1
|
Emborg ME, Metzger JM, D'Amour K, Colwell JC, Neumann LC, Zhang A, Federoff HJ. Advantages and challenges of using allogeneic vs. autologous sources for neuronal cell replacement in Parkinson's disease: Insights from non-human primate studies. Brain Res Bull 2025; 224:111297. [PMID: 40086764 PMCID: PMC12036832 DOI: 10.1016/j.brainresbull.2025.111297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/25/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Intracerebral grafting of dopamine-producing cells is proposed as a strategy to replace the typical neurons lost to Parkinson's disease (PD) and improve PD motor symptoms. Non-human primate studies have provided clues on the relationship between the host's immune response and grafting success. Herein, we discuss how the host's immune system differentially affects the graft depending on the origin of the cells and reflect on the advantages and limitations of the immune paradigms utilized to assess graft-related outcomes. We also consider new strategies to minimize or circumvent the host's immunological response and related preclinical research needed to identify the most promising new approaches to be translated into the clinic.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Medical Physics, University of Wisconsin-Madison, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, USA.
| | - Jeanette M Metzger
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | | | - Julia C Colwell
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, USA
| | - Lindsey C Neumann
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | - Ai Zhang
- Genentech, South San Francisco, CA, USA
| | - Howard J Federoff
- Kenai Therapeutics, San Diego, CA, USA; Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
2
|
DeFranco JP, Telling GC. The Evolution of Experimental Rodent Models for Prion Diseases. J Neurochem 2025; 169:e70039. [PMID: 40108932 PMCID: PMC11968085 DOI: 10.1111/jnc.70039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Prion diseases are a group of fatal, neurodegenerative diseases that affect animals and humans. These diseases are characterized by the conformational conversion of normal, host-encoded PrPC into a disease-causing prion isoform, PrPSc. Significant advancements in biological, genetic, and prion research have led to the capability of studying this pathogenetic process using recombinant proteins, ex vivo systems, in vitro models, and mammalian hosts, the latter being the gold standard for assaying prion infectivity, transmission, and strain evolution. While devoid of nucleic acid, prions encipher strain information by the conformation of their constituent infectious proteins, with diversity altering pathogenesis, host-range dynamics, and the efficacy of therapeutics. To properly study the strain properties of natural prions and develop appropriate therapeutic strategies, it is essential to utilize models that authentically recapitulate these infectious agents in experimental mammalian hosts. In this review, we examine the evolution of research on prion diseases using non-transgenic and transgenic animals, primarily focusing on rodent models. We discuss the successes and limitations of each experimental system and provide insights based on recent findings in novel gene-targeted mice.
Collapse
Affiliation(s)
- Joseph P. DeFranco
- Prion Research Center, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Glenn C. Telling
- Prion Research Center, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
3
|
Espargaró A, Álvarez-Berbel I, Busquets MA, Sabate R. In Vivo Assays for Amyloid-Related Diseases. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2024; 17:433-458. [PMID: 38598824 DOI: 10.1146/annurev-anchem-061622-023326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Amyloid-related diseases, such as Alzheimer's and Parkinson's disease, are devastating conditions caused by the accumulation of abnormal protein aggregates known as amyloid fibrils. While assays involving animal models are essential for understanding the pathogenesis and developing therapies, a wide array of standard analytical techniques exists to enhance our understanding of these disorders. These techniques provide valuable information on the formation and propagation of amyloid fibrils, as well as the pharmacokinetics and pharmacodynamics of candidate drugs. Despite ethical concerns surrounding animal use, animal models remain vital tools in the search for treatments. Regardless of the specific animal model chosen, the analytical methods used are usually standardized. Therefore, the main objective of this review is to categorize and outline the primary analytical methods used in in vivo assays for amyloid-related diseases, highlighting their critical role in furthering our understanding of these disorders and developing effective therapies.
Collapse
Affiliation(s)
- Alba Espargaró
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Irene Álvarez-Berbel
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
| | - Maria Antònia Busquets
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sabate
- 1Department of Pharmacy and Pharmaceutical Technology and Department of Physical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain;
- 2Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Han B, Liang W, Li XJ, Li S, Yan S, Tu Z. Large animal models for Huntington's disease research. Zool Res 2024; 45:275-283. [PMID: 38485497 PMCID: PMC11017086 DOI: 10.24272/j.issn.2095-8137.2023.199] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/05/2023] [Indexed: 03/19/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder for which there is currently no effective treatment available. Consequently, the development of appropriate disease models is critical to thoroughly investigate disease progression. The genetic basis of HD involves the abnormal expansion of CAG repeats in the huntingtin ( HTT) gene, leading to the expansion of a polyglutamine repeat in the HTT protein. Mutant HTT carrying the expanded polyglutamine repeat undergoes misfolding and forms aggregates in the brain, which precipitate selective neuronal loss in specific brain regions. Animal models play an important role in elucidating the pathogenesis of neurodegenerative disorders such as HD and in identifying potential therapeutic targets. Due to the marked species differences between rodents and larger animals, substantial efforts have been directed toward establishing large animal models for HD research. These models are pivotal for advancing the discovery of novel therapeutic targets, enhancing effective drug delivery methods, and improving treatment outcomes. We have explored the advantages of utilizing large animal models, particularly pigs, in previous reviews. Since then, however, significant progress has been made in developing more sophisticated animal models that faithfully replicate the typical pathology of HD. In the current review, we provide a comprehensive overview of large animal models of HD, incorporating recent findings regarding the establishment of HD knock-in (KI) pigs and their genetic therapy. We also explore the utilization of large animal models in HD research, with a focus on sheep, non-human primates (NHPs), and pigs. Our objective is to provide valuable insights into the application of these large animal models for the investigation and treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Bofeng Han
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-Human Primate Research, Guangzhou, Guangdong 510632, China
| | - Weien Liang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-Human Primate Research, Guangzhou, Guangdong 510632, China
| | - Xiao-Jiang Li
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-Human Primate Research, Guangzhou, Guangdong 510632, China
| | - Shihua Li
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-Human Primate Research, Guangzhou, Guangdong 510632, China
| | - Sen Yan
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-Human Primate Research, Guangzhou, Guangdong 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China. E-mail:
| | - Zhuchi Tu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-Human Primate Research, Guangzhou, Guangdong 510632, China. E-mail:
| |
Collapse
|
5
|
Sanfeliu C, Bartra C, Suñol C, Rodríguez-Farré E. New insights in animal models of neurotoxicity-induced neurodegeneration. Front Neurosci 2024; 17:1248727. [PMID: 38260026 PMCID: PMC10800989 DOI: 10.3389/fnins.2023.1248727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
The high prevalence of neurodegenerative diseases is an unintended consequence of the high longevity of the population, together with the lack of effective preventive and therapeutic options. There is great pressure on preclinical research, and both old and new models of neurodegenerative diseases are required to increase the pipeline of new drugs for clinical testing. We review here the main models of neurotoxicity-based animal models leading to central neurodegeneration. Our main focus was on studying how changes in neurotransmission and neuroinflammation, mainly in rodent models, contribute to harmful processes linked to neurodegeneration. The majority of the models currently in use mimic Parkinson's disease (PD) and Alzheimer's disease (AD), which are the most common neurodegenerative conditions in older adults. AD is the most common age-related dementia, whereas PD is the most common movement disorder with also cases of dementia. Several natural toxins and xenobiotic agents induce dopaminergic neurodegeneration and can reproduce neuropathological traits of PD. The literature analysis of MPTP, 6-OH-dopamine, and rotenone models suggested the latter as a useful model when specific doses of rotenone were administrated systemically to C57BL/6 mice. Cholinergic neurodegeneration is mainly modelled with the toxin scopolamine, which is a useful rodent model for the screening of protective drugs against cognitive decline and AD. Several agents have been used to model neuroinflammation-based neurodegeneration and dementia in AD, including lipopolysaccharide (LPS), streptozotocin, and monomeric C-reactive protein. The bacterial agent LPS makes a useful rodent model for testing anti-inflammatory therapies to halt the development and severity of AD. However, neurotoxin models might be more useful than genetic models for drug discovery in PD but that is not the case in AD where they cannot beat the new developments in transgenic mouse models. Overall, we should work using all available models, either in vivo, in vitro, or in silico, considering the seriousness of the moment and urgency of developing effective drugs.
