1
|
Wang X, Shi H, Wei Z, Liu P, Tian S, Song X. Improvement of Parkinson's Disease Symptoms by Butylphthalide Through Modulation of Microglial Activation. Neuromolecular Med 2025; 27:40. [PMID: 40394290 DOI: 10.1007/s12017-025-08865-x] [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: 02/07/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Microglia-mediated neuroinflammation plays an important role in the pathogenesis of Parkinson's disease (PD). Studies have shown that butylphthalide (3-n-butylphthalide or NBP) can play an anti-inflammatory role in other diseases by regulating the activation of microglia. This study investigates the neuroprotective and anti-inflammatory effects of NBP in a mouse model of Parkinson's disease (PD) induced by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The primary aim was to determine whether NBP can improve symptoms of PD by modulating microglial activation and to explore the underlying mechanisms involved. Motor function (assessed via Open Field and Pole Climbing tests), dopaminergic neuronal loss, and activation of different microglial subtypes were assessed in control, MPTP-treated, and NBP + MPTP-treated mice. A p38 phosphorylation inhibitor + MPTP group was also established to investigate potential mechanisms of NBP action. Mice treated with NBP exhibited significantly improved motor function and reduced dopaminergic neuronal loss compared to MPTP-treated mice. In PD mice, pro-inflammatory factor expression was elevated, anti-inflammatory factor expression was reduced, and the expression of arginase-1 (arg-1), a marker for M2 microglia, was decreased. NBP treatment resulted in reduced levels of pro-inflammatory factors, increased levels of anti-inflammatory factors, and elevated arg-1 expression. Additionally, inhibition of p38 phosphorylation further decreased pro-inflammatory factor expression while increasing both anti-inflammatory factor levels and arg-1 expression. The findings indicate that NBP regulates neuroinflammation and improves symptoms of PD by promoting the transformation of microglia to the M2 phenotype, likely mediated through the p38 phosphorylation pathway.
Collapse
Affiliation(s)
- Xue Wang
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huimin Shi
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zibin Wei
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ping Liu
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shujuan Tian
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Key Laboratory of Clinical Neurology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China.
- Key Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
2
|
Potemkin N, Cawood SMF, Guévremont D, Mockett B, Treece J, Stanton JAL, Williams JM. Whole Transcriptome RNA-Seq Reveals Drivers of Pathological Dysfunction in a Transgenic Model of Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04878-6. [PMID: 40186694 DOI: 10.1007/s12035-025-04878-6] [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: 11/19/2023] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
Alzheimer's disease (AD) affects more than 55 million people worldwide, yet current theories cannot fully explain its aetiology. Accordingly, gene expression profiling has been used to provide a holistic view of the biology underpinning AD. Focusing primarily on protein-coding genes, such approaches have highlighted a critical involvement of microglia-related inflammatory processes. Simultaneous investigation of transcriptional regulators and noncoding RNA (ncRNA) can offer further insight into AD biology and inform the development of disease-modifying therapies. We previously described a method for whole transcriptome sampling to simultaneously investigate protein-coding genes and ncRNA. Here, we use this technique to explore transcriptional changes in a murine model of AD (15-month-old APP/PS1 mice). We confirmed the extensive involvement of microglia-associated genes and gene networks, consistent with literature. We also report a wealth of differentially-expressed non-coding RNA - including microRNA, long non-coding RNA, small nuclear and small nucleolar RNA, and pseudogenes - many of which have been overlooked previously. Transcription factor analysis determined that six transcription factors likely regulate gene expression changes in this model (Irf8, Junb, c-Fos, Lmo2, Runx1, and Nfe2l2). We then utilised validated miRNA-target interactions, finding 60 interactions between 15 miRNA and 42 mRNA (messenger RNA) with largely consistent directionality. Furthermore, we found that eight transcription factors (Clock, Lmo2, Runx1, Nfe2l2, Egr2, c-Fos, Junb, and Nr4a1) are likely responsible for the regulation of miRNA expression. Taken together, these data indicate a complex interplay of coding and non-coding RNA, driven by a small number of specific transcription factors, contributing to transcriptional changes in 15-month-old APP/PS1 mice.
Collapse
Affiliation(s)
- Nikita Potemkin
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Sophie M F Cawood
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Diane Guévremont
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Bruce Mockett
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Jackson Treece
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Jo-Ann L Stanton
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand.
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
3
|
Zeng H, Lu H, Yang J, Hu P. An Update on Recent Drug Delivery Systems Targeting Brain Diseases via the Transnasal Pathway. Pharm Res 2024; 41:2121-2141. [PMID: 39477900 DOI: 10.1007/s11095-024-03790-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVE To explore the potential of transnasal drug delivery systems (DDS) as an effective means of bypassing the bloodbrain barrier (BBB) for enhanced central nervous system (CNS) targeting, aiming to improve therapeutic outcomes for CNS disorders while reducing systemic side effects. METHODS A review of current and emerging DDS technologies, including polymer nanoparticles, liposomes, and micelles, was conducted to assess their suitability for precision-targeted delivery to the brain through the transnasal route. RESULTS The investigated DDS demonstrate promising capabilities for CNS targeting via the nasal pathway, effectively preserving both the nasal mucosa and CNS integrity. These systems enhance drug precision within neural tissues, potentially improving therapeutic outcomes without harming adjacent tissues. CONCLUSIONS Transnasal DDS offer a promising alternative to traditional delivery methods, with significant potential to advance the treatment of cerebrovascular diseases, neurodegenerative disorders, brain tumors, and psychiatric conditions. This approach represents an evolving frontier in neurotherapeutics, with the potential to transform CNS drug delivery practices.
Collapse
Affiliation(s)
- Huiying Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511436, Guangdong, P. R. China
- College of Pharmacy, Jinan University, Guangzhou, 511436, Guangdong, P. R. China
| | - Huangjie Lu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511436, Guangdong, P. R. China
- College of Pharmacy, Jinan University, Guangzhou, 511436, Guangdong, P. R. China
| | - Jie Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511436, Guangdong, P. R. China
- College of Pharmacy, Jinan University, Guangzhou, 511436, Guangdong, P. R. China
| | - Ping Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 511436, Guangdong, P. R. China.
