101
|
Li J, Chen L, Qin Q, Wang D, Zhao J, Gao H, Yuan X, Zhang J, Zou Y, Mao Z, Xiong Y, Min Z, Yan M, Wang CY, Xue Z. Upregulated hexokinase 2 expression induces the apoptosis of dopaminergic neurons by promoting lactate production in Parkinson's disease. Neurobiol Dis 2022; 163:105605. [PMID: 34973450 DOI: 10.1016/j.nbd.2021.105605] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/28/2021] [Accepted: 12/28/2021] [Indexed: 12/29/2022] Open
Abstract
Parkinson's disease (PD) is characterized by impaired mitochondrial function and decreased ATP levels. Aerobic glycolysis and lactate production have been shown to be upregulated in dopaminergic neurons to sustain ATP levels, but the effect of upregulated glycolysis on dopaminergic neurons remains unknown. Since lactate promotes apoptosis and α-synuclein accumulation in neurons, we hypothesized that the lactate produced upon upregulated glycolysis is involved in the apoptosis of dopaminergic neurons in PD. In this study, we examined the expression of hexokinase 2 (HK2) and lactate dehydrogenase (LDH), the key enzymes in glycolysis, and lactate levels in the substantia nigra pars compacta (SNpc) of a MPTP-induced mouse model of PD and in MPP+-treated SH-SY5Y cells. We found that the expression of HK2 and LDHA and the lactate levels were markedly increased in the SNpc of MPTP-treated mice and in MPP+-treated SH-SY5Y cells. Exogenous lactate treatment led to the apoptosis of SH-SY5Y cells. Intriguingly, lactate production and the apoptosis of dopaminergic neurons were suppressed by the application of 3-bromopyruvic acid (3-Brpa), a HK2 inhibitor, or siRNA both in vivo and in vitro. 3-Brpa treatment markedly improved the motor behaviour of MPTP-treated mice in pole test and rotarod test. Mechanistically, lactate increases the activity of adenosine monophosphate-activated protein kinase (AMPK) and suppresses the phosphorylation of serine/threonine kinase 1 (Akt) and mammalian target of rapamycin (mTOR). Together, our data suggest that upregulated HK2 and LDHA and increased lactate levels prompt the apoptosis of dopaminergic neurons in PD. Inhibition of HK2 expression attenuated the apoptosis of dopaminergic neurons by downregulating lactate production and AMPK/Akt/mTOR pathway in PD.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longmin Chen
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qixiong Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danlei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwei Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongling Gao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Yuan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Zou
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijuan Mao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongjie Xiong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Min
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manli Yan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Xue
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
102
|
Hussain A, Rafeeq H, Munir N, Jabeen Z, Afsheen N, Rehman KU, Bilal M, Iqbal HMN. Dendritic Cell-Targeted Therapies to Treat Neurological Disorders. Mol Neurobiol 2022; 59:603-619. [PMID: 34743292 DOI: 10.1007/s12035-021-02622-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023]
Abstract
Dendritic cells (DCs) are the immune system's highly specialized antigen-presenting cells. When DCs are sluggish and mature, self-antigen presentation results in tolerance; however, when pathogen-associated molecular patterns stimulate mature DCs, antigen presentation results in the development of antigen-specific immunity. DCs have been identified in various vital organs of mammals (e.g., the skin, heart, lungs, intestines, and spleen), but the brain has long been thought to be devoid of DCs in the absence of neuroinflammation. However, neuroinflammation is becoming more recognized as a factor in a variety of brain illnesses. DCs are present in the brain parenchyma in trace amounts under healthy circumstances, but their numbers rise during neuroinflammation. New therapeutics are being developed that can reduce dendritic cell immunogenicity by inhibiting pro-inflammatory cytokine production and T cell co-stimulatory pathways. Additionally, innovative ways of regulating dendritic cell growth and differentiation and harnessing their tolerogenic capability are being explored. Herein, we described the function of dendritic cells in neurological disorders and discussed the potential for future therapeutic techniques that target dendritic cells and dendritic cell-related targets in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Asim Hussain
- Department of Biochemistry, Riphah International University, Faisalabad, 38040, Pakistan
| | - Hamza Rafeeq
- Department of Biochemistry, Riphah International University, Faisalabad, 38040, Pakistan
| | - Nimra Munir
- Department of Biochemistry, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Zara Jabeen
- Department of Biochemistry, Riphah International University, Faisalabad, 38040, Pakistan
| | - Nadia Afsheen
- Department of Biochemistry, Riphah International University, Faisalabad, 38040, Pakistan
| | - Khalil Ur Rehman
- Department of Biochemistry, Riphah International University, Faisalabad, 38040, Pakistan
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, 64849, Monterrey, Mexico.
| |
Collapse
|
103
|
Tu H, Yuan B, Hou X, Zhang X, Pei C, Ma Y, Yang Y, Fan Y, Qin Z, Liu C, Hu L. α-synuclein suppresses microglial autophagy and promotes neurodegeneration in a mouse model of Parkinson's disease. Aging Cell 2021; 20:e13522. [PMID: 34811872 PMCID: PMC8672776 DOI: 10.1111/acel.13522] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/18/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022] Open
Abstract
The cell‐to‐cell transfer of α‐synuclein (α‐Syn) greatly contributes to Parkinson's disease (PD) pathogenesis and underlies the spread of α‐Syn pathology. During this process, extracellular α‐Syn can activate microglia and neuroinflammation, which plays an important role in PD. However, the effect of extracellular α‐Syn on microglia autophagy is poorly understood. In the present study, we reported that extracellular α‐Syn inhibited the autophagy initiation, as indicated by LC3‐II reduction and p62 protein elevation in BV2 and cultured primary microglia. The in vitro findings were verified in microglia‐enriched population isolated from α‐Syn‐overexpressing mice induced by adeno‐associated virus (AAV2/9)‐encoded wildtype human α‐Syn injection into the substantia nigra (SN). Mechanistically, α‐Syn led to microglial autophagic impairment through activating toll‐like receptor 4 (Tlr4) and its downstream p38 and Akt‐mTOR signaling because Tlr4 knockout and inhibition of p38, Akt as well as mTOR prevented α‐Syn‐induced autophagy inhibition. Moreover, inhibition of Akt reversed the mTOR activation but failed to affect p38 phosphorylation triggered by α‐Syn. Functionally, the in vivo evidence showed that lysozyme 2 Cre (Lyz2cre)‐mediated depletion of autophagy‐related gene 5 (Atg5) in microglia aggravated the neuroinflammation and dopaminergic neuron losses in the SN and exacerbated the locomotor deficit in α‐Syn‐overexpressing mice. Taken together, the results suggest that extracellular α‐Syn, via Tlr4‐dependent p38 and Akt‐mTOR signaling cascades, disrupts microglial autophagy activity which synergistically contributes to neuroinflammation and PD development.
Collapse
Affiliation(s)
- Hai‐Yue Tu
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou Jiangsu China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Bao‐Shi Yuan
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Xiao‐Ou Hou
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou Jiangsu China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Xiao‐Jun Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Chong‐Shuang Pei
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Ya‐Ting Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Ya‐Ping Yang
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou Jiangsu China
| | - Yi Fan
- Department of Pharmacology Nanjing Medical University Nanjing Jiangsu China
| | - Zheng‐Hong Qin
- Department of Pharmacology College of Pharmaceutical Sciences Soochow University Suzhou Jiangsu China
| | - Chun‐Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou Jiangsu China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| | - Li‐Fang Hu
- Department of Neurology and Clinical Research Center of Neurological Disease The Second Affiliated Hospital of Soochow University Suzhou Jiangsu China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience Soochow University Suzhou Jiangsu China
| |
Collapse
|
104
|
Su R, Zhou T. Alpha-Synuclein Induced Immune Cells Activation and Associated Therapy in Parkinson's Disease. Front Aging Neurosci 2021; 13:769506. [PMID: 34803660 PMCID: PMC8602361 DOI: 10.3389/fnagi.2021.769506] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder closely related to immunity. An important aspect of the pathogenesis of PD is the interaction between α-synuclein and a series of immune cells. Studies have shown that accumulation of α-synuclein can induce an autoimmune response that accelerates the progression of PD. This study discusses the mechanisms underlying the interaction between α-synuclein and the immune system. During the development of PD, abnormally accumulated α-synuclein becomes an autoimmune antigen that binds to Toll-like receptors (TLRs) that activate microglia, which differentiate into the microglia type 1 (M1) subtype. The microglia activate intracellular inflammatory pathways, induce the release of proinflammatory cytokines, and promote the differentiation of cluster of differentiation 4 + (CD4 +) T cells into proinflammatory T helper type 1 (Th1) and T helper type 17 (Th17) subtypes. Given the important role of α-synuclein in the immune system of the patients with PD, identifying potential targets of immunotherapy related to α-synuclein is critical for slowing disease progression. An enhanced understanding of immune-associated mechanisms in PD can guide the development of associated therapeutic strategies in the future.
Collapse
Affiliation(s)
- Ruichen Su
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, China
| | - Tian Zhou
- School of Basic Medical Science, Nanchang University, Nanchang, China
| |
Collapse
|
105
|
Ferroptosis as a Major Factor and Therapeutic Target for Neuroinflammation in Parkinson's Disease. Biomedicines 2021; 9:biomedicines9111679. [PMID: 34829907 PMCID: PMC8615560 DOI: 10.3390/biomedicines9111679] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/16/2022] Open
Abstract
Mounting evidence suggests that ferroptosis is not just a consequence but also a fundamental contributor to the development and progression of Parkinson’s disease (PD). Ferroptosis is characterized as iron-dependent regulated cell death caused by excessive lipid peroxidation, leading to plasma membrane rupture, release of damage-associated molecular patterns, and neuroinflammation. Due to the crucial role of intracellular iron in mediating the production of reactive oxygen species and the formation of lipid peroxides, ferroptosis is intimately controlled by regulators involved in many aspects of iron metabolism, including iron uptake, storage and export, and by pathways constituting the antioxidant systems. Translational and transcriptional regulation of iron homeostasis and redox status provide an integrated network to determine the sensitivity of ferroptosis. We herein review recent advances related to ferroptosis, ranging from fundamental mechanistic discoveries and cutting-edge preclinical animal studies, to clinical trials in PD and the regulation of neuroinflammation via ferroptosis pathways. Elucidating the roles of ferroptosis in the survival of dopaminergic neurons and microglial activity can enhance our understanding of the pathogenesis of PD and provide opportunities for the development of novel prevention and treatment strategies.
Collapse
|
106
|
Heterozygous GBA D409V and ATP13a2 mutations do not exacerbate pathological α-synuclein spread in the prodromal preformed fibrils model in young mice. Neurobiol Dis 2021; 159:105513. [PMID: 34536552 DOI: 10.1016/j.nbd.2021.105513] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Autophagic dysregulation and lysosomal impairment have been implicated in the pathogenesis of Parkinson's disease, partly due to the identification of mutations in multiple genes involved in these pathways such as GBA, SNCA, ATP13a2 (also known as PARK9), TMEM175 and LRRK2. Mutations resulting in lysosomal dysfunction are proposed to contribute to Parkinson's disease by increasing α-synuclein levels, that in turn may promote aggregation of this protein. Here, we used two different genetic models-one heterozygous for a mutated form of the GBA protein (D409V), and the other heterozygous for an ATP13a2 loss-of-function mutation, to test whether these mutations exacerbate the spread of α-synuclein pathology following injection of α-synuclein preformed fibrils in the olfactory bulb of 12-week-old mice. Contrary to our hypothesis, we found that mice harboring GBA D409V+/- and ATP13a2+/- mutations did not have exacerbated behavioral impairments or histopathology (α-synuclein, LAMP2, and Iba1) when compared to their wildtype littermates. This indicates that in the young mouse brain, neither the GBA D409V mutation or ATP13a2 loss-of-function mutation accelerate the spread of α-synuclein pathology. As a consequence, we postulate that these mutations increase Parkinson's disease risk only by acting in one of the initial, upstream events in the Parkinson's disease pathogenic process. Further, the mutations, and the molecular pathways they impact, appear to play a less important role once the pathogenic process has been triggered and therefore do not specifically influence α-synuclein pathology spread.