Collapse
Affiliation(s)
- Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Clara Bartra
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Cristina Suñol
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eduard Rodríguez-Farré
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
6
|
Sabeva N, Castro W, Acosta YF, Ferchmin PA, Eterović VA, Sierra-Mercado D, Rios NP, Rivas-Tumanyan S, Martins AH. Determining the safety of the tobacco cembranoid (1S,2E,4R,6R,7E,11E)-Cembratriene-4,6-diol (4R): A translational study in nonhuman primates. Toxicol Appl Pharmacol 2024; 482:116772. [PMID: 38036230 PMCID: PMC10872440 DOI: 10.1016/j.taap.2023.116772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
The tobacco cembranoid known as (1S,2E,4R,6R,7E,11E)-2,7,11-cembratriene-4,6-diol (4R) has been shown to offer neuroprotection against conditions such as brain ischemia, systemic inflammation, Parkinson's disease, and organophosphate toxicity in rodents. Previous safety studies conducted on male and female Sprague Dawley rats revealed no significant side effects following a single injection of 4R at varying concentrations (6, 24, or 98 mg/kg of body weight). This study aimed to assess the potential of 4R for clinical trials in neurotherapy in male nonhuman primates. Ten macaques (Macacca mulatta) were randomly separated into two groups of 5 and then intravenously injected with 4R or vehicle for 11 consecutive days at a dose of 1.4 mg/kg. Throughout the study, we monitored brain activity by electroencephalogram, somatosensory evoked potentials, and transcranial motor evoked potentials on days 0, 4, 8, and 12 and found no significant changes. The spontaneous behavior of the primates remained unaffected by the treatment. Minor hematological and blood composition variations were also detected in the experimental animals but lacked clinical significance. In conclusion, our results reinforce the notion that 4R is non-toxic in nonhuman primates under the conditions of this study.
Collapse
Affiliation(s)
- Nadezhda Sabeva
- Department of Neurosciences, Universidad Central del Caribe, School of Medicine, Bayamón, PR 00956, USA
| | | | - Yancy Ferrer Acosta
- Department of Neurosciences, Universidad Central del Caribe, School of Medicine, Bayamón, PR 00956, USA; Department of Anatomy and Neurobiology University of Puerto Rico, Medical Sciences Campus, Guillermo Arbona, Área de Centro Médico Río Piedras, PR 00935, USA.
| | | | | | - Demetrio Sierra-Mercado
- Department of Anatomy and Neurobiology University of Puerto Rico, Medical Sciences Campus, Guillermo Arbona, Área de Centro Médico Río Piedras, PR 00935, USA.
| | - Naydi Pérez Rios
- Biostatistics, Epidemiology, and Research Design Core, Hispanic Alliance for Clinical and Translational Research, University of Puerto Rico, Medical Sciences Campus, Área de Centro Médico Río Piedras, PR 00935, USA.
| | - Sona Rivas-Tumanyan
- Biostatistics, Epidemiology, and Research Design Core, Hispanic Alliance for Clinical and Translational Research, University of Puerto Rico, Medical Sciences Campus, Área de Centro Médico Río Piedras, PR 00935, USA; Office of Assistant Dean for Research and Department of Surgical Sciences, School of Dental Medicine, University of Puerto Rico Medical Sciences Campus, Área de Centro Médico Río Piedras, PR 00935, USA.
| | - Antonio H Martins
- Department of Pharmacology and Toxicology, University of Puerto Rico, Medical Sciences Campus, Guillermo Arbona, Área de Centro Médico Río Piedras, PR 00935, USA.
| |
Collapse
|
7
|
Emborg ME, Gambardella JC, Zhang A, Federoff HJ. Autologous vs heterologous cell replacement strategies for Parkinson disease and other neurologic diseases. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:41-56. [PMID: 39341662 DOI: 10.1016/b978-0-323-90120-8.00010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Successful cell replacement strategies for brain repair depend on graft integration into the neural network, which is affected by the immune response to the grafted cells. Using Parkinson disease as an example, in this chapter, we consider the immune system interaction and its role in autologous vs heterologous graft survival and integration, as well as past and emerging strategies to overcome the immunologic response. We also reflect on the role of nonhuman primate research to assess novel approaches and consider the role of different stakeholders on advancing the most promising new approaches into the clinic.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States.
| | - Julia C Gambardella
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Ai Zhang
- Aspen Neuroscience, San Diego, CA, United States
| | - Howard J Federoff
- Kenai Therapeutics, San Diego, CA, United States; Georgetown University Medical Center, Georgetown, Washington, DC, United States
| |
Collapse
|
8
|
Polis B, Samson AO. Addressing the Discrepancies Between Animal Models and Human Alzheimer's Disease Pathology: Implications for Translational Research. J Alzheimers Dis 2024; 98:1199-1218. [PMID: 38517793 DOI: 10.3233/jad-240058] [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] [Indexed: 03/24/2024]
Abstract
Animal models, particularly transgenic mice, are extensively used in Alzheimer's disease (AD) research to emulate key disease hallmarks, such as amyloid plaques and neurofibrillary tangles formation. Although these models have contributed to our understanding of AD pathogenesis and can be helpful in testing potential therapeutic interventions, their reliability is dubious. While preclinical studies have shown promise, clinical trials often yield disappointing results, highlighting a notable gap and disparity between animal models and human AD pathology. Existing models frequently overlook early-stage human pathologies and other key AD characteristics, thereby limiting their application in identifying optimal therapeutic interventions. Enhancing model reliability necessitates rigorous study design, comprehensive behavioral evaluations, and biomarker utilization. Overall, a nuanced understanding of each model's neuropathology, its fidelity to human AD, and its limitations is essential for accurate interpretation and successful translation of findings. This article analyzes the discrepancies between animal models and human AD pathology that complicate the translation of findings from preclinical studies to clinical applications. We also delve into AD pathogenesis and attributes to propose a new perspective on this pathology and deliberate over the primary limitations of key experimental models. Additionally, we discuss several fundamental problems that may explain the translational failures and suggest some possible directions for more effective preclinical studies.
Collapse
Affiliation(s)
- Baruh Polis
- Bar-Ilan University Azrieli Faculty of Medicine, Safed, Israel
| | | |
Collapse
|
9
|
Sharma H, Chang KA, Hulme J, An SSA. Mammalian Models in Alzheimer's Research: An Update. Cells 2023; 12:2459. [PMID: 37887303 PMCID: PMC10605533 DOI: 10.3390/cells12202459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
A form of dementia distinct from healthy cognitive aging, Alzheimer's disease (AD) is a complex multi-stage disease that currently afflicts over 50 million people worldwide. Unfortunately, previous therapeutic strategies developed from murine models emulating different aspects of AD pathogenesis were limited. Consequently, researchers are now developing models that express several aspects of pathogenesis that better reflect the clinical situation in humans. As such, this review seeks to provide insight regarding current applications of mammalian models in AD research by addressing recent developments and characterizations of prominent transgenic models and their contributions to pathogenesis as well as discuss the advantages, limitations, and application of emerging models that better capture genetic heterogeneity and mixed pathologies observed in the clinical situation.
Collapse
Affiliation(s)
- Himadri Sharma
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| | - Keun-A Chang
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea
| | - John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| |
Collapse
|
10
|
Kaya I, Nilsson A, Luptáková D, He Y, Vallianatou T, Bjärterot P, Svenningsson P, Bezard E, Andrén PE. Spatial lipidomics reveals brain region-specific changes of sulfatides in an experimental MPTP Parkinson's disease primate model. NPJ Parkinsons Dis 2023; 9:118. [PMID: 37495571 PMCID: PMC10372136 DOI: 10.1038/s41531-023-00558-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Abstract
Metabolism of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) to the neurotoxin MPP+ in the brain causes permanent Parkinson's disease-like symptoms by destroying dopaminergic neurons in the pars compacta of the substantia nigra in humans and non-human primates. However, the complete molecular pathology underlying MPTP-induced parkinsonism remains poorly understood. We used dual polarity matrix-assisted laser desorption/ionization mass spectrometry imaging to thoroughly image numerous glycerophospholipids and sphingolipids in coronal brain tissue sections of MPTP-lesioned and control non-human primate brains (Macaca mulatta). The results revealed specific distributions of several sulfatide lipid molecules based on chain-length, number of double bonds, and importantly, hydroxylation stage. More specifically, certain long-chain hydroxylated sulfatides with polyunsaturated chains in the molecular structure were depleted within motor-related brain regions in the MPTP-lesioned animals, e.g., external and internal segments of globus pallidus and substantia nigra pars reticulata. In contrast, certain long-chain non-hydroxylated sulfatides were found to be elevated within the same brain regions. These findings demonstrate region-specific dysregulation of sulfatide metabolism within the MPTP-lesioned macaque brain. The depletion of long-chain hydroxylated sulfatides in the MPTP-induced pathology indicates oxidative stress and oligodendrocyte/myelin damage within the pathologically relevant brain regions. Hence, the presented findings improve our current understanding of the molecular pathology of MPTP-induced parkinsonism within primate brains, and provide a basis for further research regarding the role of dysregulated sulfatide metabolism in PD.
