1
|
Sun YR, Lv QK, Liu JY, Wang F, Liu CF. New perspectives on the glymphatic system and the relationship between glymphatic system and neurodegenerative diseases. Neurobiol Dis 2025; 205:106791. [PMID: 39778750 DOI: 10.1016/j.nbd.2025.106791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Neurodegenerative diseases (ND) are characterized by the accumulation of aggregated proteins. The glymphatic system, through its rapid exchange mechanisms between cerebrospinal fluid (CSF) and interstitial fluid (ISF), facilitates the movement of metabolic substances within the brain, serving functions akin to those of the peripheral lymphatic system. This emerging waste clearance mechanism offers a novel perspective on the removal of pathological substances in ND. This article elucidates recent discoveries regarding the glymphatic system and updates relevant concepts within its model. It discusses the potential roles of the glymphatic system in ND, including Alzheimer's disease (AD), Parkinson's disease (PD), and multiple system atrophy (MSA), and proposes the glymphatic system as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Yan-Rui Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake hospital affilicated to Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| |
Collapse
|
2
|
Iba M, Lee YJ, Horan-Portelance L, Chang K, Szabo M, Rissman RA, You S, Masliah E, Kim C. Microglial and neuronal fates following inhibition of CSF-1R in synucleinopathy mouse model. Brain Behav Immun 2025; 123:254-269. [PMID: 39284516 DOI: 10.1016/j.bbi.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/21/2024] [Accepted: 09/13/2024] [Indexed: 12/07/2024] Open
Abstract
Synucleinopathies are age-related neurological disorders characterized by the abnormal accumulation of α-synuclein (α-syn) in neuronal and non-neuronal cells. It has been proposed that microglial cells play an important role in synucleinopathy neuroinflammation, as well as homeostatically, such as in the clearance of α-syn aggregates in the brain. Here, we examined the effects of microglia on the pathogenesis of synucleinopathies by cell depletion in a mouse model of synucleinopathies. For this purpose, we treated non-transgenic (Non-tg) and α-synuclein transgenic (α-syn-tg) mice with pexidartinib (PLX3397), a tyrosine kinase inhibitor of colony-stimulating factor 1 receptor (CSF-1R). Neuropathological and immunoblot analysis confirmed that Iba-1 immunoreactive microglial cells were decreased by 95% following PLX3397 treatment in Non-tg and α-syn-tg mice. The level of total α-syn in the Triton X-insoluble fraction of brain homogenate was significantly decreased by microglial depletion in the α-syn-tg mice, while the level of Triton X-soluble human α-syn was not affected. Furthermore, the number of p-α-syn immunoreactive inclusions was reduced in α-syn-tg mice treated with PLX3397. Microglial depletion also ameliorated neuronal and synaptic degeneration in α-syn-tg mice, thereby resulted partially improving the motor behavioral deficit in α-syn-tg mice. Moreover, we demonstrated that microglia that survived post-PLX3397 treatment (PLX-resistant microglia) have lower expressions of CSF-1R, and microglial transcriptome analysis further elucidated that PLX-resistant microglia have unique morphology and transcriptomic signatures relative to vehicle-treated microglia of both genotypes; these include differences in definitive microglial functions such as their immune response, cell mobility, cell-cell communications, and regulation of neural homeostasis. Therefore, we suggest that microglia play a critical role in the pathogenesis of synucleinopathies, and that modulation of microglial status might be an effective therapeutic strategy for synucleinopathies.
Collapse
Affiliation(s)
- Michiyo Iba
- Molecular Neuropathology Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yeon-Joo Lee
- Departments of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liam Horan-Portelance
- Molecular Neuropathology Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Chang
- Molecular Neuropathology Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcell Szabo
- Molecular Neuropathology Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert A Rissman
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sungyong You
- Departments of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Eliezer Masliah
- Molecular Neuropathology Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA; Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Changyoun Kim
- Molecular Neuropathology Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Jorstad NL, Song JH, Exposito-Alonso D, Suresh H, Castro-Pacheco N, Krienen FM, Yanny AM, Close J, Gelfand E, Long B, Seeman SC, Travaglini KJ, Basu S, Beaudin M, Bertagnolli D, Crow M, Ding SL, Eggermont J, Glandon A, Goldy J, Kiick K, Kroes T, McMillen D, Pham T, Rimorin C, Siletti K, Somasundaram S, Tieu M, Torkelson A, Feng G, Hopkins WD, Höllt T, Keene CD, Linnarsson S, McCarroll SA, Lelieveldt BP, Sherwood CC, Smith K, Walsh CA, Dobin A, Gillis J, Lein ES, Hodge RD, Bakken TE. Comparative transcriptomics reveals human-specific cortical features. Science 2023; 382:eade9516. [PMID: 37824638 PMCID: PMC10659116 DOI: 10.1126/science.ade9516] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
The cognitive abilities of humans are distinctive among primates, but their molecular and cellular substrates are poorly understood. We used comparative single-nucleus transcriptomics to analyze samples of the middle temporal gyrus (MTG) from adult humans, chimpanzees, gorillas, rhesus macaques, and common marmosets to understand human-specific features of the neocortex. Human, chimpanzee, and gorilla MTG showed highly similar cell-type composition and laminar organization as well as a large shift in proportions of deep-layer intratelencephalic-projecting neurons compared with macaque and marmoset MTG. Microglia, astrocytes, and oligodendrocytes had more-divergent expression across species compared with neurons or oligodendrocyte precursor cells, and neuronal expression diverged more rapidly on the human lineage. Only a few hundred genes showed human-specific patterning, suggesting that relatively few cellular and molecular changes distinctively define adult human cortical structure.
Collapse
Affiliation(s)
| | - Janet H.T. Song
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - David Exposito-Alonso
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hamsini Suresh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Fenna M. Krienen
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jennie Close
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Emily Gelfand
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Brian Long
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | | | | | - Soumyadeep Basu
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
- Computer Graphics and Visualization Group, Delft University of Technology, Delft, Netherlands
| | - Marc Beaudin
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Megan Crow
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Song-Lin Ding
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Jeroen Eggermont
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
| | | | - Jeff Goldy
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Katelyn Kiick
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Thomas Kroes
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
| | | | | | | | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael Tieu
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Amy Torkelson
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - William D. Hopkins
- Keeling Center for Comparative Medicine and Research, University of Texas, MD Anderson Cancer Center, Houston, TX 78602, USA
| | - Thomas Höllt
- Computer Graphics and Visualization Group, Delft University of Technology, Delft, Netherlands
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 981915, USA
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Steven A. McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Boudewijn P. Lelieveldt
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
- Pattern Recognition and Bioinformatics group, Delft University of Technology, Delft, Netherlands
| | - Chet C. Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20037, USA
| | - Kimberly Smith
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Christopher A. Walsh
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Alexander Dobin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Ed S. Lein
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | | | | |
Collapse
|
4
|
Wang Y, Xia Y, Kou L, Yin S, Chi X, Li J, Sun Y, Wu J, Zhou Q, Zou W, Jin Z, Huang J, Xiong N, Wang T. Astrocyte-to-neuron reprogramming and crosstalk in the treatment of Parkinson's disease. Neurobiol Dis 2023:106224. [PMID: 37433411 DOI: 10.1016/j.nbd.2023.106224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023] Open
Abstract
Parkinson's disease (PD) is currently the fastest growing disabling neurological disorder worldwide, with motor and non-motor symptoms being its main clinical manifestations. The primary pathological features include a reduction in the number of dopaminergic neurons in the substantia nigra and decrease in dopamine levels in the nigrostriatal pathway. Existing treatments only alleviate clinical symptoms and do not stop disease progression; slowing down the loss of dopaminergic neurons and stimulating their regeneration are emerging therapies. Preclinical studies have demonstrated that transplantation of dopamine cells generated from human embryonic or induced pluripotent stem cells can restore the loss of dopamine. However, the application of cell transplantation is limited owing to ethical controversies and the restricted source of cells. Until recently, the reprogramming of astrocytes to replenish lost dopaminergic neurons has provided a promising alternative therapy for PD. In addition, repair of mitochondrial perturbations, clearance of damaged mitochondria in astrocytes, and control of astrocyte inflammation may be extensively neuroprotective and beneficial against chronic neuroinflammation in PD. Therefore, this review primarily focuses on the progress and remaining issues in astrocyte reprogramming using transcription factors (TFs) and miRNAs, as well as exploring possible new targets for treating PD by repairing astrocytic mitochondria and reducing astrocytic inflammation.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiulu Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
5
|
Stefanova N, Wenning GK. Multiple system atrophy: at the crossroads of cellular, molecular and genetic mechanisms. Nat Rev Neurosci 2023; 24:334-346. [PMID: 37085728 DOI: 10.1038/s41583-023-00697-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2023] [Indexed: 04/23/2023]
Abstract
Multiple system atrophy (MSA) is a rare oligodendroglial α-synucleinopathy characterized by neurodegeneration in striatonigral and olivopontocerebellar regions and autonomic brain centres. It causes complex cumulative motor and non-motor disability with fast progression and effective therapy is currently lacking. The difficulties in the diagnosis and treatment of MSA are largely related to the incomplete understanding of the pathogenesis of the disease. The MSA pathogenic landscape is complex, and converging findings from genetic and neuropathological studies as well as studies in experimental models of MSA have indicated the involvement of genetic and epigenetic changes; α-synuclein misfolding, aggregation and spreading; and α-synuclein strain specificity. These studies also indicate the involvement of myelin and iron dyshomeostasis, neuroinflammation, mitochondrial dysfunction and other cell-specific aspects that are relevant to the fast progression of MSA. In this Review, we discuss these findings and emphasize the implications of the complexity of the multifactorial pathogenic cascade for future translational research and its impact on biomarker discovery and treatment target definitions.
Collapse
Affiliation(s)
- Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria.
| | - Gregor K Wenning
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
6
|
Russo C, Valle MS, Casabona A, Malaguarnera L. Chitinase Signature in the Plasticity of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24076301. [PMID: 37047273 PMCID: PMC10094409 DOI: 10.3390/ijms24076301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Several reports have pointed out that Chitinases are expressed and secreted by various cell types of central nervous system (CNS), including activated microglia and astrocytes. These cells play a key role in neuroinflammation and in the pathogenesis of many neurodegenerative disorders. Increased levels of Chitinases, in particular Chitotriosidase (CHIT-1) and chitinase-3-like protein 1 (CHI3L1), have been found increased in several neurodegenerative disorders. Although having important biological roles in inflammation, to date, the molecular mechanisms of Chitinase involvement in the pathogenesis of neurodegenerative disorders is not well-elucidated. Several studies showed that some Chitinases could be assumed as markers for diagnosis, prognosis, activity, and severity of a disease and therefore can be helpful in the choice of treatment. However, some studies showed controversial results. This review will discuss the potential of Chitinases in the pathogenesis of some neurodegenerative disorders, such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, to understand their role as distinctive biomarkers of neuronal cell activity during neuroinflammatory processes. Knowledge of the role of Chitinases in neuronal cell activation could allow for the development of new methodologies for downregulating neuroinflammation and consequently for diminishing negative neurological disease outcomes.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Laboratory of Neuro-Biomechanics, Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Correspondence:
| | - Antonino Casabona
- Laboratory of Neuro-Biomechanics, Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
7
|
Murray KA, Hu CJ, Pan H, Lu J, Abskharon R, Bowler JT, Rosenberg GM, Williams CK, Elezi G, Balbirnie M, Faull KF, Vinters HV, Seidler PM, Eisenberg DS. Small molecules disaggregate alpha-synuclein and prevent seeding from patient brain-derived fibrils. Proc Natl Acad Sci U S A 2023; 120:e2217835120. [PMID: 36757890 PMCID: PMC9963379 DOI: 10.1073/pnas.2217835120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/31/2022] [Indexed: 02/10/2023] Open
Abstract
The amyloid aggregation of alpha-synuclein within the brain is associated with the pathogenesis of Parkinson's disease (PD) and other related synucleinopathies, including multiple system atrophy (MSA). Alpha-synuclein aggregates are a major therapeutic target for treatment of these diseases. We identify two small molecules capable of disassembling preformed alpha-synuclein fibrils. The compounds, termed CNS-11 and CNS-11g, disaggregate recombinant alpha-synuclein fibrils in vitro, prevent the intracellular seeded aggregation of alpha-synuclein fibrils, and mitigate alpha-synuclein fibril cytotoxicity in neuronal cells. Furthermore, we demonstrate that both compounds disassemble fibrils extracted from MSA patient brains and prevent their intracellular seeding. They also reduce in vivo alpha-synuclein aggregates in C. elegans. Both compounds also penetrate brain tissue in mice. A molecular dynamics-based computational model suggests the compounds may exert their disaggregating effects on the N terminus of the fibril core. These compounds appear to be promising therapeutic leads for targeting alpha-synuclein for the treatment of synucleinopathies.
