1
|
Ono S, Nakamura M, Morikawa M, Doyama T, Kunieda T, Kaneko K, Tanaka K, Takahashi M, Kondo T, Yakushiji Y. Immunotherapy-responsive parkinsonism and anosmia in anti-NMDA receptor encephalitis with seropositive NMOSD: A case report. J Neurol Sci 2025; 472:123447. [PMID: 40054030 DOI: 10.1016/j.jns.2025.123447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025]
Affiliation(s)
- Shohei Ono
- Department of Neurology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Masataka Nakamura
- Department of Neurology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan.
| | - Masayasu Morikawa
- Department of Neurology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Tatsuya Doyama
- Department of Neurology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Takenobu Kunieda
- Department of Neurology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Kimihiko Kaneko
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1, Seiryo-cho, Aoba-ku, Sendai 980-8574, Japan
| | - Keiko Tanaka
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757, Asahimachi-dori, Chuoku, Niigata 951-8585, Japan
| | - Makio Takahashi
- Department of Neurodegenerative Disorders, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Takayuki Kondo
- Department of Neurology, Kansai Medical University Medical Center, 10-15, Fumizono-cho, Moriguchi, Osaka 570-8507, Japan
| | - Yusuke Yakushiji
- Department of Neurology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
2
|
Xiang H. The interplay between α-synuclein aggregation and necroptosis in Parkinson's disease: a spatiotemporal perspective. Front Neurosci 2025; 19:1567445. [PMID: 40264913 PMCID: PMC12011736 DOI: 10.3389/fnins.2025.1567445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the death of dopaminergic neurons and the aggregation of alpha-synuclein (α-Syn). It presents with prominent motor symptoms, and by the time of diagnosis, a significant number of neurons have already been lost. Current medications can only alleviate symptoms but cannot halt disease progression. Studies have confirmed that both dopaminergic neuronal loss and α-Syn aggregation are associated with necroptosis mechanisms. Necroptosis, a regulated form of cell death, has been recognized as an underexplored hotspot in PD pathogenesis research. In this review, we propose a spatiotemporal model of PD progression, highlighting the interactions between α-Syn aggregation, mitochondrial dysfunction, oxidative stress, neuroinflammation and necroptosis. These processes not only drive motor symptoms but also contribute to early non-motor symptoms, offering insights into potential diagnostic markers. Finally, we touch upon the therapeutic potential of necroptosis inhibition in enhancing current PD treatments, such as L-Dopa. This review aims to provide a new perspective on the pathogenesis of PD and to identify avenues for the development of more effective therapeutic strategies.
Collapse
Affiliation(s)
- Haoran Xiang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Department of Neurology, Yichang Central People’s Hospital, Yichang, Hubei, China
| |
Collapse
|
3
|
Pandey PP, Kumar MS. Exploring the therapeutic potential of steroidal alkaloids in managing Alzheimer's disease. Steroids 2024; 209:109468. [PMID: 38959993 DOI: 10.1016/j.steroids.2024.109468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Steroidal alkaloids are secondary metabolites that are often found in plants, fungi and sponges. These compounds are considered as a source of bioactive compounds for the treatment of chronic diseases, such as neurological disorder like Alzheimer's disease (AD). Some examples of alkaloid derivatives currently used to treat AD symptoms include galantamine, huperzine A, and other alkaloids. AD is a multifactorial disease caused by multiple factors such as inflammation, oxidative stress, and protein aggregation. Based on the various important neuroprotective activities and different pharmacological effects of steroidal alkaloids with polypharmacological modulatory effects, they can lead to the development of new drugs for the treatment of AD. There are limited studies on the involvement of steroidal alkaloids in AD. Therefore, the mechanisms and neuroprotective abilities of these compounds are still poorly understood. The purpose of this review article is to provide an overview of the mechanism, toxicity and neuroprotective benefits of steroidal alkaloids and to discuss future possibilities to improve the application of steroidal alkaloids as anti-AD agents. The therapeutic value and limitations of the steroidal alkaloid are investigated to provide new perspectives for future clinical development studies.
Collapse
Affiliation(s)
- Pratima P Pandey
- Somaiya Institute for Research and Consultancy, Somaiya Vidyavihar University, Vidyavihar (East), Mumbai 400077, India
| | - Maushmi S Kumar
- Somaiya Institute for Research and Consultancy, Somaiya Vidyavihar University, Vidyavihar (East), Mumbai 400077, India.
| |
Collapse
|
4
|
Shah S, Pushpa Tryphena K, Singh G, Kulkarni A, Pinjala P, Kumar Khatri D. Neuroprotective role of Carvacrol via Nrf2/HO-1/NLRP3 axis in Rotenone-induced PD mice model. Brain Res 2024; 1836:148954. [PMID: 38649135 DOI: 10.1016/j.brainres.2024.148954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder whose cause is unclear. Neuroinflammation is recognized as one of the major pathogenic mechanisms involved in the development and progression of PD. NLRP3 inflammasome is the most widely studied inflammatory mediator in various diseases including PD. Several phytoconstituents have shown neuroprotective role in PD. Carvacrol is a phenolic monoterpene commonly found in the essential oils derived from plants belonging to Lamiaceae family. It is well known for its anti-inflammatory and antioxidant properties and has been widely explored in several diseases. In this study, we explored the role of Carvacrol in suppressing neuroinflammation by regulating NLRP3 inflammasome through Nrf2/HO-1 axis and subsequently, inflammatory cytokines like IL-1β, IL-18 in Rotenone induced PD mice model. Three doses (25 mg/kg, 50 mg/kg, 100 mg/kg p.o.) of Carvacrol were administered to, respectively, three groups (LD, MD, HD), one hour after administration of Rotenone (1.5 mg/kg, i.p.), every day, for 21 days. Treatment with Carvacrol ameliorated the motor impairment caused by Rotenone. It alleviated neurotoxicity and reduced inflammatory cytokines. Further, Carvacrol also alleviated oxidative stress and increased antioxidant enzymes. From these results, we show that Carvacrol exerts neuroprotective effects in PD via anti-inflammatory and antioxidant mechanisms and could be a potential therapeutic option in PD.
Collapse
Affiliation(s)
- Shruti Shah
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Kamatham Pushpa Tryphena
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Gurpreet Singh
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Amrita Kulkarni
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Poojitha Pinjala
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
5
|
Bhatt V, Shukla H, Tiwari AK. Parkinson's Disease and Mitotherapy-Based Approaches towards α-Synucleinopathies. J Integr Neurosci 2024; 23:109. [PMID: 38940084 DOI: 10.31083/j.jin2306109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 06/29/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta region of the midbrain and the formation of intracellular protein aggregates known as Lewy bodies, of which a major component is the protein α-synuclein. Several studies have suggested that mitochondria play a central role in the pathogenesis of PD, encompassing both familial and sporadic forms of the disease. Mitochondrial dysfunction is attributed to bioenergetic impairment, increased oxidative stress, damage to mitochondrial DNA, and alteration in mitochondrial morphology. These alterations may contribute to improper functioning of the central nervous system and ultimately lead to neurodegeneration. The perturbation of mitochondrial function makes it a potential target, worthy of exploration for neuroprotective therapies and to improve mitochondrial health in PD. Thus, in the current review, we provide an update on mitochondria-based therapeutic approaches toward α-synucleinopathies in PD.
Collapse
Affiliation(s)
- Vidhi Bhatt
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Halak Shukla
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| |
Collapse
|
6
|
Park M, Ha J, Lee Y, Choi HS, Kim BS, Jeong YK. Low-moderate dose whole-brain γ-ray irradiation modulates the expressions of glial fibrillary acidic protein and intercellular adhesion molecule-1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease mouse model. Neurobiol Aging 2023; 132:175-184. [PMID: 37837733 DOI: 10.1016/j.neurobiolaging.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/22/2023] [Accepted: 06/24/2023] [Indexed: 10/16/2023]
Abstract
The anti-inflammatory efficacy of radiation therapy (RT) with single fractions below 1.0 Gy has been demonstrated in Alzheimer's disease mouse models. As neuroinflammation is also a major pathological feature of Parkinson's disease (PD), RT may also be effective in PD treatment. Therefore, this study aimed to investigate the anti-inflammatory effect of low-moderate dose RT (LMDRT, 0.6 Gy/single dose, for 5 days) exposure in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 30 mg/kg, intraperitoneally, for 5 consecutive days)-induced PD mouse model. Importantly, LMDRT reduced the levels of glial fibrillary acidic protein and intercellular adhesion molecule-1 (CD54) in the striatum region, which increased following MPTP administration. LMDRT also modulated inflammatory gene expression patterns in the substantia nigra region of the MPTP-treated mice. However, LMDRT had no direct effects on the severe loss of dopaminergic neurons and impaired motor behavior in the rotarod test. These results indicate that LMDRT has anti-inflammatory effects by modulating neuroinflammatory factors, including glial fibrillary acidic protein and intercellular adhesion molecule-1, but showed no behavioral improvements or neuroprotection in the MPTP-induced mouse model of PD.
Collapse
MESH Headings
- Animals
- Mice
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use
- Brain/metabolism
- Brain/radiation effects
- Disease Models, Animal
- Dopaminergic Neurons/pathology
- Glial Fibrillary Acidic Protein/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Intercellular Adhesion Molecule-1/pharmacology
- Intercellular Adhesion Molecule-1/therapeutic use
- Mice, Inbred C57BL
- Parkinson Disease/metabolism
- Parkinson Disease/radiotherapy
- Substantia Nigra/metabolism
Collapse
Affiliation(s)
- Mijeong Park
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jimin Ha
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Division of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Yuri Lee
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Division of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Hoon-Seong Choi
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Byoung Soo Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Youn Kyoung Jeong
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Bérard M, Martínez-Drudis L, Sheta R, El-Agnaf OMA, Oueslati A. Non-invasive systemic viral delivery of human alpha-synuclein mimics selective and progressive neuropathology of Parkinson's disease in rodent brains. Mol Neurodegener 2023; 18:91. [PMID: 38012703 PMCID: PMC10683293 DOI: 10.1186/s13024-023-00683-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Alpha-synuclein (α-syn) aggregation into proteinaceous intraneuronal inclusions, called Lewy bodies (LBs), is the neuropathological hallmark of Parkinson's disease (PD) and related synucleinopathies. However, the exact role of α-syn inclusions in PD pathogenesis remains elusive. This lack of knowledge is mainly due to the absence of optimal α-syn-based animal models that recapitulate the different stages of neurodegeneration. METHODS Here we describe a novel approach for a systemic delivery of viral particles carrying human α-syn allowing for a large-scale overexpression of this protein in the mouse brain. This approach is based on the use of a new generation of adeno-associated virus (AAV), AAV-PHP.eB, with an increased capacity to cross the blood-brain barrier, thus offering a viable tool for a non-invasive and large-scale gene delivery in the central nervous system. RESULTS Using this model, we report that widespread overexpression of human α-syn induced selective degeneration of dopaminergic (DA) neurons, an exacerbated neuroinflammatory response in the substantia nigra and a progressive manifestation of PD-like motor impairments. Interestingly, biochemical analysis revealed the presence of insoluble α-syn oligomers in the midbrain. Together, our data demonstrate that a single non-invasive systemic delivery of viral particles overexpressing α-syn prompted selective and progressive neuropathology resembling the early stages of PD. CONCLUSIONS Our new in vivo model represents a valuable tool to study the role of α-syn in PD pathogenesis and in the selective vulnerability of nigral DA neurons; and offers the opportunity to test new strategies targeting α-syn toxicity for the development of disease-modifying therapies for PD and related disorders.
Collapse
Affiliation(s)
- Morgan Bérard
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Laura Martínez-Drudis
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Razan Sheta
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Omar M A El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, 34110, Qatar
| | - Abid Oueslati
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada.
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada.
| |
Collapse
|
8
|
Pozojevic J, Spielmann M. Single-Cell Sequencing in Neurodegenerative Disorders. Mol Diagn Ther 2023; 27:553-561. [PMID: 37552451 PMCID: PMC10435411 DOI: 10.1007/s40291-023-00668-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/09/2023]
Abstract
Neurodegenerative disorders are typically characterized by late onset progressive damage to specific (sub)populations of cells of the nervous system that are essential for mobility, coordination, strength, sensation, and cognition. Addressing this selective cellular vulnerability has become feasible with the emergence of single-cell-omics technologies, which now represent the state-of-the-art approach to profile heterogeneity of complex tissues including human post-mortem brain at unprecedented resolution. In this review, we briefly recapitulate the experimental workflow of single-cell RNA sequencing and summarize the recent knowledge acquired with it in the most common neurodegenerative diseases: Parkinson's, Alzheimer's, Huntington's disease, and multiple sclerosis. We also discuss the possibility of applying single-cell approaches in the diagnostics and therapy of neurodegenerative disorders, as well as the limitations. While we are currently at the point of deeply exploring the transcriptomic changes in the affected cells, further technological developments hold a promise of manipulating the affected pathways once we understand them better.