Collapse
Affiliation(s)
- Ibrahim Kaya
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Dominika Luptáková
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Yachao He
- Section of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Theodosia Vallianatou
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Patrik Bjärterot
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Svenningsson
- Section of Neurology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Erwan Bezard
- University of Bordeaux, CNRS, IMN, UMR 5293, F-33000, Bordeaux, France
| | - Per E Andrén
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
11
|
Metzger JM, Wang Y, Neuman SS, Snow KJ, Murray SA, Lutz CM, Bondarenko V, Felton J, Gimse K, Xie R, Li D, Zhao Y, Flowers MT, Simmons HA, Roy S, Saha K, Levine JE, Emborg ME, Gong S. Efficient in vivo neuronal genome editing in the mouse brain using nanocapsules containing CRISPR-Cas9 ribonucleoproteins. Biomaterials 2023; 293:121959. [PMID: 36527789 PMCID: PMC9868115 DOI: 10.1016/j.biomaterials.2022.121959] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Genome editing of somatic cells via clustered regularly interspaced short palindromic repeats (CRISPR) offers promise for new therapeutics to treat a variety of genetic disorders, including neurological diseases. However, the dense and complex parenchyma of the brain and the post-mitotic state of neurons make efficient genome editing challenging. In vivo delivery systems for CRISPR-Cas proteins and single guide RNA (sgRNA) include both viral vectors and non-viral strategies, each presenting different advantages and disadvantages for clinical application. We developed non-viral and biodegradable PEGylated nanocapsules (NCs) that deliver preassembled Cas9-sgRNA ribonucleoproteins (RNPs). Here, we show that the RNP NCs led to robust genome editing in neurons following intracerebral injection into the healthy mouse striatum. Genome editing was predominantly observed in medium spiny neurons (>80%), with occasional editing in cholinergic, calretinin, and parvalbumin interneurons. Glial activation was minimal and was localized along the needle tract. Our results demonstrate that the RNP NCs are capable of safe and efficient neuronal genome editing in vivo.
Collapse
Affiliation(s)
- Jeanette M Metzger
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yuyuan Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Samuel S Neuman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Kathy J Snow
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | | | - Viktoriya Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jesi Felton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Kirstan Gimse
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Dongdong Li
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yi Zhao
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Matthew T Flowers
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Subhojit Roy
- Departments of Pathology and Neuroscience, University of California-San Diego, San Diego, CA, 92093, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Marina E Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Shaoqin Gong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| |
Collapse
|
12
|
Nainu F, Mamada SS, Harapan H, Emran TB. Inflammation-Mediated Responses in the Development of Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:39-70. [PMID: 36949305 DOI: 10.1007/978-981-19-7376-5_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Since its first description over a century ago, neurodegenerative diseases (NDDs) have impaired the lives of millions of people worldwide. As one of the major threats to human health, NDDs are characterized by progressive loss of neuronal structure and function, leading to the impaired function of the CNS. While the precise mechanisms underlying the emergence of NDDs remains elusive, association of neuroinflammation with the emergence of NDDs has been suggested. The immune system is tightly controlled to maintain homeostatic milieu and failure in doing so has been shown catastrophic. Here, we review current concepts on the cellular and molecular drivers responsible in the induction of neuroinflammation and how such event further promotes neuronal damage leading to neurodegeneration. Experimental data generated from cell culture and animal studies, gross and molecular pathologies of human CNS samples, and genome-wide association study are discussed to provide deeper insights into the mechanistic details of neuroinflammation and its roles in the emergence of NDDs.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Sukamto S Mamada
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Harapan Harapan
- School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| |
Collapse
|
13
|
Gomez Limia C, Baird M, Schwartz M, Saxena S, Meyer K, Wein N. Emerging Perspectives on Gene Therapy Delivery for Neurodegenerative and Neuromuscular Disorders. J Pers Med 2022; 12:1979. [PMID: 36556200 PMCID: PMC9788053 DOI: 10.3390/jpm12121979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022] Open
Abstract
Neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD) and Parkinson's Disease (PD), are a group of heterogeneous diseases that mainly affect central nervous system (CNS) functions. A subset of NDDs exhibit CNS dysfunction and muscle degeneration, as observed in Gangliosidosis 1 (GM1) and late stages of PD. Neuromuscular disorders (NMDs) are a group of diseases in which patients show primary progressive muscle weaknesses, including Duchenne Muscular Dystrophy (DMD), Pompe disease, and Spinal Muscular Atrophy (SMA). NDDs and NMDs typically have a genetic component, which affects the physiological functioning of critical cellular processes, leading to pathogenesis. Currently, there is no cure or efficient treatment for most of these diseases. More than 200 clinical trials have been completed or are currently underway in order to establish safety, tolerability, and efficacy of promising gene therapy approaches. Thus, gene therapy-based therapeutics, including viral or non-viral delivery, are very appealing for the treatment of NDDs and NMDs. In particular, adeno-associated viral vectors (AAV) are an attractive option for gene therapy for NDDs and NMDs. However, limitations have been identified after systemic delivery, including the suboptimal capacity of these therapies to traverse the blood-brain barrier (BBB), degradation of the particles during the delivery, high reactivity of the patient's immune system during the treatment, and the potential need for redosing. To circumvent these limitations, several preclinical and clinical studies have suggested intrathecal (IT) delivery to target the CNS and peripheral organs via cerebrospinal fluid (CSF). CSF administration can vastly improve the delivery of small molecules and drugs to the brain and spinal cord as compared to systemic delivery. Here, we review AAV biology and vector design elements, different therapeutic routes of administration, and highlight CSF delivery as an attractive route of administration. We discuss the different aspects of neuromuscular and neurodegenerative diseases, such as pathogenesis, the landscape of mutations, and the biological processes associated with the disease. We also describe the hallmarks of NDDs and NMDs as well as discuss current therapeutic approaches and clinical progress in viral and non-viral gene therapy and enzyme replacement strategies for those diseases.
Collapse
Affiliation(s)
- Cintia Gomez Limia
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Megan Baird
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Maura Schwartz
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Smita Saxena
- Department of Neurology, Inselspital, 3010 Bern, Switzerland
| | - Kathrin Meyer
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
14
|
Levin LA, Patrick C, Choudry NB, Sharif NA, Goldberg JL. Neuroprotection in neurodegenerations of the brain and eye: Lessons from the past and directions for the future. Front Neurol 2022; 13:964197. [PMID: 36034312 PMCID: PMC9412944 DOI: 10.3389/fneur.2022.964197] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Background Neurological and ophthalmological neurodegenerative diseases in large part share underlying biology and pathophysiology. Despite extensive preclinical research on neuroprotection that in many cases bridges and unifies both fields, only a handful of neuroprotective therapies have succeeded clinically in either. Main body Understanding the commonalities among brain and neuroretinal neurodegenerations can help develop innovative ways to improve translational success in neuroprotection research and emerging therapies. To do this, analysis of why translational research in neuroprotection fails necessitates addressing roadblocks at basic research and clinical trial levels. These include optimizing translational approaches with respect to biomarkers, therapeutic targets, treatments, animal models, and regulatory pathways. Conclusion The common features of neurological and ophthalmological neurodegenerations are useful for outlining a path forward that should increase the likelihood of translational success in neuroprotective therapies.