- College of Pharmacy, Jinan University, Guangzhou, 511436, Guangdong, P. R. China.
| |
Collapse
|
4
|
Hamidpour SK, Amiri M, Ketabforoush AHME, Saeedi S, Angaji A, Tavakol S. Unraveling Dysregulated Cell Signaling Pathways, Genetic and Epigenetic Mysteries of Parkinson's Disease. Mol Neurobiol 2024; 61:8928-8966. [PMID: 38573414 DOI: 10.1007/s12035-024-04128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
Parkinson's disease (PD) is a prevalent and burdensome neurodegenerative disorder that has been extensively researched to understand its complex etiology, diagnosis, and treatment. The interplay between genetic and environmental factors in PD makes its pathophysiology difficult to comprehend, emphasizing the need for further investigation into genetic and epigenetic markers involved in the disease. Early diagnosis is crucial for optimal management of the disease, and the development of novel diagnostic biomarkers is ongoing. Although many efforts have been made in the field of recognition and interpretation of the mechanisms involved in the pathophysiology of the disease, the current knowledge about PD is just the tip of the iceberg. By scrutinizing genetic and epigenetic patterns underlying PD, new avenues can be opened for dissecting the pathology of the disorder, leading to more precise and efficient diagnostic and therapeutic approaches. This review emphasizes the importance of studying dysregulated cell signaling pathways and molecular processes associated with genes and epigenetic alterations in understanding PD, paving the way for the development of novel therapeutic strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Shayesteh Kokabi Hamidpour
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Mobina Amiri
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | | | - Saeedeh Saeedi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Abdolhamid Angaji
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran.
- Department of Research and Development, Tavakol BioMimetic Technologies Company, Tehran, Iran.
| |
Collapse
|
5
|
Shaheen N, Shaheen A, Osama M, Nashwan AJ, Bharmauria V, Flouty O. MicroRNAs regulation in Parkinson's disease, and their potential role as diagnostic and therapeutic targets. NPJ Parkinsons Dis 2024; 10:186. [PMID: 39369002 PMCID: PMC11455891 DOI: 10.1038/s41531-024-00791-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 09/15/2024] [Indexed: 10/07/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression by binding to target messenger RNA (mRNA) molecules and promoting their degradation or blocking their translation. Parkinson's disease (PD) is a neurodegenerative disorder caused by the loss of dopaminergic neurons in the substantia nigra. There is increasing evidence to suggest that miRNAs play a role in the pathogenesis of PD. Studies have identified several miRNAs that are dysregulated in the brains of PD patients, and animal models of the disease. MiRNA expression dysregulation contributes to the onset and progression of PD by modulating neuroinflammation, oxidative stress, and protein aggregation genes. Moreover, miRNAs have emerged as potential therapeutic targets for PD. This review elucidates the changes in miRNA expression profiles associated with PD, emphasising their potential as diagnostic biomarkers and therapeutic targets, and detailing specific miRNAs implicated in PD and their downstream targets. Integrated Insights into miRNA Function, Microglial Activation, Diagnostic, and Treatment Prospects in PD Note: This figure is an original figure created by the authors.
Collapse
Affiliation(s)
- Nour Shaheen
- Alexandria University, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Ahmed Shaheen
- Alexandria University, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Mahmoud Osama
- Department of Neurosurgery, Nasser Institute for Research and Treatment, Cairo, Egypt
| | | | - Vishal Bharmauria
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
- Center for Vision Research and Center for Integrative and Applied Neuroscience, York University, Toronto, ON, Canada
- Tampa Human Neurophysiology Lab, Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, USA
| | - Oliver Flouty
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA.
- Tampa Human Neurophysiology Lab, Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
6
|
Saadh MJ, Muhammad FA, Singh A, Mustafa MA, Al Zuhairi RAH, Ghildiyal P, Hashim G, Alsaikhan F, Khalilollah S, Akhavan-Sigari R. MicroRNAs Modulating Neuroinflammation in Parkinson's disease. Inflammation 2024:10.1007/s10753-024-02125-z. [PMID: 39162871 DOI: 10.1007/s10753-024-02125-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/20/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Parkinson's disease (PD) is one of the most frequent age-associated neurodegenerative disorder. Presence of α-synuclein-containing aggregates in the substantia nigra pars compacta (SNpc) and loss of dopaminergic (DA) neurons are among the characteristic of PD. One of the hallmarks of PD pathophysiology is chronic neuroinflammation. Activation of glial cells and elevated levels of pro-inflammatory factors are confirmed as frequent features of the PD brain. Chronic secretion of pro-inflammatory cytokines by activated astrocytes and microglia exacerbates DA neuron degeneration in the SNpc. MicroRNAs (miRNAs) are among endogenous non-coding small RNA with the ability to perform post-transcriptional regulation in target genes. In that regard, the capability of miRNAs for modulating inflammatory signaling is the center of attention in many investigations. MiRNAs could enhance or limit inflammatory signaling, exacerbating or ameliorating the pathological consequences of extreme neuroinflammation. This review summarizes the importance of inflammation in the pathophysiology of PD. Besides, we discuss the role of miRNAs in promoting or protecting neural cell injury in the PD model by controlling the inflammatory pathway. Modifying the neuroinflammation by miRNAs could be considered a primary therapeutic strategy for PD.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Anamika Singh
- Department of Biotechnology and Genetics, Jain (Deemed-to-Be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Mohammed Ahmed Mustafa
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur,, Jamshedpur,, India, Jharkhand, 831001
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ghassan Hashim
- Department of Nursing, Al-Zahrawi University College, Karbala, Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warszawa, Poland
| |
Collapse
|
7
|
Xu K, Li Y, Zhou Y, Zhang Y, Shi Y, Zhang C, Bai Y, Wang S. Neuroinflammation in Parkinson's disease: focus on the relationship between miRNAs and microglia. Front Cell Neurosci 2024; 18:1429977. [PMID: 39131043 PMCID: PMC11310010 DOI: 10.3389/fncel.2024.1429977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/11/2024] [Indexed: 08/13/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder that affects the central nervous system (CNS). Neuroinflammation is a crucial factor in the pathological advancement of PD. PD is characterized by the presence of activated microglia and increased levels of proinflammatory factors, which play a crucial role in its pathology. During the immune response of PD, microglia regulation is significantly influenced by microRNA (miRNA). The excessive activation of microglia, persistent neuroinflammation, and abnormal polarization of macrophages in the brain can be attributed to the dysregulation of certain miRNAs. Additionally, there are miRNAs that possess the ability to inhibit neuroinflammation. miRNAs, which are small non-coding epigenetic regulators, have the ability to modulate microglial activity in both normal and abnormal conditions. They also have a significant impact on promoting communication between neurons and microglia.