Collapse
|
107
|
Dauer Née Joppe K, Tatenhorst L, Caldi Gomes L, Zhang S, Parvaz M, Carboni E, Roser AE, El DeBakey H, Bähr M, Vogel-Mikuš K, Wang Ip C, Becker S, Zweckstetter M, Lingor P. Brain iron enrichment attenuates α-synuclein spreading after injection of preformed fibrils. J Neurochem 2021; 159:554-573. [PMID: 34176164 DOI: 10.1111/jnc.15461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022]
Abstract
Regional iron accumulation and α-synuclein (α-syn) spreading pathology within the central nervous system are common pathological findings in Parkinson's disease (PD). Whereas iron is known to bind to α-syn, facilitating its aggregation and regulating α-syn expression, it remains unclear if and how iron also modulates α-syn spreading. To elucidate the influence of iron on the propagation of α-syn pathology, we investigated α-syn spreading after stereotactic injection of α-syn preformed fibrils (PFFs) into the striatum of mouse brains after neonatal brain iron enrichment. C57Bl/6J mouse pups received oral gavage with 60, 120, or 240 mg/kg carbonyl iron or vehicle between postnatal days 10 and 17. At 12 weeks of age, intrastriatal injections of 5-µg PFFs were performed to induce seeding of α-syn aggregates. At 90 days post-injection, PFFs-injected mice displayed long-term memory deficits, without affection of motor behavior. Interestingly, quantification of α-syn phosphorylated at S129 showed reduced α-syn pathology and attenuated spreading to connectome-specific brain regions after brain iron enrichment. Furthermore, PFFs injection caused intrastriatal microglia accumulation, which was alleviated by iron in a dose-dependent way. In primary cortical neurons in a microfluidic chamber model in vitro, iron application did not alter trans-synaptic α-syn propagation, possibly indicating an involvement of non-neuronal cells in this process. Our study suggests that α-syn PFFs may induce cognitive deficits in mice independent of iron. However, a redistribution of α-syn aggregate pathology and reduction of striatal microglia accumulation in the mouse brain may be mediated via iron-induced alterations of the brain connectome.
Collapse
Affiliation(s)
- Karina Dauer Née Joppe
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Lucas Caldi Gomes
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Shuyu Zhang
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Mojan Parvaz
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Eleonora Carboni
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Anna-Elisa Roser
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Hazem El DeBakey
- Department of Neurology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Goettingen, Goettingen, Germany
| | - Katarina Vogel-Mikuš
- Biotechnical faculty, University of Ljubljana, Ljubljana, Slovenia
- Jozef Stefan Institute, Ljubljana, Slovenia
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Stefan Becker
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Markus Zweckstetter
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Research group Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Paul Lingor
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
108
|
Zhou T, Zhang M, Xie Y, Chen Y, Peng S, Jing X, Lei M, Tao E, Liang Y. Effects of miRNAs in exosomes derived from α-synuclein overexpressing SH-SY5Y cells on autophagy and inflammation of microglia. Cell Signal 2021; 89:110179. [PMID: 34715309 DOI: 10.1016/j.cellsig.2021.110179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/19/2021] [Accepted: 10/23/2021] [Indexed: 12/27/2022]
Abstract
Our previous study has revealed that GFP-α-synuclein overexpressing SH-SY5Y cells-derived exosomes (GFP-SNCA Exo) decrease autophagy in microglia via their load of miRNAs. However, it is unclear whether GFP-SNCA Exo can affect microglial inflammation via modulation of autophagy. In order to investigate the effects of miRNAs carried by GFP-SNCA Exo on autophagy and inflammation of microglia. SH-SY5Y cells were transfected with lentivirus expressing α-synuclein and then their exosomes were collected. Western blot and laser confocal images showed that α-synuclein transferred between SH-SY5Y cells and microglia through exosomes. Differentially expressed miRNAs between GFP-SNCA Exo and the vector exosomes were detected by microarray analysis. After bioinformatics analysis of the differentially expressed miRNAs, we found that their target genes were enriched in the MAPK and autophagy-associated signaling pathway. The expression of P62, p-JNK/JNK, and p-ERK/ERK and the release of IL-6 significantly increased whereas LC3 II/I decreased in microglia exposed to GFP-SNCA Exo for 48 h when compared to the control group. But rapamycin could reverse the increasing expression of p-JNK/JNK, p-ERK/ERK and the release of IL-6 induced by GFP-SNCA Exo. Dual immunofluorescence staining for LC3B and LAMP1 showed that the fluorescence density of LC3B decreased and the fluorescence of LC3B and LAMP1 were not co-located in microglia after 48 h co-culture with GFP-SNCA Exo compared with the control group, which indicated that these exosomes decreased autophagy and impaired the autophagy flux in recipient microglia. Taken together, our results indicate that GFP-SNCA Exo activate the MAPK signaling pathway and inflammation by decreasing autophagy in microglia.
Collapse
Affiliation(s)
- Tianen Zhou
- Department of Emergency, The First People's Hospital of Foshan, Foshan 528000, China
| | - Meng Zhang
- Department of General Practice, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yingyu Xie
- Department of Neurology, Shantou Central Hospital, Shantou 515000, China
| | - Ying Chen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Sudan Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiuna Jing
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Enxiang Tao
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| | - Yanran Liang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
109
|
Scheiblich H, Dansokho C, Mercan D, Schmidt SV, Bousset L, Wischhof L, Eikens F, Odainic A, Spitzer J, Griep A, Schwartz S, Bano D, Latz E, Melki R, Heneka MT. Microglia jointly degrade fibrillar alpha-synuclein cargo by distribution through tunneling nanotubes. Cell 2021; 184:5089-5106.e21. [PMID: 34555357 PMCID: PMC8527836 DOI: 10.1016/j.cell.2021.09.007] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 06/05/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022]
Abstract
Microglia are the CNS resident immune cells that react to misfolded proteins through pattern recognition receptor ligation and activation of inflammatory pathways. Here, we studied how microglia handle and cope with α-synuclein (α-syn) fibrils and their clearance. We found that microglia exposed to α-syn establish a cellular network through the formation of F-actin-dependent intercellular connections, which transfer α-syn from overloaded microglia to neighboring naive microglia where the α-syn cargo got rapidly and effectively degraded. Lowering the α-syn burden attenuated the inflammatory profile of microglia and improved their survival. This degradation strategy was compromised in cells carrying the LRRK2 G2019S mutation. We confirmed the intercellular transfer of α-syn assemblies in microglia using organotypic slice cultures, 2-photon microscopy, and neuropathology of patients. Together, these data identify a mechanism by which microglia create an "on-demand" functional network in order to improve pathogenic α-syn clearance.
Collapse
Affiliation(s)
- Hannah Scheiblich
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Cira Dansokho
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Dilek Mercan
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Luc Bousset
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, 92265 Fontenay-aux-Roses, France
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Frederik Eikens
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Alexandru Odainic
- Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Jasper Spitzer
- Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Angelika Griep
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Stephanie Schwartz
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Institute of Innate Immunity, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Ronald Melki
- Institut François Jacob, MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, 92265 Fontenay-aux-Roses, France
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, 01605 Worcester, MA, USA.
| |
Collapse
|
110
|
Venezia S, Kaufmann WA, Wenning GK, Stefanova N. Toll-like receptor 4 deficiency facilitates α-synuclein propagation and neurodegeneration in a mouse model of prodromal Parkinson's disease. Parkinsonism Relat Disord 2021; 91:59-65. [PMID: 34530328 DOI: 10.1016/j.parkreldis.2021.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 01/24/2023]
Abstract
The evidence linking innate immunity mechanisms and neurodegenerative diseases is growing, but the specific mechanisms are incompletely understood. Experimental data suggest that microglial TLR4 mediates the uptake and clearance of α-synuclein also termed synucleinophagy. The accumulation of misfolded α-synuclein throughout the brain is central to Parkinson's disease (PD). The distribution and progression of the pathology is often attributed to the propagation of α-synuclein. Here, we apply a classical α-synuclein propagation model of prodromal PD in wild type and TLR4 deficient mice to study the role of TLR4 in the progression of the disease. Our data suggest that TLR4 deficiency facilitates the α-synuclein seed spreading associated with reduced lysosomal activity of microglia. Three months after seed inoculation, more pronounced proteinase K-resistant α-synuclein inclusion pathology is observed in mice with TLR4 deficiency. The facilitated propagation of α-synuclein is associated with early loss of dopamine transporter (DAT) signal in the striatum and loss of dopaminergic neurons in substantia nigra pars compacta of TLR4 deficient mice. These new results support TLR4 signaling as a putative target for disease modification to slow the progression of PD and related disorders.
Collapse
Affiliation(s)
- Serena Venezia
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | - Walter A Kaufmann
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Gregor K Wenning
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria
| | - Nadia Stefanova
- Laboratory for Translational Neurodegeneration Research, Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Austria.
| |
Collapse
|
111
|
Brendza R, Lin H, Stark K, Foreman O, Tao J, Pierce A, Ngu H, Shen K, Easton AE, Bhangale T, Chang D, Bingol B, Friedman BA. Genetic ablation of Gpnmb does not alter synuclein-related pathology. Neurobiol Dis 2021; 159:105494. [PMID: 34464706 DOI: 10.1016/j.nbd.2021.105494] [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: 03/14/2021] [Revised: 08/03/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022] Open
Abstract
The gene GPNMB is known to play roles in phagocytosis and tissue repair, and is upregulated in microglia in many mouse models of neurodegenerative disease as well as in human patients. Nearby genomic variants are associated with both elevated Parkinson's disease (PD) risk and higher expression of this gene, suggesting that inhibiting GPNMB activity might be protective in Parkinson's disease. We tested this hypothesis in three different mouse models of neurological diseases: a remyelination model and two models of alpha-synuclein pathology. We found that Gpnmb deletion had no effect on histological, cellular, behavioral, neurochemical or gene expression phenotypes in any of these models. These data suggest that Gpnmb does not play a major role in the development of pathology or functional defects in these models and that further work is necessary to study its role in the development or progression of Parkinson's disease.
Collapse
Affiliation(s)
- Robert Brendza
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Han Lin
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Kimberly Stark
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Oded Foreman
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Janet Tao
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Andrew Pierce
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Hai Ngu
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Kimberle Shen
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Amy E Easton
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | - Tushar Bhangale
- Department of Human Genetics, Genentech, Inc., South San Francisco, CA, USA
| | - Diana Chang
- Department of Human Genetics, Genentech, Inc., South San Francisco, CA, USA
| | - Baris Bingol
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA.
| | - Brad A Friedman
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
112
|
Barth M, Bacioglu M, Schwarz N, Novotny R, Brandes J, Welzer M, Mazzitelli S, Häsler LM, Schweighauser M, Wuttke TV, Kronenberg-Versteeg D, Fog K, Ambjørn M, Alik A, Melki R, Kahle PJ, Shimshek DR, Koch H, Jucker M, Tanriöver G. Microglial inclusions and neurofilament light chain release follow neuronal α-synuclein lesions in long-term brain slice cultures. Mol Neurodegener 2021; 16:54. [PMID: 34380535 PMCID: PMC8356412 DOI: 10.1186/s13024-021-00471-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/06/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Proteopathic brain lesions are a hallmark of many age-related neurodegenerative diseases including synucleinopathies and develop at least a decade before the onset of clinical symptoms. Thus, understanding of the initiation and propagation of such lesions is key for developing therapeutics to delay or halt disease progression. METHODS Alpha-synuclein (αS) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An αS seed-recognizing human antibody was tested for blocking seeding and/or spreading of the αS lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. RESULTS To study initial stages of α-synucleinopathies, we induced αS inclusions in murine hippocampal slice cultures by seeded aggregation. Induction of αS inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesions at 2-3 weeks. The amount of αS inclusions was dependent on the type of αS seed and on the culture's genetic background (wildtype vs A53T-αS genotype). Formation of αS inclusions could be monitored by neurofilament light chain protein release into the culture medium, a fluid biomarker of neurodegeneration commonly used in clinical settings. Local microinjection of αS seeds resulted in spreading of αS inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an αS seed-recognizing human antibody. We then applied parameters of the murine cultures to surgical resection-derived adult human long-term neocortical slice cultures from 22 to 61-year-old donors. Similarly, in these human slice cultures, proof-of-principle induction of αS lesions was achieved at 1week post-seeding in combination with viral A53T-αS expressions. CONCLUSION The successful translation of these brain cultures from mouse to human with the first reported induction of human αS lesions in a true adult human brain environment underlines the potential of this model to study proteopathic lesions in intact mouse and now even aged human brain environments.