Collapse
Affiliation(s)
- Leonard A. Levin
- Departments of Ophthalmology and Visual Sciences, Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Nozhat B. Choudry
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| | - Najam A. Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
15
|
de Groot NG, de Groot N, de Vos-Rouweler AJM, Louwerse A, Bruijnesteijn J, Bontrop RE. Dynamic evolution of Mhc haplotypes in cynomolgus macaques of different geographic origins. Immunogenetics 2022; 74:409-429. [PMID: 35084546 PMCID: PMC8792142 DOI: 10.1007/s00251-021-01249-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/26/2021] [Indexed: 11/05/2022]
Abstract
The major histocompatibility complex (MHC) plays a key role in immune defense, and the Mhc genes of cynomolgus macaque display a high degree of polymorphism. Based on their geographic distribution, different populations of cynomolgus macaques are recognized. Here we present the characterization of the Mhc class I and II repertoire of a large pedigreed group of cynomolgus macaques originating from the mainland north of the isthmus of Kra (N = 42). Segregation analyses resulted in the definition of 81 unreported Mafa-A/B/DRB/DQ/DP haplotypes, which include 32 previously unknown DRB regions. In addition, we report 13 newly defined Mafa-A/B/DRB/DQ/DP haplotypes in a group of cynomolgus macaques originating from the mainland south of the isthmus of Kra/Maritime Southeast Asia (N = 16). A relatively high level of sharing of Mafa-A (51%) and Mafa-B (40%) lineage groups is observed between the populations native to the north and the south of isthmus of Kra. At the allelic level, however, the Mafa-A/B haplotypes seem to be characteristic of a population. An overall comparison of all currently known data revealed that each geographic population has its own specific combinations of Mhc class I and II haplotypes. This illustrates the dynamic evolution of the cynomolgus macaque Mhc region, which was most likely generated by recombination and maintained by selection due to the differential pathogenic pressures encountered in different geographic areas.
Collapse
Affiliation(s)
- Natasja G de Groot
- Comparative Genetics and Refinement, Biomedical Primate Research Centre, 2288 GJ, Rijswijk, The Netherlands.
| | - Nanine de Groot
- Comparative Genetics and Refinement, Biomedical Primate Research Centre, 2288 GJ, Rijswijk, The Netherlands
| | | | - Annet Louwerse
- Animal Science Department, Biomedical Primate Research Centre, 2288 GJ, Rijswijk, The Netherlands
| | - Jesse Bruijnesteijn
- Comparative Genetics and Refinement, Biomedical Primate Research Centre, 2288 GJ, Rijswijk, The Netherlands
| | - Ronald E Bontrop
- Comparative Genetics and Refinement, Biomedical Primate Research Centre, 2288 GJ, Rijswijk, The Netherlands
- Theoretical Biology and Bioinformatics, Utrecht University, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|
16
|
Niewold P, Ijsselsteijn ME, Verreck FAW, Ottenhoff THM, Joosten SA. An imaging mass cytometry immunophenotyping panel for non-human primate tissues. Front Immunol 2022; 13:915157. [PMID: 35911721 PMCID: PMC9334813 DOI: 10.3389/fimmu.2022.915157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
It has recently become clear that spatial organization contributes to cellular function and that expanding our knowledge on cellular organization is essential to further our understanding of processes in health and disease. Imaging mass cytometry enables high dimensional imaging of tissue while preserving spatial context and is therefore a suitable tool to unravel spatial relationships between cells. As availability of human tissue collected over the course of disease or infection is limited, preclinical models are a valuable source of such material. Non-human primate models are used for translational research as their anatomy, physiology and immune system closely resemble those of humans due to close evolutionary proximity. Tissue from non-human primate studies is often preserved large archives encompassing a range of conditions and organs. However, knowledge on antibody clones suitable for FFPE tissue of non-human primate origin is very limited. Here, we present an imaging mass cytometry panel development pipeline which enables the selection and incorporation of antibodies for imaging of non-human primate tissue. This has resulted in an 18-marker backbone panel which enables visualization of a broad range of leukocyte subsets in rhesus and cynomolgus macaque tissues. This high-dimensional imaging mass cytometry panel can be used to increase our knowledge of cellular organization within tissues and its effect on outcome of disease.
Collapse
Affiliation(s)
- Paula Niewold
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, Netherlands
| | | | - Frank A. W. Verreck
- Section of Tuberculosis (TB) Research and Immunology, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, Netherlands
- *Correspondence: Simone A. Joosten,
| |
Collapse
|
17
|
Bashirzade AA, Zabegalov KN, Volgin AD, Belova AS, Demin KA, de Abreu MS, Babchenko VY, Bashirzade KA, Yenkoyan KB, Tikhonova MA, Amstislavskaya TG, Kalueff AV. Modeling neurodegenerative disorders in zebrafish. Neurosci Biobehav Rev 2022; 138:104679. [PMID: 35490912 DOI: 10.1016/j.neubiorev.2022.104679] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/11/2022] [Accepted: 04/24/2022] [Indexed: 12/15/2022]
Abstract
Neurodegeneration is a major cause of Alzheimer's, Parkinson's, Huntington's, multiple and amyotrophic lateral sclerosis, pontocerebellar hypoplasia, dementia and other related brain disorders. Their complex pathogenesis commonly includes genetic and neurochemical deficits, misfolded protein toxicity, demyelination, apoptosis and mitochondrial dysfunctions. Albeit differing in specific underlying mechanisms, neurodegenerative disorders typically display evolutionarily conserved mechanisms across taxa. Here, we review the role of zebrafish models in recapitulating major human and rodent neurodegenerative conditions, demonstrating this species as a highly relevant experimental model for research on neurodegenerative diseases, and discussing how these fish models can further clarify the underlying genetic, neurochemical, neuroanatomical and behavioral pathogenic mechanisms.
Collapse
Affiliation(s)
- Alim A Bashirzade
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | | | - Andrey D Volgin
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Alisa S Belova
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Granov Scientific Research Center of Radiology and Surgical Technologies, St. Petersburg, Russia; Almazov Medical Research Center, St. Petersburg, Russia
| | | | - Vladislav Ya Babchenko
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Kseniya A Bashirzade
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia
| | - Konstantin B Yenkoyan
- Neuroscience Laboratory, COBRAIN Center, M Heratsi Yerevan State Medical University, Yerevan, Armenia; COBRAIN Center - Scientific Educational Center for Fundamental Brain Research, Yerevan, Armenia
| | - Maria A Tikhonova
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Tamara G Amstislavskaya
- Novosibirsk State University, Institute of Medicine and Psychology, Novosibirsk, Russia; Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia
| | - Allan V Kalueff
- The Russian Academy of Sciences, Moscow, Russia; Ural Federal University, Yekaterinburg, Russia; COBRAIN Center - Scientific Educational Center for Fundamental Brain Research, Yerevan, Armenia.
| |
Collapse
|
18
|
Alpha-synuclein and tau are abundantly expressed in the ENS of the human appendix and monkey cecum. PLoS One 2022; 17:e0269190. [PMID: 35687573 PMCID: PMC9187115 DOI: 10.1371/journal.pone.0269190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/16/2022] [Indexed: 11/20/2022] Open
Abstract
α-Synuclein (α-syn) proteinopathy in the neurons of the Enteric Nervous System (ENS) is proposed to have a critical role in Parkinson's disease (PD) onset and progression. Interestingly, the ENS of the human appendix harbors abundant α-syn and appendectomy has been linked to a decreased risk and delayed onset of PD, suggesting that the appendix may influence PD pathology. Common marmosets and rhesus macaques lack a distinct appendix (a narrow closed-end appendage with a distinct change in diameter at the junction with the cecum), yet the cecal microanatomy of these monkeys is similar to the human appendix. Sections of human appendix (n = 3) and ceca from common marmosets (n = 4) and rhesus macaques (n = 3) were evaluated to shed light on the microanatomy and the expression of PD-related proteins. Analysis confirmed that the human appendix and marmoset and rhesus ceca present thick walls comprised of serosa, muscularis externa, submucosa, and mucosa plus abundant lymphoid tissue. Across all three species, the myenteric plexus of the ENS was located within the muscularis externa with nerve fibers innervating all layers of the appendix/ceca. Expression of α-syn and tau in the appendix/cecum was present within myenteric ganglia and along nerve fibers of the muscularis externa and mucosa in all species. In the myenteric ganglia α-syn, p-α-syn, tau and p-tau immunoreactivities (ir) were not significantly different across species. The percent area above threshold of α-syn-ir and tau-ir in the nerve fibers of the muscularis externa and mucosa were greater in the human appendix than in the NHP ceca (α-syn-ir p<0.05; tau-ir p<0.05). Overall, this study provides critical translational evidence that the common marmoset and rhesus macaque ceca are remarkably similar to the human appendix and, thus, that these NHP species are suitable for studying the development of PD linked to α-syn and tau pathological changes in the ENS.