Collapse
Affiliation(s)
- Ke Xu
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yuan Li
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Zhou
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yu Zhang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yue Shi
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chengguang Zhang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yan Bai
- Institute of Acupuncture and Moxibustion, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Shun Wang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Zhang X, Zhang Y, Wang B, Xie C, Wang J, Fang R, Dong H, Fan G, Wang M, He Y, Shen C, Duan Y, Zhao J, Liu Z, Li Q, Ma Y, Yu M, Wang J, Fei J, Xiao L, Huang F. Pyroptosis-mediator GSDMD promotes Parkinson's disease pathology via microglial activation and dopaminergic neuronal death. Brain Behav Immun 2024; 119:129-145. [PMID: 38552923 DOI: 10.1016/j.bbi.2024.03.038] [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: 11/22/2023] [Revised: 03/02/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
GSDMD-mediated pyroptosis occurs in the nigrostriatal pathway in Parkinson's disease animals, yet the role of GSDMD in neuroinflammation and death of dopaminergic neurons in Parkinson's disease remains elusive. Here, our in vivo and in vitro studies demonstrated that GSDMD, as a pyroptosis executor, contributed to glial reaction and death of dopaminergic neurons across different Parkinson's disease models. The ablation of the Gsdmd attenuated Parkinson's disease damage by reducing dopaminergic neuronal death, microglial activation, and detrimental transformation. Disulfiram, an inhibitor blocking GSDMD pore formation, efficiently curtailed pyroptosis, thereby lessening the pathology of Parkinson's disease. Additionally, a modification in GSDMD was identified in the blood of Parkinson's disease patients in contrast to healthy subjects. Therefore, the detected alteration in GSDMD within the blood of Parkinson's disease patients and the protective impact of disulfiram could be promising for the diagnostic and therapeutic approaches against Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yunhe Zhang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Boya Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Chuantong Xie
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Rong Fang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Guangchun Fan
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mengze Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yongtao He
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Chenye Shen
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yufei Duan
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jiayin Zhao
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Qing Li
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jian Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Pudong, Shanghai 201203, China.
| | - Lei Xiao
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| |
Collapse
|
9
|
Ren Y, Wu X, Bai T, Yang N, Yuan Y, Xu L, Wen Y, Wen Y, Wang Z, Zhou L, Zou F, Li W. CDK5-USP30 signaling pathway regulates MAVS-mediated inflammation via suppressing mitophagy in MPTP/MPP + PD model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116446. [PMID: 38772138 DOI: 10.1016/j.ecoenv.2024.116446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
The discovery of MPTP, an industrial chemical and contaminant of illicit narcotics, which causes parkinsonism in humans, non-human primates and rodents, has led to environmental pollutants exposure being convicted as key candidate in Parkinson's disease (PD) pathogenesis. Though MPTP-induced mitochondrial dysfunction and neuroinflammation are mainly responsible for the causative issue of MPTP neurotoxicity, the underlying mechanism involved remains unclear. Here, we reveal a novel signaling mechanism of CDK5-USP30-MAVS regulating MPTP/MPP+ induced PD. MPP+ (the toxic metabolite of MPTP) treatment not only led to the increased protein levels of USP30 but also to mitophagy inhibition, mitochondrial dysfunction, and MAVS-mediated inflammation in BV2 microglial cells. Both mitophagy stimulation (Urolithin A administration) and USP30 knockdown relieved MAVS-mediated inflammation via restoring mitophagy and mitochondrial function in MPP+-induced cell model. Notably, MPTP/MPP+-induced CDK5 activation regulated USP30 phosphorylation at serine 216 to stabilize USP30. Moreover, CDK5-USP30 pathway promoted MAVS-mediated inflammation in MPTP/MPP+-induced PD model. Inhibition of CDK5 not only had a protective effect on MPP+-induced cell model of PD via suppressing the upregulation of USP30 and the activation of MAVS inflammation pathway in vitro, but also prevented neurodegeneration in vivo and alleviated movement impairment in MPTP mouse model of PD. Overall, our study reveal that CDK5 blocks mitophagy through phosphorylating USP30 and activates MAVS inflammation pathway in MPTP/MPP+-induced PD model, which suggests that CDK5-USP30-MAVS signaling pathway represents a valuable treatment strategy for PD induced by environmental neurotoxic pollutants in relation to MPTP.