Collapse
Affiliation(s)
- Melanie Barth
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Graduate Training Center of Neuroscience, University of Tuebingen, 72076 Tuebingen, Germany
| | - Mehtap Bacioglu
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Graduate Training Center of Neuroscience, University of Tuebingen, 72076 Tuebingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Renata Novotny
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Janine Brandes
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Marc Welzer
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Graduate Training Center of Neuroscience, University of Tuebingen, 72076 Tuebingen, Germany
| | - Sonia Mazzitelli
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Lisa M. Häsler
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Manuel Schweighauser
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Thomas V. Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Neurosurgery, University of Tuebingen, 72076 Tuebingen, Germany
| | - Deborah Kronenberg-Versteeg
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Karina Fog
- Division of Neuroscience, H. Lundbeck A/S, 2500 Valby, Denmark
| | - Malene Ambjørn
- Division of Neuroscience, H. Lundbeck A/S, 2500 Valby, Denmark
| | - Ania Alik
- MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, 92265 Fontenay-aux-Roses, France
| | - Ronald Melki
- MIRCen, CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, 92265 Fontenay-aux-Roses, France
| | - Philipp J. Kahle
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Derya R. Shimshek
- Neuroscience Research, Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Henner Koch
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Epileptology, Neurology, RWTH Aachen University, Aachen, Germany
| | - Mathias Jucker
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Gaye Tanriöver
- DZNE, German Center for Neurodegenerative Diseases, 72076 Tuebingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
113
|
Ma X, Wang Q, Yuan W, Wang Y, Zhou F, Kang K, Tong X, Liu Z. Electroacupuncture Alleviates Neuroinflammation and Motor Dysfunction by Regulating Intestinal Barrier Function in a Mouse Model of Parkinson Disease. J Neuropathol Exp Neurol 2021; 80:844-855. [PMID: 34343334 DOI: 10.1093/jnen/nlab046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Gastrointestinal dysfunction is the main nonmotor characteristic of Parkinson disease (PD), manipulation of gastrointestinal function by altering gut-brain axis is a potentially novel entry point for the treatment of PD. Acupuncture has been reported to confer beneficial effects in the gastrointestinal diseases. Therefore, this study aimed to explore the effects and mechanism of acupuncture on the pathophysiology and gastrointestinal function of PD. A PD mouse model was established by rotenone, and electroacupuncture was used to regulate the gastrointestinal function. Rotenone was found to induce the types of brain pathologies and gastrointestinal dysfunction that are similar to those observed with PD. Electroacupuncture significantly increased the spontaneous activity of mice with PD and increased the expression of tyrosine hydroxylase, while reducing the expression of Iba-1 in substantia nigra (SN), suggesting that motor dysfunction and neurological damage was alleviated. In addition, electroacupuncture significantly reduced the deposition of α-synuclein in both colon and SN, reduced intestinal inflammation, and exerted protective effects on enteric nervous system and intestinal barrier. In conclusion, electroacupuncture confers beneficial effects on the gastrointestinal system of mice with PD and can alleviate neuroinflammation and neuropathic injury by inhibiting intestinal inflammation, promoting intestinal barrier repair and reducing α-synuclein deposition in the colon.
Collapse
Affiliation(s)
- Xue Ma
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Qiang Wang
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Wei Yuan
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Yuan Wang
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Feng Zhou
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Kaiwen Kang
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Xiaopeng Tong
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Zhibin Liu
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| |
Collapse
|
114
|
Gou J, Liang S, Cheng W, Wu S, Ye Z, Ma Y, Yin Y, Wang H. Neuroprotective effect of combined use of nicotine and celecoxib by inhibiting neuroinflammation in ischemic rats. Brain Res Bull 2021; 175:234-243. [PMID: 34333049 DOI: 10.1016/j.brainresbull.2021.07.022] [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: 10/18/2020] [Revised: 06/15/2021] [Accepted: 07/27/2021] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The contribution of neuroinflammation in cognitive impairment is increasingly recognized. Non-steroidal anti-inflammatory drugs had been proven that it could improve cognitive impairment in large dose but with more side effect, which limited the application. The main objective of this study was to investigate whether the combined use of nicotine and celecoxib could obtain synergistic neuroprotective effect in ischemic rats. METHODS Twenty adult Sprague-Dawley (SD) rats underwent ischemic model surgery by injecting endothelin-1 into the left thalamus, which were classified into four groups with different interventions: nicotine (1.5 mg/kg/d), celecoxib (15 mg/kg/d), nicotine (1.5 mg/kg/d) +celecoxib (15 mg/kg/d), or saline after surgery. The other five SD rats also underwent same surgery by injecting saline instead of endothelin-1, as the control group. Morris water maze (MWM) test was adopted to assess the cognition. Micro PET/CT with 2-[18F]-A-85380 were performed for α4β2-nAChRs detection in vivo. Western blot, real-time PCR and immunohistochemical staining were adopted to detect the expression of α4β2-nAChRs and inflammatory factors which included TNF-α, IL-1β, IL-6 in brain tissue. Microglial activation in the brain was monitored by immunofluorescence with IBA1 staining. RESULTS The MWM test showed rats given with nicotine or celecoxib alone showed much better memory than rats with saline, no difference was observed between nicotine and celecoxib. The rat memory was recovered most significant when the nicotine and celecoxib were combined (p < 0.05). Micro-PET/CT showed much more tracer uptake in the left thalamus and whole brain in rats given with nicotine, or nicotine + celecoxib (nico + cele group) than saline treated rats, whereas the rats given celecoxib did not. Compared with saline treated rats, we found the proteins of α4nAChR and β2nAChR in rats given nicotine or nico + cele increased significantly, and mRNA/proteins of TNF-α, IL-1β and IL-6 decreased at the same time. The α 4nAChR and β 2nAChR proteins in rats given celecoxib is the same as saline treated rats, whereas the inflammatory factors decreased obviously compared with saline treated rats. Microglial activation was confirmed in saline treated rats, which was inhibited in rats give nicotine, celecoxib or both. CONCLUSIONS The study revealed the combined use of nicotine and celecoxib may improve the cognitive function in ischemic rats, with a better effect than either alone. Both nicotine and celecoxib can inhibit inflammation, but through different mechanisms: nicotine can activate α4β2-nAChRs while celecoxib is cyclooxygenase-2 inhibitor. Our findings suggest the combined application of two drugs with different anti-inflammation mechanism could attenuate cognitive impairment more effectively in ischemic rats, which may hold therapeutic potential in the clinical practice.
Collapse
Affiliation(s)
- Jinyu Gou
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Sheng Liang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Cheng
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuqi Wu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyi Ye
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Ma
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
115
|
Gentzel RC, Toolan D, Jinn S, Schachter JB, Ma L, Kahle PJ, Smith SM, Marcus JN. Intracranial administration of alpha-synuclein fibrils in A30P-synuclein transgenic mice causes robust synucleinopathy and microglial induction. Neurobiol Aging 2021; 106:12-25. [PMID: 34225000 DOI: 10.1016/j.neurobiolaging.2021.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/12/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022]
Abstract
Synucleinopathies are neurodegenerative disorders involving pathological alpha-synuclein (αSyn) protein, including dementia with Lewy bodies, multiple system atrophy and Parkinson's disease (PD). Current in vivo models of synucleinopathy include transgenic mice overexpressing αSyn variants and methods based on administration of aggregated, exogenous αSyn. Combining these techniques offers the ability to study consequences of introducing pathological αSyn into primed neuronal environments likely to develop synucleinopathy. Herein, we characterize the impacts pre-formed fibrils (PFFs) of recombinant, human αSyn have in mice overexpressing human A30P αSyn, a mutation associated with autosomal dominant PD. A30P mouse brain contains detergent insoluble αSyn biochemically similar to PD brain, and these mice develop Lewy-like synucleinopathy with age. Administration of PFFs in A30P mice resulted in regionally-specific accumulations of phosphorylated synuclein, microglial induction and a motor phenotype that differed from PFF-induced effects in wildtype mice. Surprisingly, PFF-induced losses of tyrosine hydroxylase were similar in A30P and wildtype mice. Thus, the PFF-A30P model recapitulates key aspects of synucleinopathy with induction of microglia, creating an appropriate system for evaluating neurodegenerative therapeutics.
Collapse
Affiliation(s)
- Renee C Gentzel
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Dawn Toolan
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Sarah Jinn
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Joel B Schachter
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA; Currently at Takeda Pharmaceutics, Inc., San Diego, CA, USA
| | - Lei Ma
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Philipp J Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research and, German Center for Neurodegenerative Diseases, University of Tübingen, Germany
| | - Sean M Smith
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jacob N Marcus
- Neuroscience Discovery, Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
116
|
Jo S, Kim YJ, Park KW, Hwang YS, Lee SH, Kim BJ, Chung SJ. Association of NO2 and Other Air Pollution Exposures With the Risk of Parkinson Disease. JAMA Neurol 2021; 78:800-808. [PMID: 33999109 DOI: 10.1001/jamaneurol.2021.1335] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance The development of Parkinson disease (PD) may be promoted by exposure to air pollution. Objective To investigate the potential association between exposure to particulate matters (PM2.5 and PM10), nitrogen dioxide (NO2), ozone (O3), sulfur dioxide (SO2), and carbon monoxide (CO) and the risk of incident PD. Design, Setting, and Participants This retrospective cohort study used data from the Korean National Health Insurance Service. Among the 1 021 208 Korean individuals in the database, those who had lived in Seoul from January 2002 to December 2006 (n = 176 875) were screened for eligibility. A total of 78 830 adults older than 40 years without PD and who lived in Seoul between January 2002 and December 2006 were included in this study. Individuals diagnosed with PD before 2006 (n = 159) and individuals 40 years or younger (n = 97 886) were excluded. Each participant was followed up with annually from January 2007 to December 2015, thereby adding up to 757 704 total person-years of follow-up. Data were analyzed from January to September 2020. Exposures Individual exposure levels to PM2.5, PM10, NO2, O3, SO2, and CO were estimated based on the participants' residential address at the district level. To evaluate long-term exposure to air pollution, time-varying 5-year mean air pollutant exposure was calculated for each participant. Main Outcomes and Measures The outcome measure was the association between air pollution and the risk of incident PD measured as hazard ratios after adjusting for demographic factors, socioeconomic factors, and medical comorbidities. Results At baseline, the mean (SD) age of the 78 830 participants was 54.4 (10.7) years, and 41 070 (52.1%) were female. A total of 338 individuals with newly diagnosed PD were identified during the study period. Exposure to NO2 was associated with an increase in risk of PD (hazard ratio for highest vs lowest quartile, 1.41; 95% CI, 1.02-1.95; P for trend = .045). No statistically significant associations between exposure to PM2.5, PM10, O3, SO2, or CO and PD incidence were found. Conclusions and Relevance In this large cohort study, a statistically significant association between NO2 exposure and PD risk was identified. This finding suggests the role of air pollutants in PD development, advocating for the need to implement a targeted public health policy.