Collapse
Affiliation(s)
- Jelena Pozojevic
- Institute of Human Genetics, Universitätsklinikum Schleswig-Holstein, University of Lübeck and University of Kiel, 23562, Lübeck, Germany
| | - Malte Spielmann
- Institute of Human Genetics, Universitätsklinikum Schleswig-Holstein, University of Lübeck and University of Kiel, 23562, Lübeck, Germany.
- Human Molecular Genomics Group, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, 23562, Lübeck, Germany.
| |
Collapse
|
9
|
Saucier J, Comeau D, Robichaud GA, Chamard-Witkowski L. Reactive gliosis and neuroinflammation: prime suspects in the pathophysiology of post-acute neuroCOVID-19 syndrome. Front Neurol 2023; 14:1221266. [PMID: 37693763 PMCID: PMC10492094 DOI: 10.3389/fneur.2023.1221266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/27/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction As the repercussions from the COVID-19 pandemic continue to unfold, an ever-expanding body of evidence suggests that infection also elicits pathophysiological manifestations within the central nervous system (CNS), known as neurological symptoms of post-acute sequelae of COVID infection (NeuroPASC). Although the neurological impairments and repercussions associated with NeuroPASC have been well described in the literature, its etiology remains to be fully characterized. Objectives This mini-review explores the current literature that elucidates various mechanisms underlining NeuroPASC, its players, and regulators, leading to persistent neuroinflammation of affected individuals. Specifically, we provide some insights into the various roles played by microglial and astroglial cell reactivity in NeuroPASC and how these cell subsets potentially contribute to neurological impairment in response to the direct or indirect mechanisms of CNS injury. Discussion A better understanding of the mechanisms and biomarkers associated with this maladaptive neuroimmune response will thus provide better diagnostic strategies for NeuroPASC and reveal new potential mechanisms for therapeutic intervention. Altogether, the elucidation of NeuroPASC pathogenesis will improve patient outcomes and mitigate the socioeconomic burden of this syndrome.
Collapse
Affiliation(s)
- Jacob Saucier
- Centre de Formation Médicale du Nouveau-Brunswick, Moncton, NB, Canada
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Dominique Comeau
- Centre de médecine de précision du Nouveau-Brunswick, Vitality Health Network, Moncton, NB, Canada
| | - Gilles A. Robichaud
- Centre de médecine de précision du Nouveau-Brunswick, Vitality Health Network, Moncton, NB, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Ludivine Chamard-Witkowski
- Centre de Formation Médicale du Nouveau-Brunswick, Moncton, NB, Canada
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de médecine de précision du Nouveau-Brunswick, Vitality Health Network, Moncton, NB, Canada
- Department of Neurology, Dr. Georges-L.-Dumont University Hospital Centre, Vitality Health Network, Moncton, NB, Canada
| |
Collapse
|
10
|
Wang J, Ge J, Jin L, Deng B, Tang W, Yu H, Zhang X, Liu X, Xue L, Zuo C, Chen X. Characterization of neuroinflammation pattern in anti-LGI1 encephalitis based on TSPO PET and symptom clustering analysis. Eur J Nucl Med Mol Imaging 2023; 50:2394-2408. [PMID: 36929211 DOI: 10.1007/s00259-023-06190-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/05/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE TSPO PET with radioligand [18F]DPA-714 is an emerging molecular imaging technique that reflects cerebral inflammation and microglial activation, and it has been recently used in central nervous system diseases. In this study, we aimed to investigate the neuroinflammation pattern of anti-leucine-rich glioma-inactivated 1 (LGI1) protein autoimmune encephalitis (AIE) and to evaluate its possible correlation with clinical phenotypes. METHODS Twenty patients with anti-LGI1 encephalitis from the autoimmune encephalitis cohort in Huashan Hospital and ten with other AIE and non-inflammatory diseases that underwent TSPO PET imaging were included in the current study. Increased regional [18F]DPA-714 retention in anti-LGI1 encephalitis was detected on a voxel basis using statistic parametric mapping analysis. Multiple correspondence analysis and hierarchical clustering were conducted for discriminate subgroups in anti-LGI1 encephalitis. Standardized uptake value ratios normalized to the cerebellum (SUVRc) were calculated for semiquantitative analysis of TSPO PET features between different LGI1-AIE subgroups. RESULTS Increased regional retention of [18F]DPA-714 was identified in the bilateral hippocampus, caudate nucleus, and frontal cortex in LGI1-AIE patients. Two subgroups of LGI1-AIE patients were distinguished based on the top seven common symptoms. Patients in cluster 1 had a high frequency of facio-brachial dystonic seizures than those in cluster 2 (p = 0.004), whereas patients in cluster 2 had a higher frequency of general tonic-clonic (GTC) seizures than those in cluster 1 (p < 0.001). Supplementary motor area and superior frontal gyrus showed higher [18F]DPA-714 retention in cluster 2 patients compared with those in cluster 1 (p = 0.024; p = 0.04, respectively). CONCLUSIONS Anti-LGI1 encephalitis had a distinctive molecular imaging pattern presented by TSPO PET scan. LGI1-AIE patients with higher retention of [18F]DPA-714 in the frontal cortex were more prone to present with GTC seizures. Further studies are required for verifying its value in clinical application.
Collapse
Affiliation(s)
- Jingguo Wang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Jingjie Ge
- Department of Nuclear Medicine/PET Center, Huashan Hospital, Fudan University, 518 East Wuzhong Road, Shanghai, 200235, China
| | - Lei Jin
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Bo Deng
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Weijun Tang
- Department of Radiology, Huashan Hospital, Shanghai, 200040, China
| | - Hai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Xiang Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Xiaoni Liu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Le Xue
- Department of Nuclear Medicine, the Second Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Chuantao Zuo
- Department of Nuclear Medicine/PET Center, Huashan Hospital, Fudan University, 518 East Wuzhong Road, Shanghai, 200235, China.
| | - Xiangjun Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
- National Center for Neurological Disorders, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
- Human Phenome Institute, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Gao J, Wang N, Zong F, Dong J, Lin Y, Zhang H, Zhang F. TIPE2 regulates the response of BV2 cells to lipopolysaccharide by the crosstalk between PI3K/AKT signaling and microglia M1/M2 polarization. Int Immunopharmacol 2023; 120:110389. [PMID: 37245300 DOI: 10.1016/j.intimp.2023.110389] [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: 02/16/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Tumor necrosis factor (TNF)-α-induced protein 8-like 2 (TIPE2) is a crucial negative regulator of both adaptive and innate immunity, which helps maintain the dynamic balance of the immune system by negatively regulating the signaling of T-cell receptors (TCR) and Toll-like receptors (TLR). In this study, we aimed to investigate the role and molecular mechanism of TIPE2 using a lipopolysaccharide (LPS)-induced inflammatory injury model in BV2 cells. Specifically, we constructed a BV2 cell line of TIPE2-overexpression or TIPE2-knockdown via lentiviral transfection. Our results demonstrated that overexpression of TIPE2 downregulated the expression of pro-inflammatory cytokines IL-1β and IL-6, which was reversed by knockdown of TIPE2 in the inflammation model of BV2 cells. In addition, overexpression of TIPE2 resulted in the conversion of BV2 cells to the M2 phenotype, while the knockdown of TIPE2 promoted the transformation of BV2 cells to the M1 phenotype. Notably, our co-culture experiments with neuronal cells SH-SY5Y showed that the overexpression of TIPE2 in inflammation-injured BV2 cells exhibited a protective effect on the neuronal cells. Finally, western blot analysis demonstrated that TIPE2 significantly reduced the expression of p-PI3K, p-AKT, p-p65, and p-IκBα in LPS treated BV2 cells, and inhibited the activation of NF-κB through the dephosphorylation of PI3K/AKT. These results suggest that TIPE2 plays an important role in mediating neuroinflammatory responses and may be involved in neuroprotection by modulating the phenotypic changes of BV2 cells and regulating the pro-inflammatory responses through the PI3K/AKT and NF-κB signaling pathways. In conclusion, our study provides new insights into the crucial role of TIPE2 in regulating neuroinflammatory responses and highlights its potential as a therapeutic target for neuroprotection.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Naidong Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Fangjiao Zong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Jiahao Dong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Yuanyuan Lin
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Hanting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| |
Collapse
|
12
|
Schneider JS. GM1 Ganglioside as a Disease-Modifying Therapeutic for Parkinson's Disease: A Multi-Functional Glycosphingolipid That Targets Multiple Parkinson's Disease-Relevant Pathogenic Mechanisms. Int J Mol Sci 2023; 24:9183. [PMID: 37298133 PMCID: PMC10252733 DOI: 10.3390/ijms24119183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder affecting millions of patients worldwide. Many therapeutics are available for treating PD symptoms but there is no disease-modifying therapeutic that has been unequivocally shown to slow or stop the progression of the disease. There are several factors contributing to the failure of many putative disease-modifying agents in clinical trials and these include the choice of patients and clinical trial designs for disease modification trials. Perhaps more important, however, is the choice of therapeutic, which for the most part, has not taken into account the multiple and complex pathogenic mechanisms and processes involved in PD. This paper discusses some of the factors contributing to the lack of success in PD disease-modification trials, which have mostly investigated therapeutics with a singular mechanism of action directed at one of the many PD pathogenic processes, and suggests that an alternative strategy for success may be to employ multi-functional therapeutics that target multiple PD-relevant pathogenic mechanisms. Evidence is presented that the multi-functional glycosphingolipid GM1 ganglioside may be just such a therapeutic.
Collapse
Affiliation(s)
- Jay S Schneider
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
13
|
Inhibition of Microglial GSK3β Activity Is Common to Different Kinds of Antidepressants: A Proposal for an In Vitro Screen to Detect Novel Antidepressant Principles. Biomedicines 2023; 11:biomedicines11030806. [PMID: 36979785 PMCID: PMC10045655 DOI: 10.3390/biomedicines11030806] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Depression is a major public health concern. Unfortunately, the present antidepressants often are insufficiently effective, whilst the discovery of more effective antidepressants has been extremely sluggish. The objective of this review was to combine the literature on depression with the pharmacology of antidepressant compounds, in order to formulate a conceivable pathophysiological process, allowing proposals how to accelerate the discovery process. Risk factors for depression initiate an infection-like inflammation in the brain that involves activation microglial Toll-like receptors and glycogen synthase kinase-3β (GSK3β). GSK3β activity alters the balance between two competing transcription factors, the pro-inflammatory/pro-oxidative transcription factor NFκB and the neuroprotective, anti-inflammatory and anti-oxidative transcription factor NRF2. The antidepressant activity of tricyclic antidepressants is assumed to involve activation of GS-coupled microglial receptors, raising intracellular cAMP levels and activation of protein kinase A (PKA). PKA and similar kinases inhibit the enzyme activity of GSK3β. Experimental antidepressant principles, including cannabinoid receptor-2 activation, opioid μ receptor agonists, 5HT2 agonists, valproate, ketamine and electrical stimulation of the Vagus nerve, all activate microglial pathways that result in GSK3β-inhibition. An in vitro screen for NRF2-activation in microglial cells with TLR-activated GSK3β activity, might therefore lead to the detection of totally novel antidepressant principles with, hopefully, an improved therapeutic efficacy.
Collapse
|
14
|
Dong-Chen X, Yong C, Yang X, Chen-Yu S, Li-Hua P. Signaling pathways in Parkinson's disease: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:73. [PMID: 36810524 PMCID: PMC9944326 DOI: 10.1038/s41392-023-01353-3] [Citation(s) in RCA: 148] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 01/16/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, and its treatment remains a big challenge. The pathogenesis of PD may be related to environmental and genetic factors, and exposure to toxins and gene mutations may be the beginning of brain lesions. The identified mechanisms of PD include α-synuclein aggregation, oxidative stress, ferroptosis, mitochondrial dysfunction, neuroinflammation, and gut dysbiosis. The interactions among these molecular mechanisms complicate the pathogenesis of PD and pose great challenges to drug development. At the same time, the diagnosis and detection of PD are also one of obstacles to the treatment of PD due to its long latency and complex mechanism. Most conventional therapeutic interventions for PD possess limited effects and have serious side effects, heightening the need to develop novel treatments for this disease. In this review, we systematically summarized the pathogenesis, especially the molecular mechanisms of PD, the classical research models, clinical diagnostic criteria, and the reported drug therapy strategies, as well as the newly reported drug candidates in clinical trials. We also shed light on the components derived from medicinal plants that are newly identified for their effects in PD treatment, with the expectation to provide the summary and outlook for developing the next generation of drugs and preparations for PD therapy.