Collapse
Affiliation(s)
- Kevin A. Murray
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Carolyn J. Hu
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Hope Pan
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Jiahui Lu
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Romany Abskharon
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Jeannette T. Bowler
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Gregory M. Rosenberg
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Christopher K. Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA90095
| | - Gazmend Elezi
- Pasarow Mass Spectrometry Laboratory, David Geffen School of Medicine, UCLA, Los Angeles, CA90095
| | - Melinda Balbirnie
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| | - Kym F. Faull
- Pasarow Mass Spectrometry Laboratory, David Geffen School of Medicine, UCLA, Los Angeles, CA90095
| | - Harry V. Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA90095
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA90095
| | - Paul M. Seidler
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA90089
| | - David S. Eisenberg
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, CA90095
- HHMI, UCLA, Los Angeles, CA90095
| |
Collapse
|
8
|
Fanciulli A, Leys F, Lehner F, Sidoroff V, Ruf VC, Raccagni C, Mahlknecht P, Kuipers DJS, van IJcken WFJ, Stockner H, Musacchio T, Volkmann J, Monoranu CM, Stankovic I, Breedveld G, Ferraro F, Fevga C, Windl O, Herms J, Kiechl S, Poewe W, Seppi K, Stefanova N, Scholz SW, Bonifati V, Wenning GK. A multiplex pedigree with pathologically confirmed multiple system atrophy and Parkinson's disease with dementia. Brain Commun 2022; 4:fcac175. [PMID: 35855480 PMCID: PMC9291376 DOI: 10.1093/braincomms/fcac175] [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: 02/14/2022] [Revised: 05/12/2022] [Accepted: 07/01/2022] [Indexed: 02/03/2023] Open
Abstract
Multiple system atrophy is considered a sporadic disease, but neuropathologically confirmed cases with a family history of parkinsonism have been occasionally described. Here we report a North-Bavarian (colloquially, Lion’s tail region) six-generation pedigree, including neuropathologically confirmed multiple system atrophy and Parkinson’s disease with dementia. Between 2012 and 2020, we examined all living and consenting family members of age and calculated the risk of prodromal Parkinson’s disease in those without overt parkinsonism. The index case and one paternal cousin with Parkinson’s disease with dementia died at follow-up and underwent neuropathological examination. Genetic analysis was performed in both and another family member with Parkinson’s disease. The index case was a female patient with cerebellar variant multiple system atrophy and a positive maternal and paternal family history for Parkinson’s disease and dementia in multiple generations. The families of the index case and her spouse were genealogically related, and one of the spouse's siblings met the criteria for possible prodromal Parkinson’s disease. Neuropathological examination confirmed multiple system atrophy in the index case and advanced Lewy body disease, as well as tau pathology in her cousin. A comprehensive analysis of genes known to cause hereditary forms of parkinsonism or multiple system atrophy lookalikes was unremarkable in the index case and the other two affected family members. Here, we report an extensive European pedigree with multiple system atrophy and Parkinson`s disease suggesting a complex underlying α-synucleinopathy as confirmed on neuropathological examination. The exclusion of known genetic causes of parkinsonism or multiple system atrophy lookalikes suggests that variants in additional, still unknown genes, linked to α-synucleinopathy lesions underlie such neurodegenerative clustering.
Collapse
Affiliation(s)
| | - Fabian Leys
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Fabienne Lehner
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Victoria Sidoroff
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Viktoria C Ruf
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Cecilia Raccagni
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Philipp Mahlknecht
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Demy J S Kuipers
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Heike Stockner
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Musacchio
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | - Camelia Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Iva Stankovic
- Neurology Clinic, Clinical Center of Serbia, University of Belgrade, Belgrade, Serbia
| | - Guido Breedveld
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Federico Ferraro
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Christina Fevga
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Otto Windl
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Werner Poewe
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
9
|
Bluhm A, Schrempel S, Schilling S, von Hörsten S, Schulze A, Roßner S, Hartlage-Rübsamen M. Immunohistochemical Demonstration of the pGlu79 α-Synuclein Fragment in Alzheimer’s Disease and Its Tg2576 Mouse Model. Biomolecules 2022; 12:biom12071006. [PMID: 35883562 PMCID: PMC9312983 DOI: 10.3390/biom12071006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
The deposition of β-amyloid peptides and of α-synuclein proteins is a neuropathological hallmark in the brains of Alzheimer’s disease (AD) and Parkinson’s disease (PD) subjects, respectively. However, there is accumulative evidence that both proteins are not exclusive for their clinical entity but instead co-exist and interact with each other. Here, we investigated the presence of a newly identified, pyroglutamate79-modified α-synuclein variant (pGlu79-aSyn)—along with the enzyme matrix metalloproteinase-3 (MMP-3) and glutaminyl cyclase (QC) implicated in its formation—in AD and in the transgenic Tg2576 AD mouse model. In the human brain, pGlu79-aSyn was detected in cortical pyramidal neurons, with more distinct labeling in AD compared to control brain tissue. Using immunohistochemical double and triple labelings and confocal laser scanning microscopy, we demonstrate an association of pGlu79-aSyn, MMP-3 and QC with β-amyloid plaques. In addition, pGlu79-aSyn and QC were present in amyloid plaque-associated reactive astrocytes that were also immunoreactive for the chaperone heat shock protein 27 (HSP27). Our data are consistent for the transgenic mouse model and the human clinical condition. We conclude that pGlu79-aSyn can be generated extracellularly or within reactive astrocytes, accumulates in proximity to β-amyloid plaques and induces an astrocytic protein unfolding mechanism involving HSP27.
Collapse
Affiliation(s)
- Alexandra Bluhm
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
| | - Sarah Schrempel
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
| | - Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, 06120 Halle (Saale), Germany; (S.S.); (A.S.)
- Faculty of Applied Biosciences and Process Engineering, Anhalt University of Applied Sciences, 06366 Köthen, Germany
| | - Stephan von Hörsten
- Department for Experimental Therapy, University Clinics Erlangen and Preclinical Experimental Center, University of Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Anja Schulze
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, 06120 Halle (Saale), Germany; (S.S.); (A.S.)
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
- Correspondence: ; Tel.: +49-341-9725758
| | - Maike Hartlage-Rübsamen
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
| |
Collapse
|
10
|
Peripheral Blood Inflammatory Cytokines are Associated with Rapid Eye Movement Sleep Behavior Disorder in Parkinson’s Disease. Neurosci Lett 2022; 782:136692. [PMID: 35605904 DOI: 10.1016/j.neulet.2022.136692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022]
|
11
|
Padhi P, Worth C, Zenitsky G, Jin H, Sambamurti K, Anantharam V, Kanthasamy A, Kanthasamy AG. Mechanistic Insights Into Gut Microbiome Dysbiosis-Mediated Neuroimmune Dysregulation and Protein Misfolding and Clearance in the Pathogenesis of Chronic Neurodegenerative Disorders. Front Neurosci 2022; 16:836605. [PMID: 35281490 PMCID: PMC8914070 DOI: 10.3389/fnins.2022.836605] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
The human gut microbiota is a complex, dynamic, and highly diverse community of microorganisms. Beginning as early as in utero fetal development and continuing through birth to late-stage adulthood, the crosstalk between the gut microbiome and brain is essential for modulating various metabolic, neurodevelopmental, and immune-related pathways. Conversely, microbial dysbiosis - defined as alterations in richness and relative abundances - of the gut is implicated in the pathogenesis of several chronic neurological and neurodegenerative disorders. Evidence from large-population cohort studies suggests that individuals with neurodegenerative conditions have an altered gut microbial composition as well as microbial and serum metabolomic profiles distinct from those in the healthy population. Dysbiosis is also linked to psychiatric and gastrointestinal complications - comorbidities often associated with the prodromal phase of Parkinson's disease (PD) and Alzheimer's disease (AD). Studies have identified potential mediators that link gut dysbiosis and neurological disorders. Recent findings have also elucidated the potential mechanisms of disease pathology in the enteric nervous system prior to the onset of neurodegeneration. This review highlights the functional pathways and mechanisms, particularly gut microbe-induced chronic inflammation, protein misfolding, propagation of disease-specific pathology, defective protein clearance, and autoimmune dysregulation, linking gut microbial dysbiosis and neurodegeneration. In addition, we also discuss how pathogenic transformation of microbial composition leads to increased endotoxin production and fewer beneficial metabolites, both of which could trigger immune cell activation and enteric neuronal dysfunction. These can further disrupt intestinal barrier permeability, aggravate the systemic pro-inflammatory state, impair blood-brain barrier permeability and recruit immune mediators leading to neuroinflammation and neurodegeneration. Continued biomedical advances in understanding the microbiota-gut-brain axis will extend the frontier of neurodegenerative disorders and enable the utilization of novel diagnostic and therapeutic strategies to mitigate the pathological burden of these diseases.
Collapse
Affiliation(s)
- Piyush Padhi
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | - Carter Worth
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Gary Zenitsky
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | - Huajun Jin
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | - Kumar Sambamurti
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Vellareddy Anantharam
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | - Arthi Kanthasamy
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
12
|
Kim C, Hovakimyan A, Zagorski K, Antonyan T, Petrushina I, Davtyan H, Chailyan G, Hasselmann J, Iba M, Adame A, Rockenstein E, Szabo M, Blurton-Jones M, Cribbs DH, Ghochikyan A, Masliah E, Agadjanyan MG. Efficacy and immunogenicity of MultiTEP-based DNA vaccines targeting human α-synuclein: prelude for IND enabling studies. NPJ Vaccines 2022; 7:1. [PMID: 35013319 PMCID: PMC8748802 DOI: 10.1038/s41541-021-00424-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Accumulation of misfolded proteins such as amyloid-β (Aβ), tau, and α-synuclein (α-Syn) in the brain leads to synaptic dysfunction, neuronal damage, and the onset of relevant neurodegenerative disorder/s. Dementia with Lewy bodies (DLB) and Parkinson's disease (PD) are characterized by the aberrant accumulation of α-Syn intracytoplasmic Lewy body inclusions and dystrophic Lewy neurites resulting in neurodegeneration associated with inflammation. Cell to cell propagation of α-Syn aggregates is implicated in the progression of PD/DLB, and high concentrations of anti-α-Syn antibodies could inhibit/reduce the spreading of this pathological molecule in the brain. To ensure sufficient therapeutic concentrations of anti-α-Syn antibodies in the periphery and CNS, we developed four α-Syn DNA vaccines based on the universal MultiTEP platform technology designed especially for the elderly with immunosenescence. Here, we are reporting on the efficacy and immunogenicity of these vaccines targeting three B-cell epitopes of hα-Syn aa85-99 (PV-1947D), aa109-126 (PV-1948D), aa126-140 (PV-1949D) separately or simultaneously (PV-1950D) in a mouse model of synucleinopathies mimicking PD/DLB. All vaccines induced high titers of antibodies specific to hα-Syn that significantly reduced PD/DLB-like pathology in hα-Syn D line mice. The most significant reduction of the total and protein kinase resistant hα-Syn, as well as neurodegeneration, were observed in various brain regions of mice vaccinated with PV-1949D and PV-1950D in a sex-dependent manner. Based on these preclinical data, we selected the PV-1950D vaccine for future IND enabling preclinical studies and clinical development.