Collapse
Affiliation(s)
- Yixian Ren
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China; Key Laboratory of Occupational Environment and Health, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou, China
| | - Xian Wu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Tianyao Bai
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Nanfei Yang
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuyu Yuan
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lingling Xu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yue Wen
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ying Wen
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhi Wang
- Key Laboratory of Occupational Environment and Health, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou, China
| | - Liping Zhou
- Key Laboratory of Occupational Environment and Health, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Wenjun Li
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
10
|
Xu Y, Wen L, Tang Y, Zhao Z, Xu M, Wang T, Chen Z. Sodium butyrate activates the K ATP channels to regulate the mechanism of Parkinson's disease microglia model inflammation. Immun Inflamm Dis 2024; 12:e1194. [PMID: 38501544 PMCID: PMC10949401 DOI: 10.1002/iid3.1194] [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: 04/26/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a common neurodegenerative disorder. Microglia-mediated neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. Activation of adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels can protect dopaminergic neurons from damage. Sodium butyrate (NaB) shows anti-inflammatory and neuroprotective effects in some animal models of brain injury and regulates the KATP channels in islet β cells. In this study, we aimed to verify the anti-inflammatory effect of NaB on PD and further explored potential molecular mechanisms. METHODS We established an in vitro PD model in BV2 cells using 1-methyl-4-phenylpyridinium (MPP+ ). The effects of MPP+ and NaB on BV2 cell viability were detected by cell counting kit-8 assays. The morphology of BV2 cells with or without MPP+ treatment was imaged via an optical microscope. The expression of Iba-1 was examined by the immunofluorescence staining. The intracellular ATP content was estimated through the colorimetric method, and Griess assay was conducted to measure the nitric oxide production. The expression levels of pro-inflammatory cytokines and KATP channel subunits were evaluated by reverse transcription-quantitative polymerase chain reaction and western blot analysis. RESULTS NaB (5 mM) activated the KATP channels through elevating Kir6.1 and Kir6.1 expression in MPP+ -challenged BV2 cells. Both NaB and pinacidil (a KATP opener) suppressed the MPP+ -induced activation of BV2 cells and reduced the production of nitrite and pro-inflammatory cytokines in MPP+ -challenged BV2 cells. CONCLUSION NaB treatment alleviates the MPP+ -induced inflammatory responses in microglia via activation of KATP channels.
Collapse
Affiliation(s)
- Ye Xu
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Laofu Wen
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Yunyi Tang
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Zhenqiang Zhao
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Miaojing Xu
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Department of Neurology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Tan Wang
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Zhibin Chen
- Department of NeurologyThe First Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| |
Collapse
|
11
|
Wu Y, Meng X, Cheng WY, Yan Z, Li K, Wang J, Jiang T, Zhou F, Wong KH, Zhong C, Dong Y, Gao S. Can pluripotent/multipotent stem cells reverse Parkinson's disease progression? Front Neurosci 2024; 18:1210447. [PMID: 38356648 PMCID: PMC10864507 DOI: 10.3389/fnins.2024.1210447] [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: 04/22/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by continuous and selective degeneration or death of dopamine neurons in the midbrain, leading to dysfunction of the nigrostriatal neural circuits. Current clinical treatments for PD include drug treatment and surgery, which provide short-term relief of symptoms but are associated with many side effects and cannot reverse the progression of PD. Pluripotent/multipotent stem cells possess a self-renewal capacity and the potential to differentiate into dopaminergic neurons. Transplantation of pluripotent/multipotent stem cells or dopaminergic neurons derived from these cells is a promising strategy for the complete repair of damaged neural circuits in PD. This article reviews and summarizes the current preclinical/clinical treatments for PD, their efficacies, and the advantages/disadvantages of various stem cells, including pluripotent and multipotent stem cells, to provide a detailed overview of how these cells can be applied in the treatment of PD, as well as the challenges and bottlenecks that need to be overcome in future translational studies.
Collapse
Affiliation(s)
- Yongkang Wu
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Xiangtian Meng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wai-Yin Cheng
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Zhichao Yan
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keqin Li
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tianfang Jiang
- Department of Neurology, Shanghai Eighth People’s Hospital Affiliated to Jiangsu University, Shanghai, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Ka-Hing Wong
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Shane Gao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Mohammed OA, Elballal MS, El-Husseiny AA, Khidr EG, El Tabaa MM, Elazazy O, Abd-Elmawla MA, Elesawy AE, Ibrahim HM, Abulsoud AI, El-Dakroury WA, Abdel Mageed SS, Elrebehy MA, Nomier Y, Abdel-Reheim MA, El-Husseiny HM, Mahmoud AMA, Saber S, Doghish AS. Unraveling the role of miRNAs in the diagnosis, progression, and therapeutic intervention of Parkinson's disease. Pathol Res Pract 2024; 253:155023. [PMID: 38081104 DOI: 10.1016/j.prp.2023.155023] [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: 11/24/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
Parkinson's disease (PD) is a debilitating neurological disorder characterized by the impairment of the motor system, resulting in symptoms such as resting tremor, cogwheel rigidity, bradykinesia, difficulty with gait, and postural instability. The occurrence of striatal dopamine insufficiency can be attributed to a notable decline in dopaminergic neurons inside the substantia nigra pars compacta. Additionally, the development of Lewy bodies serves as a pathological hallmark of PD. While current therapy approaches for PD aim to preserve dopaminergic neurons or replenish dopamine levels in the brain, it is important to acknowledge that achieving complete remission of the condition remains elusive. MicroRNAs (miRNAs, miR) are a class of small, non-coding ribonucleic acids involved in regulating gene expression at the post-transcriptional level. The miRNAs play a crucial part in the underlying pathogenic mechanisms of several neurodegenerative illnesses, including PD. The aim of this review is to explore the role of miRNAs in regulating genes associated with the onset and progression of PD, investigate the potential of miRNAs as a diagnostic tool, assess the effectiveness of targeting specific miRNAs as an alternative therapeutic strategy to impede disease advancement, and discuss the utilization of newly developed nanoparticles for delivering miRNAs as neurodegenerative therapies.
Collapse
Affiliation(s)
- Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829 Cairo, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897 Menoufia, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mai A Abd-Elmawla
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Henwa M Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt.