Collapse
Affiliation(s)
- Xu Dong-Chen
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Chen Yong
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Xu Yang
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - ShenTu Chen-Yu
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Peng Li-Hua
- College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, P. R. China. .,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P. R. China.
| |
Collapse
|
15
|
Huang B, Hu G, Zong X, Yang S, He D, Gao X, Liu D. α-Cyperone protects dopaminergic neurons and inhibits neuroinflammation in LPS-induced Parkinson's disease rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Int Immunopharmacol 2023; 115:109698. [PMID: 36634417 DOI: 10.1016/j.intimp.2023.109698] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Our previous study showed that α-Cyperone inhibited the inflammatory response triggered by activated microglia and protected dopaminergic neuron in in vitro cell model of Parkinson's disease (PD). It is unclear the effect of α-Cyperone in animal models of PD. In this study, our results indicated that α-Cyperone ameliorated motor dysfunction, protected dopaminergic neurons, and inhibited the reduction of dopamine and its metabolites in lipopolysaccharide (LPS)-induced PD rat model. Moreover, α-Cyperone suppressed the activation of microglia and the expression of neuroinflammatory factor (TNF-α, IL-6, IL-1β, iNOS, COX-2 and ROS). Furthermore, the molecular mechanism research revealed that α-Cyperone inhibited neuroinflammation and oxidative stress to exert protective effect in microglia by activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Moreover, α-Cyperone upregulated the expression of antioxidant enzymes (GCLC, GCLM and NQO1) in microglia. In conclusion, our study demonstrates α-Cyperone alleviates dopaminergic neurodegeneration by inhibiting neuroinflammation and oxidative stress in LPS-induced PD rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bingxu Huang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Guiqiu Hu
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaofeng Zong
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuo Yang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dewei He
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Xiyu Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dianfeng Liu
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China.
| |
Collapse
|
16
|
Isenbrandt A, Coulombe K, Morissette M, Bourque M, Lamontagne-Proulx J, Di Paolo T, Soulet D. Three-Dimensional Analysis of Sex- and Gonadal Status- Dependent Microglial Activation in a Mouse Model of Parkinson's Disease. Pharmaceuticals (Basel) 2023; 16:152. [PMID: 37259303 PMCID: PMC9967417 DOI: 10.3390/ph16020152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/03/2025] Open
Abstract
Parkinson's disease (PD) is characterized by neurodegeneration and neuroinflammation. PD prevalence and incidence are higher in men than in women and modulation of gonadal hormones could have an impact on the disease course. This was investigated in male and female gonadectomized (GDX) and SHAM operated (SHAM) mice. Dutasteride (DUT), a 5α-reductase inhibitor, was administered to these mice for 10 days to modulate their gonadal sex hormones. On the fifth day of DUT treatment, mice received 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to model PD. We have previously shown in these mice the toxic effect of MPTP in SHAM and GDX males and in GDX females on dopamine markers and astrogliosis whereas SHAM females were protected by their female sex hormones. In SHAM males, DUT protected against MPTP toxicity. In the present study, microglial density and the number of doublets, representative of a microglial proliferation, were increased by the MPTP lesion only in male mice and prevented by DUT in SHAM males. A three-dimensional morphological microglial analysis showed that MPTP changed microglial morphology from quiescent to activated only in male mice and was not prevented by DUT. In conclusion, microgliosis can be modulated by sex hormone-dependent and independent factors in a mice model of PD.
Collapse
Affiliation(s)
- Amandine Isenbrandt
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Katherine Coulombe
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Jérôme Lamontagne-Proulx
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculté de Pharmacie, Pavillon Ferdinand-Vandry, Université Laval, 1050, Avenue de la Médecine, Québec, QC G1V 0A6, Canada
| |
Collapse
|
17
|
Augustin A, Guennec AL, Umamahesan C, Kendler‐Rhodes A, Tucker RM, Chekmeneva E, Takis P, Lewis M, Balasubramanian K, DeSouza N, Mullish BH, Taylor D, Ryan S, Whelan K, Ma Y, Ibrahim MAA, Bjarnason I, Hayee BH, Charlett A, Dobbs SM, Dobbs RJ, Weller C. Faecal metabolite deficit, gut inflammation and diet in Parkinson's disease: Integrative analysis indicates inflammatory response syndrome. Clin Transl Med 2023; 13:e1152. [PMID: 36588088 PMCID: PMC9806009 DOI: 10.1002/ctm2.1152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Gut-brain axis is widely implicated in the pathophysiology of Parkinson's disease (PD). We take an integrated approach to considering the gut as a target for disease-modifying intervention, using continuous measurements of disease facets irrespective of diagnostic divide. METHODS We characterised 77 participants with diagnosed-PD, 113 without, by dietary/exogenous substance intake, faecal metabolome, intestinal inflammation, serum cytokines/chemokines, clinical phenotype including colonic transit time. Complete-linkage hierarchical cluster analysis of metabolites discriminant for PD-status was performed. RESULTS Longer colonic transit was linked to deficits in faecal short-chain-fatty acids outside PD, to a 'tryptophan-containing metabolite cluster' overall. Phenotypic cluster analysis aggregated colonic transit with brady/hypokinesia, tremor, sleep disorder and dysosmia, each individually associated with tryptophan-cluster deficit. Overall, a faster pulse was associated with deficits in a metabolite cluster including benzoic acid and an imidazole-ring compound (anti-fungals) and vitamin B3 (anti-inflammatory) and with higher serum CCL20 (chemotactic for lymphocytes/dendritic cells towards mucosal epithelium). The faster pulse in PD was irrespective of postural hypotension. The benzoic acid-cluster deficit was linked to (well-recognised) lower caffeine and alcohol intakes, tryptophan-cluster deficit to higher maltose intake. Free-sugar intake was increased in PD, maltose intake being 63% higher (p = .001). Faecal calprotectin was 44% (95% CI 5%, 98%) greater in PD [p = .001, adjusted for proton-pump inhibitors (p = .001)], with 16% of PD-probands exceeding a cut-point for clinically significant inflammation compatible with inflammatory bowel disease. Higher maltose intake was associated with exceeding this calprotectin cut-point. CONCLUSIONS Emerging picture is of (i) clinical phenotype being described by deficits in microbial metabolites essential to gut health; (ii) intestinal inflammation; (iii) a systemic inflammatory response syndrome.
Collapse
Affiliation(s)
- Aisha Augustin
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- The Maudsley HospitalLondonUK
| | | | - Chianna Umamahesan
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- The Maudsley HospitalLondonUK
| | | | - Rosalind M. Tucker
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- The Maudsley HospitalLondonUK
| | - Elena Chekmeneva
- National Phenome CentreImperial College LondonLondonUK
- Section of Bioanalytical ChemistryImperial College LondonLondonUK
| | - Panteleimon Takis
- National Phenome CentreImperial College LondonLondonUK
- Section of Bioanalytical ChemistryImperial College LondonLondonUK
| | - Matthew Lewis
- National Phenome CentreImperial College LondonLondonUK
- Section of Bioanalytical ChemistryImperial College LondonLondonUK
| | | | | | - Benjamin H Mullish
- Department of MetabolismDigestion and ReproductionImperial College, LondonUK
| | - David Taylor
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- The Maudsley HospitalLondonUK
| | | | - Kevin Whelan
- Nutritional SciencesKing's College LondonLondonUK
| | - Yun Ma
- Institute of Liver StudiesKing's College HospitalLondonUK
| | | | | | | | - André Charlett
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- Statistics, Modelling and EconomicsUK Health Security AgencyLondonUK
| | - Sylvia M. Dobbs
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- GastroenterologyKing's College HospitalLondonUK
| | - R. John Dobbs
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
- GastroenterologyKing's College HospitalLondonUK
| | - Clive Weller
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| |
Collapse
|
18
|
The complex role of inflammation and gliotransmitters in Parkinson's disease. Neurobiol Dis 2023; 176:105940. [PMID: 36470499 PMCID: PMC10372760 DOI: 10.1016/j.nbd.2022.105940] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Our understanding of the role of innate and adaptive immune cell function in brain health and how it goes awry during aging and neurodegenerative diseases is still in its infancy. Inflammation and immunological dysfunction are common components of Parkinson's disease (PD), both in terms of motor and non-motor components of PD. In recent decades, the antiquated notion that the central nervous system (CNS) in disease states is an immune-privileged organ, has been debunked. The immune landscape in the CNS influences peripheral systems, and peripheral immunological changes can alter the CNS in health and disease. Identifying immune and inflammatory pathways that compromise neuronal health and survival is critical in designing innovative and effective strategies to limit their untoward effects on neuronal health.
Collapse
|
19
|
Gordon J, Lockard G, Monsour M, Alayli A, Choudhary H, Borlongan CV. Sequestration of Inflammation in Parkinson's Disease via Stem Cell Therapy. Int J Mol Sci 2022; 23:ijms231710138. [PMID: 36077534 PMCID: PMC9456021 DOI: 10.3390/ijms231710138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease is the second most common neurodegenerative disease. Insidious and progressive, this disorder is secondary to the gradual loss of dopaminergic signaling and worsening neuroinflammation, affecting patients’ motor capabilities. Gold standard treatment includes exogenous dopamine therapy in the form of levodopa–carbidopa, or surgical intervention with a deep brain stimulator to the subcortical basal ganglia. Unfortunately, these therapies may ironically exacerbate the already pro-inflammatory environment. An alternative approach may involve cell-based therapies. Cell-based therapies, whether endogenous or exogenous, often have anti-inflammatory properties. Alternative strategies, such as exercise and diet modifications, also appear to play a significant role in facilitating endogenous and exogenous stem cells to induce an anti-inflammatory response, and thus are of unique interest to neuroinflammatory conditions including Parkinson’s disease. Treating patients with current gold standard therapeutics and adding adjuvant stem cell therapy, alongside the aforementioned lifestyle modifications, may ideally sequester inflammation and thus halt neurodegeneration.
Collapse
Affiliation(s)
- Jonah Gordon
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Gavin Lockard
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Molly Monsour
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Adam Alayli
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Hassan Choudhary
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Correspondence:
| |
Collapse
|
20
|
Mehdi A, Lamiae B, Samira B, Ramchoun M, Abdelouahed K, Tamas F, Hicham B. Pomegranate ( Punica granatum L.) Attenuates Neuroinflammation Involved in Neurodegenerative Diseases. Foods 2022; 11:2570. [PMID: 36076756 PMCID: PMC9455244 DOI: 10.3390/foods11172570] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 12/03/2022] Open
Abstract
Food scientists have studied the many health benefits of polyphenols against pernicious human diseases. Evidence from scientific studies has shown that earlier healthy lifestyle changes, particularly in nutrition patterns, can reduce the burden of age-related diseases. In this context, a large number of plant-derived components belonging to the class of polyphenols have been reported to possess neuroprotective benefits. In this review, we examined studies on the effect of dietary polyphenols, notably from Punica granatum L., on neurodegenerative disease, including Alzheimer's disease, which is symptomatically characterized by impairment of cognitive functions. Clinical trials are in favor of the role of some polyphenols in maintaining neuronal homeostasis and attenuating clinical presentations of the disease. However, discrepancies in study design often bring inconsistent findings on the same component and display differences in their effectiveness due to interindividual variability, bioavailability in the body after administration, molecular structures, cross-blood-brain barrier, and signaling pathways such as nuclear factor kappa B (NF-κB). Based on preclinical and clinical trials, it appears that pomegranate may prove valuable in treating neurodegenerative disorders, including Alzheimer's disease (AD) and Parkinson's disease (PD). Therefore, due to the lack of information on human clinical trials, future in-depth studies, focusing on human beings, of several bioactive components of pomegranate's polyphenols and their synergic effects should be carried out to evaluate their curative treatment.