Collapse
Affiliation(s)
- Changyoun Kim
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - Hayk Davtyan
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Jonathan Hasselmann
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - Michiyo Iba
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Marcell Szabo
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, USA
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA.
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA.
| |
Collapse
|
13
|
Li H, Knight WC, Xu J. Striatal oxidative damages and neuroinflammation correlate with progression and survival of Lewy body and Alzheimer diseases. Neural Regen Res 2021; 17:867-874. [PMID: 34472487 PMCID: PMC8530139 DOI: 10.4103/1673-5374.322463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Neurodegenerative diseases are a class of chronic and complex disorders featuring progressive loss of neurons in distinct brain areas. The mechanisms responsible for the disease progression in neurodegeneration are not fully illustrated. In this observational study, we have examined diverse biochemical parameters in the caudate and putamen of patients with Lewy body diseases (LBDs) and Alzheimer disease (AD), shedding some light on the involvement of oxidative damage and neuroinflammation in advanced neurodegeneration. We performed Spearman and Mantel-Cox analyses to investigate how oxidative stress and neuroinflammation exert comprehensive effects on disease progression and survival. Disease progression in LBDs correlated positively with poly (ADP-Ribose) and triggering receptors expressed on myeloid cell 2 levels in the striatum of LBD cohorts, indicating that potential parthanatos was a dominant feature of worsening disease progression and might contribute to switching microglial inflammatory phenotypes. Disease progression in AD corresponds negatively with 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxo-dG) and myeloperoxidase concentrations in the striatum, suggesting that possible mitochondria dysfunction may be involved in the progression of AD via a mechanism of β-amyloid entering the mitochondria and subsequent free radicals generation. Patients with lower striatal 8-oxo-dG and myeloperoxidase levels had a survival advantage in AD. The age of onset also affected disease progression. Tissue requests for the postmortem biochemistry, genetics, and autoradiography studies were approved by the Washington University Alzheimer’s Disease Research Center (ADRC) Biospecimens Committee (ethics approval reference number: T1705, approval date: August 6, 2019). Recombinant DNA and Hazardous Research Materials were approved by the Washington University Environmental Health & Safety Biological Safety Committee (approval code: 3739, approval date: February 25, 2020). Radioactive Material Authorization was approved by the Washington University Environmental Health & Safety Radiation Safety Committee (approval code: 1056, approval date: September 18, 2019).
Collapse
Affiliation(s)
- Huifangjie Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - William C Knight
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jinbin Xu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
14
|
Current experimental disease-modifying therapeutics for multiple system atrophy. J Neural Transm (Vienna) 2021; 128:1529-1543. [PMID: 34398313 PMCID: PMC8528757 DOI: 10.1007/s00702-021-02406-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/08/2021] [Indexed: 02/06/2023]
Abstract
Multiple system atrophy (MSA) is a challenging neurodegenerative disorder with a difficult and often inaccurate early diagnosis, still lacking effective treatment. It is characterized by a highly variable clinical presentation with parkinsonism, cerebellar ataxia, autonomic dysfunction, and pyramidal signs, with a rapid progression and an aggressive clinical course. The definite MSA diagnosis is only possible post-mortem, when the presence of distinctive oligodendroglial cytoplasmic inclusions (GCIs), mainly composed of misfolded and aggregated α-Synuclein (α-Syn) is demonstrated. The process of α-Syn accumulation and aggregation within oligodendrocytes is accepted one of the main pathological events underlying MSA. However, MSA is considered a multifactorial disorder with multiple pathogenic events acting together including neuroinflammation, oxidative stress, and disrupted neurotrophic support, among others. The discussed here treatment approaches are based on our current understanding of the pathogenesis of MSA and the results of preclinical and clinical therapeutic studies conducted over the last 2 decades. We summarize leading disease-modifying approaches for MSA including targeting α-Syn pathology, modulation of neuroinflammation, and enhancement of neuroprotection. In conclusion, we outline some challenges related to the need to overcome the gap in translation between preclinical and clinical studies towards a successful disease modification in MSA.
Collapse
|
15
|
Bender H, Fietz SA, Richter F, Stanojlovic M. Alpha-Synuclein Pathology Coincides With Increased Number of Early Stage Neural Progenitors in the Adult Hippocampus. Front Cell Dev Biol 2021; 9:691560. [PMID: 34307368 PMCID: PMC8293917 DOI: 10.3389/fcell.2021.691560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Alpha-synuclein pathology driven impairment in adult neurogenesis was proposed as a potential cause of, or at least contributor to, memory impairment observed in both patients and animal models of Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB). Mice overexpressing wild-type alpha-synuclein under the Thy-1 promoter (Thy1-aSyn, line 61) uniquely replicate early cognitive deficits together with multiple other characteristic motor and non-motor symptoms, alpha-synuclein pathology and dopamine loss. Here we report overt intracellular accumulation of phosphorylated alpha-synuclein in the hippocampus of these transgenic mice. To test whether this alters adult neurogenesis and total number of mature neurons, we employed immunohistochemistry and an unbiased stereology approach to quantify the distinct neural progenitor cells and neurons in the hippocampal granule cell layer and subgranular zone of 6 (prodromal stage) and 16-month (dopamine loss) old Thy1-aSyn mice. Surprisingly, we observed an increase in the number of early stage, i.e., Pax6 expressing, progenitors whereas the numbers of late stage, i.e., Tbr2 expressing, progenitors and neurons were not altered. Astroglia marker was increased in the hippocampus of transgenic mice, but this was not specific to the regions where adult neurogenesis takes place, arguing against a commitment of additional early stage progenitors to the astroglia lineage. Together, this uncovers a novel aspect of alpha-synuclein pathology in adult neurogenesis. Studying its mechanisms in Thy1-aSyn mice could lead to discovery of effective therapeutic interventions for cognitive dysfunction in PD and DLB.
Collapse
Affiliation(s)
- Hannah Bender
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hanover, Germany.,Center for Systems Neuroscience, Hanover, Germany
| | - Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hanover, Germany
| |
Collapse
|
16
|
Samidurai M, Palanisamy BN, Bargues-Carot A, Hepker M, Kondru N, Manne S, Zenitsky G, Jin H, Anantharam V, Kanthasamy AG, Kanthasamy A. PKC Delta Activation Promotes Endoplasmic Reticulum Stress (ERS) and NLR Family Pyrin Domain-Containing 3 (NLRP3) Inflammasome Activation Subsequent to Asynuclein-Induced Microglial Activation: Involvement of Thioredoxin-Interacting Protein (TXNIP)/Thioredoxin (Trx) Redoxisome Pathway. Front Aging Neurosci 2021; 13:661505. [PMID: 34276337 PMCID: PMC8283807 DOI: 10.3389/fnagi.2021.661505] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022] Open
Abstract
A classical hallmark of Parkinson's disease (PD) pathogenesis is the accumulation of misfolded alpha-synuclein (αSyn) within Lewy bodies and Lewy neurites, although its role in microglial dysfunction and resultant dopaminergic (DAergic) neurotoxicity is still elusive. Previously, we identified that protein kinase C delta (PKCδ) is activated in post mortem PD brains and experimental Parkinsonism and that it participates in reactive microgliosis; however, the relationship between PKCδ activation, endoplasmic reticulum stress (ERS) and the reactive microglial activation state in the context of α-synucleinopathy is largely unknown. Herein, we show that oxidative stress, mitochondrial dysfunction, NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, and PKCδ activation increased concomitantly with ERS markers, including the activating transcription factor 4 (ATF-4), serine/threonine-protein kinase/endoribonuclease inositol-requiring enzyme 1α (p-IRE1α), p-eukaryotic initiation factor 2 (eIF2α) as well as increased generation of neurotoxic cytokines, including IL-1β in aggregated αSynagg-stimulated primary microglia. Importantly, in mouse primary microglia-treated with αSynagg we observed increased expression of Thioredoxin-interacting protein (TXNIP), an endogenous inhibitor of the thioredoxin (Trx) pathway, a major antioxidant protein system. Additionally, αSynagg promoted interaction between NLRP3 and TXNIP in these cells. In vitro knockdown of PKCδ using siRNA reduced ERS and led to reduced expression of TXNIP and the NLRP3 activation response in αSynagg-stimulated mouse microglial cells (MMCs). Additionally, attenuation of mitochondrial reactive oxygen species (mitoROS) via mito-apocynin and amelioration of ERS via the eIF2α inhibitor salubrinal (SAL) reduced the induction of the ERS/TXNIP/NLRP3 signaling axis, suggesting that mitochondrial dysfunction and ERS may act in concert to promote the αSynagg-induced microglial activation response. Likewise, knockdown of TXNIP by siRNA attenuated the αSynagg-induced NLRP3 inflammasome activation response. Finally, unilateral injection of αSyn preformed fibrils (αSynPFF) into the striatum of wild-type mice induced a significant increase in the expression of nigral p-PKCδ, ERS markers, and upregulation of the TXNIP/NLRP3 inflammasome signaling axis prior to delayed loss of TH+ neurons. Together, our results suggest that inhibition of ERS and its downstream signaling mediators TXNIP and NLRP3 might represent novel therapeutic avenues for ameliorating microglia-mediated neuroinflammation in PD and other synucleinopathies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Arthi Kanthasamy
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| |
Collapse
|
17
|
Mavroeidi P, Xilouri M. Neurons and Glia Interplay in α-Synucleinopathies. Int J Mol Sci 2021; 22:4994. [PMID: 34066733 PMCID: PMC8125822 DOI: 10.3390/ijms22094994] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of the neuronal presynaptic protein alpha-synuclein within proteinaceous inclusions represents the key histophathological hallmark of a spectrum of neurodegenerative disorders, referred to by the umbrella term a-synucleinopathies. Even though alpha-synuclein is expressed predominantly in neurons, pathological aggregates of the protein are also found in the glial cells of the brain. In Parkinson's disease and dementia with Lewy bodies, alpha-synuclein accumulates mainly in neurons forming the Lewy bodies and Lewy neurites, whereas in multiple system atrophy, the protein aggregates mostly in the glial cytoplasmic inclusions within oligodendrocytes. In addition, astrogliosis and microgliosis are found in the synucleinopathy brains, whereas both astrocytes and microglia internalize alpha-synuclein and contribute to the spread of pathology. The mechanisms underlying the pathological accumulation of alpha-synuclein in glial cells that under physiological conditions express low to non-detectable levels of the protein are an area of intense research. Undoubtedly, the presence of aggregated alpha-synuclein can disrupt glial function in general and can contribute to neurodegeneration through numerous pathways. Herein, we summarize the current knowledge on the role of alpha-synuclein in both neurons and glia, highlighting the contribution of the neuron-glia connectome in the disease initiation and progression, which may represent potential therapeutic target for a-synucleinopathies.
Collapse
Affiliation(s)
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
18
|
Boeri L, Perottoni S, Izzo L, Giordano C, Albani D. Microbiota-Host Immunity Communication in Neurodegenerative Disorders: Bioengineering Challenges for In Vitro Modeling. Adv Healthc Mater 2021; 10:e2002043. [PMID: 33661580 PMCID: PMC11468246 DOI: 10.1002/adhm.202002043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Human microbiota communicates with its host by secreting signaling metabolites, enzymes, or structural components. Its homeostasis strongly influences the modulation of human tissue barriers and immune system. Dysbiosis-induced peripheral immunity response can propagate bacterial and pro-inflammatory signals to the whole body, including the brain. This immune-mediated communication may contribute to several neurodegenerative disorders, as Alzheimer's disease. In fact, neurodegeneration is associated with dysbiosis and neuroinflammation. The interplay between the microbial communities and the brain is complex and bidirectional, and a great deal of interest is emerging to define the exact mechanisms. This review focuses on microbiota-immunity-central nervous system (CNS) communication and shows how gut and oral microbiota populations trigger immune cells, propagating inflammation from the periphery to the cerebral parenchyma, thus contributing to the onset and progression of neurodegeneration. Moreover, an overview of the technological challenges with in vitro modeling of the microbiota-immunity-CNS axis, offering interesting technological hints about the most advanced solutions and current technologies is provided.