Collapse
|
19
|
Yoo HS, Shanmugalingam U, Smith PD. Potential roles of branched-chain amino acids in neurodegeneration. Nutrition 2022; 103-104:111762. [DOI: 10.1016/j.nut.2022.111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/12/2022] [Accepted: 05/31/2022] [Indexed: 10/31/2022]
|
20
|
Tello JA, Williams HE, Eppler RM, Steinhilb ML, Khanna M. Animal Models of Neurodegenerative Disease: Recent Advances in Fly Highlight Innovative Approaches to Drug Discovery. Front Mol Neurosci 2022; 15:883358. [PMID: 35514431 PMCID: PMC9063566 DOI: 10.3389/fnmol.2022.883358] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases represent a formidable challenge to global health. As advances in other areas of medicine grant healthy living into later decades of life, aging diseases such as Alzheimer's disease (AD) and other neurodegenerative disorders can diminish the quality of these additional years, owed largely to the lack of efficacious treatments and the absence of durable cures. Alzheimer's disease prevalence is predicted to more than double in the next 30 years, affecting nearly 15 million Americans, with AD-associated costs exceeding $1 billion by 2050. Delaying onset of AD and other neurodegenerative diseases is critical to improving the quality of life for patients and reducing the burden of disease on caregivers and healthcare systems. Significant progress has been made to model disease pathogenesis and identify points of therapeutic intervention. While some researchers have contributed to our understanding of the proteins and pathways that drive biological dysfunction in disease using in vitro and in vivo models, others have provided mathematical, biophysical, and computational technologies to identify potential therapeutic compounds using in silico modeling. The most exciting phase of the drug discovery process is now: by applying a target-directed approach that leverages the strengths of multiple techniques and validates lead hits using Drosophila as an animal model of disease, we are on the fast-track to identifying novel therapeutics to restore health to those impacted by neurodegenerative disease.
Collapse
Affiliation(s)
- Judith A. Tello
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Haley E. Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Robert M. Eppler
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - Michelle L. Steinhilb
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
- Department of Molecular Pathobiology, New York University, New York, NY, United States
| |
Collapse
|
21
|
Haney MJ, Yuan H, Shipley ST, Wu Z, Zhao Y, Pate K, Frank JE, Massoud N, Stewart PW, Perlmutter JS, Batrakova EV. Biodistribution of Biomimetic Drug Carriers, Mononuclear Cells, and Extracellular Vesicles, in Nonhuman Primates. Adv Biol (Weinh) 2022; 6:e2101293. [PMID: 34939369 PMCID: PMC8825682 DOI: 10.1002/adbi.202101293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/23/2021] [Indexed: 02/03/2023]
Abstract
Discovery of novel drug delivery systems to the brain remains a key task for successful treatment of neurodegenerative disorders. Herein, the biodistribution of immunocyte-based carriers, peripheral blood mononuclear cells (PBMCs), and monocyte-derived EVs are investigated in adult rhesus macaques using longitudinal PET/MRI imaging. 64 Cu-labeled drug carriers are introduced via different routes of administration: intraperitoneal (IP), intravenous (IV), or intrathecal (IT) injection. Whole body PET/MRI (or PET/CT) images are acquired at 1, 24, and 48 h post injection of 64 Cu-labeled drug carriers, and standardized uptake values (SUVmean and SUVmax ) in the main organs are estimated. The brain retention for both types of carriers increases based on route of administration: IP < IV < IT. Importantly, a single IT injection of PBMCs produces higher brain retention compared to IT injection of EVs. In contrast, EVs show superior brain accumulation compared to the cells when administered via IP and IV routes, respectively. Finally, a comprehensive chemistry panel of blood samples demonstrates no cytotoxic effects of either carrier. Overall, living cells and EVs have a great potential to be used for drug delivery to the brain. When identifying the ideal drug carrier, the route of administration could make big differences in CNS drug delivery.
Collapse
Affiliation(s)
- Matthew J. Haney
- Center for NanotechFnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA;,Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hong Yuan
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steven T. Shipley
- Division of Comparative Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Zhanhong Wu
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yuling Zhao
- Center for NanotechFnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA;,Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kelly Pate
- Division of Comparative Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan E. Frank
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nicole Massoud
- Division of Comparative Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paul W. Stewart
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joel S. Perlmutter
- School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Elena V. Batrakova
- Center for NanotechFnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA;,Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA;,Correspondence should be addressed to E.V.B. (), UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7362, Phone: 919-537-3712
| |
Collapse
|
22
|
A Novel, Automated, and Real-Time Method for the Analysis of Non-Human Primate Behavioral Patterns Using a Depth Image Sensor. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12010471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
By virtue of their upright locomotion, similar to that of humans, motion analysis of non-human primates has been widely used in order to better understand musculoskeletal biomechanics and neuroscience problems. Given the difficulty of conducting a marker-based infrared optical tracking system for the behavior analysis of primates, a 2-dimensional (D) video analysis has been applied. Distinct from a conventional marker-based optical tracking system, a depth image sensor system provides 3-D information on movement without any skin markers. The specific aim of this study was to develop a novel algorithm to analyze the behavioral patterns of non-human primates in a home cage using a depth image sensor. The behavioral patterns of nine monkeys in their home cage, including sitting, standing, and pacing, were captured using a depth image sensor. Thereafter, these were analyzed by observers’ manual assessment and the newly written automated program. We confirmed that the measurement results from the observers’ manual assessments and the automated program with depth image analysis were statistically identical.
Collapse
|
23
|
Haver HN, Scaglione KM. Dictyostelium discoideum as a Model for Investigating Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:759532. [PMID: 34776869 PMCID: PMC8578527 DOI: 10.3389/fncel.2021.759532] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/07/2021] [Indexed: 12/28/2022] Open
Abstract
The social amoeba Dictyostelium discoideum is a model organism that is used to investigate many cellular processes including chemotaxis, cell motility, cell differentiation, and human disease pathogenesis. While many single-cellular model systems lack homologs of human disease genes, Dictyostelium's genome encodes for many genes that are implicated in human diseases including neurodegenerative diseases. Due to its short doubling time along with the powerful genetic tools that enable rapid genetic screening, and the ease of creating knockout cell lines, Dictyostelium is an attractive model organism for both interrogating the normal function of genes implicated in neurodegeneration and for determining pathogenic mechanisms that cause disease. Here we review the literature involving the use of Dictyostelium to interrogate genes implicated in neurodegeneration and highlight key questions that can be addressed using Dictyostelium as a model organism.
Collapse
Affiliation(s)
- Holly N. Haver
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - K. Matthew Scaglione
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
- Department of Neurology, Duke University, Durham, NC, United States
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, NC, United States
| |
Collapse
|
24
|
Stonebarger GA, Bimonte-Nelson HA, Urbanski HF. The Rhesus Macaque as a Translational Model for Neurodegeneration and Alzheimer's Disease. Front Aging Neurosci 2021; 13:734173. [PMID: 34539388 PMCID: PMC8446616 DOI: 10.3389/fnagi.2021.734173] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/12/2021] [Indexed: 12/01/2022] Open
Abstract
A major obstacle to progress in understanding the etiology of normative and pathological human brain aging is the availability of suitable animal models for experimentation. The present article will highlight our current knowledge regarding human brain aging and neurodegeneration, specifically in the context of Alzheimer's disease (AD). Additionally, it will examine the use of the rhesus macaque monkey as a pragmatic translational animal model in which to study underlying causal mechanisms. Specifically, the discussion will focus on behavioral and protein-level brain changes that occur within the central nervous system (CNS) of aged monkeys, and compare them to the changes observed in humans during clinically normative aging and in AD.
Collapse
Affiliation(s)
- Gail A. Stonebarger
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
25
|
Darusman HS, Saepuloh U, Mariya SS, Sajuthi D, Schapiro SJ, Hau J. Increased expression of GAPDH in cynomolgus monkeys with spontaneous cognitive decline and amyloidopathy reminiscent of an Alzheimer's-type disease is reflected in the circulation. Am J Primatol 2021; 83:e23296. [PMID: 34196425 DOI: 10.1002/ajp.23296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023]
Abstract
Previous studies of aging cynomolgus monkeys from our group identified spontaneous age-associated cognitive declines associated with biomarkers and brain lesions reminiscent of Alzheimer's Disease (AD), in a proportion of aged monkeys. However, the molecular mechanisms that underlie the spontaneous amyloid disorders and cognitive declines observed in these affected monkeys have yet to be investigated in detail. Using reverse transcriptase quantitative real time PCR techniques, normalized to the ACTB housekeeping gene, we analyzed the expression patterns of a number of genes which have been implicated in amyloid and tau abnormalities, in well-characterized aged cynomolgus monkeys with cognitive decline. A significantly increased expression of the genes coding for glyceraldehyde 3-phosphate dehydrogenase (GAPDH), was found in aged-cognitive decline monkeys compared to age-matched healthy controls. GAPDH has been implicated in several neurodegenerative diseases and interacts with beta amyloid precursor proteins. These findings provide support for the utilization of cynomolgus macaques in translational preclinical research as valid spontaneous models in experimental investigations of the relationships among aging, cognitive decline, and the neuropathy of AD.