Collapse
Affiliation(s)
- Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ye-Jee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kye Won Park
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yun Su Hwang
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Hyun Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bum Joon Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
117
|
Bergkvist L, Johnson ME, Mercado G, Steiner JA, Meyerdirk L, Schulz E, Madaj Z, Ma J, Becker K, Li Y, Brundin P. An extended release GLP-1 analogue increases α-synuclein accumulation in a mouse model of prodromal Parkinson's disease. Exp Neurol 2021; 341:113693. [PMID: 33727096 PMCID: PMC10461454 DOI: 10.1016/j.expneurol.2021.113693] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/17/2021] [Accepted: 03/10/2021] [Indexed: 11/26/2022]
Abstract
The repurposing of drugs developed to treat type 2 diabetes for the treatment of Parkinson's disease (PD) was encouraged by the beneficial effect exerted by the glucagon-like peptide 1 (GLP-1) analogue exenatide in a phase 2 clinical trial. The effects of GLP-1 analogues have been investigated extensively using rodent toxin models of PD. However, many of the toxin-based models used lack robust α-synuclein (α-syn) pathology, akin to the Lewy bodies and neurites seen in PD. One prior study has reported a protective effect of a GLP-1 analogue on midbrain dopamine neurons following injection of α-syn preformed fibrils (PFF) into the striatum. Here, we used olfactory bulb injections of PFF as a model of prodromal PD and monitored the effect of a long-acting GLP-1 analogue on the propagation of α-syn pathology in the olfactory system. Thirteen weeks after PFF injection, mice treated with long-acting the GLP-1 analogue had a significant increase in pathological α-syn in brain regions connected to the olfactory bulb, accompanied by signs of microglia activation. Our results suggest that the nature of the neuronal insult and intrinsic properties of the targeted neuronal population markedly influence the effect of GLP-1 analogues.
Collapse
Affiliation(s)
- Liza Bergkvist
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Michaela E Johnson
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Gabriela Mercado
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jennifer A Steiner
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Lindsay Meyerdirk
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Emily Schulz
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Zachary Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Jiyan Ma
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Katelyn Becker
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Patrik Brundin
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
118
|
Alpha-Synuclein as a Prominent Actor in the Inflammatory Synaptopathy of Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22126517. [PMID: 34204581 PMCID: PMC8234932 DOI: 10.3390/ijms22126517] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/04/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is considered the most common disorder of synucleinopathy, which is characterised by intracellular inclusions of aggregated and misfolded α-synuclein (α-syn) protein in various brain regions, and the loss of dopaminergic neurons. During the early prodromal phase of PD, synaptic alterations happen before cell death, which is linked to the synaptic accumulation of toxic α-syn specifically in the presynaptic terminals, affecting neurotransmitter release. The oligomers and protofibrils of α-syn are the most toxic species, and their overexpression impairs the distribution and activation of synaptic proteins, such as the SNARE complex, preventing neurotransmitter exocytosis and neuronal synaptic communication. In the last few years, the role of the immune system in PD has been increasingly considered. Microglial and astrocyte activation, the gene expression of proinflammatory factors, and the infiltration of immune cells from the periphery to the central nervous system (CNS) represent the main features of the inflammatory response. One of the actors of these processes is α-syn accumulation. In light of this, here, we provide a systematic review of PD-related α-syn and inflammation inter-players.
Collapse
|
119
|
Zheng T, Zhang Z. Activated microglia facilitate the transmission of α-synuclein in Parkinson's disease. Neurochem Int 2021; 148:105094. [PMID: 34097990 DOI: 10.1016/j.neuint.2021.105094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta and abnormal aggregates of α-synuclein protein called Lewy bodies. To date, there is no drug that can definitely slow down or stop the progression of this disease. The discovery of the cell-to-cell transmission of pathologic α-synuclein seeds offers the possibility to explore novel treatment strategies to prevent the spread of α-synuclein, with the purpose of slowing down the progression of PD in its tracks. Although recent studies have made tremendous progress in understanding how α-synuclein spreads throughout the brain, neuroinflammation seems to play a crucial role in the development of α-synuclein pathology in PD. The activation of microglia, one of the hallmarks of the neuroinflammatory process, is suggested to influence the neuron-to-neuron transmission of α-synuclein. This review summarizes how activated microglia facilitate this process, and focuses on the following mechanisms including the activation of microglia in PD, the reduced ability of activated microglia to clear α-synuclein and increased migratory capacity of microglia in PD, as well as the cooperation between microglia and exosomes in mediating α-synuclein release and propagation. In conclusion, this article help collate information on microglia in-relation to PD.
Collapse
Affiliation(s)
- Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China
| | - Zhengxiang Zhang
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China.
| |
Collapse
|
120
|
Leites EP, Morais VA. The PINK1-Mediated Crosstalk between Neural Cells and the Underlying Link to Parkinson's Disease. Cells 2021; 10:1395. [PMID: 34198743 PMCID: PMC8228719 DOI: 10.3390/cells10061395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial dysfunction has a fundamental role in the development of idiopathic and familiar forms of Parkinson's disease (PD). The nuclear-encoded mitochondrial kinase PINK1, linked to familial PD, is responsible for diverse mechanisms of mitochondrial quality control, ATP production, mitochondrial-mediated apoptosis and neuroinflammation. The main pathological hallmark of PD is the loss of dopaminergic neurons. However, novel discoveries have brought forward the concept that a disruption in overall brain homeostasis may be the underlying cause of this neurodegeneration disease. To sustain this, astrocytes and microglia cells lacking PINK1 have revealed increased neuroinflammation and deficits in physiological roles, such as decreased wound healing capacity and ATP production, which clearly indicate involvement of these cells in the physiopathology of PD. PINK1 executes vital functions within mitochondrial regulation that have a detrimental impact on the development and progression of PD. Hence, in this review, we aim to broaden the horizon of PINK1-mediated phenotypes occurring in neurons, astrocytes and microglia and, ultimately, highlight the importance of the crosstalk between these neural cells that is crucial for brain homeostasis.
Collapse
Affiliation(s)
| | - Vanessa Alexandra Morais
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Lisboa, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| |
Collapse
|
121
|
Ray B, Mahalakshmi AM, Tuladhar S, Bhat A, Srinivasan A, Pellegrino C, Kannan A, Bolla SR, Chidambaram SB, Sakharkar MK. "Janus-Faced" α-Synuclein: Role in Parkinson's Disease. Front Cell Dev Biol 2021; 9:673395. [PMID: 34124057 PMCID: PMC8194081 DOI: 10.3389/fcell.2021.673395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/15/2021] [Indexed: 01/03/2023] Open
Abstract
Parkinson's disease (PD) is a pathological condition characterized by the aggregation and the resultant presence of intraneuronal inclusions termed Lewy bodies (LBs) and Lewy neurites which are mainly composed of fibrillar α-synuclein (α-syn) protein. Pathogenic aggregation of α-syn is identified as the major cause of LBs deposition. Several mutations in α-syn showing varied aggregation kinetics in comparison to the wild type (WT) α-syn are reported in PD (A30P, E46K, H 50Q, G51D, A53E, and A53T). Also, the cell-to-cell spread of pathological α-syn plays a significant role in PD development. Interestingly, it has also been suggested that the pathology of PD may begin in the gastrointestinal tract and spread via the vagus nerve (VN) to brain proposing the gut-brain axis of α-syn pathology in PD. Despite multiple efforts, the behavior and functions of this protein in normal and pathological states (specifically in PD) is far from understood. Furthermore, the etiological factors responsible for triggering aggregation of this protein remain elusive. This review is an attempt to collate and present latest information on α-syn in relation to its structure, biochemistry and biophysics of aggregation in PD. Current advances in therapeutic efforts toward clearing the pathogenic α-syn via autophagy/lysosomal flux are also reviewed and reported.
Collapse
Affiliation(s)
- Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Sunanda Tuladhar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Asha Srinivasan
- Division of Nanoscience & Technology, Faculty of Life Sciences, JSS Academy of Higher Education & Research, Mysuru, India
| | - Christophe Pellegrino
- Institut National de la Santé et de la Recherche Médicale, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Anbarasu Kannan
- Department of Protein Chemistry and Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Srinivasa Rao Bolla
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Nur-Sultan City, Kazakhstan
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- Special Interest Group – Brain, Behaviour, and Cognitive Neurosciences Research, JSS Academy of Higher Education & Research, Mysuru, India
| | | |
Collapse
|
122
|
Zilberman A, Cornelison RC. Microphysiological models of the central nervous system with fluid flow. Brain Res Bull 2021; 174:72-83. [PMID: 34029679 DOI: 10.1016/j.brainresbull.2021.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022]
Abstract
There are over 1,000 described neurological and neurodegenerative disorders affecting nearly 100 million Americans - roughly one third of the U.S. population. Collectively, treatment of neurological conditions is estimated to cost $800 billion every year. Lowering this societal burden will require developing better model systems in which to study these diverse disorders. Microphysiological systems are promising tools for modeling healthy and diseased neural tissues to study mechanisms and treatment of neuropathology. One major benefit of microphysiological systems is the ability to incorporate biophysical forces, namely the forces derived from biological fluid flow. Fluid flow in the central nervous system (CNS) is a complex but important element of physiology, and pathologies as diverse as traumatic or ischemic injury, cancer, neurodegenerative disease, and natural aging have all been found to alter flow pathways. In this review, we summarize recent advances in three-dimensional microphysiological systems for studying the biology and therapy of CNS disorders and highlight the ability and growing need to incorporate biological fluid flow in these miniaturized model systems.
Collapse
Affiliation(s)
- Aleeza Zilberman
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, United States
| | - R Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, United States.
| |
Collapse
|
123
|
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
|
124
|
Wang Q, Luo Y, Chaudhuri KR, Reynolds R, Tan EK, Pettersson S. The role of gut dysbiosis in Parkinson's disease: mechanistic insights andtherapeutic options. Brain 2021; 144:2571-2593. [PMID: 33856024 DOI: 10.1093/brain/awab156] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/23/2021] [Accepted: 03/23/2021] [Indexed: 12/02/2022] Open
Abstract
Parkinson's disease is a common neurodegenerative disease in which gastrointestinal symptoms may appear prior to motor symptoms. The gut microbiota of patients with Parkinson's disease shows unique changes, which may be used as early biomarkers of disease. Alteration in gut microbiota composition may be related to the cause or effect of motor or non-motor symptoms, but the specific pathogenic mechanisms are unclear. The gut microbiota and its metabolites have been suggested to be involved in the pathogenesis of Parkinson's disease by regulating neuroinflammation, barrier function and neurotransmitter activity. There is bidirectional communication between the enteric nervous system and the central nervous system, and the microbiota-gut-brain axis may provide a pathway for the transmission of α-synuclein. We highlight recent discoveries and alterations of the gut microbiota in Parkinson's disease, and highlight current mechanistic insights on the microbiota-gut-brain axis in disease pathophysiology. We discuss the interactions between production and transmission of α-synuclein and gut inflammation and neuroinflammation. In addition, we also draw attention to diet modification, use of probiotics and prebiotics and fecal microbiota transplantation as potential therapeutic approaches that may lead to a new treatment paradigm for Parkinson's disease.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - K Ray Chaudhuri
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS, UK
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Sven Pettersson
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore.,LKC School of Medicine, NTU, Singapore.,Sunway University, Department of Medical Sciences, Kuala Lumpur, Malaysia
| |
Collapse
|
125
|
George S, Tyson T, Rey NL, Sheridan R, Peelaerts W, Becker K, Schulz E, Meyerdirk L, Burmeister AR, von Linstow CU, Steiner JA, Galvis MLE, Ma J, Pospisilik JA, Labrie V, Brundin L, Brundin P. T Cells Limit Accumulation of Aggregate Pathology Following Intrastriatal Injection of α-Synuclein Fibrils. JOURNAL OF PARKINSONS DISEASE 2021; 11:585-603. [PMID: 33579871 PMCID: PMC8150548 DOI: 10.3233/jpd-202351] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND α-Synuclein (α-syn) is the predominant protein in Lewy-body inclusions, which are pathological hallmarks of α-synucleinopathies, such as Parkinson's disease (PD) and multiple system atrophy (MSA). Other hallmarks include activation of microglia, elevation of pro-inflammatory cytokines, as well as the activation of T and B cells. These immune changes point towards a dysregulation of both the innate and the adaptive immune system. T cells have been shown to recognize epitopes derived from α-syn and altered populations of T cells have been found in PD and MSA patients, providing evidence that these cells can be key to the pathogenesis of the disease.ObjectiveTo study the role of the adaptive immune system with respect to α-syn pathology. METHODS We injected human α-syn preformed fibrils (PFFs) into the striatum of immunocompromised mice (NSG) and assessed accumulation of phosphorylated α-syn pathology, proteinase K-resistant α-syn pathology and microgliosis in the striatum, substantia nigra and frontal cortex. We also assessed the impact of adoptive transfer of naïve T and B cells into PFF-injected immunocompromised mice. RESULTS Compared to wildtype mice, NSG mice had an 8-fold increase in phosphorylated α-syn pathology in the substantia nigra. Reconstituting the T cell population decreased the accumulation of phosphorylated α-syn pathology and resulted in persistent microgliosis in the striatum when compared to non-transplanted mice. CONCLUSION Our work provides evidence that T cells play a role in the pathogenesis of experimental α-synucleinopathy.