Collapse
Affiliation(s)
- Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Diego Albani
- Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSvia Mario Negri 2Milan20156Italy
| |
Collapse
|
19
|
Carter B, Justin HS, Gulick D, Gamsby JJ. The Molecular Clock and Neurodegenerative Disease: A Stressful Time. Front Mol Biosci 2021; 8:644747. [PMID: 33889597 PMCID: PMC8056266 DOI: 10.3389/fmolb.2021.644747] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Circadian rhythm dysfunction occurs in both common and rare neurodegenerative diseases. This dysfunction manifests as sleep cycle mistiming, alterations in body temperature rhythms, and an increase in symptomatology during the early evening hours known as Sundown Syndrome. Disruption of circadian rhythm homeostasis has also been implicated in the etiology of neurodegenerative disease. Indeed, individuals exposed to a shifting schedule of sleep and activity, such as health care workers, are at a higher risk. Thus, a bidirectional relationship exists between the circadian system and neurodegeneration. At the heart of this crosstalk is the molecular circadian clock, which functions to regulate circadian rhythm homeostasis. Over the past decade, this connection has become a focal point of investigation as the molecular clock offers an attractive target to combat both neurodegenerative disease pathogenesis and circadian rhythm dysfunction, and a pivotal role for neuroinflammation and stress has been established. This review summarizes the contributions of molecular clock dysfunction to neurodegenerative disease etiology, as well as the mechanisms by which neurodegenerative diseases affect the molecular clock.
Collapse
Affiliation(s)
- Bethany Carter
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Hannah S Justin
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Danielle Gulick
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joshua J Gamsby
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
20
|
Tinaz S, Kamel S, Aravala SS, Sezgin M, Elfil M, Sinha R. Distinct neural circuits are associated with subclinical neuropsychiatric symptoms in Parkinson's disease. J Neurol Sci 2021; 423:117365. [PMID: 33636663 DOI: 10.1016/j.jns.2021.117365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/24/2021] [Accepted: 02/18/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Parkinson's disease (PD) can present with neuropsychiatric symptoms (here, anxiety, depression, and apathy) at any stage of the disease. We investigated the neural correlates of subclinical neuropsychiatric symptoms in relation to motor and cognitive symptoms in a high-functioning PD cohort. METHODS Brain morphometry of the cognitively intact, early-stage (Hoehn & Yahr 2) PD group (n = 48) was compared to matched controls (n = 37). Whole-brain, pairwise, resting-state functional connectivity measures were correlated with neuropsychiatric symptom, motor exam, and global cognitive scores of the PD group. RESULTS Factor analysis of highly collinear anxiety, depression, and apathy scores revealed a single principal component (i.e., composite neuropsychiatric symptom score) explaining 71.6% of variance. There was no collinearity between the neuropsychiatric, motor, and cognitive scores. Compared to controls, PD group showed only subcortical changes including amygdala and nucleus accumbens atrophy, and greater pallidal volume. Reduced functional connectivity in the limbic cortical-striatal circuits and increased functional connectivity between the cerebellum and occipito-temporal regions were associated with a more impaired neuropsychiatric profile. This functional connectivity pattern was distinct from those associated with motor deficits and global cognitive functioning. The individual components of the neuropsychiatric symptoms also exhibited unique connectivity patterns. LIMITATIONS Patients were scanned in "on-medication" state only and a control group with similar neuropsychiatric symptoms was not included. CONCLUSION Abnormal functional connectivity of distinct neural circuits is present even at the subclinical stage of neuropsychiatric symptoms in PD. Neuropsychiatric phenotyping is important and may facilitate early interventions to "reorganize" these circuits and delay/prevent clinical symptom onset.
Collapse
Affiliation(s)
- Sule Tinaz
- Yale University School of Medicine, Department of Neurology, Division of Movement Disorders, 15 York St, LCI 710, New Haven, CT 06510, USA; Yale University School of Medicine, Clinical Neurosciences Imaging Center, 789 Howard Ave, New Haven, CT 06519, USA.
| | - Serageldin Kamel
- Yale University School of Medicine, Department of Neurology, Division of Movement Disorders, 15 York St, LCI 710, New Haven, CT 06510, USA
| | - Sai S Aravala
- Yale University School of Medicine, Department of Neurology, Division of Movement Disorders, 15 York St, LCI 710, New Haven, CT 06510, USA
| | - Mine Sezgin
- Yale University School of Medicine, Department of Neurology, Division of Movement Disorders, 15 York St, LCI 710, New Haven, CT 06510, USA; Istanbul University Faculty of Medicine, Department of Neurology, Millet Street, Fatih, Istanbul 34093, Turkey
| | - Mohamed Elfil
- Yale University School of Medicine, Department of Neurology, Division of Movement Disorders, 15 York St, LCI 710, New Haven, CT 06510, USA
| | - Rajita Sinha
- Yale School of Medicine, Yale Stress Center, 2 Church St South, Suite 209, New Haven, CT 06519, USA; Yale School of Medicine, Department of Psychiatry, 300 George St, New Haven, CT 06511, USA; Yale School of Medicine, Department of Neuroscience, 333 Cedar St, SHM-L-200, New Haven, CT 06510, USA
| |
Collapse
|
21
|
La Vitola P, Balducci C, Baroni M, Artioli L, Santamaria G, Castiglioni M, Cerovic M, Colombo L, Caldinelli L, Pollegioni L, Forloni G. Peripheral inflammation exacerbates α-synuclein toxicity and neuropathology in Parkinson's models. Neuropathol Appl Neurobiol 2021; 47:43-60. [PMID: 32696999 DOI: 10.1111/nan.12644] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
AIMS Parkinson's disease and related disorders are devastating neurodegenerative pathologies. Since α-synuclein was identified as a main component of Lewy bodies and neurites, efforts have been made to clarify the pathogenic mechanisms of α-synuclein's detrimental effects. α-synuclein oligomers are the most harmful species and may recruit and activate glial cells. Inflammation is emerging as a bridge between genetic susceptibility and environmental factors co-fostering Parkinson's disease. However, direct evidence linking inflammation to the harmful activities of α-synuclein oligomers or to the Parkinson's disease behavioural phenotype is lacking. METHODS To clarify whether neuroinflammation influences Parkinson's disease pathogenesis, we developed: (i) a 'double-hit' approach in C57BL/6 naive mice where peripherally administered lipopolysaccharides were followed by intracerebroventricular injection of an inactive oligomer dose; (ii) a transgenic 'double-hit' model where lipopolysaccharides were given to A53T α-synuclein transgenic Parkinson's disease mice. RESULTS Lipopolysaccharides induced a long-lasting neuroinflammatory response which facilitated the detrimental cognitive activities of oligomers. LPS-activated microglia and astrocytes responded differently to the oligomers with microglia activating further and acquiring a pro-inflammatory M1 phenotype, while astrocytes atrophied. In the transgenic 'double-hit' A53T mouse model, lipopolysaccharides aggravated cognitive deficits and increased microgliosis. Again, astrocytes responded differently to the double challenge. These findings indicate that peripherally induced neuroinflammation potentiates the α-synuclein oligomer's actions and aggravates cognitive deficits in A53T mice. CONCLUSIONS The fine management of both peripheral and central inflammation may offer a promising therapeutic approach to prevent or slow down some behavioural aspects in α-synucleinopathies.
Collapse
Affiliation(s)
- P La Vitola
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - C Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Baroni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Artioli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - G Santamaria
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Castiglioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M Cerovic
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - L Caldinelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - L Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - G Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
22
|
Stanojlovic M, Pallais JP, Kotz CM. Inhibition of Orexin/Hypocretin Neurons Ameliorates Elevated Physical Activity and Energy Expenditure in the A53T Mouse Model of Parkinson's Disease. Int J Mol Sci 2021; 22:E795. [PMID: 33466831 PMCID: PMC7830608 DOI: 10.3390/ijms22020795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
Aside from the classical motor symptoms, Parkinson's disease also has various non-classical symptoms. Interestingly, orexin neurons, involved in the regulation of exploratory locomotion, spontaneous physical activity, and energy expenditure, are affected in Parkinson's. In this study, we hypothesized that Parkinson's-disease-associated pathology affects orexin neurons and therefore impairs functions they regulate. To test this, we used a transgenic animal model of Parkinson's, the A53T mouse. We measured body composition, exploratory locomotion, spontaneous physical activity, and energy expenditure. Further, we assessed alpha-synuclein accumulation, inflammation, and astrogliosis. Finally, we hypothesized that chemogenetic inhibition of orexin neurons would ameliorate observed impairments in the A53T mice. We showed that aging in A53T mice was accompanied by reductions in fat mass and increases in exploratory locomotion, spontaneous physical activity, and energy expenditure. We detected the presence of alpha-synuclein accumulations in orexin neurons, increased astrogliosis, and microglial activation. Moreover, loss of inhibitory pre-synaptic terminals and a reduced number of orexin cells were observed in A53T mice. As hypothesized, this chemogenetic intervention mitigated the behavioral disturbances induced by Parkinson's disease pathology. This study implicates the involvement of orexin in early Parkinson's-disease-associated impairment of hypothalamic-regulated physiological functions and highlights the importance of orexin neurons in Parkinson's disease symptomology.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany
| | - Jean Pierre Pallais
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, 321 Church St SE, Minneapolis, MN 55455, USA; (J.P.P.); (C.M.K.)
| | - Catherine M. Kotz
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, 321 Church St SE, Minneapolis, MN 55455, USA; (J.P.P.); (C.M.K.)
- Minneapolis VA Health Care System, GRECC, 1 Veterans Dr, Minneapolis, MN 55417, USA
| |
Collapse
|
23
|
Hu Y, Guo P, Lian TH, Zuo LJ, Yu SY, Liu L, Jin Z, Yu QJ, Wang RD, Li LX, Piao YS, Zhang W. Clinical Characteristics, Iron Metabolism and Neuroinflammation: New Insight into Excessive Daytime Sleepiness in Parkinson's Disease. Neuropsychiatr Dis Treat 2021; 17:2041-2051. [PMID: 34188474 PMCID: PMC8232841 DOI: 10.2147/ndt.s272110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To investigate the clinical characteristics, iron metabolism and neuroinflammation in Parkinson's disease (PD) patients with excessive daytime sleepiness (EDS). METHODS We studied 379 patients with PD and 30 age-matched controls. All subjects were evaluated by Epworth sleepiness scale (ESS) and a series of rating scales and were divided into PD-EDS and PD-NEDS groups according to ESS score. The concentrations of iron and iron-related proteins and inflammatory cytokines in both cerebrospinal fluid (CSF) and serum were examined. RESULTS 1. The occurrence rate of EDS in total PD patients was 16.09%. 2. PD-EDS group had significantly severer disease stages, more severe motor and non-motor features of the disease. 3. In CSF, the concentrations of iron and IL-1β in the PD-EDS group were significantly higher and ferritin concentration was prominently lower when compared with the PD-NEDS group and the control group; ESS score was significantly associated with high concentrations of iron and IL-1β and low concentration of ferritin in the PD group. Iron concentration was positively correlated with IL-1β concentration in the PD-EDS group. 4. In serum, no changes were observed in iron and iron-related proteins and inflammatory cytokines among the three groups. CONCLUSION EDS was a common symptom in PD patients. PD patients with EDS had more severe motor and some non-motor symptoms. Overloaded iron-relevant inflammation in the brain might be an underlying mechanism of PD-EDS.