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Yousra Nomier
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Oman
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Hussein M El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya 13736, Egypt
| | - Abdulla M A Mahmoud
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
13
|
Khish NS, Ghiasizadeh P, Rasti A, Moghimi O, Zadeh AZ, Bahiraee A, Ebrahimi R. Regulatory Non-coding RNAs Involved in Oxidative Stress and Neuroinflammation: An Intriguing Crosstalk in Parkinson's Disease. Curr Med Chem 2024; 31:5576-5597. [PMID: 37592769 DOI: 10.2174/0929867331666230817102135] [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: 02/04/2023] [Revised: 05/23/2023] [Accepted: 06/01/2023] [Indexed: 08/19/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the accumulation of α-synuclein and the degeneration of dopaminergic neurons in the substantia nigra. Although the molecular bases for PD development are not fully recognized, extensive evidence has suggested that the development of PD is strongly associated with neuroinflammation. It is noteworthy that while neuroinflammation might not be a primary factor in all patients with PD, it seems to be a driving force for disease progression, and therefore, exploring the role of pathways involved in neuroinflammation is of great importance. Besides, the importance of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and competing endogenous RNAs (ceRNAs), has been widely studied with a focus on the pathogenesis of PD. However, there is no comprehensive review regarding the role of neuroinflammation- related ncRNAs as prospective biomarkers and therapeutic targets involved in the pathogenesis of PD, even though the number of studies connecting ncRNAs to neuroinflammatory pathways and oxidative stress has markedly increased in the last few years. Hence, the present narrative review intended to describe the crosstalk between regulatory ncRNAs and neuroinflammatory targets with respect to PD to find and propose novel combining biomarkers or therapeutic targets in clinical settings.
Collapse
Affiliation(s)
- Naser Salari Khish
- Department of Biology, Payam Noor University International, Center of Gheshm, Hormozgan, Iran
| | - Pooran Ghiasizadeh
- Student Research Committee, Arak University of Medical Science, Arak, Iran
| | - Abolhasan Rasti
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Omid Moghimi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Arash Zeynali Zadeh
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Reyhane Ebrahimi
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
14
|
Zhang D, Zhang J, Wang Y, Wang G, Tang P, Liu Y, Zhang Y, Ouyang L. Targeting epigenetic modifications in Parkinson's disease therapy. Med Res Rev 2023; 43:1748-1777. [PMID: 37119043 DOI: 10.1002/med.21962] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 01/10/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
Parkinson's disease (PD) is a multifactorial disease due to a complex interplay between genetic and epigenetic factors. Recent efforts shed new light on the epigenetic mechanisms involved in regulating pathways related to the development of PD, including DNA methylation, posttranslational modifications of histones, and the presence of microRNA (miRNA or miR). Epigenetic regulators are potential therapeutic targets for neurodegenerative disorders. In the review, we aim to summarize mechanisms of epigenetic regulation in PD, and describe how the DNA methyltransferases, histone deacetylases, and histone acetyltransferases that mediate the key processes of PD are attractive therapeutic targets. We discuss the use of inhibitors and/or activators of these regulators in PD models or patients, and how these small molecule epigenetic modulators elicit neuroprotective effects. Further more, given the importance of miRNAs in PD, their contributions to the underlying mechanisms of PD will be discussed as well, together with miRNA-based therapies.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Jifa Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Yuxi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Pan Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Yun Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Yiwen Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Sichuan, Chengdu, China
| |
Collapse
|
15
|
Szelągowski A, Kozakiewicz M. A Glance at Biogenesis and Functionality of MicroRNAs and Their Role in the Neuropathogenesis of Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7759053. [PMID: 37333462 PMCID: PMC10270766 DOI: 10.1155/2023/7759053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/11/2023] [Accepted: 05/20/2023] [Indexed: 06/20/2023]
Abstract
MicroRNAs (miRNAs) are short, noncoding RNA transcripts. Mammalian miRNA coding sequences are located in introns and exons of genes encoding various proteins. As the central nervous system is the largest source of miRNA transcripts in living organisms, miRNA molecules are an integral part of the regulation of epigenetic activity in physiological and pathological processes. Their activity depends on many proteins that act as processors, transporters, and chaperones. Many variants of Parkinson's disease have been directly linked to specific gene mutations which in pathological conditions are cumulated resulting in the progression of neurogenerative changes. These mutations can often coexist with specific miRNA dysregulation. Dysregulation of different extracellular miRNAs has been confirmed in many studies on the PD patients. It seems reasonable to conduct further research on the role of miRNAs in the pathogenesis of Parkinson's disease and their potential use in future therapies and diagnosis of the disease. This review presents the current state of knowledge about the biogenesis and functionality of miRNAs in the human genome and their role in the neuropathogenesis of Parkinson's disease (PD)-one of the most common neurodegenerative disorders. The article also describes the process of miRNA formation which can occur in two ways-the canonical and noncanonical one. However, the main focus was on miRNA's use in in vitro and in vivo studies in the context of pathophysiology, diagnosis, and treatment of PD. Some issues, especially those regarding the usefulness of miRNAs in PD's diagnostics and especially its treatment, require further research. More standardization efforts and clinical trials on miRNAs are needed.
Collapse
Affiliation(s)
- Adam Szelągowski
- Nicolaus Copernicus University in Toruń Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Geriatrics, Bydgoszcz, Poland
| | - Mariusz Kozakiewicz
- Nicolaus Copernicus University in Toruń Ludwik Rydygier Collegium Medicum in Bydgoszcz, Faculty of Health Sciences, Department of Geriatrics, Bydgoszcz, Poland
| |
Collapse
|
16
|
Guedes BFS, Cardoso SM, Esteves AR. The Impact of microRNAs on Mitochondrial Function and Immunity: Relevance to Parkinson's Disease. Biomedicines 2023; 11:biomedicines11051349. [PMID: 37239020 DOI: 10.3390/biomedicines11051349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's Disease (PD), the second most common neurodegenerative disorder, is characterised by the severe loss of dopaminergic neurons in the Substantia Nigra pars compacta (SNpc) and by the presence of Lewy bodies. PD is diagnosed upon the onset of motor symptoms, such as bradykinesia, resting tremor, rigidity, and postural instability. It is currently accepted that motor symptoms are preceded by non-motor features, such as gastrointestinal dysfunction. In fact, it has been proposed that PD might start in the gut and spread to the central nervous system. Growing evidence reports that the gut microbiota, which has been found to be altered in PD patients, influences the function of the central and enteric nervous systems. Altered expression of microRNAs (miRNAs) in PD patients has also been reported, many of which regulate key pathological mechanisms involved in PD pathogenesis, such as mitochondrial dysfunction and immunity. It remains unknown how gut microbiota regulates brain function; however, miRNAs have been highlighted as important players. Remarkably, numerous studies have depicted the ability of miRNAs to modulate and be regulated by the host's gut microbiota. In this review, we summarize the experimental and clinical studies implicating mitochondrial dysfunction and immunity in PD. Moreover, we gather recent data on miRNA involvement in these two processes. Ultimately, we discuss the reciprocal crosstalk between gut microbiota and miRNAs. Studying the bidirectional interaction of gut microbiome-miRNA might elucidate the aetiology and pathogenesis of gut-first PD, which could lead to the application of miRNAs as potential biomarkers or therapeutical targets for PD.