Collapse
Affiliation(s)
- Sonia George
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Trevor Tyson
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Nolwen L Rey
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.,Laboratory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Rachael Sheridan
- Flow Cytometry Core Facility, Van Andel Institute, Grand Rapids, MI, USA
| | - Wouter Peelaerts
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Katelyn Becker
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Emily Schulz
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Lindsay Meyerdirk
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Amanda R Burmeister
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Jennifer A Steiner
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Jiyan Ma
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Viviane Labrie
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.,Michigan State University - College of Human Medicine, Department of Psychiatry, Grand Rapids, MI, USA
| | - Lena Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.,Michigan State University - College of Human Medicine, Department of Psychiatry, Grand Rapids, MI, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA.,Michigan State University - College of Human Medicine, Department of Psychiatry, Grand Rapids, MI, USA
| |
Collapse
|
126
|
Duffy SS, Hayes JP, Fiore NT, Moalem-Taylor G. The cannabinoid system and microglia in health and disease. Neuropharmacology 2021; 190:108555. [PMID: 33845074 DOI: 10.1016/j.neuropharm.2021.108555] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022]
Abstract
Recent years have yielded significant advances in our understanding of microglia, the immune cells of the central nervous system (CNS). Microglia are key players in CNS development, immune surveillance, and the maintenance of proper neuronal function throughout life. In the healthy brain, homeostatic microglia have a unique molecular signature. In neurological diseases, microglia become activated and adopt distinct transcriptomic signatures, including disease-associated microglia (DAM) implicated in neurodegenerative disorders. Homeostatic microglia synthesise the endogenous cannabinoids 2-arachidonoylglycerol and anandamide and express the cannabinoid receptors CB1 and CB2 at constitutively low levels. Upon activation, microglia significantly increase their synthesis of endocannabinoids and upregulate their expression of CB2 receptors, which promote a protective microglial phenotype by enhancing their production of neuroprotective factors and reducing their production of pro-inflammatory factors. Here, we summarise the effects of the microglial cannabinoid system in the CNS demyelinating disease multiple sclerosis, the neurodegenerative diseases Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis, chronic inflammatory and neuropathic pain, and psychiatric disorders including depression, anxiety and schizophrenia. We discuss the therapeutic potential of cannabinoids in regulating microglial activity and highlight the need to further investigate their specific microglia-dependent immunomodulatory effects.
Collapse
Affiliation(s)
- Samuel S Duffy
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Jessica P Hayes
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Nathan T Fiore
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia
| | - Gila Moalem-Taylor
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, UNSW Sydney, NSW, 2052, Australia.
| |
Collapse
|
127
|
Xia Y, Zhang G, Kou L, Yin S, Han C, Hu J, Wan F, Sun Y, Wu J, Li Y, Huang J, Xiong N, Zhang Z, Wang T. Reactive microglia enhance the transmission of exosomal alpha-synuclein via toll-like receptor 2. Brain 2021; 144:2024-2037. [PMID: 33792662 DOI: 10.1093/brain/awab122] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 02/14/2021] [Accepted: 03/08/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence suggests that microglial activation is strongly linked to the initiation and progression of Parkinson's disease (PD). Cell-to-cell propagation of α-synuclein (α-syn) pathology is a highlighted feature of PD, and the focus of such research has been primarily on neurons. However, recent studies as well as the data contained herein suggest that microglia, the primary phagocytes in the brain, play a direct role in the spread of α-syn pathology. Recent data revealed that plasma exosomes derived from PD patients (PD-EXO) carry pathological α-syn and target microglia preferentially. Hence, PD-EXO is likely a key tool for investigating the role of microglia in α-syn transmission. We showed that intrastriatal injection of PD-EXO resulted in the propagation of exosomal α-syn from microglia to neurons following microglia activation. Toll-like receptor 2 (TLR2) in microglia was activated by exosomal α-syn and acted as a crucial mediator of PD-EXO-induced microglial activation. Additionally, partial microglia depletion resulted in a significant decrease of exogenous α-syn in the substantia nigra (SN). Furthermore, exosomal α-syn internalization was initiated by binding to TLR2 of microglia. Excessive α-syn phagocytosis may induce the inflammatory responses of microglia and provide the seed for microglia-to-neuron transmission. Consistently, TLR2 silencing in microglia mitigated α-syn pathology in vivo. Overall, the present data support the idea that the interaction of exosomal α-syn and microglial TLR2 contribute to excessive α-syn phagocytosis and microglial activation, which lead to the further propagation and spread of α-syn pathology, thereby highlighting the pivotal roles of reactive microglia in α-syn transmission.
Collapse
Affiliation(s)
- Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Han
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of Science and Technology of China, Hefei, China
| | - Junjie Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunna Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
128
|
Harms AS, Ferreira SA, Romero-Ramos M. Periphery and brain, innate and adaptive immunity in Parkinson's disease. Acta Neuropathol 2021; 141:527-545. [PMID: 33555429 PMCID: PMC7952334 DOI: 10.1007/s00401-021-02268-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 12/29/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where alpha-synuclein plays a central role in the death and dysfunction of neurons, both, in central, as well as in the peripheral nervous system. Besides the neuronal events observed in patients, PD also includes a significant immune component. It is suggested that the PD-associated immune response will have consequences on neuronal health, thus opening immunomodulation as a potential therapeutic strategy in PD. The immune changes during the disease occur in the brain, involving microglia, but also in the periphery with changes in cells of the innate immune system, particularly monocytes, as well as those of adaptive immunity, such as T-cells. This realization arises from multiple patient studies, but also from data in animal models of the disease, providing strong evidence for innate and adaptive immune system crosstalk in the central nervous system and periphery in PD. Here we review the data showing that alpha-synuclein plays a crucial role in the activation of the innate and adaptive immune system. We will also describe the studies suggesting that inflammation in PD includes early changes in innate and adaptive immune cells that develop dynamically through time during disease, contributing to neuronal degeneration and symptomatology in patients. This novel finding has contributed to the definition of PD as a multisystem disease that should be approached in a more integratory manner rather than a brain-focused classical approach.
Collapse
Affiliation(s)
- Ashley S Harms
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara A Ferreira
- Department of Biomedicine and CNS Disease Modelling Group, Aarhus University, Høegh-Guldbergsgade 10, 8000C, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine and CNS Disease Modelling Group, Aarhus University, Høegh-Guldbergsgade 10, 8000C, Aarhus, Denmark.
| |
Collapse
|
129
|
Song N, Chen L, Xie J. Alpha-Synuclein Handling by Microglia: Activating, Combating, and Worsening. Neurosci Bull 2021; 37:751-753. [PMID: 33743127 DOI: 10.1007/s12264-021-00651-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/14/2020] [Indexed: 11/25/2022] Open
Affiliation(s)
- Ning Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| | - Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
130
|
Role of microgliosis, oxidative stress and associated neuroinflammation in the pathogenesis of Parkinson's disease: The therapeutic role of Nrf2 activators. Neurochem Int 2021; 145:105014. [PMID: 33689805 DOI: 10.1016/j.neuint.2021.105014] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 12/24/2022]
Abstract
Microglial cells are the resident immune cells of the central nervous system. They are essential for normal functioning, maintenance of tissue integrity, clearance of dying neurons, elimination of pathogens, development and maintenance of homeostasis of the CNS. Many studies have consistently reported that oxidative stress and associated neuroinflammation mediated by microglial cells have a degenerating effect on dopaminergic neurons. In Parkinson's disease, the microglial cells by a process called microgliosis undergo rapid proliferation, accumulate at the site of tissue injury and undergo phenotypic and functional changes that result in the release of massive amounts of free radicals causing inflammation and neurodegeneration of dopaminergic neurons. Following the discovery of the irrefutable role oxidative stress and associated neuroinflammation, several proven antioxidants were tested for possible protective and therapeutic potential in Parkinson's disease but the results so far have not been encouraging and equivocal. Consequently, it is rational to look for endogenous targets that enhance the oxidative defense mechanism against free radicals and protect dopaminergic neurons from neuroinflammation and neurodegeneration. One such target is a nuclear factor-erythroid -2-related factor 2 (Nrf2). Nrf2 is a redox-sensitive transcription factor located in the cytoplasm of the cells that helps cells adapt to oxidative stress and inflammation by upregulating the expression of almost 200 cytoprotective genes. Fractalkine exists in a transmembrane form and a soluble form and is a cytokine that links microglial cells and Nrf2. The fractalkine receptors, expressed exclusively by microglial cells, on activation by fractalkine protects dopaminergic neurons from degeneration caused by free radicals and pro-inflammatory mediators through increased expression of Nrf2 dependent genes. The current anti Parkinsonism drugs do not cure the disease and also cause several debilitating motor and non-motor adverse drug effects. So it becomes imperative to explore novel targets and discover novel therapeutic agents to treat Parkinson's disease in a better way and improve the quality of life of patients with Parkinson's disease.
Collapse
|
131
|
Nassir CMNCM, Ghazali MM, Hashim S, Idris NS, Yuen LS, Hui WJ, Norman HH, Gau CH, Jayabalan N, Na Y, Feng L, Ong LK, Abdul Hamid H, Ahamed HN, Mustapha M. Diets and Cellular-Derived Microparticles: Weighing a Plausible Link With Cerebral Small Vessel Disease. Front Cardiovasc Med 2021; 8:632131. [PMID: 33718454 PMCID: PMC7943466 DOI: 10.3389/fcvm.2021.632131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
Cerebral small vessel disease (CSVD) represents a spectrum of pathological processes of various etiologies affecting the brain microcirculation that can trigger neuroinflammation and the subsequent neurodegenerative cascade. Prevalent with aging, CSVD is a recognized risk factor for stroke, vascular dementia, Alzheimer disease, and Parkinson disease. Despite being the most common neurodegenerative condition with cerebrocardiovascular axis, understanding about it remains poor. Interestingly, modifiable risk factors such as unhealthy diet including high intake of processed food, high-fat foods, and animal by-products are known to influence the non-neural peripheral events, such as in the gastrointestinal tract and cardiovascular stress through cellular inflammation and oxidation. One key outcome from such events, among others, includes the cellular activations that lead to elevated levels of endogenous cellular-derived circulating microparticles (MPs). MPs can be produced from various cellular origins including leukocytes, platelets, endothelial cells, microbiota, and microglia. MPs could act as microthrombogenic procoagulant that served as a plausible culprit for the vulnerable end-artery microcirculation in the brain as the end-organ leading to CSVD manifestations. However, little attention has been paid on the potential role of MPs in the onset and progression of CSVD spectrum. Corroboratively, the formation of MPs is known to be influenced by diet-induced cellular stress. Thus, this review aims to appraise the body of evidence on the dietary-related impacts on circulating MPs from non-neural peripheral origins that could serve as a plausible microthrombosis in CSVD manifestation as a precursor of neurodegeneration. Here, we elaborate on the pathomechanical features of MPs in health and disease states; relevance of dietary patterns on MP release; preclinical studies pertaining to diet-based MPs contribution to disease; MP level as putative surrogates for early disease biomarkers; and lastly, the potential of MPs manipulation with diet-based approach as a novel preventive measure for CSVD in an aging society worldwide.