Collapse
Affiliation(s)
- Yang Hu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Peng Guo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Teng-Hong Lian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Li-Jun Zuo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Shu-Yang Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Li Liu
- Department of Internal Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Zhao Jin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Qiu-Jin Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Rui-Dan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Li-Xia Li
- Department of Internal Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Ying-Shan Piao
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Wei Zhang
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, 100070, People's Republic of China.,Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, Beijing, 100069, People's Republic of China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, People's Republic of China.,Beijing Key Laboratory on Parkinson's Disease, Beijing, 100053, People's Republic of China
| |
Collapse
|
24
|
Forloni G, La Vitola P, Cerovic M, Balducci C. Inflammation and Parkinson's disease pathogenesis: Mechanisms and therapeutic insight. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:175-202. [PMID: 33453941 DOI: 10.1016/bs.pmbts.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, β amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Pietro La Vitola
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Claudia Balducci
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
25
|
Peña-Díaz S, Pujols J, Pinheiro F, Santos J, Pallarés I, Navarro S, Conde-Gimenez M, García J, Salvatella X, Dalfó E, Sancho J, Ventura S. Inhibition of α-Synuclein Aggregation and Mature Fibril Disassembling With a Minimalistic Compound, ZPDm. Front Bioeng Biotechnol 2020; 8:588947. [PMID: 33178678 PMCID: PMC7597392 DOI: 10.3389/fbioe.2020.588947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/22/2020] [Indexed: 11/13/2022] Open
Abstract
Synucleinopathies are a group of disorders characterized by the accumulation of α-Synuclein amyloid inclusions in the brain. Preventing α-Synuclein aggregation is challenging because of the disordered nature of the protein and the stochastic nature of fibrillogenesis, but, at the same time, it is a promising approach for therapeutic intervention in these pathologies. A high-throughput screening initiative allowed us to discover ZPDm, the smallest active molecule in a library of more than 14.000 compounds. Although the ZPDm structure is highly related to that of the previously described ZPD-2 aggregation inhibitor, we show here that their mechanisms of action are entirely different. ZPDm inhibits the aggregation of wild-type, A30P, and H50Q α-Synuclein variants in vitro and interferes with α-Synuclein seeded aggregation in protein misfolding cyclic amplification assays. However, ZPDm distinctive feature is its strong potency to dismantle preformed α-Synuclein amyloid fibrils. Studies in a Caenorhabditis elegans model of Parkinson's Disease, prove that these in vitro properties are translated into a significant reduction in the accumulation of α-Synuclein inclusions in ZPDm treated animals. Together with previous data, the present work illustrates how different chemical groups on top of a common molecular scaffold can result in divergent but complementary anti-amyloid activities.
Collapse
Affiliation(s)
- Samuel Peña-Díaz
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Pujols
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisca Pinheiro
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jaime Santos
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irantzu Pallarés
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susanna Navarro
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Conde-Gimenez
- Department of Biochemistry and Molecular and Cell Biology, Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, and Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
| | - Jesús García
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Xavier Salvatella
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,ICREA, Barcelona, Spain
| | - Esther Dalfó
- Medicine, M2, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Barcelona, Spain
| | - Javier Sancho
- Department of Biochemistry and Molecular and Cell Biology, Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, and Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
26
|
Kim C, Beilina A, Smith N, Li Y, Kim M, Kumaran R, Kaganovich A, Mamais A, Adame A, Iba M, Kwon S, Lee WJ, Shin SJ, Rissman RA, You S, Lee SJ, Singleton AB, Cookson MR, Masliah E. LRRK2 mediates microglial neurotoxicity via NFATc2 in rodent models of synucleinopathies. Sci Transl Med 2020; 12:eaay0399. [PMID: 33055242 PMCID: PMC8100991 DOI: 10.1126/scitranslmed.aay0399] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/04/2019] [Accepted: 03/31/2020] [Indexed: 12/15/2022]
Abstract
Synucleinopathies are neurodegenerative disorders characterized by abnormal α-synuclein deposition that include Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The pathology of these conditions also includes neuronal loss and neuroinflammation. Neuron-released α-synuclein has been shown to induce neurotoxic, proinflammatory microglial responses through Toll-like receptor 2, but the molecular mechanisms involved are poorly understood. Here, we show that leucine-rich repeat kinase 2 (LRRK2) plays a critical role in the activation of microglia by extracellular α-synuclein. Exposure to α-synuclein was found to enhance LRRK2 phosphorylation and activity in mouse primary microglia. Furthermore, genetic and pharmacological inhibition of LRRK2 markedly diminished α-synuclein-mediated microglial neurotoxicity via lowering of tumor necrosis factor-α and interleukin-6 expression in mouse cultures. We determined that LRRK2 promoted a neuroinflammatory cascade by selectively phosphorylating and inducing nuclear translocation of the immune transcription factor nuclear factor of activated T cells, cytoplasmic 2 (NFATc2). NFATc2 activation was seen in patients with synucleinopathies and in a mouse model of synucleinopathy, where administration of an LRRK2 pharmacological inhibitor restored motor behavioral deficits. Our results suggest that modulation of LRRK2 and its downstream signaling mediator NFATc2 might be therapeutic targets for treating synucleinopathies.
Collapse
Affiliation(s)
- Changyoun Kim
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Alexandria Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nathan Smith
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Biochemistry and Redox Biology Center, University of Nebraska, Lincoln, NE 68588, USA
| | - Yan Li
- Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Minhyung Kim
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ravindran Kumaran
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alice Kaganovich
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anthony Adame
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michiyo Iba
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Somin Kwon
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Won-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Soo-Jean Shin
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Robert A Rissman
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sungyong You
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eliezer Masliah
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Lemos M, Venezia S, Refolo V, Heras-Garvin A, Schmidhuber S, Giese A, Leonov A, Ryazanov S, Griesinger C, Galabova G, Staffler G, Wenning GK, Stefanova N. Targeting α-synuclein by PD03 AFFITOPE® and Anle138b rescues neurodegenerative pathology in a model of multiple system atrophy: clinical relevance. Transl Neurodegener 2020; 9:38. [PMID: 32972456 PMCID: PMC7513530 DOI: 10.1186/s40035-020-00217-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/08/2020] [Indexed: 12/20/2022] Open
Abstract
Background Misfolded oligomeric α-synuclein plays a pivotal role in the pathogenesis of α-synucleinopathies including Parkinson’s disease and multiple system atrophy, and its detection parallels activation of microglia and a loss of neurons in the substantia nigra pars compacta. Here we aimed to analyze the therapeutic efficacy of PD03, a new AFFITOPE® immunotherapy approach, either alone or in combination with Anle138b, in a PLP-α-syn mouse model. Methods The PLP-α-syn mice were treated with PD03 immunotherapy, Anle138b, or a combination of two. Five months after study initiation, the mice underwent behavioral testing and were sacrificed for neuropathological analysis. The treatment groups were compared to the vehicle group with regard to motor performance, nigral neuronal loss, microglial activation and α-synuclein pathology. Results The PLP-α-syn mice receiving the PD03 or Anle138b single therapy showed improvement of gait deficits and preservation of nigral dopaminergic neurons associated with the reduced α-synuclein oligomer levels and decreased microglial activation. The combined therapy with Anle138b and PD03 resulted in lower IgG binding in the brain as compared to the single immunotherapy with PD03. Conclusions PD03 and Anle138b can selectively target oligomeric α-synuclein, resulting in attenuation of neurodegeneration in the PLP-α-syn mice. Both approaches are potential therapies that should be developed further for disease modification in α-synucleinopathies.
Collapse
Affiliation(s)
- Miguel Lemos
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, 6020, Innsbruck, Austria
| | - Serena Venezia
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, 6020, Innsbruck, Austria
| | - Violetta Refolo
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, 6020, Innsbruck, Austria
| | - Antonio Heras-Garvin
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, 6020, Innsbruck, Austria
| | | | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Andrei Leonov
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Sergey Ryazanov
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | | | - Gergana Galabova
- AFFIRIS AG, Vienna, Austria.,Present Address: Origenis GmbH, Munich, Germany
| | | | - Gregor Karl Wenning
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, 6020, Innsbruck, Austria
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Innsbruck Medical University, 6020, Innsbruck, Austria.
| |
Collapse
|
28
|
Lemos M, Stefanova N. Histone Deacetylase 6 and the Disease Mechanisms of α-Synucleinopathies. Front Synaptic Neurosci 2020; 12:586453. [PMID: 33041780 PMCID: PMC7518386 DOI: 10.3389/fnsyn.2020.586453] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/24/2020] [Indexed: 12/31/2022] Open
Abstract
The abnormal accumulation of α-Synuclein (α-Syn) is a prominent pathological feature in a group of diseases called α-Synucleinopathies, such as Parkinson’s disease, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). The formation of Lewy bodies (LBs) and glial cytoplasmic inclusions (GCIs) in neurons and oligodendrocytes, respectively, is highly investigated. However, the molecular mechanisms behind α-Syn improper folding and aggregation remain unclear. Histone deacetylase 6 (HDAC6) is a Class II deacetylase, containing two active catalytic domains and a ubiquitin-binding domain. The properties of HDAC6 and its exclusive cytoplasmic localization allow HDAC6 to modulate the microtubule dynamics, acting as a specific α-tubulin deacetylase. Also, HDAC6 can bind ubiquitinated proteins, facilitating the formation of the aggresome, a cellular defense mechanism to cope with higher levels of misfolded proteins. Several studies report that the aggresome shares similarities in size and composition with LBs and GCIs. HDAC6 is found to co-localize with α-Syn in neurons and in oligodendrocytes, together with other aggresome-related proteins. The involvement of HDAC6 in several neurodegenerative diseases is already under discussion, however, the results obtained by modulating HDAC6 activity are not entirely conclusive. The main goal of this review is to summarize and critically discuss previous in vitro and in vivo data regarding the specific role of HDAC6 in the context of α-Syn accumulation and protein aggregation in α-Synucleinopathies.
Collapse
Affiliation(s)
- Miguel Lemos
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
29
|
Salado-Manzano C, Perpiña U, Straccia M, Molina-Ruiz FJ, Cozzi E, Rosser AE, Canals JM. Is the Immunological Response a Bottleneck for Cell Therapy in Neurodegenerative Diseases? Front Cell Neurosci 2020; 14:250. [PMID: 32848630 PMCID: PMC7433375 DOI: 10.3389/fncel.2020.00250] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders such as Parkinson's (PD) and Huntington's disease (HD) are characterized by a selective detrimental impact on neurons in a specific brain area. Currently, these diseases have no cures, although some promising trials of therapies that may be able to slow the loss of brain cells are underway. Cell therapy is distinguished by its potential to replace cells to compensate for those lost to the degenerative process and has shown a great potential to replace degenerated neurons in animal models and in clinical trials in PD and HD patients. Fetal-derived neural progenitor cells, embryonic stem cells or induced pluripotent stem cells are the main cell sources that have been tested in cell therapy approaches. Furthermore, new strategies are emerging, such as the use of adult stem cells, encapsulated cell lines releasing trophic factors or cell-free products, containing an enriched secretome, which have shown beneficial preclinical outcomes. One of the major challenges for these potential new treatments is to overcome the host immune response to the transplanted cells. Immune rejection can cause significant alterations in transplanted and endogenous tissue and requires immunosuppressive drugs that may produce adverse effects. T-, B-lymphocytes and microglia have been recognized as the main effectors in striatal graft rejection. This review aims to summarize the preclinical and clinical studies of cell therapies in PD and HD. In addition, the precautions and strategies to ensure the highest quality of cell grafts, the lowest risk during transplantation and the reduction of a possible immune rejection will be outlined. Altogether, the wide-ranging possibilities of advanced therapy medicinal products (ATMPs) could make therapeutic treatment of these incurable diseases possible in the near future.