Collapse
Affiliation(s)
- Beatriz F S Guedes
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
17
|
Wang M, Liu M, Ma Z. Cannabinoid type 2 receptor activation inhibits MPP +-induced M1 differentiation of microglia through activating PI3K/Akt/Nrf2 signal pathway. Mol Biol Rep 2023; 50:4423-4433. [PMID: 36977807 DOI: 10.1007/s11033-023-08395-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Growing evidence indicates that cannabinoid type 2 (CB2) receptor activation inhibits neuroinflammation in the pathogenesis of Parkinson's disease (PD). Nonetheless, the precise mechanisms of CB2 receptor-mediated neuroprotection have not been fully elucidated. The differentiation of microglia from the M1 to M2 phenotype plays a vital role in neuroinflammation. METHODS In the present study, we investigated the effect of CB2 receptor activation on the M1/M2 phenotypic transformation of microglia treated with 1-methyl-4-phenylpyridinium (MPP+). The M1 phenotype microglia markers, including inducible nitric oxide (iNOS), interleukin 6 (IL-6), and CD86, and the M2 phenotype microglia markers, including arginase-1 (Arg-1), IL-10, and CD206, were detected by western blots and flow cytometry. The levels of phosphoinositide-3-kinase (PI3K)/Akt and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined by Western blots. Subsequent addition of Nrf2 inhibitors initially revealed the specific mechanism by which CB2 receptors affect phenotypic changes in microglia. RESULTS Our results showed that pretreatment with JWH133 significantly inhibited the MPP+-induced up-regulation of M1 phenotype microglia markers. Meanwhile, JWH133 increased the levels of M2 phenotype microglia markers. JWH133-mediated effects were blocked by co-treatment with AM630. Mechanism studies found that MPP+ treatment downregulated PI3K, Akt phosphorylated proteins, and nuclear Nrf2 protein. JWH133 pretreatment promoted PI3K/Akt activation and facilitated nuclear translocation of Nrf2, which was reversed by the PI3K inhibitor. Further studies showed that Nrf2 inhibitors inverted the effect of JWH133 on microglia polarization. CONCLUSION The results indicate that CB2 receptor activation promotes MPP+-induced microglia transformation from M1 to M2 phenotype through PI3K/Akt/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Mengya Wang
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disorders, Qingdao University, Qingdao, 266071, China
| | - Man Liu
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disorders, Qingdao University, Qingdao, 266071, China
| | - Zegang Ma
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
18
|
Non-coding RNAs as key players in the neurodegenerative diseases: Multi-platform strategies and approaches for exploring the Genome's dark matter. J Chem Neuroanat 2023; 129:102236. [PMID: 36709005 DOI: 10.1016/j.jchemneu.2023.102236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
A growing amount of evidence in the last few years has begun to unravel that non-coding RNAs have a myriad of functions in gene regulation. Intensive investigation on non-coding RNAs (ncRNAs) has led to exploring their broad role in neurodegenerative diseases (NDs) owing to their regulatory role in gene expression. RNA sequencing technologies and transcriptome analysis has unveiled significant dysregulation of ncRNAs attributed to their biogenesis, upregulation, downregulation, aberrant epigenetic regulation, and abnormal transcription. Despite these advances, the understanding of their potential as therapeutic targets and biomarkers underpinning detailed mechanisms is still unknown. Advancements in bioinformatics and molecular technologies have improved our knowledge of the dark matter of the genome in terms of recognition and functional validation. This review aims to shed light on ncRNAs biogenesis, function, and potential role in NDs. Further deepening of their role is provided through a focus on the most recent platforms, experimental approaches, and computational analysis to investigate ncRNAs. Furthermore, this review summarizes and evaluates well-studied miRNAs, lncRNAs and circRNAs concerning their potential role in pathogenesis and use as biomarkers in NDs. Finally, a perspective on the main challenges and novel methods for the future and broad therapeutic use of ncRNAs is offered.
Collapse
|
19
|
Abrishamdar M, Jalali MS, Rashno M. MALAT1 lncRNA and Parkinson's Disease: The role in the Pathophysiology and Significance for Diagnostic and Therapeutic Approaches. Mol Neurobiol 2022; 59:5253-5262. [PMID: 35665903 DOI: 10.1007/s12035-022-02899-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/24/2022] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder. PD is characterized by progressive loss of dopamine-producing neurons in the substantia nigra (SN) region of brain tissue followed by the α-synuclein-based Lewy bodies' formation. These conditions are manifested by various motor and non-motor symptoms such as resting tremor, limb rigidity, bradykinesia and posture instability, cognitive impairment, sleep disorders, and emotional and memory dysfunctions. Long non-coding RNAs (lncRNAs) are closely related to protein-coding genes and are involved in various biological processes. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) lncRNA is involved in different pathways, including alternative splicing, transcriptional regulation, and post-transcriptional regulation, and also interacts with RNAs as a miRNA sponge. MALAT1 is highly expressed in brain tissues and several lines of evidence suggested it is probably involved in synapse generation and other neurophysiological pathways. This narrative review discussed all aspects of MALAT1-associated mechanisms involved in the PD pathogenesis, i.e., perturbed α-synuclein homeostasis, apoptosis and autophagy, and neuro-inflammation. Lastly, the possible applications of MALAT1 as a diagnostic biomarker and its importance to developing therapeutic strategies were highlighted. The literature search was conducted using neurodegeneration, neurodegenerative disorders, Parkinson's disease, lncRNA, and MALAT1 as search items in Google Scholar, Web of Knowledge, PubMed, and Scopus up to December 2021.