Collapse
Affiliation(s)
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Sabarisah Hashim
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Nur Suhaila Idris
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Lee Si Yuen
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Wong Jia Hui
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Haziq Hazman Norman
- Anatomy Unit, International Medical School (IMS), Management and Science University (MSU), Shah Alam, Malaysia
| | - Chuang Huei Gau
- Department of Psychology and Counselling, Faculty of Arts and Social Science, Universiti Tunku Abdul Rahman (UTAR), Kampar, Malaysia
| | - Nanthini Jayabalan
- Translational Neuroscience Lab, University of Queensland (UQ), Centre for Clinical Research, The University of Queensland, Herston, QLD, Australia
| | - Yuri Na
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Linqing Feng
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, National Health and Medical Research Council (NHMRC), Heidelberg, VIC, Australia
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Haja Nazeer Ahamed
- Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, India
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Hospital Universiti Sains Malaysia, Jalan Raja Perempuan Zainab II, Kubang Kerian, Malaysia
| |
Collapse
|
132
|
Zhang XY, Guo Z, Li TP, Sun T. Dietary capsaicin normalizes CGRP peptidergic DRG neurons in experimental diabetic peripheral neuropathy. Sci Rep 2021; 11:1704. [PMID: 33462325 PMCID: PMC7814129 DOI: 10.1038/s41598-021-81427-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022] Open
Abstract
Diabetic sensory neuropathy leads to impairment of peripheral sensory nerves and downregulation of calcitonin gene-related peptide (CGRP) in a functionally specific subset of peripheral sensory neurons mediating pain. Whether CGRP plays a neuroprotective role in peripheral sensory nerve is unclear. We evaluated alterations in noxious thermal sensation and downregulation of CGRP in the 8 weeks after induction of diabetes in rats. We supplemented capsaicin in the diet of the animals to upregulate CGRP and reversed the downregulation of the neuropeptide in the dorsal root ganglion (DRG) neurons dissociated from the diabetic animals, via gene transfection and exogenous CGRP, to test disease-preventing and disease-limiting effects of CGRP. Significant preservation of the nociceptive sensation, CGRP in spinal cord and DRG neurons, and number of CGRP-expressing neurons was found in the diabetic animals given capsaicin. Improvement in the survival of the neurons and the outgrowth of neurites was achieved in the neurons transfected by LV-CGRP or by exogenous CGRP, paralleling the correction of abnormalities of intracellular reactive oxygen species and mitochondrial transmembrane potentials. The results suggest that downregulation of CGRP impairs viability, regeneration and function of peripheral sensory neurons while capsaicin normalizes the CGRP peptidergic DRG neurons and function of the sensory nerves.
Collapse
Affiliation(s)
- Xiao-Yi Zhang
- Department of Anesthesiology, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Zheng Guo
- Department of Anesthesiology, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China. .,Department of Anesthesiology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China. .,Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| | - Tu-Ping Li
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Tao Sun
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| |
Collapse
|
133
|
Metabotropic glutamate receptor 5 inhibits α-synuclein-induced microglia inflammation to protect from neurotoxicity in Parkinson's disease. J Neuroinflammation 2021; 18:23. [PMID: 33461598 PMCID: PMC7814625 DOI: 10.1186/s12974-021-02079-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Background Microglia activation induced by α-synuclein (α-syn) is one of the most important factors in Parkinson’s disease (PD) pathogenesis. However, the molecular mechanisms by which α-syn exerts neuroinflammation and neurotoxicity remain largely elusive. Targeting metabotropic glutamate receptor 5 (mGluR5) has been an attractive strategy to mediate microglia activation for neuroprotection, which might be an essential regulator to modulate α-syn-induced neuroinflammation for the treatment of PD. Here, we showed that mGluR5 inhibited α-syn-induced microglia inflammation to protect from neurotoxicity in vitro and in vivo. Methods Co-immunoprecipitation assays were utilized to detect the interaction between mGluR5 and α-syn in microglia. Griess, ELISA, real-time PCR, western blotting, and immunofluorescence assays were used to detect the regulation of α-syn-induced inflammatory signaling, cytokine secretion, and lysosome-dependent degradation. Results α-syn selectively interacted with mGluR5 but not mGluR3, and α-syn N terminal deletion region was essential for binding to mGluR5 in co-transfected HEK293T cells. The interaction between these two proteins was further detected in BV2 microglia, which was inhibited by the mGluR5 specific agonist CHPG without effect by its selective antagonist MTEP. Moreover, in both BV2 cells and primary microglia, activation of mGluR5 by CHPG partially inhibited α-syn-induced inflammatory signaling and cytokine secretion and also inhibited the microglia activation to protect from neurotoxicity. We further found that α-syn overexpression decreased mGluR5 expression via a lysosomal pathway, as evidenced by the lysosomal inhibitor, NH4Cl, by blocking mGluR5 degradation, which was not evident with the proteasome inhibitor, MG132. Additionally, co-localization of mGluR5 with α-syn was detected in lysosomes as merging with its marker, LAMP-1. Consistently, in vivo experiments with LPS- or AAV-α-syn-induced rat PD model also confirmed that α-syn accelerated lysosome-dependent degradation of mGluR5 involving a complex, to regulate neuroinflammation. Importantly, the binding is strengthened with LPS or α-syn overexpression but alleviated by urate, a potential clinical biomarker for PD. Conclusions These findings provided evidence for a novel mechanism by which the association of α-syn with mGluR5 was attributed to α-syn-induced microglia activation via modulation of mGluR5 degradation and its intracellular signaling. This may be a new molecular target for an effective therapeutic strategy for PD pathology. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02079-1.
Collapse
|
134
|
Choi B, Ahn MH, Hong S, Barcelon EE, Sangshetti J, Arote RB, Lee SJ. Development of novel, biocompatible, polyester amines for microglia-targeting gene delivery. RSC Adv 2021; 11:36792-36800. [PMID: 35494387 PMCID: PMC9043621 DOI: 10.1039/d1ra06277h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/07/2021] [Indexed: 11/21/2022] Open
Abstract
Recent progress in personalized medicine and gene delivery has created exciting opportunities in therapeutics for central nervous system (CNS) disorders. Despite the interest in gene-based therapies, successful delivery of nucleic acids for treatment of CNS disorders faces major challenges. Here we report the facile synthesis of a novel, biodegradable, microglia-targeting polyester amine (PEA) carrier based on hydrophilic triethylene glycol dimethacrylate (TG) and low-molecular weight polyethylenimine (LMW-PEI). This nanocarrier, TG-branched PEI (TGP), successfully condensed double-stranded DNA into a size smaller than 200 nm. TGP nanoplexes were nontoxic in primary mixed glial cells and showed elevated transfection efficiency compared with PEI-25K and lipofector-EZ. After intrathecal and intracranial administration, PEA nanoplexes delivered genes specifically to microglia in the spinal cord and brain, respectively, proposing TGP as a novel microglia-specific gene delivery nanocarrier. The microglia-specific targeting of the TGP nanocarrier offers a new therapeutic strategy to modulate CNS disorders involving aberrant microglia activation while minimizing off-target side effects. A novel microglia-targeting polyester amine nanocarrier allows microglia-specific gene delivery for the treatment of CNS disorder involving microglia dysfunction.![]()
Collapse
Affiliation(s)
- Boomin Choi
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Min-Hye Ahn
- Department of Molecular Genetics and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Seojin Hong
- Department of Molecular Genetics and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Ellane Eda Barcelon
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiprakash Sangshetti
- Department of Medicinal Chemistry, Y. B. Chavan College of Pharmacy, Dr Rafiq Zakaria Campus, Rauza Baugh, Aurangabad, MS, India
| | - Rohidas B. Arote
- Department of Molecular Genetics and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Joong Lee
- Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
135
|
Jimenez-Ferrer I, Bäckström F, Dueñas-Rey A, Jewett M, Boza-Serrano A, Luk KC, Deierborg T, Swanberg M. The MHC class II transactivator modulates seeded alpha-synuclein pathology and dopaminergic neurodegeneration in an in vivo rat model of Parkinson's disease. Brain Behav Immun 2021; 91:369-382. [PMID: 33223048 DOI: 10.1016/j.bbi.2020.10.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/24/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Abnormal folding, aggregation and spreading of alpha-synuclein (αsyn) is a mechanistic hypothesis for the progressive neuropathology in Parkinson's disease (PD). Spread of αsyn between cells is supported by clinical, neuropathological and experimental evidence. It has been proposed that a pro-inflammatory micro-environment in response to αsyn can promote its aggregation. We have previously shown that allelic differences in the major histocompatibility complex class two transactivator (Mhc2ta) gene, located in the VRA4 locus, alter MHCII expression levels, microglial activation and antigen presentation capacity in rats upon human αsyn over-expression. In addition, Mhc2ta regulated dopaminergic neurodegeneration and the extent of motor impairment. The purpose of this study was to determine whether Mhc2ta regulates αsyn aggregation, propagation and dopaminergic pathology in an αsyn pre-formed fibril (PFF)-seeded in vivo model of PD. METHODS The DA and DA.VRA4 congenic rat strains share background genome but display differential microglial antigen presenting capacity due to different Mhc2ta alleles in the VRA4 locus. PFFs of human αsyn or BSA solution were injected unilaterally to the striatum of DA and DA.VRA4 rats two weeks after ipsilateral administration of recombinant adeno-associated virus (rAAV) vectors carrying human αsyn or GFP to the substantia nigra pars compacta. Behavioural assessment was performed at 2, 5 and 8 weeks while histological evaluation of αsyn pathology, inflammation and neurodegeneration as well as determination of serum cytokine profiles were performed at 8 weeks. RESULTS rAAV-mediated expression of human αsyn in nigral dopaminergic neurons combined with striatal PFF administration induced enhanced αsyn pathology in DA.VRA4 compared to DA rats. Mhc2ta thus significantly regulated the seeding, propagation and toxicity of αsyn in vivo. This was reflected in terms of wider extent and anatomical distribution of αsyn inclusions, ranging from striatum to the forebrain, midbrain, hindbrain and cerebellum in DA.VRA4. Compared to DA rats, DA.VRA4 also displayed enhanced motor impairment and dopaminergic neurodegeneration as well as higher levels of the proinflammatory cytokines IL-2 and TNFα in serum. CONCLUSIONS We conclude that the key regulator of MHCII expression, Mhc2ta, modulates neuroinflammation, αsyn-seeded Lewy-like pathology, dopaminergic neurodegeneration and motor impairment. This makes Mhc2ta and microglial antigen presentation promising therapeutic targets for reducing the progressive neuropathology and clinical manifestations in PD.
Collapse
Affiliation(s)
- Itzia Jimenez-Ferrer
- Translational Neurogenetics Unit, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| | - Filip Bäckström
- Translational Neurogenetics Unit, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| | - Alfredo Dueñas-Rey
- Translational Neurogenetics Unit, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| | - Michael Jewett
- Translational Neurogenetics Unit, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| | | | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Institute on Aging and Centre for Neurodegenerative Disease Research, Philadelphia, PA, USA
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Maria Swanberg
- Translational Neurogenetics Unit, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden.
| |
Collapse
|
136
|
Ma J, Gao J, Niu M, Zhang X, Wang J, Xie A. P2X4R Overexpression Upregulates Interleukin-6 and Exacerbates 6-OHDA-Induced Dopaminergic Degeneration in a Rat Model of PD. Front Aging Neurosci 2020; 12:580068. [PMID: 33328961 PMCID: PMC7671967 DOI: 10.3389/fnagi.2020.580068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/28/2020] [Indexed: 01/12/2023] Open
Abstract
The pathogenesis of Parkinson’s disease (PD) remains elusive. Current thinking suggests that the activation of microglia and the subsequent release of inflammatory factors, including interleukin-6 (IL-6), are involved in the pathogenesis of PD. P2X4 receptor (P2X4R) is a member of the P2X superfamily of ion channels activated by ATP. To study the possible effect of the ATP-P2X4R signal axis on IL-6 in PD, lentivirus carrying the P2X4R-overexpression gene or empty vector was injected into the substantia nigra (SN) of rats, followed by treatment of 6-hydroxydopamine (6-OHDA) or saline 1 week later. The research found the relative expression of P2X4R in the 6-OHDA-induced PD rat models was notably higher than that in the normal. And P2X4R overexpression could upregulate the expression of IL-6, reduce the amount of dopaminergic (DA) neurons in the SN of PD rats, suggesting that P2X4R may mediate the production of IL-6 to damage DA neurons in the SN. Our data revealed the important role of P2X4R in modulating IL-6, which leads to neuroinflammation involved in PD pathogenesis.