Collapse
Affiliation(s)
- Alami Mehdi
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23000, Morocco
| | - Benchagra Lamiae
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23000, Morocco
| | - Boulbaroud Samira
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23000, Morocco
| | - Mhamed Ramchoun
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23000, Morocco
| | - Khalil Abdelouahed
- Department of Medicine, Geriatrics Service, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Fulop Tamas
- Department of Medicine, Geriatrics Service, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Berrougui Hicham
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23000, Morocco
- Department of Medicine, Geriatrics Service, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| |
Collapse
|
21
|
Giri B, Seamon M, Banerjee A, Chauhan S, Purohit S, Morgan J, Baban B, Wakade C. Emerging urinary alpha-synuclein and miRNA biomarkers in Parkinson's disease. Metab Brain Dis 2022; 37:1687-1696. [PMID: 33881722 DOI: 10.1007/s11011-021-00735-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases after Alzheimer's disease (AD), afflicting adults above the age of sixty irrespective of gender, race, ethnicity, and social status. PD is characterized by motor dysfunctions, displaying resting tremor, rigidity, bradykinesia, and postural imbalance. Non-motor symptoms, including rapid eye movement (REM) behavior disorder, constipation, and loss of sense of smell, typically occur many years before the appearance of the PD motor symptoms that lead to a diagnosis. The loss of dopaminergic neurons in the substantia nigra, which leads to the motor symptoms seen in PD, is associated with the deposition of aggregated, misfolded α-Synuclein (α-Syn, SNCA) proteins forming Lewy Bodies. Additionally, dysregulation of miRNA (a short form of mRNA) may contribute to the developing pathophysiology in PD and other diseases such as cancer. Overexpression of α-Syn and miRNA in human samples has been found in PD, AD, and dementia. Therefore, evaluating these molecules in urine, present either in the free form or in association with extracellular vesicles of biological fluids, may lead to early biomarkers for clinical diagnosis. Collection of urine is non-invasive and thus beneficial, particularly in geriatric populations, for biomarker analysis. Considering the expression and function of α-Syn and miRNA, we predict that they can be used as early biomarkers in the diagnosis and prognosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Banabihari Giri
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA.
| | - Marissa Seamon
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
- Department of Neuroscience, Augusta University, Augusta, GA, USA
| | - Aditi Banerjee
- Brain Peds Division, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Sneha Chauhan
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA
| | - Sharad Purohit
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
| | - John Morgan
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Parkinson's Foundation Center of Excellence, Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Babak Baban
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation (CERSI), Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Chandramohan Wakade
- Department of Physical Therapy, College of Allied Health Sciences, Augusta University, 987 St. Sebastian Way, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, USA
- Department of Neuroscience, Augusta University, Augusta, GA, USA
- Parkinson's Foundation Center of Excellence, Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| |
Collapse
|
22
|
Liu C, She Y, Huang J, Liu Y, Li W, Zhang C, Zhang T, Yu L. HMGB1-NLRP3-P2X7R pathway participates in PM 2.5-induced hippocampal neuron impairment by regulating microglia activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113664. [PMID: 35605331 DOI: 10.1016/j.ecoenv.2022.113664] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 06/15/2023]
Abstract
Neuroinflammation is a key mechanism underlying the cognitive impairment induced by PM2.5, and activated microglia plays an important role in this process. However, the mechanisms by which activated microglia induced by PM2.5 impair hippocampal neurons have not been fully elucidated. In this study, we focused on the role of HMGB1-NLRP3-P2X7R pathway which mediated the microglia activation in hippocampal neurons impairment induced by PM2.5 using a co-culture model of microglia and hippocampal neurons. We found that PM2.5 resulted in activated microglia and HMGB1-NLRP3 inflammatory pathway, and elevated proinflammatory cytokines of IL-18 and IL-1β in a dose-dependent manner. Notably, we next utilized previously reported pharmacological inhibitors or siRNA for HMGB1 and found that they significantly inhibited the activation of downstream NLRP3 and MAPK pathways derived from PM2.5 exposure, and down-regulated IL-18 and IL-1β in microglia. Furthermore, we employed co-cultured hippocampal neurons and microglia and found that reducing HMGB1 significantly decreased neuron impairment, apoptosis related protein of cl-caspase3, synaptic damage, and neurotransmitter receptor of 5-HT2A, along with notably elevated presynaptic and postsynaptic proteins of SYP and PSD-95, as well as learning and memory related proteins of p-CREB and BDNF. The neuronal impairment induced by PM2.5 could not be prevented in the case of simultaneous employment of HMGB1 siRNA and NLRP3 agonist. After silencing NLRP3 alone in microglia, hippocampal neurons demonstrated decreased excessive autophagy and up-regulated synaptic protein of GAP43 as well as learning and memory related protein of NCAM1. Therefore, we further studied how hippocampal neurons affected microglia under PM2.5 exposure, Further investigation indicated that silencing HMGB1 could affect the activation of P2X7R and reduce the release of ATP from hippocampal neurons, thus protecting the interaction between microglia and hippocampal neurons. The present work suggests that regulation of HMGB1-NLRP3-P2X7R pathway can inhibit the microglia activation induced by PM2.5 to alleviate hippocampal neuron impairment and stabilize the microenvironment between microglia and neurons. This contributes to maintaining the normal function of hippocampal neurons and alleviating the cognitive impairment derived from PM2.5 exposure.
Collapse
Affiliation(s)
- Chong Liu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Yingjie She
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Jia Huang
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Yongping Liu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Wanwei Li
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Tianliang Zhang
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China.
| | - Li Yu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China.
| |
Collapse
|
23
|
Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022; 11:2091. [PMID: 35805174 PMCID: PMC9266143 DOI: 10.3390/cells11132091] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.
Collapse
Affiliation(s)
| | - Shelley B. Hooks
- Hooks Lab, Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
24
|
Sola P, Krishnamurthy PT, Kumari M, Byran G, Gangadharappa HV, Garikapati KK. Neuroprotective approaches to halt Parkinson's disease progression. Neurochem Int 2022; 158:105380. [PMID: 35718278 DOI: 10.1016/j.neuint.2022.105380] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
One of the most significant threats in Parkinson's disease (PD) is neurodegeneration. Neurodegeneration at both nigral as well as non-nigral regions of the brain is considered responsible for disease progression in PD. The key factors that initiate neurodegeneration are oxidative stress, neuroinflammation, mitochondrial complex-1 inhibition, and abnormal α-synuclein (SNCA) protein aggregations. Nigral neurodegeneration results in motor symptoms (tremor, bradykinesia, rigidity, shuffling gait, and postural instability) whereas; non-nigral neurodegeneration is responsible for non-motor symptoms (depression, cognitive dysfunctions, sleep disorders, hallucination, and psychosis). The available therapies for PD aim at increasing dopamine levels. The medications such as Monoamine oxidase B (MAO-B) inhibitors, catechol o-methyltransferase (COMT) inhibitors, Dopamine precursor (Levodopa), dopamine agonists, and dopamine reuptake inhibitors drastically improve the motor symptoms and quality of life only in the early stages of the disease. However, dopa resistant motor symptoms (abnormality in posture, speech impediment, gait, and balance problems), dopa resistant non-motor signs (sleep problems, autonomic dysfunction, mood, and cognitive impairment, pain), and drug-related side effects (motor fluctuations, psychosis, and dyskinesias) are considered responsible for the failure of these therapies. Further, none of the treatments, alone or in combination, are capable of halting the disease progression in the long run. Therefore, there is a need to develop safe and efficient neuroprotective agents, which can slow or stop the disease progression for the better management of PD. In this review, an effort has been made to discuss the various mechanisms responsible for progressive neurodegeneration (disease progression) in PD and also multiple strategies available for halting disease progression.
Collapse
Affiliation(s)
- Piyong Sola
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India.
| | - Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Gowramma Byran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | | | - Kusuma Kumari Garikapati
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| |
Collapse
|
25
|
Grotemeyer A, McFleder RL, Wu J, Wischhusen J, Ip CW. Neuroinflammation in Parkinson's Disease - Putative Pathomechanisms and Targets for Disease-Modification. Front Immunol 2022; 13:878771. [PMID: 35663989 PMCID: PMC9158130 DOI: 10.3389/fimmu.2022.878771] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive and debilitating chronic disease that affects more than six million people worldwide, with rising prevalence. The hallmarks of PD are motor deficits, the spreading of pathological α-synuclein clusters in the central nervous system, and neuroinflammatory processes. PD is treated symptomatically, as no causally-acting drug or procedure has been successfully established for clinical use. Various pathways contributing to dopaminergic neuron loss in PD have been investigated and described to interact with the innate and adaptive immune system. We discuss the possible contribution of interconnected pathways related to the immune response, focusing on the pathophysiology and neurodegeneration of PD. In addition, we provide an overview of clinical trials targeting neuroinflammation in PD.
Collapse
Affiliation(s)
| | | | - Jingjing Wu
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Jörg Wischhusen
- Section for Experimental Tumor Immunology, Department of Obstetrics and Gynecology, University Hospital of Würzburg, Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Smajić S, Prada-Medina CA, Landoulsi Z, Ghelfi J, Delcambre S, Dietrich C, Jarazo J, Henck J, Balachandran S, Pachchek S, Morris CM, Antony P, Timmermann B, Sauer S, Pereira SL, Schwamborn JC, May P, Grünewald A, Spielmann M. Single-cell sequencing of human midbrain reveals glial activation and a Parkinson-specific neuronal state. Brain 2022; 145:964-978. [PMID: 34919646 PMCID: PMC9050543 DOI: 10.1093/brain/awab446] [Citation(s) in RCA: 264] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/21/2021] [Accepted: 11/18/2021] [Indexed: 11/29/2022] Open
Abstract
Idiopathic Parkinson's disease is characterized by a progressive loss of dopaminergic neurons, but the exact disease aetiology remains largely unknown. To date, Parkinson's disease research has mainly focused on nigral dopaminergic neurons, although recent studies suggest disease-related changes also in non-neuronal cells and in midbrain regions beyond the substantia nigra. While there is some evidence for glial involvement in Parkinson's disease, the molecular mechanisms remain poorly understood. The aim of this study was to characterize the contribution of all cell types of the midbrain to Parkinson's disease pathology by single-nuclei RNA sequencing and to assess the cell type-specific risk for Parkinson's disease using the latest genome-wide association study. We profiled >41 000 single-nuclei transcriptomes of post-mortem midbrain from six idiopathic Parkinson's disease patients and five age-/sex-matched controls. To validate our findings in a spatial context, we utilized immunolabelling of the same tissues. Moreover, we analysed Parkinson's disease-associated risk enrichment in genes with cell type-specific expression patterns. We discovered a neuronal cell cluster characterized by CADPS2 overexpression and low TH levels, which was exclusively present in idiopathic Parkinson's disease midbrains. Validation analyses in laser-microdissected neurons suggest that this cluster represents dysfunctional dopaminergic neurons. With regard to glial cells, we observed an increase in nigral microglia in Parkinson's disease patients. Moreover, nigral idiopathic Parkinson's disease microglia were more amoeboid, indicating an activated state. We also discovered a reduction in idiopathic Parkinson's disease oligodendrocyte numbers with the remaining cells being characterized by a stress-induced upregulation of S100B. Parkinson's disease risk variants were associated with glia- and neuron-specific gene expression patterns in idiopathic Parkinson's disease cases. Furthermore, astrocytes and microglia presented idiopathic Parkinson's disease-specific cell proliferation and dysregulation of genes related to unfolded protein response and cytokine signalling. While reactive patient astrocytes showed CD44 overexpression, idiopathic Parkinson's disease microglia revealed a pro-inflammatory trajectory characterized by elevated levels of IL1B, GPNMB and HSP90AA1. Taken together, we generated the first single-nuclei RNA sequencing dataset from the idiopathic Parkinson's disease midbrain, which highlights a disease-specific neuronal cell cluster as well as 'pan-glial' activation as a central mechanism in the pathology of the movement disorder. This finding warrants further research into inflammatory signalling and immunomodulatory treatments in Parkinson's disease.