Collapse
Affiliation(s)
- M Abrishamdar
- Persian Gulf Physiology Research Center, Department of Physiology, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - M S Jalali
- Persian Gulf Physiology Research Center, Department of Physiology, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - M Rashno
- Department of Immunulogy, Cellular and Molecular Research Center, Medicine Faculty, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
20
|
Zhang H, Liu X, Liu Y, Liu J, Gong X, Li G, Tang M. Crosstalk between regulatory non-coding RNAs and oxidative stress in Parkinson’s disease. Front Aging Neurosci 2022; 14:975248. [PMID: 36016854 PMCID: PMC9396353 DOI: 10.3389/fnagi.2022.975248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease is the second most common neurodegenerative disease after Alzheimer’s disease, which imposes an ever-increasing burden on society. Many studies have indicated that oxidative stress may play an important role in Parkinson’s disease through multiple processes related to dysfunction or loss of neurons. Besides, several subtypes of non-coding RNAs are found to be involved in this neurodegenerative disorder. However, the interplay between oxidative stress and regulatory non-coding RNAs in Parkinson’s disease remains to be clarified. In this article, we comprehensively survey and overview the role of regulatory ncRNAs in combination with oxidative stress in Parkinson’s disease. The interaction between them is also summarized. We aim to provide readers with a relatively novel insight into the pathogenesis of Parkinson’s disease, which would contribute to the development of pre-clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Gang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
- *Correspondence: Gang Li Min Tang
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- *Correspondence: Gang Li Min Tang
| |
Collapse
|
21
|
The Role of Non-Coding RNAs in the Pathogenesis of Parkinson’s Disease: Recent Advancement. Pharmaceuticals (Basel) 2022; 15:ph15070811. [PMID: 35890110 PMCID: PMC9315906 DOI: 10.3390/ph15070811] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Parkinson’s disease (PD) is a prevalent neurodegenerative aging disorder that manifests as motor and non-motor symptoms, and its etiopathogenesis is influenced by non-coding RNAs (ncRNAs). Signal pathway and gene sequence studies have proposed that alteration of ncRNAs is relevant to the occurrence and development of PD. Furthermore, many studies on brain tissues and body fluids from patients with PD indicate that variations in ncRNAs and their target genes could trigger or exacerbate neurodegenerative pathogenesis and serve as potential non-invasive biomarkers of PD. Numerous ncRNAs have been considered regulators of apoptosis, α-syn misfolding and aggregation, mitochondrial dysfunction, autophagy, and neuroinflammation in PD etiology, and evidence is mounting for the determination of the role of competing endogenous RNA (ceRNA) mechanisms in disease development. In this review, we discuss the current knowledge regarding the regulation and function of ncRNAs as well as ceRNA networks in PD pathogenesis, focusing on microRNAs, long ncRNAs, and circular RNAs to increase the understanding of the disease and propose potential target identification and treatment in the early stages of PD.
Collapse
|
22
|
Zhou S, Chen R, She Y, Liu X, Zhao H, Li C, Jia Y. A new perspective on depression and neuroinflammation: Non-coding RNA. J Psychiatr Res 2022; 148:293-306. [PMID: 35193033 DOI: 10.1016/j.jpsychires.2022.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 12/19/2022]
Abstract
The high incidence and relapse rate of depression, as well comorbidity with other diseases, has made depression one of the primary causes of years of life lived with disability. Moreover, the unknown biological mechanism of depression has made treatment difficult. Neuroinflammation is important in the pathogenesis of depression. Neuroinflammation may affect depression by regulating the production of immune factors, immune cell activation, neuron generation, synaptic plasticity, and neurotransmission. Non-coding RNAs (ncRNAs) may be a breakthrough link between depression and neuroinflammation, as ncRNAs participate in these biological changes. We summarize the functions and mechanisms of ncRNAs in neuroinflammation and depression, and predict ncRNAs that may regulate the occurrence and progression of depression through neuritis. These findings not only broaden our understanding of the genetic regulation of depression and neuroinflammation but also provide a new perspective of the underlying mechanism and aid in the design of novel prevention, diagnosis, and treatment strategies.
Collapse
Affiliation(s)
- Shanyao Zhou
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China
| | - Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China.
| | - Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China
| | - Xuanjun Liu
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China
| | - Hui Zhao
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China
| | - Cheng Li
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China.
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
23
|
Li S, Bi G, Han S, Huang R. MicroRNAs Play a Role in Parkinson’s Disease by Regulating Microglia Function: From Pathogenetic Involvement to Therapeutic Potential. Front Mol Neurosci 2022; 14:744942. [PMID: 35126050 PMCID: PMC8814625 DOI: 10.3389/fnmol.2021.744942] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease (PD) is a clinically common neurodegenerative disease of the central nervous system (CNS) characterized by loss of dopamine neurons in the substantia nigra. Microglia (MG), as an innate immune cell in the CNS, are involved in a variety of immunity and inflammatory responses in the CNS. A number of studies have shown that the overactivation of MG is one of the critical pathophysiological mechanisms underlying PD. MicroRNAs (miRNAs) are considered to be an important class of gene expression regulators and are involved in a variety of physiological and pathological mechanisms, including immunity and inflammation. In addition, miRNAs can affect the progress of PD by regulating the expression of various MG genes and the polarization state of the MG. Here, we summarize recent articles and describe the important role of MG pathological polarization in the progression of PD, the diverse mechanisms responsible for how miRNAs regulate MG, and the potential therapeutic prospects of miRNAs for PD. We also propose that the regulation of miRNAs may be a novel protective approach against the pathogenesis of PD.