Collapse
Affiliation(s)
- Jiangnan Ma
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinzhao Gao
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengyue Niu
- Department of Neurology, Ruijin Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Xiaona Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Wang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
137
|
Calderón-Garcidueñas L, González-Maciel A, Reynoso-Robles R, Hammond J, Kulesza R, Lachmann I, Torres-Jardón R, Mukherjee PS, Maher BA. Quadruple abnormal protein aggregates in brainstem pathology and exogenous metal-rich magnetic nanoparticles (and engineered Ti-rich nanorods). The substantia nigrae is a very early target in young urbanites and the gastrointestinal tract a key brainstem portal. ENVIRONMENTAL RESEARCH 2020; 191:110139. [PMID: 32888951 DOI: 10.1016/j.envres.2020.110139] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/21/2020] [Accepted: 08/22/2020] [Indexed: 06/11/2023]
Abstract
Fine particulate air pollution (PM2.5) exposures are linked with Alzheimer's and Parkinson's diseases (AD,PD). AD and PD neuropathological hallmarks are documented in children and young adults exposed lifelong to Metropolitan Mexico City air pollution; together with high frontal metal concentrations (especially iron)-rich nanoparticles (NP), matching air pollution combustion- and friction-derived particles. Here, we identify aberrant hyperphosphorylated tau, ɑ synuclein and TDP-43 in the brainstem of 186 Mexico City 27.29 ± 11.8y old residents. Critically, substantia nigrae (SN) pathology seen in mitochondria, endoplasmic reticulum and neuromelanin (NM) is co-associated with the abundant presence of exogenous, Fe-, Al- and Ti-rich NPs.The SN exhibits early and progressive neurovascular unit damage and mitochondria and NM are associated with metal-rich NPs including exogenous engineered Ti-rich nanorods, also identified in neuroenteric neurons. Such reactive, cytotoxic and magnetic NPs may act as catalysts for reactive oxygen species formation, altered cell signaling, and protein misfolding, aggregation and fibril formation. Hence, pervasive, airborne and environmental, metal-rich and magnetic nanoparticles may be a common denominator for quadruple misfolded protein neurodegenerative pathologies affecting urbanites from earliest childhood. The substantia nigrae is a very early target and the gastrointestinal tract (and the neuroenteric system) key brainstem portals. The ultimate neural damage and neuropathology (Alzheimer's, Parkinson's and TDP-43 pathology included) could depend on NP characteristics and the differential access and targets achieved via their portals of entry. Thus where you live, what air pollutants you are exposed to, what you are inhaling and swallowing from the air you breathe,what you eat, how you travel, and your occupational longlife history are key. Control of NP sources becomes critical.
Collapse
Affiliation(s)
| | | | | | - Jessica Hammond
- Centre for Environmental Magnetism and Paleomagnetism, Lancaster Environment Centre, University of Lancaster, Lancaster, LA1 4YQ, UK
| | - Randy Kulesza
- Auditory Research Center, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | | | - Ricardo Torres-Jardón
- Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, UNAM, Mexico City, 04510, Mexico
| | | | - Barbara A Maher
- Centre for Environmental Magnetism and Paleomagnetism, Lancaster Environment Centre, University of Lancaster, Lancaster, LA1 4YQ, UK
| |
Collapse
|
138
|
Picca A, Guerra F, Calvani R, Coelho-Júnior HJ, Landi F, Bernabei R, Romano R, Bucci C, Marzetti E. Extracellular Vesicles and Damage-Associated Molecular Patterns: A Pandora's Box in Health and Disease. Front Immunol 2020; 11:601740. [PMID: 33304353 PMCID: PMC7701251 DOI: 10.3389/fimmu.2020.601740] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Sterile inflammation develops as part of an innate immunity response to molecules released upon tissue injury and collectively indicated as damage-associated molecular patterns (DAMPs). While coordinating the clearance of potential harmful stimuli, promotion of tissue repair, and restoration of tissue homeostasis, a hyper-activation of such an inflammatory response may be detrimental. The complex regulatory pathways modulating DAMPs generation and trafficking are actively investigated for their potential to provide relevant insights into physiological and pathological conditions. Abnormal circulating extracellular vesicles (EVs) stemming from altered endosomal-lysosomal system have also been reported in several age-related conditions, including cancer and neurodegeneration, and indicated as a promising route for therapeutic purposes. Along this pathway, mitochondria may dispose altered components to preserve organelle homeostasis. However, whether a common thread exists between DAMPs and EVs generation is yet to be clarified. A deeper understanding of the highly complex, dynamic, and variable intracellular and extracellular trafficking of DAMPs and EVs, including those of mitochondrial origin, is needed to unveil relevant pathogenic pathways and novel targets for drug development. Herein, we describe the mechanisms of generation of EVs and mitochondrial-derived vesicles along the endocytic pathway and discuss the involvement of the endosomal-lysosomal in cancer and neurodegeneration (i.e., Alzheimer's and Parkinson's disease).
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | | | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Bernabei
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Romano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
139
|
Gou DH, Huang TT, Li W, Gao XD, Haikal C, Wang XH, Song DY, Liang X, Zhu L, Tang Y, Ding C, Li JY. Inhibition of copper transporter 1 prevents α-synuclein pathology and alleviates nigrostriatal degeneration in AAV-based mouse model of Parkinson's disease. Redox Biol 2020; 38:101795. [PMID: 33232911 PMCID: PMC7691620 DOI: 10.1016/j.redox.2020.101795] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
The formation of α-synuclein aggregates is a major pathological hallmark of Parkinson's disease. Copper promotes α-synuclein aggregation and toxicity in vitro. The level of copper and copper transporter 1, which is the only known high-affinity copper importer in the brain, decreases in the substantia nigra of Parkinson's disease patients. However, the relationship between copper, copper transporter 1 and α-synuclein pathology remains elusive. Here, we aim to decipher the molecular mechanisms of copper and copper transporter 1 underlying Parkinson's disease pathology. We employed yeast and mammalian cell models expressing human α-synuclein, where exogenous copper accelerated intracellular α-synuclein inclusions and silencing copper transporter 1 reduced α-synuclein aggregates in vitro, suggesting that copper transporter 1 might inhibit α-synuclein pathology. To study our hypothesis in vivo, we generated a new transgenic mouse model with copper transporter 1 conditional knocked-out specifically in dopaminergic neuron. Meanwhile, we unilaterally injected adeno-associated viral human-α-synuclein into the substantia nigra of these mice. Importantly, we found that copper transporter 1 deficiency significantly reduced S129-phosphorylation of α-synuclein, prevented dopaminergic neuronal loss, and alleviated motor dysfunction caused by α-synuclein overexpression in vivo. Overall, our data indicated that inhibition of copper transporter 1 alleviated α-synuclein mediated pathologies and provided a novel therapeutic strategy for Parkinson's disease and other synucleinopathies. Ctr1 deficiency reduces α-synuclein aggregates in vitro. Ctr1 deficiency inhibits the level of pathological α-synuclein in vivo. Ctr1 deficiency prevents nigrostriatal neurodegeneration in vivo. Ctr1 deficiency alleviates motor dysfunction caused by α-synuclein in vivo.
Collapse
Affiliation(s)
- De-Hai Gou
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Ting-Ting Huang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Wen Li
- Institute of Health Sciences, China Medical University, Shenyang, 110122, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 22184, Lund, Sweden
| | - Xin-Di Gao
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Caroline Haikal
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 22184, Lund, Sweden
| | - Xin-He Wang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Dong-Yan Song
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Xin Liang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Lin Zhu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Chen Ding
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China.
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China; Institute of Health Sciences, China Medical University, Shenyang, 110122, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 22184, Lund, Sweden.
| |
Collapse
|
140
|
Henrich MT, Geibl FF, Lakshminarasimhan H, Stegmann A, Giasson BI, Mao X, Dawson VL, Dawson TM, Oertel WH, Surmeier DJ. Determinants of seeding and spreading of α-synuclein pathology in the brain. SCIENCE ADVANCES 2020; 6:eabc2487. [PMID: 33177086 PMCID: PMC7673735 DOI: 10.1126/sciadv.abc2487] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/22/2020] [Indexed: 05/22/2023]
Abstract
In Parkinson's disease (PD), fibrillar forms of α-synuclein are hypothesized to propagate through synaptically coupled networks, causing Lewy pathology (LP) and neurodegeneration. To more rigorously characterize the determinants of spreading, preformed α-synuclein fibrils were injected into the mouse pedunculopontine nucleus (PPN), a brain region that manifests LP in PD patients and the distribution of developing α-synuclein pathology compared to that ascertained by anterograde and retrograde connectomic mapping. Within the PPN, α-synuclein pathology was cell-specific, being robust in PD-vulnerable cholinergic neurons but not in neighboring noncholinergic neurons. While nearly all neurons projecting to PPN cholinergics manifested α-synuclein pathology, the kinetics, magnitude, and persistence of the propagated pathology were unrelated to the strength of those connections. Thus, neuronal phenotype governs the somatodendritic uptake of pathological α-synuclein, and while the afferent connectome restricts the subsequent spreading of pathology, its magnitude and persistence is not a strict function of the strength of coupling.
Collapse
Affiliation(s)
- Martin T Henrich
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurology, Philipps University Marburg, Marburg 35043, Germany
| | - Fanni F Geibl
- Department of Neurology, Philipps University Marburg, Marburg 35043, Germany
| | - Harini Lakshminarasimhan
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Anna Stegmann
- Department of Neurology, Philipps University Marburg, Marburg 35043, Germany
| | - Benoit I Giasson
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, Marburg 35043, Germany
| | - D James Surmeier
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
141
|
Microglia in Prion Diseases: Angels or Demons? Int J Mol Sci 2020; 21:ijms21207765. [PMID: 33092220 PMCID: PMC7589037 DOI: 10.3390/ijms21207765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Prion diseases are rare transmissible neurodegenerative disorders caused by the accumulation of a misfolded isoform (PrPSc) of the cellular prion protein (PrPC) in the central nervous system (CNS). Neuropathological hallmarks of prion diseases are neuronal loss, astrogliosis, and enhanced microglial proliferation and activation. As immune cells of the CNS, microglia participate both in the maintenance of the normal brain physiology and in driving the neuroinflammatory response to acute or chronic (e.g., neurodegenerative disorders) insults. Microglia involvement in prion diseases, however, is far from being clearly understood. During this review, we summarize and discuss controversial findings, both in patient and animal models, suggesting a neuroprotective role of microglia in prion disease pathogenesis and progression, or—conversely—a microglia-mediated exacerbation of neurotoxicity in later stages of disease. We also will consider the active participation of PrPC in microglial functions, by discussing previous reports, but also by presenting unpublished results that support a role for PrPC in cytokine secretion by activated primary microglia.