Collapse
Affiliation(s)
- Huda S Darusman
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia.,Faculty of Veterinary Medicine, IPB University, Bogor, Indonesia
| | - Uus Saepuloh
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia
| | - Sela S Mariya
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia
| | - Dondin Sajuthi
- Primate Research Center, Institute of Research and Community Service, Bogor Agricultural University (IPB University), Bogor, Indonesia.,Faculty of Veterinary Medicine, IPB University, Bogor, Indonesia
| | - Steven J Schapiro
- Department of Comparative Medicine, UTMD Anderson Cancer Center, Bastrop, Texas, USA.,Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jann Hau
- Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Erikson DW, Blue SW, Kaucher AV, Shnitko TA. LC-MS/MS measurement of endogenous oxytocin in the posterior pituitary and CSF of macaques: A pilot study. Peptides 2021; 140:170544. [PMID: 33811949 PMCID: PMC8462972 DOI: 10.1016/j.peptides.2021.170544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022]
Abstract
Oxytocin (OT) is a nanopeptide released into systemic circulation via the posterior pituitary (peripheral) and into the central nervous system via widespread OTergic pathways (central). Central OT plays a significant role in variety of functions from social and executive cognition to immune regulation. Many ongoing studies explore its therapeutic potential for variety of neuropsychiatric disorders. Measures of peripheral OT levels are most frequently used as an indicator of its concentration in the central nervous system in humans and animal models. In this study, LC-MS/MS was used to measure OT in pituitary samples collected from adult male macaque monkeys in order to explore the correlation between individual levels of OT in the CSF (central) and pituitary (peripheral). We quantified individual differences in the levels of OT in the pituitaries (44-151 ng/mg) and CSF (41-66 pg/mL) of these monkeys. A positive correlation between these two measures was identified. These preliminary results allow for future analyses to determine whether LC-MS/MS measures of peripheral OT can be used as markers of OT levels in the brain of nonhuman primates that serve as valuable models for many human neuropsychiatric disorders.
Collapse
Affiliation(s)
- D W Erikson
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006-3448, USA
| | - S W Blue
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006-3448, USA
| | - A V Kaucher
- Endocrine Technologies Core, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006-3448, USA
| | - T A Shnitko
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006-3448, USA.
| |
Collapse
|
27
|
Koizumi M, Nogami N, Owari K, Kawanobe A, Nakatani T, Seki K. Motility Profile of Captive-Bred Marmosets Revealed by a Long-Term In-Cage Monitoring System. Front Syst Neurosci 2021; 15:645308. [PMID: 33935661 PMCID: PMC8081884 DOI: 10.3389/fnsys.2021.645308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/26/2021] [Indexed: 11/13/2022] Open
Abstract
A quantitative evaluation of motility is crucial for studies employing experimental animals. Here, we describe the development of an in-cage motility monitoring method for new world monkeys using off-the-shelf components, and demonstrate its capability for long-term operation (e.g., a year). Based on this novel system, we characterized the motility of the common marmoset over different time scales (seconds, hours, days, and weeks). Monitoring of seven young animals belonging to two different age groups (sub-adult and young-adult) over a 231-day period revealed: (1) strictly diurnal activity (97.3% of movement during daytime), (2) short-cycle (∼20 s) transition in activity, and (3) bimodal diurnal activity including a "siesta" break. Additionally, while the mean duration of short-cycle activity, net daily activity, and diurnal activity changed over the course of development, 24-h periodicity remained constant. Finally, the method allowed for detection of progressive motility deterioration in a transgenic marmoset. Motility measurement offers a convenient way to characterize developmental and pathological changes in animals, as well as an economical and labor-free means for long-term evaluation in a wide range of basic and translational studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Kazuhiko Seki
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
28
|
Exploring the conservation of Alzheimer-related pathways between H. sapiens and C. elegans: a network alignment approach. Sci Rep 2021; 11:4572. [PMID: 33633188 PMCID: PMC7907373 DOI: 10.1038/s41598-021-83892-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder with an –as of yet– unclear etiology and pathogenesis. Research to unveil disease processes underlying AD often relies on the use of neurodegenerative disease model organisms, such as Caenorhabditis elegans. This study sought to identify biological pathways implicated in AD that are conserved in Homo sapiens and C. elegans. Protein–protein interaction networks were assembled for amyloid precursor protein (APP) and Tau in H. sapiens—two proteins whose aggregation is a hallmark in AD—and their orthologs APL-1 and PTL-1 for C. elegans. Global network alignment was used to compare these networks and determine similar, likely conserved, network regions. This comparison revealed that two prominent pathways, the APP-processing and the Tau-phosphorylation pathways, are highly conserved in both organisms. While the majority of interactions between proteins in those pathways are known to be associated with AD in human, they remain unexamined in C. elegans, signifying the need for their further investigation. In this work, we have highlighted conserved interactions related to AD in humans and have identified specific proteins that can act as targets for experimental studies in C. elegans, aiming to uncover the underlying mechanisms of AD.
Collapse
|
29
|
Zhang X, Li CX, Yan Y, Nair G, Rilling JK, Herndon JG, Preuss TM, Hu X, Li L. In-vivo diffusion MRI protocol optimization for the chimpanzee brain and examination of aging effects on the primate optic nerve at 3T. Magn Reson Imaging 2020; 77:194-203. [PMID: 33359631 DOI: 10.1016/j.mri.2020.12.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/30/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diffusion MRI (dMRI) data acquisition protocols are well-established on modern high-field clinical scanners for human studies. However, these protocols are not suitable for the chimpanzee (or other large-brained mammals) because of its substantial difference in head geometry and brain volume compared with humans. Therefore, an optimal dMRI data acquisition protocol dedicated to chimpanzee neuroimaging is needed. METHODS A multi-shot (4 segments) double spin-echo echo-planar imaging (MS-EPI) sequence and a single-shot double spin-echo EPI (SS-EPI) sequence were optimized separately for in vivo dMRI data acquisition of chimpanzees using a clinical 3T scanner. Correction for severe susceptibility-induced image distortion and signal drop-off of the chimpanzee brain was performed and evaluated using FSL software. DTI indices in different brain regions and probabilistic tractography were compared. A separate DTI data set from n=34 chimpanzees (13 to 56 years old) was collected using the optimal protocol. Age-related changes in diffusivity indices of optic nerve fibers were evaluated. RESULTS The SS-EPI sequence acquired dMRI data of the chimpanzee brain with approximately doubled the SNR as the MS-EPI sequence given the same scan time. The quality of white matter fiber tracking from the SS-EPI data was much higher than that from MS-EPI data. However, quantitative analysis of DTI indices showed no difference in most ROIs between the SS-EPI and MS-EPI sequences. The progressive evolution of diffusivity indices of optic nerves indicated mild changes in fiber bundles of chimpanzees aged 40 years and above. CONCLUSION The single-shot EPI-based acquisition protocol provided better image quality of dMRI for chimpanzee brains and is recommended for in vivo dMRI study or clinical diagnosis of chimpanzees (or other large animals) using a clinical scanner. Also, the tendency of FA decrease or diffusivity increase in the optic nerve of aged chimpanzees was seen but did not show significant age-related changes, suggesting aging may have less impact on optic nerve fiber integrity of chimpanzees, in contrast to previous results for both macaque monkeys and humans.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America; Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America.
| | - Chun-Xia Li
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Yumei Yan
- Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Govind Nair
- qMRI Core Facility, NINDS, NIH, Bethesda, MD 20892, United States of America
| | - James K Rilling
- Department of Anthropology, Emory University, Atlanta, GA, United States of America; Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - James G Herndon
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Todd M Preuss
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Xiaoping Hu
- Dept of Bioengineering, University of California, Riverside, CA, United States of America
| | - Longchuan Li
- Marcus Autism Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, United States of America.