Collapse
Affiliation(s)
- Cristina Salado-Manzano
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Unai Perpiña
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Francisco J. Molina-Ruiz
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Emanuele Cozzi
- Department of Cardio-Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
- Transplant Immunology Unit, Padua University Hospital, Padua, Italy
| | - Anne E. Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Josep M. Canals
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| |
Collapse
|
30
|
Ferrer I, Andrés-Benito P, Zelaya MV, Aguirre MEE, Carmona M, Ausín K, Lachén-Montes M, Fernández-Irigoyen J, Santamaría E, del Rio JA. Familial globular glial tauopathy linked to MAPT mutations: molecular neuropathology and seeding capacity of a prototypical mixed neuronal and glial tauopathy. Acta Neuropathol 2020; 139:735-771. [PMID: 31907603 PMCID: PMC7096369 DOI: 10.1007/s00401-019-02122-9] [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: 10/21/2019] [Revised: 12/23/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Globular glial tauopathy (GGT) is a progressive neurodegenerative disease involving the grey matter and white matter (WM) and characterized by neuronal deposition of hyper-phosphorylated, abnormally conformed, truncated, oligomeric 4Rtau in neurons and in glial cells forming typical globular astrocyte and oligodendrocyte inclusions (GAIs and GOIs, respectively) and coiled bodies. Present studies centre on four genetic GGT cases from two unrelated families bearing the P301T mutation in MAPT and one case of sporadic GGT (sGGT) and one case of GGT linked to MAPT K317M mutation, for comparative purposes. Clinical and neuropathological manifestations and biochemical profiles of phospho-tau are subjected to individual variations in patients carrying the same mutation, even in carriers of the same family, independently of the age of onset, gender, and duration of the disease. Immunohistochemistry, western blotting, transcriptomic, proteomics and phosphoproteomics, and intra-cerebral inoculation of brain homogenates to wild-type (WT) mice were the methods employed. In GGT cases linked to MAPT P301T mutation, astrocyte markers GFAP, ALDH1L1, YKL40 mRNA and protein, GJA1 mRNA, and AQ4 protein are significantly increased; glutamate transporter GLT1 (EAAT2) and glucose transporter (SLC2A1) decreased; mitochondrial pyruvate carrier 1 (MPC1) increased, and mitochondrial uncoupling protein 5 (UCP5) almost absent in GAIs in frontal cortex (FC). Expression of oligodendrocyte markers OLIG1 and OLIG2mRNA, and myelin-related genes MBP, PLP1, CNP, MAG, MAL, MOG, and MOBP are significantly decreased in WM; CNPase, PLP1, and MBP antibodies reveal reduction and disruption of myelinated fibres; and SMI31 antibodies mark axonal damage in the WM. Altered expression of AQ4, GLUC-t, and GLT-1 is also observed in sGGT and in GGT linked to MAPT K317M mutation. These alterations point to primary astrogliopathy and oligodendrogliopathy in GGT. In addition, GGT linked to MAPT P301T mutation proteotypes unveil a proteostatic imbalance due to widespread (phospho)proteomic dearrangement in the FC and WM, triggering a disruption of neuron projection morphogenesis and synaptic transmission. Identification of hyper-phosphorylation of variegated proteins calls into question the concept of phospho-tau-only alteration in the pathogenesis of GGT. Finally, unilateral inoculation of sarkosyl-insoluble fractions of GGT homogenates from GGT linked to MAPT P301T, sGGT, and GGT linked to MAPT K317M mutation in the hippocampus, corpus callosum, or caudate/putamen in wild-type mice produces seeding, and time- and region-dependent spreading of phosphorylated, non-oligomeric, and non-truncated 4Rtau and 3Rtau, without GAIs and GOIs but only of coiled bodies. These experiments prove that host tau strains are important in the modulation of cellular vulnerability and phenotypes of phospho-tau aggregates.
Collapse
|
31
|
Zhang F, Niu L, Liu X, Liu Y, Li S, Yu H, Le W. Rapid Eye Movement Sleep Behavior Disorder and Neurodegenerative Diseases: An Update. Aging Dis 2020; 11:315-326. [PMID: 32257544 PMCID: PMC7069464 DOI: 10.14336/ad.2019.0324] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022] Open
Abstract
Rapid eye movement sleep behavior disorder (RBD) is a sleep behavior disorder characterized by abnormal behaviors and loss of muscle atonia during rapid eye movement (REM) sleep. RBD is generally considered to be associated with synucleinopathies, such as Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), and usually precedes years before the first symptom of these diseases. It is believed that RBD predicts the neurodegeneration in synucleinopathy. However, increasing evidences have shown that RBD is also found in non-synucleinopathy neurodegenerative diseases, including Alzheimer’s disease (AD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), etc. Sleep disturbance such as RBD may be an early sign of neurodegeneration in these diseases, and also serve as an assessment of cognitive impairments. In this review, we updated the clinical characteristics, diagnosis, and possible mechanisms of RBD in neurogenerative diseases. A better understanding of RBD in these neurogenerative diseases will provide biomarkers and novel therapeutics for the early diagnosis and treatment of the diseases.
Collapse
Affiliation(s)
- Feng Zhang
- 1Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,2Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Long Niu
- 1Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,2Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xinyao Liu
- 1Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,2Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yufei Liu
- 1Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,2Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- 1Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,2Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Huan Yu
- 3Sleep and Wake Disorders Center and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weidong Le
- 1Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,2Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
32
|
Kery R, Chen APF, Kirschen GW. Genetic targeting of astrocytes to combat neurodegenerative disease. Neural Regen Res 2020; 15:199-211. [PMID: 31552885 PMCID: PMC6905329 DOI: 10.4103/1673-5374.265541] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Astrocytes, glial cells that interact extensively with neurons and other support cells throughout the central nervous system, have recently come under the spotlight for their potential contribution to, or potential regenerative role in a host of neurodegenerative disorders. It is becoming increasingly clear that astrocytes, in concert with microglial cells, activate intrinsic immunological pathways in the setting of neurodegenerative injury, although the direct and indirect consequences of such activation are still largely unknown. We review the current literature on the astrocyte’s role in several neurodegenerative diseases, as well as highlighting recent advances in genetic manipulation of astrocytes that may prove critical to modulating their response to neurological injury, potentially combatting neurodegenerative damage.
Collapse
Affiliation(s)
- Rachel Kery
- Medical Scientist Training Program (MSTP), Stony Brook Medicine; Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Allen P F Chen
- Medical Scientist Training Program (MSTP), Stony Brook Medicine; Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Gregory W Kirschen
- Medical Scientist Training Program (MSTP), Stony Brook Medicine, Stony Brook, NY, USA
| |
Collapse
|
33
|
Li Y, Niu M, Zhao A, Kang W, Chen Z, Luo N, Zhou L, Zhu X, Lu L, Liu J. CXCL12 is involved in α-synuclein-triggered neuroinflammation of Parkinson's disease. J Neuroinflammation 2019; 16:263. [PMID: 31831012 PMCID: PMC6909602 DOI: 10.1186/s12974-019-1646-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/18/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The mechanisms underlying the pathogenesis and progression of Parkinson's disease (PD) remain elusive, but recent opinions and perspectives have focused on whether the inflammation process induced by microglia contributes to α-synuclein-mediated toxicity. Migration of microglia to the substantia nigra (SN) could precede neurodegeneration in A53T mice. We hypothesized that CXCL12 could be a mediator in the α-synuclein-induced migration of microglia. METHODS After establishing appropriate animal and cell culture models, we explored the relationship between α-synuclein and CXCL12 in A53T mice, primary microglia, and BV-2 cell lines. We also explored the mechanisms of these interactions and the signaling processes involved in neuroinflammation. RESULTS We confirmed the positive correlation between α-synuclein and CXCL12 in the postmortem brain tissue of PD patients and the upregulated CXCR4 expression in SN microglia of A53T mice. In addition, as expected, α-synuclein increased the production of CXCL12 in microglia via TLR4/IκB-α/NF-κB signaling. Importantly, CXCL12/CXCR4/FAK/Src/Rac1 signaling was shown to be involved in α-synuclein-induced microglial accumulation. CONCLUSIONS Our study suggests that CXCL12 could be a novel target for the prevention of α-synuclein-triggered ongoing microglial responses. Blocking CXCL12/CXCR4 may be a potential therapeutic approach for PD progression.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Mengyue Niu
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.,Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Aonan Zhao
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wenyan Kang
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Zhichun Chen
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Ningdi Luo
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Liche Zhou
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Liming Lu
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Jun Liu
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
34
|
Zhang H, Wu J, Shen FF, Yuan YS, Li X, Ji P, Zhu L, Sun L, Ding J, Niu Q, Zhang KZ. Activated Schwann cells and increased inflammatory cytokines IL-1β, IL-6, and TNF-α in patients' sural nerve are lack of tight relationship with specific sensory disturbances in Parkinson's disease. CNS Neurosci Ther 2019; 26:518-526. [PMID: 31828965 PMCID: PMC7163790 DOI: 10.1111/cns.13282] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 01/05/2023] Open
Abstract
Aims Neuroinflammation is one of the most important processes in the pathogenesis of Parkinson's disease (PD). Sensory disturbances are common in patients with PD, but the underlying pathophysiological mechanisms remain unknown. This study aimed to characterize the activation of Schwann cells (SCs) and the increase of expression of inflammatory cytokines IL‐1β, IL‐6, and TNF‐α in the sural nerve of PD, and further explore whether peripheral nerve inflammation is the cause of PD sensory disturbances. Methods A total of 14 patients with PD (including 5 with sensory disturbances and 9 without sensory disturbances) and 6 controls were included. The excitation and conduction function of sural nerve was detected by sural nerve electrophysiological examination. With sural nerve biopsy samples, ultrastructural changes of sural nerve were observed by electron microscopy; Schwann cell biomarker glial fibrillary acid protein (GFAP) and inflammatory cytokines including interleukin‐1beta (IL‐1β), interleukin 6 (IL‐6), and tumor necrosis factor‐alpha (TNF‐α) were detected by immunohistochemistry, and the outcome of immunostaining slice was semiquantitatively counted; double immunofluorescence was used to identify the locus immunoreactive for inflammatory cytokines. Results Compared with healthy controls, nerve conduction velocity (NCV) slowed down and sensory nerve action potential (SNAP) amplitude decreased in PD patients, accompanied by axonal degeneration and demyelinating lesions, and expression of GFAP and inflammatory cytokines was increased. Inflammatory cytokines were significantly colocalized with GFAP and slightly colocalized with NF. These indicators did not differ significantly between PD patients with and without sensory disturbances. Conclusion Our study results suggest that peripheral sensory nerve injury exists in PD patients, accompanied by Schwann cell activation and inflammation, thus demonstrate peripheral nerve inflammation participates in the pathophysiological process of PD but it is not necessarily related to the patient's sensory disturbance.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei-Fei Shen
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong-Sheng Yuan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pan Ji
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Zhu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Sun
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Ding
- Department of Neurology, The First People's Hospital of Changzhou, Changzhou, China
| | - Qi Niu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ke-Zhong Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Earls RH, Menees KB, Chung J, Barber J, Gutekunst CA, Hazim MG, Lee JK. Intrastriatal injection of preformed alpha-synuclein fibrils alters central and peripheral immune cell profiles in non-transgenic mice. J Neuroinflammation 2019; 16:250. [PMID: 31796095 PMCID: PMC6889316 DOI: 10.1186/s12974-019-1636-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the accumulation of alpha-synuclein (α-syn) inclusions, the major component of Lewy bodies. Extracellular α-syn aggregates act as a damage-associated molecular pattern (DAMP) and the presence of autoantibodies against α-syn species in the cerebrospinal fluid and the serum of PD patients implicate the involvement of innate and adaptive immune responses. In non-transgenic (Tg) mice, intrastriatal injection of preformed fibril (PFF) α-syn results in widespread pathologic α-syn inclusions in the CNS. While the PFF model has been broadly utilized to study the mechanistic relationship between α-syn transmission and other neuropathological phenotypes, the immune phenotypes in this model are not clearly demonstrated. This study aimed to characterize the immune phenotypes during pathologic α-syn propagation by utilizing PFF α-syn-injected non-tg mice. Here, we showed that pathologic α-syn inclusions are prevalent in various brain regions and the gut at 5 months post injection (p.i.), preceding the degeneration of dopaminergic neurons in substantia nigra (SN). We discovered a distinct inflammatory response involving both activation of microglia and astrocytes and infiltration of B, CD4+ T, CD8+ T, and natural killer cells in the brain at 5 months p.i. Moreover, PFF α-syn-injected mice display significant alterations in the frequency and number of leukocyte subsets in the spleen and lymph nodes with minimum alterations in the blood. Our data provide primary evidence that intracerebral-initiated synucleinopathies in non-tg mice alter immune cell profiles both in the CNS and peripheral lymphoid organs. Furthermore, our data provides support for utilizing this mouse model to assess the mechanistic connection between immune responses and synuclein pathology.