Collapse
|
142
|
Fouka M, Mavroeidi P, Tsaka G, Xilouri M. In Search of Effective Treatments Targeting α-Synuclein Toxicity in Synucleinopathies: Pros and Cons. Front Cell Dev Biol 2020; 8:559791. [PMID: 33015057 PMCID: PMC7500083 DOI: 10.3389/fcell.2020.559791] [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: 05/07/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA) and Dementia with Lewy bodies (DLB) represent pathologically similar, progressive neurodegenerative disorders characterized by the pathological aggregation of the neuronal protein α-synuclein. PD and DLB are characterized by the abnormal accumulation and aggregation of α-synuclein in proteinaceous inclusions within neurons named Lewy bodies (LBs) and Lewy neurites (LNs), whereas in MSA α-synuclein inclusions are mainly detected within oligodendrocytes named glial cytoplasmic inclusions (GCIs). The presence of pathologically aggregated α-synuclein along with components of the protein degradation machinery, such as ubiquitin and p62, in LBs and GCIs is considered to underlie the pathogenic cascade that eventually leads to the severe neurodegeneration and neuroinflammation that characterizes these diseases. Importantly, α-synuclein is proposed to undergo pathogenic misfolding and oligomerization into higher-order structures, revealing self-templating conformations, and to exert the ability of "prion-like" spreading between cells. Therefore, the manner in which the protein is produced, is modified within neural cells and is degraded, represents a major focus of current research efforts in the field. Given that α-synuclein protein load is critical to disease pathogenesis, the identification of means to limit intracellular protein burden and halt α-synuclein propagation represents an obvious therapeutic approach in synucleinopathies. However, up to date the development of effective therapeutic strategies to prevent degeneration in synucleinopathies is limited, due to the lack of knowledge regarding the precise mechanisms underlying the observed pathology. This review critically summarizes the recent developed strategies to counteract α-synuclein toxicity, including those aimed to increase protein degradation, to prevent protein aggregation and cell-to-cell propagation, or to engage antibodies against α-synuclein and discuss open questions and unknowns for future therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
143
|
C. elegans Models to Study the Propagation of Prions and Prion-Like Proteins. Biomolecules 2020; 10:biom10081188. [PMID: 32824215 PMCID: PMC7464663 DOI: 10.3390/biom10081188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
A hallmark common to many age-related neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), is that patients develop proteinaceous deposits in their central nervous system (CNS). The progressive spreading of these inclusions from initially affected sites to interconnected brain areas is reminiscent of the behavior of bona fide prions in transmissible spongiform encephalopathies (TSEs), hence the term prion-like proteins has been coined. Despite intensive research, the exact mechanisms that facilitate the spreading of protein aggregation between cells, and the associated loss of neurons, remain poorly understood. As population demographics in many countries continue to shift to higher life expectancy, the incidence of neurodegenerative diseases is also rising. This represents a major challenge for healthcare systems and patients’ families, since patients require extensive support over several years and there is still no therapy to cure or stop these diseases. The model organism Caenorhabditis elegans offers unique opportunities to accelerate research and drug development due to its genetic amenability, its transparency, and the high degree of conservation of molecular pathways. Here, we will review how recent studies that utilize this soil dwelling nematode have proceeded to investigate the propagation and intercellular transmission of prions and prion-like proteins and discuss their relevance by comparing their findings to observations in other model systems and patients.
Collapse
|
144
|
Fatoba O, Itokazu T, Yamashita T. Microglia as therapeutic target in central nervous system disorders. J Pharmacol Sci 2020; 144:102-118. [PMID: 32921391 DOI: 10.1016/j.jphs.2020.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic microglial activation is associated with the pathogenesis of several CNS disorders. Microglia show phenotypic diversity and functional complexity in diseased CNS. Thus, understanding the pathology-specific heterogeneity of microglial behavior is crucial for the future development of microglia-modulating therapy for variety of CNS disorders. This review summarizes up-to-date knowledge on how microglia contribute to CNS homeostasis during development and throughout adulthood. We discuss the heterogeneity of microglial phenotypes in the context of CNS disorders with an emphasis on neurodegenerative diseases, demyelinating diseases, CNS trauma, and epilepsy. We conclude this review with a discussion about the disease-specific heterogeneity of microglial function and how it could be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Oluwaseun Fatoba
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
145
|
Picca A, Calvani R, Coelho-Junior HJ, Landi F, Bernabei R, Marzetti E. Mitochondrial Dysfunction, Oxidative Stress, and Neuroinflammation: Intertwined Roads to Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9080647. [PMID: 32707949 PMCID: PMC7466131 DOI: 10.3390/antiox9080647] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress develops as a response to injury and reflects a breach in the cell’s antioxidant capacity. Therefore, the fine-tuning of reactive oxygen species (ROS) generation is crucial for preserving cell’s homeostasis. Mitochondria are a major source and an immediate target of ROS. Under different stimuli, including oxidative stress and impaired quality control, mitochondrial constituents (e.g., mitochondrial DNA, mtDNA) are displaced toward intra- or extracellular compartments. However, the mechanisms responsible for mtDNA unloading remain largely unclear. While shuttling freely within the cell, mtDNA can be delivered into the extracellular compartment via either extrusion of entire nucleoids or the generation and release of extracellular vesicles. Once discarded, mtDNA may act as a damage-associated molecular pattern (DAMP) and trigger an innate immune inflammatory response by binding to danger-signal receptors. Neuroinflammation is associated with a large array of neurological disorders for which mitochondrial DAMPs could represent a common thread supporting disease progression. The exploration of non-canonical pathways involved in mitochondrial quality control and neurodegeneration may unveil novel targets for the development of therapeutic agents. Here, we discuss these processes in the setting of two common neurodegenerative diseases (Alzheimer’s and Parkinson’s disease) and Down syndrome, the most frequent progeroid syndrome.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Correspondence: (R.C.); (R.B.); Tel.: +39-06-3015-5559 (R.C. & R.B.); Fax: +39-06-3051-911 (R.C. & R.B.)
| | | | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Roberto Bernabei
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Correspondence: (R.C.); (R.B.); Tel.: +39-06-3015-5559 (R.C. & R.B.); Fax: +39-06-3051-911 (R.C. & R.B.)
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (F.L.); (E.M.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
146
|
Pajares M, I. Rojo A, Manda G, Boscá L, Cuadrado A. Inflammation in Parkinson's Disease: Mechanisms and Therapeutic Implications. Cells 2020; 9:cells9071687. [PMID: 32674367 PMCID: PMC7408280 DOI: 10.3390/cells9071687] [Citation(s) in RCA: 407] [Impact Index Per Article: 81.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder primarily characterized by the death of dopaminergic neurons that project from the substantia nigra pars compacta. Although the molecular bases for PD development are still little defined, extensive evidence from human samples and animal models support the involvement of inflammation in onset or progression. However, the exact trigger for this response remains unclear. Here, we provide a systematic review of the cellular mediators, i.e., microglia, astroglia and endothelial cells. We also discuss the genetic and transcriptional control of inflammation in PD and the immunomodulatory role of dopamine and reactive oxygen species. Finally, we summarize the preclinical and clinical approaches targeting neuroinflammation in PD.
Collapse
Affiliation(s)
- Marta Pajares
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Ana I. Rojo
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Gina Manda
- Department Cellular and Molecular Medicine, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Cardiovasculares (CIBERcv), ISCIII, 28029 Madrid, Spain
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
- Department Cellular and Molecular Medicine, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Correspondence: ; Tel.: +34-915854383; Fax: +34-915854401
| |
Collapse
|
147
|
Tomov N. Glial cells in intracerebral transplantation for Parkinson's disease. Neural Regen Res 2020; 15:1173-1178. [PMID: 31960796 PMCID: PMC7047789 DOI: 10.4103/1673-5374.270296] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/02/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
In the last few decades, intracerebral transplantation has grown from a dubious neuroscientific topic to a plausible modality for treatment of neurological disorders. The possibility for cell replacement opens a new field of perspectives in the therapy of neurodegenerative disorders, ischemia, and neurotrauma, with the most lessons learned from intracerebral transplantation in Parkinson's disease. Multiple animal studies and a few small-scale clinical trials have proven the concept of intracerebral grafting, but still have to provide a uniform and highly efficient approach to the procedure, suitable for clinical application. The success of intracerebral transplantation is highly dependent on the integration of the grafted cells with the host brain. In this process, glial cells are clearly more than passive bystanders. They provide transplanted cells with mechanical support, trophics, mediate synapse formation, and participate in graft vascularization. At the same time, glial cells mediate scarring, graft rejection, and neuroinflammation, which can be detrimental. We can use this information to try to understand the mechanisms behind the glial reaction to intracerebral transplantation. Recognizing and utilizing glial reactivity can move translational research forward and provide an insight not only to post-transplantation events but also to mechanisms of neuronal death and degeneration. Knowledge about glial reactivity to transplanted cells could also be a key for optimization of transplantation protocols, which ultimately should contribute to greater patient benefit.
Collapse
Affiliation(s)
- Nikola Tomov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| |
Collapse
|
148
|
Impact of α-synuclein pathology on transplanted hESC-derived dopaminergic neurons in a humanized α-synuclein rat model of PD. Proc Natl Acad Sci U S A 2020; 117:15209-15220. [PMID: 32541058 PMCID: PMC7334514 DOI: 10.1073/pnas.2001305117] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Preclinical assessment of the therapeutic potential of dopamine (DA) neuron replacement in Parkinson's disease (PD) has primarily been performed in the 6-hydroxydopamine toxin model. While this is a good model to assess graft function, it does not reflect the pathological features or progressive nature of the disease. In this study, we establish a humanized transplantation model of PD that better recapitulates the main disease features, obtained by coinjection of preformed human α-synuclein (α-syn) fibrils and adeno-associated virus (AAV) expressing human wild-type α-syn unilaterally into the rat substantia nigra (SN). This model gives rise to DA neuron dysfunction and progressive loss of DA neurons from the SN and terminals in the striatum, accompanied by extensive α-syn pathology and a prominent inflammatory response, making it an interesting and relevant model in which to examine long-term function and integrity of transplanted neurons in a PD-like brain. We transplanted DA neurons derived from human embryonic stem cells (hESCs) into the striatum and assessed their survival, growth, and function over 6 to 18 wk. We show that the transplanted cells, even in the presence of ongoing pathology, are capable of innervating the DA-depleted striatum. However, on closer examination of the grafts, we found evidence of α-syn pathology in the form of inclusions of phosphorylated α-syn in a small fraction of the grafted DA neurons, indicating host-to-graft transfer of α-syn pathology, a phenomenon that has previously been observed in PD patients receiving fetal tissue grafts but has not been possible to demonstrate and study in toxin-based animal models.
Collapse
|
149
|
Belloli S, Morari M, Murtaj V, Valtorta S, Moresco RM, Gilardi MC. Translation Imaging in Parkinson's Disease: Focus on Neuroinflammation. Front Aging Neurosci 2020; 12:152. [PMID: 32581765 PMCID: PMC7289967 DOI: 10.3389/fnagi.2020.00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the appearance of α-synuclein insoluble aggregates known as Lewy bodies. Neurodegeneration is accompanied by neuroinflammation mediated by cytokines and chemokines produced by the activated microglia. Several studies demonstrated that such an inflammatory process is an early event, and contributes to oxidative stress and mitochondrial dysfunctions. α-synuclein fibrillization and aggregation activate microglia and contribute to disease onset and progression. Mutations in different genes exacerbate the inflammatory phenotype in the monogenic compared to sporadic forms of PD. Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) with selected radiopharmaceuticals allow in vivo imaging of molecular modifications in the brain of living subjects. Several publications showed a reduction of dopaminergic terminals and dopamine (DA) content in the basal ganglia, starting from the early stages of the disease. Moreover, non-dopaminergic neuronal pathways are also affected, as shown by in vivo studies with serotonergic and glutamatergic radiotracers. The role played by the immune system during illness progression could be investigated with PET ligands that target the microglia/macrophage Translocator protein (TSPO) receptor. These agents have been used in PD patients and rodent models, although often without attempting correlations with other molecular or functional parameters. For example, neurodegeneration and brain plasticity can be monitored using the metabolic marker 2-Deoxy-2-[18F]fluoroglucose ([18F]-FDG), while oxidative stress can be probed using the copper-labeled diacetyl-bis(N-methyl-thiosemicarbazone) ([Cu]-ATSM) radioligand, whose striatal-specific binding ratio in PD patients seems to correlate with a disease rating scale and motor scores. Also, structural and functional modifications during disease progression may be evaluated by Magnetic Resonance Imaging (MRI), using different parameters as iron content or cerebral volume. In this review article, we propose an overview of in vivo clinical and non-clinical imaging research on neuroinflammation as an emerging marker of early PD. We also discuss how multimodal-imaging approaches could provide more insights into the role of the inflammatory process and related events in PD development.
Collapse
Affiliation(s)
- Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Michele Morari
- Section of Pharmacology, Department of Medical Sciences, National Institute for Neuroscience, University of Ferrara, Ferrara, Italy
| | - Valentina Murtaj
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Maria Carla Gilardi
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
150
|
Lazdon E, Stolero N, Frenkel D. Microglia and Parkinson's disease: footprints to pathology. J Neural Transm (Vienna) 2020; 127:149-158. [DOI: 10.1007/s00702-020-02154-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/26/2020] [Indexed: 12/11/2022]
|