally MPTP-treated rhesus monkey. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2001:215-25. [PMID: 11205142 DOI: 10.1007/978-3-7091-6301-6_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
The propargylamine CGP 3466B prevents dopamine cell death both in vitro and in rodent models of Parkinson's disease. The present study investigates the efficacy of this compound to prevent the behavioral consequences of dopaminergic cell death in the best animal model of Parkinson's disease, the bilaterally MPTP-treated monkey. Rhesus monkeys were bilaterally treated with MPTP, using a two-step procedure: 2.50 mg MPTP was infused into the left carotid artery followed by a second bolus of 1.25 mg into the right carotid artery, 8 weeks later. Subcutaneous injection of either 0.014 mg/kg CGP 3466B (n = 4) or its solvent (distilled water; n = 4), twice daily for fourteen days, started two hours after the second MPTP infusion. A Parkinson rating scale was assessed for the evaluation of the effects. After the first MPTP treatment, the monkeys developed mild to moderate parkinsonian symptoms. The second MPTP treatment strongly increased the severity of Parkinson scores in all control monkeys, as assessed on day 3, 7, 14, 21, 28 and 35 after the second MPTP treatment. In contrast, CGP 3466B nearly completely prevented the increase of parkinsonian symptoms after the second MPTP treatment. The therapeutic effects of CGP 3466B were still present after a washout period of 3 weeks, implying that the effects were not symptomatic. These data are the first to show that the systemic administration of CGP 3466B is able to prevent the development of MPTP-induced motor symptoms in primates. This compound may have great value for inhibiting the progression of the neurodegenerative process in patients with Parkinson's disease.
Collapse
Affiliation(s)
- G Andringa
- Department of Psychoneuropharmacology, Faculty of Medicine, University of Nijmegen, The Netherlands
| | | |
Collapse
|