Collapse
Affiliation(s)
- Rachael H Earls
- Department of Physiology and Pharmacology, University of Georgia College of Veterinary Medicine, 501 D.W. Brooks Drive, Athens, GA, 30602, USA
| | - Kelly B Menees
- Department of Physiology and Pharmacology, University of Georgia College of Veterinary Medicine, 501 D.W. Brooks Drive, Athens, GA, 30602, USA
| | - Jaegwon Chung
- Department of Physiology and Pharmacology, University of Georgia College of Veterinary Medicine, 501 D.W. Brooks Drive, Athens, GA, 30602, USA
| | - James Barber
- Department of Infectious Disease, University of Georgia College of Veterinary Medicine, Athens, GA, 30602, USA
| | - Claire-Anne Gutekunst
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Manuel G Hazim
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, University of Georgia College of Veterinary Medicine, 501 D.W. Brooks Drive, Athens, GA, 30602, USA.
| |
Collapse
|
36
|
Stanojlovic M, Pallais JP, Lee MK, Kotz CM. Pharmacological and chemogenetic orexin/hypocretin intervention ameliorates Hipp-dependent memory impairment in the A53T mice model of Parkinson's disease. Mol Brain 2019; 12:87. [PMID: 31666100 PMCID: PMC6822428 DOI: 10.1186/s13041-019-0514-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/18/2019] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD), classically defined as a progressive motor disorder accompanied with dopaminergic neuron loss and presence of Lewy bodies, is the second most common neurodegenerative disease. PD also has various non-classical symptoms, including cognitive impairments. In addition, inflammation and astrogliosis are recognized as an integral part of PD pathology. The hippocampus (Hipp) is a brain region involved in cognition and memory, and the neuropeptide orexin has been shown to enhance learning and memory. Previous studies show impairments in Hipp-dependent memory in a transgenic mouse model of Parkinson's disease (A53T mice), and we hypothesized that increasing orexin tone will reverse this. To test this, we subjected 3, 5, and 7-month old A53T mice to a Barnes maze and a contextual object recognition test to determine Hipp dependent memory. Inflammation and astrogliosis markers in the Hipp were assessed by immuno-fluorescence densitometry. The data show that early cognitive impairment is coupled with an increase in expression of inflammatory and astrogliosis markers. Next, in two separate experiments, mice were given intra-hippocampal injections of orexin or chemogenetic viral injections of an orexin neuron specific Designer Receptor Exclusively Activated by Designer Drug (DREADD). For the pharmacological approach mice were intracranially treated with orexin A, whereas the chemogenetic approach utilized clozapine N-oxide (CNO). Both pharmacological orexin A intervention as well as chemogenetic activation of orexin neurons ameliorated Hipp-dependent early memory impairment observed in A53T mice. This study implicates orexin in PD-associated cognitive impairment and suggests that exogenous orexin treatment and/or manipulation of endogenous orexin levels may be a potential strategy for addressing early cognitive loss in PD.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA.
| | - Jean Pierre Pallais
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience (ITN), University of Minnesota, Minneapolis, MN, USA
| | - Catherine M Kotz
- Integrative Biology and Physiology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
- Minneapolis VA Health Care System, GRECC, Minneapolis, MN, USA
| |
Collapse
|
37
|
Stanojlovic M, Pallais JP, Kotz CM. Chemogenetic Modulation of Orexin Neurons Reverses Changes in Anxiety and Locomotor Activity in the A53T Mouse Model of Parkinson's Disease. Front Neurosci 2019; 13:702. [PMID: 31417337 PMCID: PMC6682689 DOI: 10.3389/fnins.2019.00702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/21/2019] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PD symptomology is recognized as heterogeneous and in addition to motor function decline includes cognitive, mood, sleep, and metabolic disorders. Previous studies showed early reductions in anxiety and locomotion in the A53T mice model of PD. Since inflammation and astrogliosis are an integral part of PD pathology and impair proper neuronal function, we were keen to investigate if behavioral changes in A53T mice are accompanied by increased inflammation and astrogliosis in the hippocampus (Hipp) and motor cortex (mCtx) brain regions involved in the regulation of anxiety and locomotion, respectively. To test this, we used 3-, 5-, and 7-month-old A53T mice to examine anxiety-like behavior, locomotion, and expression of inflammation and astrogliosis markers in the Hipp and mCtx. Further, we examined the presence of alpha-synuclein accumulation in orexin neurons and orexin neuronal loss. The data show early reductions in anxiety-like behavior as well as increased locomotor activity, which was accompanied by inflammation and astrogliosis in the Hipp and mCtx. Due to the persistence of the orexin neuron population in A53T mice and the involvement of orexin in anxiety and locomotor regulation, we hypothesized that chemogenetic modulation of orexin neurons would reverse the observed reductions in anxiety-like behavior and the increases in locomotor activity in these animals. We showed that chemogenetic activation of orexin neurons in A53T mice restores anxiety-like behavior back to control levels without affecting locomotor activity, whereas the inhibition of orexin neurons reverses the elevated locomotor activity without any effects on anxiety-like behavior. This study exemplifies the complex role of orexin neurons in this model of PD and demonstrates the novel finding that changes in locomotor and anxiety-like behavior are accompanied by inflammation and astrogliosis. Together, these data suggest that the orexin system may play a significant role in early and late stages of PD.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Catherine M Kotz
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States.,Minneapolis VA Health Care System, Geriatric Research, Education and Clinical Center, Minneapolis, MN, United States
| |
Collapse
|
38
|
Hermann JK, Capadona JR. Understanding the Role of Innate Immunity in the Response to Intracortical Microelectrodes. Crit Rev Biomed Eng 2019; 46:341-367. [PMID: 30806249 DOI: 10.1615/critrevbiomedeng.2018027166] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intracortical microelectrodes exhibit enormous potential for researching the nervous system, steering assistive devices and functional electrode stimulation systems for severely paralyzed individuals, and augmenting the brain with computing power. Unfortunately, intracortical microelectrodes often fail to consistently record signals over clinically useful periods. Biological mechanisms, such as the foreign body response to intracortical microelectrodes and self-perpetuating neuroinflammatory cascades, contribute to the inconsistencies and decline in recording performance. Unfortunately, few studies have directly correlated microelectrode performance with the neuroinflammatory response to the implanted devices. However, of those select studies that have, the role of the innate immune system remains among the most likely links capable of corroborating the results of different studies, across laboratories. Therefore, the overall goal of this review is to highlight the role of innate immunity signaling in the foreign body response to intracortical microelectrodes and hypothesize as to appropriate strategies that may become the most relevant in enabling brain-dwelling electrodes of any geometry, or location, for a range of clinical applications.
Collapse
Affiliation(s)
- John K Hermann
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland, OH 44106; Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland, OH 44106; Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702
| |
Collapse
|
39
|
Stanojlovic M, Pallais Yllescas JP, Vijayakumar A, Kotz C. Early Sociability and Social Memory Impairment in the A53T Mouse Model of Parkinson's Disease Are Ameliorated by Chemogenetic Modulation of Orexin Neuron Activity. Mol Neurobiol 2019; 56:8435-8450. [PMID: 31250383 DOI: 10.1007/s12035-019-01682-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/14/2019] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is a multi-layered progressive neurodegenerative disease. Signature motor system impairments are accompanied by a variety of other symptoms such as mood, sleep, metabolic, and cognitive disorders. Interestingly, social cognition impairments can be observed from the earliest stages of the disease, prior to the onset of the motor symptoms. In this study, we investigated age-related reductions in sociability and social memory in the A53T mouse model of PD. Since inflammation and astrogliosis are an integral part of PD pathology and impair proper neuronal function, we examined astrogliosis and inflammation markers and parvalbumin expression in medial pre-frontal cortex (mPFC), part of the brain responsible for social cognition regulation. Finally, we used DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) for the stimulation and inhibition of orexin neuronal activity to modulate sociability and social memory in A53T mice. We observed that social cognition impairment in A53T mice is accompanied by an increase in astrogliosis and inflammation markers, in addition to loss of parvalbumin neurons and inhibitory pre-synaptic terminals in the mPFC. Moreover, DREADD-induced activation of orexin neurons restores social cognition in the A53T mouse model of PD. SIGNIFICANCE STATEMENT: Social cognition is severely affected in the early stages of Parkinson's disease. In this study, we identified the A53T mouse as a model of social cognitive impairment in PD. Observed alterations in sociability and social memory are accompanied by loss of parvalbumin positive neurons and loss of inhibitory input to mPFC. Stimulating orexin neurons using a chemogenetic approach (DREADDs) ameliorated social cognitive impairment. This study identifies a role for orexin neurons in social cognition in PD and suggests potential therapeutic targets for PD-related social cognition impairments.
Collapse
Affiliation(s)
- Milos Stanojlovic
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.
| | | | - Aarthi Vijayakumar
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Catherine Kotz
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA.,GRECC, Minneapolis VA Health Care System, Minneapolis, MN, USA
| |
Collapse
|
40
|
Jellinger KA. Neuropathology and pathogenesis of extrapyramidal movement disorders: a critical update-I. Hypokinetic-rigid movement disorders. J Neural Transm (Vienna) 2019; 126:933-995. [PMID: 31214855 DOI: 10.1007/s00702-019-02028-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
Abstract
Extrapyramidal movement disorders include hypokinetic rigid and hyperkinetic or mixed forms, most of them originating from dysfunction of the basal ganglia (BG) and their information circuits. The functional anatomy of the BG, the cortico-BG-thalamocortical, and BG-cerebellar circuit connections are briefly reviewed. Pathophysiologic classification of extrapyramidal movement disorder mechanisms distinguish (1) parkinsonian syndromes, (2) chorea and related syndromes, (3) dystonias, (4) myoclonic syndromes, (5) ballism, (6) tics, and (7) tremor syndromes. Recent genetic and molecular-biologic classifications distinguish (1) synucleinopathies (Parkinson's disease, dementia with Lewy bodies, Parkinson's disease-dementia, and multiple system atrophy); (2) tauopathies (progressive supranuclear palsy, corticobasal degeneration, FTLD-17; Guamian Parkinson-dementia; Pick's disease, and others); (3) polyglutamine disorders (Huntington's disease and related disorders); (4) pantothenate kinase-associated neurodegeneration; (5) Wilson's disease; and (6) other hereditary neurodegenerations without hitherto detected genetic or specific markers. The diversity of phenotypes is related to the deposition of pathologic proteins in distinct cell populations, causing neurodegeneration due to genetic and environmental factors, but there is frequent overlap between various disorders. Their etiopathogenesis is still poorly understood, but is suggested to result from an interaction between genetic and environmental factors. Multiple etiologies and noxious factors (protein mishandling, mitochondrial dysfunction, oxidative stress, excitotoxicity, energy failure, and chronic neuroinflammation) are more likely than a single factor. Current clinical consensus criteria have increased the diagnostic accuracy of most neurodegenerative movement disorders, but for their definite diagnosis, histopathological confirmation is required. We present a timely overview of the neuropathology and pathogenesis of the major extrapyramidal movement disorders in two parts, the first one dedicated to hypokinetic-rigid forms and the second to hyperkinetic disorders.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
41
|
Morenas-Rodríguez E, Alcolea D, Suárez-Calvet M, Muñoz-Llahuna L, Vilaplana E, Sala I, Subirana A, Querol-Vilaseca M, Carmona-Iragui M, Illán-Gala I, Ribosa-Nogué R, Blesa R, Haass C, Fortea J, Lleó A. Different pattern of CSF glial markers between dementia with Lewy bodies and Alzheimer's disease. Sci Rep 2019; 9:7803. [PMID: 31127154 PMCID: PMC6534578 DOI: 10.1038/s41598-019-44173-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
The role of innate immunity in dementia with Lewy bodies (DLB) has been little studied. We investigated the levels in cerebrospinal fluid (CSF) of glial proteins YKL-40, soluble TREM2 (sTREM2) and progranulin in DLB and their relationship with Alzheimer's disease (AD) biomarkers. We included patients with DLB (n = 37), prodromal DLB (prodDLB, n = 23), AD dementia (n = 50), prodromal AD (prodAD, n = 53), and cognitively normal subjects (CN, n = 44). We measured levels of YKL-40, sTREM2, progranulin, Aβ1-42, total tau (t-tau) and phosphorylated tau (p-tau) in CSF. We stratified the group DLB according to the ratio t-tau/Aβ1-42 (≥0.52, indicative of AD pathology) and the A/T classification. YKL-40, sTREM2 and progranulin levels did not differ between DLB groups and CN. YKL-40 levels were higher in AD and prodAD compared to CN and to DLB and prodDLB. Patients with DLB with a CSF profile suggestive of AD copathology had higher levels of YKL-40, but not sTREM2 or PGRN, than those without. T+ DLB patients had also higher YKL-40 levels than T-. Of these glial markers, only YKL-40 correlated with t-tau and p-tau in DLB and in prodDLB. In contrast, in prodAD, sTREM2 and PGRN also correlated with t-tau and p-tau. In conclusion, sTREM2 and PGRN are not increased in the CSF of DLB patients. YKL-40 is only increased in DLB patients with an AD biomarker profile, suggesting that the increase is driven by AD-related neurodegeneration. These data suggest a differential glial activation between DLB and AD.