Collapse
Affiliation(s)
- Semra Smajić
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | | | - Zied Landoulsi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Jenny Ghelfi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Carola Dietrich
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | - Javier Jarazo
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- OrganoTherapeutics SARL-S, L-4362 Esch-sur-Alzette, Luxembourg
| | - Jana Henck
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | | | - Sinthuja Pachchek
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Christopher M. Morris
- Newcastle Brain Tissue Resource, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, NE1 7RU Newcastle upon Tyne, UK
| | - Paul Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Bernd Timmermann
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
| | - Sascha Sauer
- Max-Delbrück-Centrum für Molekulare Medizin, Genomics Group, D-13125 Berlin, Germany
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- OrganoTherapeutics SARL-S, L-4362 Esch-sur-Alzette, Luxembourg
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, D-23562 Lübeck, Germany
| | - Malte Spielmann
- Max Planck Institute for Molecular Genetics, D-14195 Berlin, Germany
- Institute of Human Genetics, Kiel University, D-42118 Kiel, Germany
- Institute of Human Genetics, University of Lübeck, D-23562 Lübeck, Germany
| |
Collapse
|
27
|
Mabrouk R. Principal Component Analysis versus Subject’s Residual Profile Analysis for Neuroinflammation Investigation in Parkinson Patients: A PET Brain Imaging Study. J Imaging 2022; 8:jimaging8030056. [PMID: 35324611 PMCID: PMC8954189 DOI: 10.3390/jimaging8030056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 12/10/2022] Open
Abstract
Dysfunction of neurons in the central nervous system is the primary pathological feature of Parkinson’s disease (PD). Despite different triggering, emerging evidence indicates that neuroinflammation revealed through microglia activation is critical for PD. Moreover, recent investigations sought a potential relationship between Lrrk2 genetic mutation and microglia activation. In this paper, neuroinflammation in sporadic PD, Lrrk2-PD and unaffected Lrrk2 mutation carriers were investigated. The principal component analysis (PCA) and the subject’s residual profile (SRP) techniques were performed on multiple groups and regions of interest in 22 brain-regions. The 11C-PBR28 binding profiles were compared in four genotypes depending on groups, i.e., HC, sPD, Lrrk2-PD and UC, using the PCA and SPR scores. The genotype effect was found as a principal feature of group-dependent 11C-PBR28 binding, and preliminary evidence of a MAB-Lrrk2 mutation interaction in manifest Parkinson’s and subjects at risk was found.
Collapse
Affiliation(s)
- Rostom Mabrouk
- Department of Computer Science, Bishop's University, Sherbrooke, QC J1M 1Z7, Canada
| |
Collapse
|
28
|
Zhao Y, Zhang Y, Zhang J, Yang G. Plasma proteome profiling using tandem mass tag labeling technology reveals potential biomarkers for Parkinson's disease: a preliminary study. Proteomics Clin Appl 2021; 16:e2100010. [PMID: 34791804 DOI: 10.1002/prca.202100010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 11/08/2022]
Abstract
PURPOSE Parkinson's disease (PD) is the second most frequently occurring progressive neurodegenerative disorder. Biomarkers are useful indicators for tracking disease progression, early diagnosis, and intervention of disease progression. We aimed to develop plasma biomarker panel which maybe aid to predict the onset and progression of PD. EXPERIMENTAL DESIGN Tandem mass tag (TMT) mass spectrometry was applied using an Orbitrap Lumos mass spectrometer to analyze plasma protein expression in patients diagnosed with PD and healthy controls. RESULTS In total, 555 proteins were quantified. Using a cut-off of p < 0.05 and a fold change of >1.2 for the variation in expression, 25 proteins were differentially expressed between the PD and control groups. Sixteen proteins were upregulated and nine were downregulated. Several proteins, including Chitinase-3-like protein 1 (CHI3L1) and thymosin beta-4 (TMSB4X) were implicated in PD pathogenesis. CONCLUSIONS The data from the TMT-based proteomic profiling of plasma samples in PD may help advance the understanding of the molecular mechanisms of PD and identify potential novel biomarkers of PD for further characterization.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Yidan Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Jian Zhang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Guofeng Yang
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| |
Collapse
|
29
|
McGregor BA, Schommer J, Guo K, Raihan MO, Ghribi O, Hur J, Porter JE. Alpha-Synuclein-induced DNA Methylation and Gene Expression in Microglia. Neuroscience 2021; 468:186-198. [PMID: 34082066 PMCID: PMC12037068 DOI: 10.1016/j.neuroscience.2021.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022]
Abstract
Synucleinopathy disorders are characterized by aggregates of α-synuclein (α-syn), which engage microglia to elicit a neuroinflammatory response. Here, we determined the gene expression and DNA methylation changes in microglia induced by aggregate α-syn. Transgenic murine Thy-1 promoter (mThy1)-Asyn mice overexpressing human α-syn are a model of synucleinopathy. Microglia from 3 and 13-month-old mice were used to isolate nucleic acids for methylated DNA and RNA-sequencing. α-Syn-regulated changes in gene expression and genomic methylation were determined and examined for functional enrichment followed by network analysis to further elucidate possible connections within the data. Microglial DNA isolated from our 3-month cohort had 5315 differentially methylated gene (DMG) changes, while RNA levels demonstrated a change in 119 differentially expressed genes (DEGs) between mThy1-Asyn mice and wild-type littermate controls. The 3-month DEGs and DMGs were highly associated with adhesion and migration signaling, suggesting a phenotypic transition from resting to active microglia. We observed 3742 DMGs and 3766 DEGs in 13-month mThy1-Asyn mice. These genes were often related to adhesion, migration, cell cycle, cellular metabolism, and immune response. Network analysis also showed increased cell mobility and inflammatory functions at 3 months, shifting to cell cycle, immune response, and metabolism changes at 13 months. We observed significant α-syn-induced methylation and gene expression changes in microglia. Our data suggest that α-syn overexpression initiates microglial activation leading to neuroinflammation and cellular metabolic stresses, which is associated with disease progression.
Collapse
Affiliation(s)
- Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Jared Schommer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Md Obayed Raihan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| |
Collapse
|
30
|
Microglia in Neurodegenerative Events-An Initiator or a Significant Other? Int J Mol Sci 2021; 22:ijms22115818. [PMID: 34072307 PMCID: PMC8199265 DOI: 10.3390/ijms22115818] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
A change in microglia structure, signaling, or function is commonly associated with neurodegeneration. This is evident in the patient population, animal models, and targeted in vitro assays. While there is a clear association, it is not evident that microglia serve as an initiator of neurodegeneration. Rather, the dynamics imply a close interaction between the various cell types and structures in the brain that orchestrate the injury and repair responses. Communication between microglia and neurons contributes to the physiological phenotype of microglia maintaining cells in a surveillance state and allows the cells to respond to events occurring in their environment. Interactions between microglia and astrocytes is not as well characterized, nor are interactions with other members of the neurovascular unit; however, given the influence of systemic factors on neuroinflammation and disease progression, such interactions likely represent significant contributes to any neurodegenerative process. In addition, they offer multiple target sites/processes by which environmental exposures could contribute to neurodegenerative disease. Thus, microglia at least play a role as a significant other with an equal partnership; however, claiming a role as an initiator of neurodegeneration remains somewhat controversial.
Collapse
|
31
|
Microglial Extracellular Vesicles as Vehicles for Neurodegeneration Spreading. Biomolecules 2021; 11:biom11060770. [PMID: 34063832 PMCID: PMC8224033 DOI: 10.3390/biom11060770] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Microglial cells are the neuroimmune competent cells of the central nervous system. In the adult, microglia are responsible for screening the neuronal parenchyma searching for alterations in homeostasis. Chronic neuroinflammation plays a role in neurodegenerative disease. Indeed, microglia-mediated neuroinflammation is involved in the onset and progression of several disorders in the brain and retina. Microglial cell reactivity occurs in an orchestrated manner and propagates across the neural parenchyma spreading the neuroinflammatory signal from cell to cell. Extracellular vesicles are important vehicles of intercellular communication and act as message carriers across boundaries. Extracellular vesicles can be subdivided in several categories according to their cellular origin (apoptotic bodies, microvesicles and exosomes), each presenting, different but sometimes overlapping functions in cell communication. Mounting evidence suggests a role for extracellular vesicles in regulating microglial cell action. Herein, we explore the role of microglial extracellular vesicles as vehicles for cell communication and the mechanisms that trigger their release. In this review we covered the role of microglial extracellular vesicles, focusing on apoptotic bodies, microvesicles and exosomes, in the context of neurodegeneration and the impact of these vesicles derived from other cells in microglial cell reactivity.
Collapse
|
32
|
Behl T, Kaur G, Sehgal A, Bhardwaj S, Singh S, Buhas C, Judea-Pusta C, Uivarosan D, Munteanu MA, Bungau S. Multifaceted Role of Matrix Metalloproteinases in Neurodegenerative Diseases: Pathophysiological and Therapeutic Perspectives. Int J Mol Sci 2021; 22:ijms22031413. [PMID: 33573368 PMCID: PMC7866808 DOI: 10.3390/ijms22031413] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegeneration is the pathological condition, in which the nervous system or neuron loses its structure, function, or both, leading to progressive degeneration or the death of neurons, and well-defined associations of tissue system, resulting in clinical manifestations. Neuroinflammation has been shown to precede neurodegeneration in several neurodegenerative diseases (NDs). No drug is yet known to delay or treat neurodegeneration. Although the etiology and potential causes of NDs remain widely indefinable, matrix metalloproteinases (MMPs) evidently have a crucial role in the progression of NDs. MMPs, a protein family of zinc (Zn2+)-containing endopeptidases, are pivotal agents that are involved in various biological and pathological processes in the central nervous system (CNS). The current review delineates the several emerging evidence demonstrating the effects of MMPs in the progression of NDs, wherein they regulate several processes, such as (neuro)inflammation, microglial activation, amyloid peptide degradation, blood brain barrier (BBB) disruption, dopaminergic apoptosis, and α-synuclein modulation, leading to neurotoxicity and neuron death. Published papers to date were searched via PubMed, MEDLINE, etc., while using selective keywords highlighted in our manuscript. We also aim to shed a light on pathophysiological effect of MMPs in the CNS and focus our attention on its detrimental and beneficial effects in NDs, with a special focus on Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), multiple sclerosis (MS), and Huntington's disease (HD), and discussed various therapeutic strategies targeting MMPs, which could serve as potential modulators in NDs. Over time, several agents have been developed in order to overcome challenges and open up the possibilities for making selective modulators of MMPs to decipher the multifaceted functions of MMPs in NDs. There is still a greater need to explore them in clinics.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| | - Gagandeep Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
| | - Shaveta Bhardwaj
- Department of Pharmacology, GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana 141104, Punjab, India;
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh 140401, Punjab, India; (G.K.); (A.S.); (S.S.)
| | - Camelia Buhas
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (C.J.-P.)
| | - Claudia Judea-Pusta
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (C.J.-P.)
| | - Diana Uivarosan
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Mihai Alexandru Munteanu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| |
Collapse
|
33
|
Guo JJ, Yue F, Song DY, Bousset L, Liang X, Tang J, Yuan L, Li W, Melki R, Tang Y, Chan P, Guo C, Li JY. Intranasal administration of α-synuclein preformed fibrils triggers microglial iron deposition in the substantia nigra of Macaca fascicularis. Cell Death Dis 2021; 12:81. [PMID: 33441545 PMCID: PMC7807015 DOI: 10.1038/s41419-020-03369-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
Iron deposition is present in main lesion areas in the brains of patients with Parkinson's disease (PD) and an abnormal iron content may be associated with dopaminergic neuronal cytotoxicity and degeneration in the substantia nigra of the midbrain. However, the cause of iron deposition and its role in the pathological process of PD are unclear. In the present study, we investigated the effects of the nasal mucosal delivery of synthetic human α-synuclein (α-syn) preformed fibrils (PFFs) on the pathogenesis of PD in Macaca fascicularis. We detected that iron deposition was clearly increased in a time-dependent manner from 1 to 17 months in the substantia nigra and globus pallidus, highly contrasting to other brain regions after treatments with α-syn PFFs. At the cellular level, the iron deposits were specifically localized in microglia but not in dopaminergic neurons, nor in other types of glial cells in the substantia nigra, whereas the expression of transferrin (TF), TF receptor 1 (TFR1), TF receptor 2 (TFR2), and ferroportin (FPn) was increased in dopaminergic neurons. Furthermore, no clear dopaminergic neuron loss was observed in the substantia nigra, but with decreased immunoreactivity of tyrosine hydroxylase (TH) and appearance of axonal swelling in the putamen. The brain region-enriched and cell-type-dependent iron localizations indicate that the intranasal α-syn PFFs treatment-induced iron depositions in microglia in the substantia nigra may appear as an early cellular response that may initiate neuroinflammation in the dopaminergic system before cell death occurs. Our data suggest that the inhibition of iron deposition may be a potential approach for the early prevention and treatment of PD.