| |
Collapse
|
30
|
Kelberman M, Keilholz S, Weinshenker D. What's That (Blue) Spot on my MRI? Multimodal Neuroimaging of the Locus Coeruleus in Neurodegenerative Disease. Front Neurosci 2020; 14:583421. [PMID: 33122996 PMCID: PMC7573566 DOI: 10.3389/fnins.2020.583421] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
The locus coeruleus (LC) has long been underappreciated for its role in the pathophysiology of Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative disorders. While AD and PD are distinct in clinical presentation, both are characterized by prodromal protein aggregation in the LC, late-stage degeneration of the LC, and comorbid conditions indicative of LC dysfunction. Many of these early studies were limited to post-mortem histological techniques due to the LC’s small size and location deep in the brainstem. Thus, there is a growing interest in utilizing in vivo imaging of the LC as a predictor of preclinical neurodegenerative processes and biomarker of disease progression. Simultaneously, neuroimaging in animal models of neurodegenerative disease holds promise for identifying early alterations to LC circuits, but has thus far been underutilized. While still in its infancy, a handful of studies have reported effects of single gene mutations and pathology on LC function in disease using various neuroimaging techniques. Furthermore, combining imaging and optogenetics or chemogenetics allows for interrogation of network connectivity in response to changes in LC activity. The purpose of this article is twofold: (1) to review what magnetic resonance imaging (MRI) and positron emission tomography (PET) have revealed about LC dysfunction in neurodegenerative disease and its potential as a biomarker in humans, and (2) to explore how animal models can be used to test hypotheses derived from clinical data and establish a mechanistic framework to inform LC-focused therapeutic interventions to alleviate symptoms and impede disease progression.
Collapse
Affiliation(s)
- Michael Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Shella Keilholz
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
31
|
Kim S, Lee Y, Jeon CY, Kim K, Jeon Y, Jin YB, Oh S, Lee C. Quantitative magnetic susceptibility assessed by 7T magnetic resonance imaging in Alzheimer's disease caused by streptozotocin administration. Quant Imaging Med Surg 2020; 10:789-797. [PMID: 32269937 DOI: 10.21037/qims.2020.02.08] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Streptozotocin treatment has emerged as an alternative model of sporadic Alzheimer's disease (SAD). Streptozotocin-induced alterations in iron and calcium levels reflect magnetic susceptibility changes, while susceptibility distribution in the cerebral regions has not been reported yet. This study aimed to investigate susceptibility distribution in the limbic system after streptozotocin administration to cynomolgus monkeys for exploring informative SAD biomarkers. Quantitative susceptibility mapping (QSM) using 7T magnetic resonance imaging (MRI) was utilized to quantitatively compare the susceptibility distributions in monkeys with sporadic Alzheimer disease and age-matched healthy controls. Compared to healthy controls, overall susceptibility values differed in the SAD models. Notable substantial susceptibility changes were observed in the hypothalamus with a 4.38-time decrease (AD: -47.45±12.19 ppb, healthy controls: 14.02±9.51 ppb) and in the posterior parts of the corpus callosum with a 2.83-times increase (AD: 31.49±15.90 ppb; healthy controls: 11.13±4.02 ppb). These susceptibility alterations may reflect neuronal death, and could serve as key biomarkers in the SAD. These results may be useful for specifying AD pathologies such as cognitive and non-cognitive symptoms.
Collapse
Affiliation(s)
- Sangwoo Kim
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Korea.,Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Korea
| | - Keunil Kim
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Korea.,Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Youngjae Jeon
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Korea.,Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Korea
| | - Sukhoon Oh
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Chulhyun Lee
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Korea.,Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| |
Collapse
|
32
|
Vermilyea SC, Babinski A, Tran N, To S, Guthrie S, Kluss JH, Schmidt JK, Wiepz GJ, Meyer MG, Murphy ME, Cookson MR, Emborg ME, Golos TG. In Vitro CRISPR/Cas9-Directed Gene Editing to Model LRRK2 G2019S Parkinson's Disease in Common Marmosets. Sci Rep 2020; 10:3447. [PMID: 32103062 PMCID: PMC7044232 DOI: 10.1038/s41598-020-60273-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/04/2020] [Indexed: 11/09/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) G2019S is a relatively common mutation, associated with 1-3% of Parkinson's disease (PD) cases worldwide. G2019S is hypothesized to increase LRRK2 kinase activity. Dopaminergic neurons derived from induced pluripotent stem cells of PD patients carrying LRRK2 G2019S are reported to have several phenotypes compared to wild type controls, including increased activated caspase-3 and reactive oxygen species (ROS), autophagy dysfunction, and simplification of neurites. The common marmoset is envisioned as a candidate nonhuman primate species for comprehensive modeling of genetic mutations. Here, we report our successful use of CRISPR/Cas9 with repair template-mediated homology directed repair to introduce the LRRK2 G2019S mutation, as well as a truncation of the LRRK2 kinase domain, into marmoset embryonic and induced pluripotent stem cells. We found that, similar to humans, marmoset LRRK2 G2019S resulted in elevated kinase activity. Phenotypic evaluation after dopaminergic differentiation demonstrated LRRK2 G2019S-mediated increased intracellular ROS, decreased neuronal viability, and reduced neurite complexity. Importantly, these phenotypes were not observed in clones with LRRK2 truncation. These results demonstrate the feasibility of inducing monogenic mutations in common marmosets and support the use of this species for generating a novel genetic-based model of PD that expresses physiological levels of LRRK2 G2019S.
Collapse
Affiliation(s)
- Scott C Vermilyea
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Alexander Babinski
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nina Tran
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Samantha To
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott Guthrie
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jillian H Kluss
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Gregory J Wiepz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael G Meyer
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Megan E Murphy
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Marina E Emborg
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA.
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Departments of Comparative Biosciences and Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
33
|
Li X, Yang W, Li X, Chen M, Liu C, Li J, Yu S. Alpha-synuclein oligomerization and dopaminergic degeneration occur synchronously in the brain and colon of MPTP-intoxicated parkinsonian monkeys. Neurosci Lett 2019; 716:134640. [PMID: 31759083 DOI: 10.1016/j.neulet.2019.134640] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/01/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023]
Abstract
Dopaminergic (DAergic) degeneration and abnormal α-synuclein (α-syn) expression, phosphorylation and aggregation are observed in both the nigrostriatal system (NSS) and enteric nervous system (ENS) of patients with Parkinson's disease (PD). Whether these alterations in α-syn and DAergic neurons occur synchronously in the two nervous systems or follow a process that spreads from the gut to the brain remains a subject of debate. Here, in MPTP-intoxicated cynomolgus monkeys, we showed a parallel DAergic degeneration in the colon as well as in the substantia nigra and striatum (SN/STR), as indicated by reduced expression of tyrosine hydroxylase (TH) and dopamine transporter (DAT). In addition, we observed a simultaneous increase in the concentrations of total, phosphorylated, and oligomeric α-syn in the colon and SN/STR. Moreover, we identified that the above changes in α-syn were associated with an increase in the expression of polo-like kinase 2 (PLK2), an enzyme that promotes α-syn phosphorylation, and a decrease in the activity of protein phosphatase 2A (PP2A), an enzyme that facilitates α-syn dephosphorylation. Because the colonic ENS can be readily analyzed using routine biopsies, the shared pathological features between the colonic ENS and the brain NSS found in this study provide useful information for assessing and understanding the neuropathology in PD patients using colonic biopsies.
Collapse
Affiliation(s)
- Xuran Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Weiwei Yang
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xin Li
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Min Chen
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Chengwei Liu
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jie Li
- Department of Neurology, Beijing Daxing District Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, China
| | - Shun Yu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China.
| |
Collapse
|
34
|
Chen ZZ, Niu YY. Stem cell therapy for Parkinson's disease using non-human primate models. Zool Res 2019; 40:349-357. [PMID: 31343853 PMCID: PMC6755115 DOI: 10.24272/j.issn.2095-8137.2019.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/26/2019] [Indexed: 12/23/2022] Open
Abstract
Stem cell therapy (SCT) for Parkinson's disease (PD) has received considerable attention in recent years. Non-human primate (NHP) models of PD have played an instrumental role in the safety and efficacy of emerging PD therapies and facilitated the translation of initiatives for human patients. NHP models of PD include primates with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonism, who are responsive to dopamine replacement therapies, similar to human PD patients. Extensive research in SCT has been conducted to better treat the progressive dopaminergic neurodegeneration that underlies PD. For effective application of SCT in PD, however, a number of basic parameters still need to be tested and optimized in NHP models, including preparation and storage of cells for engraftment, methods of transplantation, choice of target sites, and timelines for recovery. In this review, we discuss the current status of NHP models of PD in stem cell research. We also analyze the advances and remaining challenges for successful clinical translation of SCT for this persistent disease.