Collapse
Affiliation(s)
- Estrella Morenas-Rodríguez
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Daniel Alcolea
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Marc Suárez-Calvet
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Laia Muñoz-Llahuna
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Eduard Vilaplana
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Isabel Sala
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrea Subirana
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Marta Querol-Vilaseca
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - María Carmona-Iragui
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Ignacio Illán-Gala
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Roser Ribosa-Nogué
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Rafael Blesa
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Christian Haass
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Juan Fortea
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain
| | - Alberto Lleó
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
- Institut d'Investigacions Biomediques Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Instituto de Salud Carlos III, Barcelona, Spain.
| |
Collapse
|
42
|
Tamtaji OR, Behnam M, Pourattar MA, Jafarpour H, Asemi Z. Aquaporin 4: A key player in Parkinson's disease. J Cell Physiol 2019; 234:21471-21478. [PMID: 31127615 DOI: 10.1002/jcp.28871] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases which occur in aged people worldwide. Given that a sequence of cellular and molecular mechanisms, including oxidative stresses, apoptosis, inflammatory pathways, microglia, astrocyte activation, and aquaporin 4 (AQP4) are associated with initiation and the progression of PD. AQP4 may affect various pathways (i.e., α-synuclein, inflammatory pathways, and microglia and astrocyte activation). Few reports have evaluated the relationship between AQP4 and PD-related cellular and molecular pathways. Here, for the first time, we highlighted the relationship between AQP4 and molecular mechanisms involved in PD pathogenesis.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | | | - Hamed Jafarpour
- Student Research Committee, Mazandaran University of Medical Science, Sari, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
43
|
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
44
|
Ndayisaba A, Jellinger K, Berger T, Wenning GK. TNFα inhibitors as targets for protective therapies in MSA: a viewpoint. J Neuroinflammation 2019; 16:80. [PMID: 30975183 PMCID: PMC6458780 DOI: 10.1186/s12974-019-1477-5] [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: 12/11/2018] [Accepted: 04/02/2019] [Indexed: 01/06/2023] Open
Abstract
Multiple system atrophy (MSA) is a unique and fatal α-synucleinopathy associated with oligodendroglial inclusions and secondary neurodegeneration affecting striatum, substantia nigra, pons, and cerebellum. The pathogenesis remains elusive; however, there is emerging evidence suggesting a prominent role of neuroinflammation. Here, we critically review the relationship between αS and microglial activation depending on its aggregation state and its role in neuroinflammation to explore the potential of TNFα inhibitors as a treatment strategy for MSA and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, A-6020 Innsbruck, Austria
| | - Kurt Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Gregor K. Wenning
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstraße 35, A-6020 Innsbruck, Austria
| |
Collapse
|
45
|
de Boni L, Wüllner U. Epigenetic Analysis in Human Neurons: Considerations for Disease Modeling in PD. Front Neurosci 2019; 13:276. [PMID: 31024227 PMCID: PMC6460245 DOI: 10.3389/fnins.2019.00276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 03/08/2019] [Indexed: 12/28/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder next to Alzheimer’s disease. Most PD cases are considered to be sporadic and despite considerable scientific effort, the underlying cause(s) still remain(s) enigmatic. In particular, it is unknown to which extent epigenetic alterations contribute to the pathophysiology of this devastating disorder. This is partly due to the fact that appropriate PD models are not yet available. Moreover, epigenetic patterns and mechanisms are species specific and murine systems reflect only a few of the idiosyncrasies of human neurons. For several years now, patient-specific stem cell-derived neural and non-neural cells have been employed to overcome this limitation allowing the analysis and establishment of humanized disease models for PD. Thus, several studies tried to dissect epigenetic alterations such as aberrant DNA methylation or microRNA patterns using lund human mesencephalic cell lines or neurons derived from (patient-specific) induced pluripotent stem cells. These studies demonstrate that human neurons have the potential to be used as model systems for the study of epigenetic modifications in PD such as characterizing epigenetic changes, correlating epigenetic changes to gene expression alterations and hopefully using these insights for the development of novel therapeutics. However, more research is required to define the epigenetic (age-associated) landscape of human in vitro neurons and compare these to native neurons before they can be established as suitable models for epigenetic studies in PD. In this review, we summarize the knowledge about epigenetic studies performed on human neuronal PD models, and we discuss advantages and current limitations of these (stem cell-derived) neuronal models for the study of epigenetic alterations in PD.
Collapse
Affiliation(s)
- Laura de Boni
- Dementia Research Institute, University College London, London, United Kingdom
| | - Ullrich Wüllner
- Department of Neurology, University Hospital Bonn, German Center for Neurologic Diseases, Bonn, Germany
| |
Collapse
|
46
|
Pain S, Vergote J, Gulhan Z, Bodard S, Chalon S, Gaillard A. Inflammatory process in Parkinson disease: neuroprotection by neuropeptide Y. Fundam Clin Pharmacol 2019; 33:544-548. [PMID: 30866091 DOI: 10.1111/fcp.12464] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/26/2019] [Accepted: 03/08/2019] [Indexed: 01/27/2023]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons in the nigro-striatal pathway. Interestingly, it has already been shown that an intracerebral administration of neuropeptide Y (NPY) decreases the neurodegeneration induced by 6-hydroxydopamine (6-OHDA) in rodents and prevents loss of dopamine (DA) and DA transporter density. The etiology of idiopathic PD now suggest that chronic production of inflammatory mediators by activated microglial cells mediates the majority of DA-neuronal tissue destruction. In an animal experimental model of PD, the present study shows that NPY inhibited the activation of microglia evaluated by the binding of the translocator protein (TSPO) ligand [3H]PK11195 in striatum and substantia nigra of 6-OHDA rats. These results suggest a potential role for inflammation in the pathophysiology of the disease and a potential treatment by NPY in PD.
Collapse
Affiliation(s)
- Stéphanie Pain
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC)-INSERM U1084, Pôle Biologie-Santé, Université de Poitiers, Poitiers, France
| | - Jackie Vergote
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Zuhal Gulhan
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvie Bodard
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Afsaneh Gaillard
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC)-INSERM U1084, Pôle Biologie-Santé, Université de Poitiers, Poitiers, France
| |
Collapse
|
47
|
Cis-4-[18F]fluoro-D-proline detects neurodegeneration in patients with akinetic-rigid parkinsonism. Nucl Med Commun 2019; 40:383-387. [PMID: 30875335 DOI: 10.1097/mnm.0000000000000982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES This study aimed to investigate whether the amino acid PET tracer cis-4-[F]fluoro-D-proline [D-cis-[F]FPro] shows increased uptake in the basal ganglia of patients with neurodegenerative akinetic-rigid parkinsonism. D-Cis-[F]FPro is a sensitive PET tracer for inflammation-associated neurodegeneration in animal models. We hypothesized that D-cis-[F]FPro might also be a sensitive marker of alterations of the basal ganglia in parkinsonian syndromes. PARTICIPANTS AND METHODS Ten patients with neurodegenerative akinetic-rigid parkinsonism (five with idiopathic Parkinson's disease and five with atypical parkinsonian syndromes) were imaged with D-cis-[F]FPro and compared with 13 patients with brain tumors who had no basal ganglia involvement. PET images 20-50 min after injection were evaluated and tracer uptake in the basal ganglia was quantified using volume-of-interest analysis with basal ganglia to background ratios. The severity of disease was assessed with unified Parkinson's disease rating scale III and correlated with D-cis-[F]FPro uptake. RESULTS In patients with parkinsonism, volume-of-interest analysis showed mild, but significantly increased D-cis-[F]FPro uptake in the basal ganglia, pronounced in the lenticular nucleus. Disease severity correlated with D-cis-[F]FPro uptake in the right pallidum (r=-0.687, P=0.041). CONCLUSION Data suggest that D-cis-[F]FPro is a sensitive marker of inflammation-associated degenerative processes in parkinsonian syndromes.
Collapse
|
48
|
Mot AI, Depp C, Nave KA. An emerging role of dysfunctional axon-oligodendrocyte coupling in neurodegenerative diseases. DIALOGUES IN CLINICAL NEUROSCIENCE 2019. [PMID: 30936768 PMCID: PMC6436955 DOI: 10.31887/dcns.2018.20.4/amot] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Myelin is made by highly specialized glial cells and enables fast axonal impulse propagation. Recent studies show that oligodendrocytes in the central nervous system are, in addition to myelination, required for the integrity and survival of axons, independent of the presence or absence of myelin itself. The underlying mechanism of this support is given by glycolytic oligodendrocytes which provide axons with energy-rich metabolites. These findings represent a paradigm shift for the physiological function of axon-associated glia, and open the intriguing possibility that oligodendrocytes are important contributors to neurodegenerative diseases in which myelinated axons are lost, such as in Alzheimer disease, amyotrophic lateral sclerosis, and multiple system atrophy. Understanding the role of axon-oligodendrocyte coupling in neurodegenerative diseases may pave the way for the development of metabolism-based therapeutic approaches.
Collapse
Affiliation(s)
- Alexandra I Mot
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Gottingen, Germany
| | - Constanze Depp
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Gottingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Gottingen, Germany
| |
Collapse
|
49
|
Yang Q, Zhou J. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2018; 67:1017-1035. [DOI: 10.1002/glia.23571] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Qiao‐qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
| | - Jia‐wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
- University of Chinese Academy of Sciences Shanghai 200031 China
| |
Collapse
|
50
|
Brás IC, Lopes LV, Outeiro TF. Sensing α-Synuclein From the Outside via the Prion Protein: Implications for Neurodegeneration. Mov Disord 2018; 33:1675-1684. [DOI: 10.1002/mds.27478] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Affiliation(s)
- Inês Caldeira Brás
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
| | - Luísa V. Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; Lisboa Portugal
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration; University Medical Center Göttingen; Göttingen Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciências Médicas; Universidade Nova de Lisboa, Campo dos Mártires da Pátria; Lisboa Portugal
- Max Planck Institute for Experimental Medicine; Göttingen Germany
- Institute of Neuroscience, The Medical School; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|