Collapse
Affiliation(s)
- Jian-Jun Guo
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Feng Yue
- Beijing Key Laboratory of Parkinson's Disease, National Clinical Research Center for Geriatric Disorders, Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Dong-Yan Song
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Luc Bousset
- Laboratory of Neurodegenerative Diseases, CNRS and Institut François Jacob (MIRCen), CEA, Fontenay-aux-Roses, 92260, France
| | - Xin Liang
- Department of Histology, Chongqing Medical University, Chongqing, 400000, China
| | - Jing Tang
- Department of Histology, Chongqing Medical University, Chongqing, 400000, China
| | - Lin Yuan
- Unit of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, 110112, China
| | - Wen Li
- Unit of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, 110112, China
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, Lund, 22184, Sweden
| | - Ronald Melki
- Laboratory of Neurodegenerative Diseases, CNRS and Institut François Jacob (MIRCen), CEA, Fontenay-aux-Roses, 92260, France
| | - Yong Tang
- Department of Histology, Chongqing Medical University, Chongqing, 400000, China
| | - Piu Chan
- Beijing Key Laboratory of Parkinson's Disease, National Clinical Research Center for Geriatric Disorders, Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Chuang Guo
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
- Unit of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, 110112, China.
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, Lund, 22184, Sweden.
| |
Collapse
|
34
|
Fardell C, Schiöler L, Nissbrandt H, Torén K, Åberg M. The erythrocyte sedimentation rate in male adolescents and subsequent risk of Parkinson's disease: an observational study. J Neurol 2020; 268:1508-1516. [PMID: 33277665 PMCID: PMC7990830 DOI: 10.1007/s00415-020-10324-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
Systemic inflammation may be implicated in the pathophysiology of Parkinson’s disease (PD). Since PD occurs usually in later life, most studies of causal factors are conducted in older populations, so potentially important influences from early life cannot be adequately captured. We investigated whether the erythrocyte sedimentation rate (ESR) in early adulthood is associated with the subsequent development of PD in men. As part of Swedish national conscription testing conducted from 1968 through 1983 (N = 716,550), the erythrocyte sedimentation rate, as a measure of inflammation, was measured in 659,278 young men. The cohort was observed for subsequent PD events (N = 1513) through December 2016. Cox proportional hazards models were used to estimate the hazard ratios (HR) with 95% CI with adjustment for potential confounders. Individuals with higher ESRs were significantly less likely to be diagnosed with PD, as ESR was linearly and inversely associated with PD risk. The magnitude of the association between ESR and PD risk was similar for increases up to 15 mm/h, leveled off thereafter, and was non-significant for ESR values > 20 mm/h. The HR for PD with basic adjustments (age at conscription, year of conscription, test center and erythrocyte volume fraction) was 0.94 (95% CI 0.89–0.99, P = 0.02) per log2 increase in ESR, corresponding to a two-fold increase in ESR. Further adjustments for potential confounders (parental education, systolic and diastolic blood pressures, and IQ) scarcely altered the HR. The results suggest a prospective association between high ESR and reduced risk for PD.
Collapse
Affiliation(s)
- Camilla Fardell
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linus Schiöler
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hans Nissbrandt
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kjell Torén
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Occupational and Environmental Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Maria Åberg
- School of Public Health and Community Medicine/Primary Health Care, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Box 454, 405 30, Gothenburg, Sweden. .,Region Västra Götaland, Regionhälsan, Gothenburg, Sweden.
| |
Collapse
|
35
|
Thadathil N, Xiao J, Hori R, Alway SE, Khan MM. Brain Selective Estrogen Treatment Protects Dopaminergic Neurons and Preserves Behavioral Function in MPTP-induced Mouse Model of Parkinson's Disease. J Neuroimmune Pharmacol 2020; 16:667-678. [PMID: 33221984 DOI: 10.1007/s11481-020-09972-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons in the substantia nigra and loss of both motor and non-motor features. Several clinical and preclinical studies have provided evidence that estrogen therapy reduces the risk of PD but have limitations in terms of adverse peripheral effects. Therefore, we examined the potential beneficial effects of the brain-selective estrogen prodrug, 10β, 17β-dihydroxyestra-1,4-dien-3-one (DHED) on nigrostriatal dopaminergic neurodegeneration and behavioral abnormalities in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Wild-type mice were treated with daily subcutaneous injections of DHED (50 and 100 µg/kg) or vehicle for four weeks. To produce PD-like symptoms, mice were injected with MPTP (18 mg/kg in saline; intraperitoneally) four times at 2-hr intervals for one day. After behavioral examination, mice were sacrificed, and the brains were isolated for neurochemical and morphological examinations. MPTP injected mice exhibited loss of dopaminergic neurons and fibers in substantia nigra and striatum respectively, along with impaired motor function at day 7 post MPTP injection. These phenotypes were associated with significantly increased oxidative stress and inflammatory responses in the striatum regions. DHED treatments significantly mitigated behavioral impairments and dopaminergic neurodegeneration induced by MPTP. We further observed that DHED treatment suppressed oxidative stress and inflammation in the striatum of MPTP treated mice when compared to vehicle treated mice. In conclusions, our findings suggest that DHED protects dopaminergic neurons from MPTP toxicity in mouse model of PD and support a beneficial effect of brain-selective estrogen in attenuating neurodegeneration and motor symptoms in PD-related neurological disorders. Graphical Abstract.
Collapse
Affiliation(s)
- Nidheesh Thadathil
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - Jianfeng Xiao
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - Roderick Hori
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Stephen E Alway
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA.
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
36
|
Lavisse S, Goutal S, Wimberley C, Tonietto M, Bottlaender M, Gervais P, Kuhnast B, Peyronneau MA, Barret O, Lagarde J, Sarazin M, Hantraye P, Thiriez C, Remy P. Increased microglial activation in patients with Parkinson disease using [ 18F]-DPA714 TSPO PET imaging. Parkinsonism Relat Disord 2020; 82:29-36. [PMID: 33242662 DOI: 10.1016/j.parkreldis.2020.11.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/18/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Increasing evidence suggests that neuroinflammation is active in Parkinson disease (PD) and contributes to neurodegeneration. This process can be studied in vivo with PET and radioligands targeting TSPO, upregulated in activated microglia. Initial PET studies investigating microglial activation in PD with the [11C]-PK11195 have provided inconclusive results. Here we assess the presence and distribution of neuroinflammatory response in PD patients using [18F]-DPA714 and to correlate imaging biomarkers to dopamine transporter imaging and clinical status. METHODS PD patients (n = 24, Hoehn and Yahr I-III) and 28 healthy controls were scanned with [18F]-DPA714 and [11C]-PE2I and analyzed. They were all genotyped for TSPO polymorphism. Regional binding parameters were estimated (reference Logan graphical approach with supervised cluster analysis). Impact of TSPO genotype was analyzed using Wilcoxon signed-rank test. Differences between groups were investigated using a two-way ANOVA and Tukey post hoc tests. RESULTS PD patients showed significantly higher [18F]-DPA714 binding compared to healthy controls bilaterally in the midbrain (p < 0.001), the frontal cortex (p = 0.001), and the putamen contralateral to the more clinically affected hemibody (p = 0.038). Microglial activation in these regions did not correlate with the severity of motor symptoms, disease duration nor putaminal [11C]-PE2I uptake. However, there was a trend toward a correlation between cortical TSPO binding and disease duration (p = 0.015 uncorrected, p = 0.07 after Bonferroni correction). CONCLUSION [18F]-DPA714 binding confirmed that there is a specific topographic pattern of microglial activation in the nigro-striatal pathway and the frontal cortex of PD patients. TRIAL REGISTRATION Trial registration: INFLAPARK, NCT02319382. Registered 18 December 2014- Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT02319382.
Collapse
Affiliation(s)
- Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Sébastien Goutal
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Catriona Wimberley
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Mattéo Tonietto
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Michel Bottlaender
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, 91191, Gif-sur-Yvette, France.
| | - Philippe Gervais
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Bertrand Kuhnast
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Marie-Anne Peyronneau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France.
| | - Olivier Barret
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Julien Lagarde
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France; Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Sainte-Anne Hospital, Paris, 75014, France; Université de Paris, F-75006, France.
| | - Marie Sarazin
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4, Place du Général Leclerc, Orsay, 91401, France; Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Sainte-Anne Hospital, Paris, 75014, France; Université de Paris, F-75006, France.
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Claire Thiriez
- Centre Expert Parkinson, Neurologie, CHU Henri Mondor, AP-HP, 51 Avenue du Maréchal de Lattre de Tassigny, Créteil, France.
| | - Philippe Remy
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France; Centre Expert Parkinson, Neurologie, CHU Henri Mondor, AP-HP, 51 Avenue du Maréchal de Lattre de Tassigny, Créteil, France; IMRB, INSERM, Université Paris Est Créteil and NeurATRIS, France.
| |
Collapse
|
37
|
Boi L, Pisanu A, Palmas MF, Fusco G, Carboni E, Casu MA, Satta V, Scherma M, Janda E, Mocci I, Mulas G, Ena A, Spiga S, Fadda P, De Simone A, Carta AR. Modeling Parkinson's Disease Neuropathology and Symptoms by Intranigral Inoculation of Preformed Human α-Synuclein Oligomers. Int J Mol Sci 2020; 21:E8535. [PMID: 33198335 PMCID: PMC7696693 DOI: 10.3390/ijms21228535] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
The accumulation of aggregated α-synuclein (αSyn) is a hallmark of Parkinson's disease (PD). Current evidence indicates that small soluble αSyn oligomers (αSynOs) are the most toxic species among the forms of αSyn aggregates, and that size and topological structural properties are crucial factors for αSynOs-mediated toxicity, involving the interaction with either neurons or glial cells. We previously characterized a human αSynO (H-αSynO) with specific structural properties promoting toxicity against neuronal membranes. Here, we tested the neurotoxic potential of these H-αSynOs in vivo, in relation to the neuropathological and symptomatic features of PD. The H-αSynOs were unilaterally infused into the rat substantia nigra pars compacta (SNpc). Phosphorylated αSyn (p129-αSyn), reactive microglia, and cytokine levels were measured at progressive time points. Additionally, a phagocytosis assay in vitro was performed after microglia pre-exposure to αsynOs. Dopaminergic loss, motor, and cognitive performances were assessed. H-αSynOs triggered p129-αSyn deposition in SNpc neurons and microglia and spread to the striatum. Early and persistent neuroinflammatory responses were induced in the SNpc. In vitro, H-αSynOs inhibited the phagocytic function of microglia. H-αsynOs-infused rats displayed early mitochondrial loss and abnormalities in SNpc neurons, followed by a gradual nigrostriatal dopaminergic loss, associated with motor and cognitive impairment. The intracerebral inoculation of structurally characterized H-αSynOs provides a model of progressive PD neuropathology in rats, which will be helpful for testing neuroprotective therapies.
Collapse
Affiliation(s)
- Laura Boi
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | | | - Maria Francesca Palmas
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, UK;
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Maria Antonietta Casu
- CNR Institute of Translational Pharmacology, 09010 Cagliari, Italy; (M.A.C.); (I.M.)
| | - Valentina Satta
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Maria Scherma
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Ignazia Mocci
- CNR Institute of Translational Pharmacology, 09010 Cagliari, Italy; (M.A.C.); (I.M.)
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, 09126 Cagliari, Italy; (G.M.); (S.S.)
| | - Anna Ena
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, 09126 Cagliari, Italy; (G.M.); (S.S.)