Collapse
Affiliation(s)
- Zhen-Zhen Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming Yunnan 650500, China
- Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming Yunnan 650500
| | - Yu-Yu Niu
- Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming Yunnan 650500, China; E-mail:
| |
Collapse
|
35
|
Rios A, Soma S, Yoshida J, Nonomura S, Kawabata M, Sakai Y, Isomura Y. Differential Changes in the Lateralized Activity of Identified Projection Neurons of Motor Cortex in Hemiparkinsonian Rats. eNeuro 2019; 6:ENEURO.0110-19.2019. [PMID: 31235466 PMCID: PMC6620387 DOI: 10.1523/eneuro.0110-19.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/24/2022] Open
Abstract
In the parkinsonian state, the motor cortex and basal ganglia (BG) undergo dynamic remodeling of movement representation. One such change is the loss of the normal contralateral lateralized activity pattern. The increase in the number of movement-related neurons responding to ipsilateral or bilateral limb movements may cause motor problems, including impaired balance, reduced bimanual coordination, and abnormal mirror movements. However, it remains unknown how individual types of motor cortical neurons organize this reconstruction. To explore the effect of dopamine depletion on lateralized activity in the parkinsonian state, we used a partial hemiparkinsonian model [6-hydroxydopamine (6-OHDA) lesion] in Long-Evans rats performing unilateral movements in a right-left pedal task, while recording from primary (M1) and secondary motor cortex (M2). The lesion decreased contralateral preferred activity in both M1 and M2. In addition, this change differed among identified intratelencephalic (IT) and pyramidal tract (PT) cortical projection neurons, depending on the cortical area. We detected a decrease in lateralized activity only in PT neurons in M1, whereas in M2, this change was observed in IT neurons, with no change in the PT population. Our results suggest a differential effect of dopamine depletion in the lateralized activity of the motor cortex, and suggest possible compensatory changes in the contralateral hemisphere.
Collapse
Affiliation(s)
- Alain Rios
- Brain Science Institute, Tamagawa University, Tokyo 194-8610, Japan
- Graduate School of Brain Sciences, Tamagawa University, Tokyo 194-8610, Japan
- Physiology and Cell Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Shogo Soma
- Department of Anatomy and Neurobiology. University of California, Irvine, Irvine, CA 92697
| | - Junichi Yoshida
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Satoshi Nonomura
- Brain Science Institute, Tamagawa University, Tokyo 194-8610, Japan
- Physiology and Cell Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Masanori Kawabata
- Brain Science Institute, Tamagawa University, Tokyo 194-8610, Japan
- Graduate School of Brain Sciences, Tamagawa University, Tokyo 194-8610, Japan
- Physiology and Cell Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Yutaka Sakai
- Brain Science Institute, Tamagawa University, Tokyo 194-8610, Japan
- Graduate School of Brain Sciences, Tamagawa University, Tokyo 194-8610, Japan
| | - Yoshikazu Isomura
- Brain Science Institute, Tamagawa University, Tokyo 194-8610, Japan
- Graduate School of Brain Sciences, Tamagawa University, Tokyo 194-8610, Japan
- Physiology and Cell Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| |
Collapse
|
36
|
Vermilyea SC, Guthrie S, Hernandez I, Bondarenko V, Emborg ME. α-Synuclein Expression Is Preserved in Substantia Nigra GABAergic Fibers of Young and Aged Neurotoxin-Treated Rhesus Monkeys. Cell Transplant 2019; 28:379-387. [PMID: 30857404 PMCID: PMC6628567 DOI: 10.1177/0963689719835794] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/18/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023] Open
Abstract
α-Synuclein (α-syn) is a small presynaptic protein distributed ubiquitously in the central and peripheral nervous system. In normal conditions, α-syn is found in soluble form, while in Parkinson's disease (PD) it may phosphorylate, aggregate, and combine with other proteins to form Lewy bodies. The purpose of this study was to evaluate, in nonhuman primates, whether α-syn expression is affected by age and neurotoxin challenge. Young adult (n = 5, 5-10 years old) and aged (n = 4, 23-25 years old) rhesus monkeys received a single unilateral carotid artery injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Three months post-MPTP the animals were necropsied by transcardiac perfusion, and their brains extracted and processed with immunohistochemical methods. Quantification of tyrosine hydroxylase (TH)-positive substantia nigra (SN) neurons showed a significant 80-89% decrease in the side ipsilateral to MPTP administration in young and old animals. Optical density of TH- immunoreactivity (-ir) in the caudate and putamen presented a 60-70% loss compared with the contralateral side. α-Syn-ir was present in both ipsi- and contra- lateral MPTP-treated nigra, caudate, and putamen, mostly in fibers; its intracellular distribution was not affected by age. Comparison of α-syn-ir between MPTP-treated young and aged monkeys revealed significantly higher optical density for both the ipsi- and contralateral caudate and SN in the aged animals. TH and α-syn immunofluorescence confirmed the loss of nigral TH-ir dopaminergic neurons in the MPTP-treated side of intoxicated animals, but bilateral α-syn expression. Colabeling of GAD67 and α-syn immunofluorescence showed that α-syn expression was present mainly in GABAergic fibers. Our results demonstrate that, 3 months post unilateral intracarotid artery infusion of MPTP, α-syn expression in the SN is largely present in GABAergic fibers, regardless of age. Bilateral increase of α-syn expression in SN fibers of aged, compared with young rhesus monkeys, suggests that α-syn-ir may increase with age, but not after neurotoxin-induced dopaminergic nigral cell loss.
Collapse
Affiliation(s)
- Scott C. Vermilyea
- Neuroscience Training Program, University of Wisconsin-Madison, USA
- Preclinical Parkinson’s Research Program, Wisconsin National Primate
Research Center, University of Wisconsin-Madison, USA
| | - Scott Guthrie
- Preclinical Parkinson’s Research Program, Wisconsin National Primate
Research Center, University of Wisconsin-Madison, USA
| | - Iliana Hernandez
- Preclinical Parkinson’s Research Program, Wisconsin National Primate
Research Center, University of Wisconsin-Madison, USA
| | - Viktorya Bondarenko
- Preclinical Parkinson’s Research Program, Wisconsin National Primate
Research Center, University of Wisconsin-Madison, USA
| | - Marina E. Emborg
- Neuroscience Training Program, University of Wisconsin-Madison, USA
- Preclinical Parkinson’s Research Program, Wisconsin National Primate
Research Center, University of Wisconsin-Madison, USA
- Department of Medical Physics, University of Wisconsin-Madison, USA
| |
Collapse
|
37
|
Chansel‐Debordeaux L, Bezard E. Local transgene expression and whole-body transgenesis to model brain diseases in nonhuman primate. Animal Model Exp Med 2019; 2:9-17. [PMID: 31016282 PMCID: PMC6431118 DOI: 10.1002/ame2.12055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/10/2018] [Indexed: 12/26/2022] Open
Abstract
Animal model is an essential tool in the life sciences research, notably in understanding the pathogenesis of the diseases and for further therapeutic intervention success. Rodents have been the most frequently used animals to model human disease since the establishment of gene manipulation technique. However, they remain inadequate to fully mimic the pathophysiology of human brain disease, partially due to huge differences between rodents and humans in terms of anatomy, brain function, and social behaviors. Nonhuman primates are more suitable in translational perspective. Thus, genetically modified animals have been generated to investigate neurologic and psychiatric disorders. The classical transgenesis technique is not efficient in that model; so, viral vector-mediated transgene delivery and the new genome-editing technologies have been promoted. In this review, we summarize some of the technical progress in the generation of an ad hoc animal model of brain diseases by gene delivery and real transgenic nonhuman primate.
Collapse
Affiliation(s)
- Lucie Chansel‐Debordeaux
- Institut des Maladies NeurodégénérativesUniversity of BordeauxUMR 5293BordeauxFrance
- CNRSInstitut des Maladies NeurodégénérativesUMR 5293BordeauxFrance
- CHU BordeauxService de Biologie de la reproduction‐CECOSBordeauxFrance
| | - Erwan Bezard
- Institut des Maladies NeurodégénérativesUniversity of BordeauxUMR 5293BordeauxFrance
- CNRSInstitut des Maladies NeurodégénérativesUMR 5293BordeauxFrance
| |
Collapse
|