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
- CNR Institute of Neuroscience, 09042 Cagliari, Italy;
- Italian Neuroscience Institute (INN), 10126 Torino, Italy
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
- Italian Neuroscience Institute (INN), 10126 Torino, Italy
| |
Collapse
|
38
|
Zhang M, He Q, Chen G, Li PA. Suppression of NLRP3 Inflammasome, Pyroptosis, and Cell Death by NIM811 in Rotenone-Exposed Cells as an in vitro Model of Parkinson's Disease. NEURODEGENER DIS 2020; 20:73-83. [PMID: 33176317 DOI: 10.1159/000511207] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/28/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by the selective death of dopaminergic neurons in the substantia nigra. Recently, NLRP3 inflammasome and pyroptosis were found to be associated with PD. Cyclosporine A (CsA), an immunosuppressant, reduces neuronal death in PD. However, CsA could hardly pass through the blood-brain barrier (BBB) and high dose is associated with severe side effects and toxicity. N-methyl-4-isoleucine-cyclosporine (NIM811) is a CsA derivate that can pass through the BBB. However, little is known about its effect on PD. OBJECTIVE The objectives of this study were to explore the mechanism of rotenone-induced cell damage and to examine the protective effects of NIM811 on the neurotoxicity of a Parkinson-like in vitro model induced by rotenone. METHODS Murine hippocampal HT22 cells were cultured with the mitochondrial complex I inhibitor rotenone, a widely used pesticide that has been used for many years as a tool to induce a PD model in vitro and in vivo and proven to be reproducible. NIM811 was added to the culture media 3 h prior to the rotenone incubation. Cell viability was determined by resazurin assay, reactive oxygen species (ROS) production by dihydroethidine (DHE), and mitochondrial membrane potential by tetramethyl rhodamine methyl ester (TMRM). TUNEL and caspase-1 immunofluorescent double staining was used to detect pyroptosis. NLRP3, caspase-1, pro-caspase-1, GSDMD, and interleukin-18 (IL-18) were measured using Western blotting after 24 h of rotenone incubation. The reactivity of interleukin-1β (IL-1β) was determined by ELISA. RESULTS Our results demonstrated that rotenone caused more than 40% of cell death, increased ROS production, and reduced mitochondrial membrane potential, while NIM811 reversed these alterations. Immunofluorescent double staining showed that rotenone increased the percentage of caspase-1 and TUNEL double-labelled cells, an indication of pyroptosis, after 24 h of incubation. The protein expression of NLRP3, caspase-1, pro-caspase-1, GSDMD, IL-18, and IL-1β was significantly increased after 24 h of rotenone incubation. NIM811 suppressed rotenone-induced pyroptosis and downregulated the protein expression of NLRP3, caspase-1, pro-caspase-1, GSDMD, IL-1β, and IL-18. CONCLUSION These results provide evidence that rotenone activates the NLRP3 inflammomere and induces pyroptosis. NIM811 protects the cell from rotenone-induced damage and inhibits NLRP3 inflammasome and pyroptosis. NIM811 might serve as a potential therapeutic drug in the treatment of PD.
Collapse
Affiliation(s)
- Minghao Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA.,Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Guisheng Chen
- Department of Neurology, Neuroscience Center, General Hospital of Ningxia Medical University, Key Laboratory of Craniocerebral Diseases, Yinchuan, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), College of Arts and Sciences, North Carolina Central University, Durham, North Carolina, USA,
| |
Collapse
|
39
|
Lv R, Du L, Zhou F, Yuan X, Liu X, Zhang L. Rosmarinic Acid Alleviates Inflammation, Apoptosis, and Oxidative Stress through Regulating miR-155-5p in a Mice Model of Parkinson's Disease. ACS Chem Neurosci 2020; 11:3259-3266. [PMID: 32946211 DOI: 10.1021/acschemneuro.0c00375] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder mainly occurring in the elderly. MicroRNA-155-5p (miR-155-5p) plays a vital role in neurodegenerative disease and has been reported to be regulated by rosmarinic acid (RA). In our previous study, it was found that RA could improve motor function and alleviate inflammatory responses in a mice model of PD. This study aimed to investigate the role of miR-155-5p in RA-treated PD mice. The PD mice model was established by injecting mice with N-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) and treated with RA or/and miR-155-5p agomir. The effects of miR-155-5p agomir on motor function, microglial activation, inflammation, apoptosis, and oxidative stress were analyzed by performing a behavioral test, ionized calcium-binding adapter molecule 1 staining, quantitative real-time PCR, Western blot, enzyme-linked immunosorbent assay, tyrosine hydroxylase (TH)-terminal dUTP nick end labeling double staining, TH-cleaved-caspase 3 double staining, and assessment of antioxidative parameters in RA-treated PD mice. The interaction between miR-155-5p and suppressor of cytokine signaling 1/nuclear factor erythroid 2-related factor 2 was validated using dual-luciferase reporter assay. MiR-155-5p up-regulation inhibited the alleviation of motor deficits caused by RA in PD mice, as evidenced by increasing descending time, decreasing limb movement score, increasing the time crossing the beam, and decreasing the times of front limb use. MiR-155-5p up-regulation could elevate microglial activation, inflammation, apoptosis, and oxidative stress in RA-treated PD mice. In conclusion, RA was able to alleviate PD by regulating miR-155-5p, suggesting that miR-155-5p could be used as a therapeutic target for PD treatment.
Collapse
Affiliation(s)
- Runxiao Lv
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Lili Du
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang 110122, People's Republic of China
| | - Fenghua Zhou
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiangnan Yuan
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xueyong Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Lixin Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
40
|
Microglia, inflammation and gut microbiota responses in a progressive monkey model of Parkinson's disease: A case series. Neurobiol Dis 2020; 144:105027. [PMID: 32712266 DOI: 10.1016/j.nbd.2020.105027] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/29/2022] Open
Abstract
Inflammation has been linked to the development of nonmotor symptoms in Parkinson's disease (PD), which greatly impact patients' quality of life and can often precede motor symptoms. Suitable animal models are critical for our understanding of the mechanisms underlying disease and the associated prodromal disturbances. The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkey model is commonly seen as a "gold standard" model that closely mimics the clinical motor symptoms and the nigrostriatal dopaminergic loss of PD, however MPTP toxicity extends to other nondopaminergic regions. Yet, there are limited reports monitoring the MPTP-induced progressive central and peripheral inflammation as well as other nonmotor symptoms such as gastrointestinal function and microbiota. We report 5 cases of progressive parkinsonism in non-human primates to gain a broader understanding of MPTP-induced central and peripheral inflammatory dysfunction to understand the potential role of inflammation in prodromal/pre-motor features of PD-like degeneration. We measured inflammatory proteins in plasma and CSF and performed [18F]FEPPA PET scans to evaluate translocator proteins (TSPO) or microglial activation. Monkeys were also evaluated for working memory and executive function using various behavior tasks and for gastrointestinal hyperpermeability and microbiota composition. Additionally, monkeys were treated with a novel TNF inhibitor XPro1595 (10 mg/kg, n = 3) or vehicle (n = 2) every three days starting 11 weeks after the initiation of MPTP to determine whether XPro1595 would alter inflammation and microglial behavior in a progressive model of PD. The case studies revealed that earlier and robust [18F]FEPPA PET signals resulted in earlier and more severe parkinsonism, which was seen in male cases compared to female cases. Potential other sex differences were observed in circulating inflammation, microbiota diversity and their metabolites. Additional studies with larger group sizes of both sexes would enable confirmation and extension of these findings. If these findings reflect potential differences in humans, these sex differences have significant implications for therapeutic development of inflammatory targets in the clinic.
Collapse
|
41
|
Dhankhar J, Agrawal N, Shrivastava A. An interplay between immune response and neurodegenerative disease progression: An assessment using Drosophila as a model. J Neuroimmunol 2020; 346:577302. [PMID: 32683186 DOI: 10.1016/j.jneuroim.2020.577302] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023]
Abstract
Neurodegeneration, the slow and progressive loss of neurons in the central nervous system has become a major challenge to public health worldwide particularly with elderly people. Until recently, the brain and immune system were studied exclusively, independent of each other representing two distinct systems. Recent studies ensue crosstalk between these two systems to maintain homeostasis. Though the progressive loss of specific neuronal subsets is a hallmark of neurodegenerative disease, emerging evidences indicate that immune response also plays a critical role in disease progression. Due to conservation of mechanisms that govern neural development and innate immune activation in flies and humans, and availability of powerful genetic tools, the fruit fly Drosophila melanogaster is one of the best model organisms to investigate the immune response in neurodegenerative disease. Owing to significant homology between human and Drosophila immune system and recent reports on interplay between immune system and neurodegenerative disease progression, the main focus of the review is to develop a comprehensive understanding of how neuro-immune interactions contribute to neurodegeneration using Drosophila as a model system.
Collapse
Affiliation(s)
- Jyoti Dhankhar
- Department of Zoology, University of Delhi, Delhi, India
| | - Namita Agrawal
- Department of Zoology, University of Delhi, Delhi, India
| | | |
Collapse
|
42
|
Wang Q, Oyarzabal EA, Song S, Wilson B, Santos JH, Hong JS. Locus coeruleus neurons are most sensitive to chronic neuroinflammation-induced neurodegeneration. Brain Behav Immun 2020; 87:359-368. [PMID: 31923552 PMCID: PMC7316605 DOI: 10.1016/j.bbi.2020.01.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) develops over decades through spatiotemporal stages that ascend from the brainstem to the forebrain. The mechanism behind this caudo-rostral neurodegeneration remains largely undefined. In unraveling this phenomenon, we recently developed a lipopolysaccharide (LPS)-elicited chronic neuroinflammatory mouse model that displays sequential losses of neurons in brainstem, substantia nigra, hippocampus and cortex. In this study, we aimed to investigate the mechanisms of caudo-rostral neurodegeneration and focused our efforts on the earliest neurodegeneration of vulnerable noradrenergic locus coeruleus (NE-LC) neurons in the brainstem. We found that compared with neurons in other brain regions, NE-LC neurons in untreated mice displayed high levels of mitochondrial oxidative stress that was severely exacerbated in the presence of LPS-elicited chronic neuroinflammation. In agreement, NE-LC neurons in LPS-treated mice displayed early reduction of complex IV expression and mitochondrial swelling and loss of cristae. Mechanistically, the activation of the superoxide-generating enzyme NADPH oxidase (NOX2) on NE-LC neurons was essential for their heightened vulnerability during chronic neuroinflammation. LPS induced early and high expressions of NOX2 in NE-LC neurons. Genetic or pharmacological inactivation of NOX2 markedly reduced mitochondrial oxidative stress and dysfunction in LPS-treated mice. Furthermore, inhibition of NOX2 significantly ameliorated LPS-induced NE-LC neurodegeneration. More importantly, post-treatment with NOX2 inhibitor diphenyleneiodonium when NE-LC neurodegeneration had already begun, still showed high efficacy in protecting NE-LC neurons from degeneration in LPS-treated mice. This study strongly supports that chronic neuroinflammation and NOX2 expression among vulnerable neuronal populations contribute to caudo-rostral degeneration in PD.
Collapse
Affiliation(s)
- Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China.
| | - Esteban A. Oyarzabal
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Sheng Song
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Belinda Wilson
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Janine H. Santos
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
43
|
Gan P, Ding L, Hang G, Xia Q, Huang Z, Ye X, Qian X. Oxymatrine Attenuates Dopaminergic Neuronal Damage and Microglia-Mediated Neuroinflammation Through Cathepsin D-Dependent HMGB1/TLR4/NF-κB Pathway in Parkinson's Disease. Front Pharmacol 2020; 11:776. [PMID: 32528295 PMCID: PMC7264119 DOI: 10.3389/fphar.2020.00776] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Oxymatrine (OMT), a natural quinoxaline alkaloid extracted from the root of Sophora flavescens, presents amounts of pharmacological properties including immunomodulation, anti-inflammation, anti-oxidation, and anti-virus. Recent studies tend to focus on its effects on neuroinflammation and neuroprotection in Parkinson’s disease (PD) due to its profound anti-inflammatory effect. In this study, the neuroprotective and anti-neuroinflammatory effects of OMT were investigated in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-stimulated mice and 1-methyl-4-phenylpyridinium (MPP+)-induced mice primary microglia. Additionally, mice primary neuron-microglia co-cultures and primary microglia infected with Cathepsin D (CathD)-overexpressed lentivirus were used to clarify whether the neuroprotective effect of OMT was through a CathD-dependent pathway. Results showed that OMT dose-dependently alleviated MPTP-induced motor deficits and conferred significant dopamine (DA) neuroprotection against MPTP/MPP+-induced neurotoxicity. In addition, OMT inhibited MPTP/MPP+-induced microglia activation and the pro-inflammatory cytokines release. Further, OMT down-regulated the expression of CathD, and inhibited the activation of the HMGB1/TLR4 signaling pathway as well as the nuclear translocation of NF-κB both in vivo and in vitro. It is worth noting that overexpression of CathD reversed OMT-targeted inhibition of HMGB1/TLR4/NF-κB signaling and OMT-produced neuroprotection in reconstituted neuron-microglia co-cultures. Our findings indicated that OMT conferred DA neuroprotection and attenuated microglial-mediated neuroinflammation through CathD-dependent inhibition of HMGB1/TLR4/NF-κB signaling pathway. Our study supports a potential role for OMT in ameliorating PD, and proposes that OMT may be useful in the treatment of PD.
Collapse
Affiliation(s)
- Ping Gan
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| | - Lidong Ding
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| | - Guihua Hang
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| | - Qiaofang Xia
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| | - Zhimei Huang
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| | - Xing Ye
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| | - Xiaojuan Qian
- Department of Pharmacy, Taizhou Second People's Hospital, Taizhou, China
| |
Collapse
|
44
|
Li J, Jian L, Huang J, Xiong N, Wang T. The rs3129882/rs4248166 in HLA-DRA and rs34372695 in SYT11 are not associated with sporadic Parkinson's disease in Central Chinese population. Int J Neurosci 2020; 131:674-680. [PMID: 32253955 DOI: 10.1080/00207454.2020.1753728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a common progressive neurodegenerative disorder. Up to now, several single-nucleotide polymorphisms (SNPs) located in virulence gene sites have been reported linked to PD. Candidate gene association studies and genome-wide association studies have identified rs3129882, rs4248166 in HLA-DRA and rs34372695 in SYT11 as risk factors for familial or sporadic PD. However, the association between variants of HLA-DRA, SYT11 and PD are still controversial, especially in the Central Chinese population. We here performed a case-control study to investigate whether HLA-DRA and SYT11 genes could predispose to sporadic PD in the Chinese population. METHODS We investigate 486 PD patients and 457 age- and sex-matched controls from Central China to assess this association. RESULTS In the allele model, the odds ratio (OR) result of rs3129882 was 0.905 (p = 0.287). Moreover, no significant difference was observed in the association between rs424816 (OR = 0.864, p = 0.106) and rs34372695 (p = 1.0) with PD risk. Genotypic analysis in SNP rs3129882, rs4248166 and rs34372695 indicated no significant association with PD. Subgroup analysis of our data showed age-onset and gender were not associated with either genotype or minor allele frequencies of rs3129882 and rs4248166. Moreover, the negative results were also observed in a meta-analysis of studies of rs3129882 from mainland China and Taiwanese population. CONCLUSIONS Our results reveal that rs3129882, rs4248166 and rs34372695 do not confer significant risks for sporadic PD in the Central Chinese population.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurology, TaikangTongji (Wuhan) Hospital, Wuhan, Hubei, China
| | - Lubao Jian
- Department of Neurology, TaikangTongji (Wuhan) Hospital, Wuhan, Hubei, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
45
|
Wang L, Liu Y, Yan S, Du T, Fu X, Gong X, Zhou X, Zhang T, Wang X. Disease Progression-Dependent Expression of CD200R1 and CX3CR1 in Mouse Models of Parkinson's Disease. Aging Dis 2020; 11:254-268. [PMID: 32257540 PMCID: PMC7069458 DOI: 10.14336/ad.2019.0615] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022] Open
Abstract
Microglial activation is an important contributor to the pathogenesis of Parkinson’s disease (PD). Microglia are tightly and efficiently regulated by immune checkpoints, including CD200-CD200R1 and CX3CL1-CX3CR1. Understanding the involvement of these checkpoints in disease progression provides important insights into how microglial activation contributes to PD pathology. However, so far, studies have produced seemingly conflicting results. In this study, we demonstrate that CD200R1 expression is down-regulated at both early and late stage of PD model, and CX3CR1 expression is down-regulated in early stage and recovered in late stage. In primary cultured microglia, CD200R1 and CX3CR1 expressions are both directly regulated by LPS or α-synuclein, and CD200R1 expression is more sensitively regulated than CX3CR1. In addition, CD200 knockout causes an increase in proinflammatory cytokine production and microglial activation in the midbrain. Remarkably, DA neurons in the substantial nigra are degenerated in CD200-/- mice. Finally, activation of the CD200R with CD200Fc alleviates the neuroinflammation in microglia. Together, these results suggest that immune checkpoints play distinct functional roles in different stage of PD pathology, and the CD200-CD200R1 axis plays a significant role in nigrostriatal neuron viability and function.
Collapse
Affiliation(s)
- Le Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Yang Liu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Shuxin Yan
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tianshu Du
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xia Fu
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- 2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Xinyu Zhou
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ting Zhang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- 1Department of Neurobiology, Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China.,2Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Iarkov A, Barreto GE, Grizzell JA, Echeverria V. Strategies for the Treatment of Parkinson's Disease: Beyond Dopamine. Front Aging Neurosci 2020; 12:4. [PMID: 32076403 PMCID: PMC7006457 DOI: 10.3389/fnagi.2020.00004] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is the second-leading cause of dementia and is characterized by a progressive loss of dopaminergic neurons in the substantia nigra alongside the presence of intraneuronal α-synuclein-positive inclusions. Therapies to date have been directed to the restoration of the dopaminergic system, and the prevention of dopaminergic neuronal cell death in the midbrain. This review discusses the physiological mechanisms involved in PD as well as new and prospective therapies for the disease. The current data suggest that prevention or early treatment of PD may be the most effective therapeutic strategy. New advances in the understanding of the underlying mechanisms of PD predict the development of more personalized and integral therapies in the years to come. Thus, the development of more reliable biomarkers at asymptomatic stages of the disease, and the use of genetic profiling of patients will surely permit a more effective treatment of PD.
Collapse
Affiliation(s)
- Alexandre Iarkov
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - J Alex Grizzell
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Valentina Echeverria
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile.,Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, United States
| |
Collapse
|
47
|
Inflammation in Post-Traumatic Stress Disorder (PTSD): A Review of Potential Correlates of PTSD with a Neurological Perspective. Antioxidants (Basel) 2020; 9:antiox9020107. [PMID: 31991875 PMCID: PMC7070581 DOI: 10.3390/antiox9020107] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a chronic condition characterized by symptoms of physiological and psychosocial burden. While growing research demonstrated signs of inflammation in PTSD, specific biomarkers that may be representative of PTSD such as the detailed neural correlates underlying the inflammatory responses in relation to trauma exposure are seldom discussed. Here, we review recent studies that explored alterations in key inflammatory markers in PTSD, as well as neuroimaging-based studies that further investigated signs of inflammation within the brain in PTSD, as to provide a comprehensive summary of recent literature with a neurological perspective. A search was conducted on studies published from 2009 through 2019 in PubMed and Web of Science. Fifty original articles were selected. Major findings included elevated levels of serum proinflammatory cytokines in individuals with PTSD across various trauma types, as compared with those without PTSD. Furthermore, neuroimaging-based studies demonstrated that altered inflammatory markers are associated with structural and functional alterations in brain regions that are responsible for the regulation of stress and emotion, including the amygdala, hippocampus, and frontal cortex. Future studies that utilize both central and peripheral inflammatory markers are warranted to elucidate the underlying neurological pathway of the pathophysiology of PTSD.
Collapse
|
48
|
Rizor A, Pajarillo E, Johnson J, Aschner M, Lee E. Astrocytic Oxidative/Nitrosative Stress Contributes to Parkinson's Disease Pathogenesis: The Dual Role of Reactive Astrocytes. Antioxidants (Basel) 2019; 8:antiox8080265. [PMID: 31374936 PMCID: PMC6719180 DOI: 10.3390/antiox8080265] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide; it is characterized by dopaminergic neurodegeneration in the substantia nigra pars compacta, but its etiology is not fully understood. Astrocytes, a class of glial cells in the central nervous system (CNS), provide critical structural and metabolic support to neurons, but growing evidence reveals that astrocytic oxidative and nitrosative stress contributes to PD pathogenesis. As astrocytes play a critical role in the production of antioxidants and the detoxification of reactive oxygen and nitrogen species (ROS/RNS), astrocytic oxidative/nitrosative stress has emerged as a critical mediator of the etiology of PD. Cellular stress and inflammation induce reactive astrogliosis, which initiates the production of astrocytic ROS/RNS and may lead to oxidative/nitrosative stress and PD pathogenesis. Although the cause of aberrant reactive astrogliosis is unknown, gene mutations and environmental toxicants may also contribute to astrocytic oxidative/nitrosative stress. In this review, we briefly discuss the physiological functions of astrocytes and the role of astrocytic oxidative/nitrosative stress in PD pathogenesis. Additionally, we examine the impact of PD-related genes such as α-synuclein, protein deglycase DJ-1( DJ-1), Parkin, and PTEN-induced kinase 1 (PINK1) on astrocytic function, and highlight the impact of environmental toxicants, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, manganese, and paraquat, on astrocytic oxidative/nitrosative stress in experimental models.
Collapse
Affiliation(s)
- Asha Rizor
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA
| | - James Johnson
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine Bronx, New York, NY 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy Florida A&M University, Tallahassee, FL 32301, USA.
| |
Collapse
|
49
|
Kirkley KS, Popichak KA, Hammond SL, Davies C, Hunt L, Tjalkens RB. Genetic suppression of IKK2/NF-κB in astrocytes inhibits neuroinflammation and reduces neuronal loss in the MPTP-Probenecid model of Parkinson's disease. Neurobiol Dis 2019; 127:193-209. [PMID: 30818064 PMCID: PMC6588478 DOI: 10.1016/j.nbd.2019.02.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/20/2018] [Accepted: 02/22/2019] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammatory activation of glia is considered a pathological hallmark of Parkinson's disease (PD) and is seen in both human PD patients and in animal models of PD; however, the relative contributions of these cell types, especially astrocytes, to the progression of disease is not fully understood. The transcription factor, nuclear factor kappa B (NFκB), is an important regulator of inflammatory gene expression in glia and is activated by multiple cellular stress signals through the kinase complex, IKK2. We sought to determine the role of NFκB in modulating inflammatory activation of astrocytes in a model of PD by generating a conditional knockout mouse (hGfapcre/Ikbk2F/F) in which IKK2 is specifically deleted in astrocytes. Measurements of IKK2 revealed a 70% deletion rate of IKK2 within astrocytes, as compared to littermate controls (Ikbk2F/F). Use of this mouse in a subacute, progressive model of PD through exposure to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTPp) revealed significant protection in exposed mice to direct and progressive loss of dopaminergic neurons in the substantia nigra (SN). hGfapcre/Ikbk2F/F mice were also protected against MPTPp-induced loss in motor activity, loss of striatal proteins, and genomic alterations in nigral NFκB gene expression, but were not protected from loss of striatal catecholamines. Neuroprotection in hGfapcre/Ikbk2F/F mice was associated with inhibition of MPTPp-induced astrocytic expression of inflammatory genes and protection against nitrosative stress and apoptosis in neurons. These data indicate that deletion of IKK2 within astrocytes is neuroprotective in the MPTPp model of PD and suggests that reactive astrocytes directly contribute the potentiation of dopaminergic pathology.
Collapse
Affiliation(s)
- Kelly S Kirkley
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Katriana A Popichak
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Sean L Hammond
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Cecilia Davies
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Lindsay Hunt
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA 80523; Program in Molecular, Cellular and Integrative Neuroscience, Colorado State University, Fort Collins, CO, USA 80523.
| |
Collapse
|
50
|
Liu JQ, Chu SF, Zhou X, Zhang DY, Chen NH. Role of chemokines in Parkinson's disease. Brain Res Bull 2019; 152:11-18. [PMID: 31136787 DOI: 10.1016/j.brainresbull.2019.05.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 01/10/2023]
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder with an increasing incidence year by year, particularly as the population ages. The most common neuropathologic manifestation in patients with Parkinson's disease is dopamine neurons degeneration and loss in substantia nigra of middle brain. The main neurochemistry problem is the lack of the neurotransmitter dopamine. Clinically, PD patients may also have higher levels of glutamate, gamma-aminobutyric acid, acetylcholine and other neurotransmitters. At present, many data have shown that some chemokines are involved in regulating the release and transmission of neurotransmitters, and the growth and development of related neurons. In recent years, most of the studies relative to PD is based on immune and inflammatory mechanisms, and chemokines is also the focus on this mechanism. Chemokines are a class of cytokines that have definite chemotaxis effects on the different target cells. They might be involved in the pathogenesis of PD by inducing neuronal apoptosis and microglia activation. Clinical data has shown that the levels of chemokines in plasma and cerebrospinal fluid of PD patients have corresponding changes compared with the healthy persons. This review summarizes recent studies on chemokines and their receptors in Parkinson's disease: (i) to explore the role of chemokines in Parkinson's disease; (ii) to provide new indicators for clinical diagnosis of PD; (iii) to provide new targets for drug research and development in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Jia-Qi Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing 211198, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xin Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Da-Yong Zhang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing 211198, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|