Collapse
Affiliation(s)
- Silu Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guorong Bi
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shunchang Han
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Rui Huang,
| |
Collapse
|
24
|
Ebrahimi R, Golestani A. The emerging role of noncoding RNAs in neuroinflammation: Implications in pathogenesis and therapeutic approaches. J Cell Physiol 2021; 237:1206-1224. [PMID: 34724212 DOI: 10.1002/jcp.30624] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/28/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022]
Abstract
Noncoding RNAs (ncRNAs) are important regulators of gene expression in different cell processes. Due to their ability in monitoring neural development genes, these transcripts confer neurons with the potential to exert broad control over the expression of genes for performing neurobiological functions. Although the change of ncRNA expression in different neurodegenerative diseases has been reviewed elsewhere, only recent evidence drove our attention to unravel the involvement of these molecules in neuroinflammation within these devastating disorders. Remarkably, the interactions between ncRNAs and inflammatory pathways are not fully recognized. Therefore, this review has focused on the interplay between diverse inflammatory pathways and the related ncRNAs, including microRNAs, long noncoding RNAs, and competing endogenous RNAs in Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, epilepsy, multiple sclerosis, Huntington's disease, and prion diseases. Providing novel insights in the field of combining biomarkers is a critical step for using them as diagnostic tools and therapeutic targets in clinical settings.
Collapse
Affiliation(s)
- Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most commonly prescribed medications for psychiatric disorders, yet they leave the majority of patients without full symptom relief. Therefore, a major research challenge is to identify novel targets for the improved treatment of these disorders. SSRIs act by blocking the serotonin transporter (SERT), the high-affinity, low-capacity, uptake-1 transporter for serotonin. Other classes of antidepressant work by blocking the norepinephrine or dopamine transporters (NET and DAT), the high-affinity, low-capacity uptake-1 transporters for norepinephrine and dopamine, or by blocking combinations of SERT, NET, and DAT. It has been proposed that uptake-2 transporters, which include organic cation transporters (OCTs) and the plasma membrane monoamine transporter (PMAT), undermine the therapeutic utility of uptake-1 acting antidepressants. Uptake-2 transporters for monoamines have low affinity for these neurotransmitters, but a high capacity to transport them. Thus, activity of these transporters may limit the increase of extracellular monoamines thought to be essential for ultimate therapeutic benefit. Here preclinical evidence supporting a role for OCT2, OCT3, and PMAT in behaviors relevant to psychiatric disorders is presented. Importantly, preclinical evidence revealing these transporters as targets for the development of novel therapeutics for psychiatric disorders is discussed.
Collapse
|
26
|
Ng J, Barral S, De La Fuente Barrigon C, Lignani G, Erdem FA, Wallings R, Privolizzi R, Rossignoli G, Alrashidi H, Heasman S, Meyer E, Ngoh A, Pope S, Karda R, Perocheau D, Baruteau J, Suff N, Antinao Diaz J, Schorge S, Vowles J, Marshall LR, Cowley SA, Sucic S, Freissmuth M, Counsell JR, Wade-Martins R, Heales SJR, Rahim AA, Bencze M, Waddington SN, Kurian MA. Gene therapy restores dopamine transporter expression and ameliorates pathology in iPSC and mouse models of infantile parkinsonism. Sci Transl Med 2021; 13:eaaw1564. [PMID: 34011628 PMCID: PMC7612279 DOI: 10.1126/scitranslmed.aaw1564] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/20/2020] [Accepted: 02/20/2021] [Indexed: 12/11/2022]
Abstract
Most inherited neurodegenerative disorders are incurable, and often only palliative treatment is available. Precision medicine has great potential to address this unmet clinical need. We explored this paradigm in dopamine transporter deficiency syndrome (DTDS), caused by biallelic loss-of-function mutations in SLC6A3, encoding the dopamine transporter (DAT). Patients present with early infantile hyperkinesia, severe progressive childhood parkinsonism, and raised cerebrospinal fluid dopamine metabolites. The absence of effective treatments and relentless disease course frequently leads to death in childhood. Using patient-derived induced pluripotent stem cells (iPSCs), we generated a midbrain dopaminergic (mDA) neuron model of DTDS that exhibited marked impairment of DAT activity, apoptotic neurodegeneration associated with TNFα-mediated inflammation, and dopamine toxicity. Partial restoration of DAT activity by the pharmacochaperone pifithrin-μ was mutation-specific. In contrast, lentiviral gene transfer of wild-type human SLC6A3 complementary DNA restored DAT activity and prevented neurodegeneration in all patient-derived mDA lines. To progress toward clinical translation, we used the knockout mouse model of DTDS that recapitulates human disease, exhibiting parkinsonism features, including tremor, bradykinesia, and premature death. Neonatal intracerebroventricular injection of human SLC6A3 using an adeno-associated virus (AAV) vector provided neuronal expression of human DAT, which ameliorated motor phenotype, life span, and neuronal survival in the substantia nigra and striatum, although off-target neurotoxic effects were seen at higher dosage. These were avoided with stereotactic delivery of AAV2.SLC6A3 gene therapy targeted to the midbrain of adult knockout mice, which rescued both motor phenotype and neurodegeneration, suggesting that targeted AAV gene therapy might be effective for patients with DTDS.
Collapse
Affiliation(s)
- Joanne Ng
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK.
| | | | - Gabriele Lignani
- Clinical and Experimental Epilepsy, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Fatma A Erdem
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
- Institute of Pharmacology and Gaston H. Glock Laboratories for Exploratory Drug Research, Centre of Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rebecca Wallings
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Riccardo Privolizzi
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Giada Rossignoli
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Haya Alrashidi
- Genetics and Genomic Medicine, GOS-Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Sonja Heasman
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Esther Meyer
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Adeline Ngoh
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
| | - Simon Pope
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Rajvinder Karda
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
| | - Dany Perocheau
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
| | - Julien Baruteau
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
- Genetics and Genomic Medicine, GOS-Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Natalie Suff
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
- Department of Women and Children's Health, King's College London, London, WC2R 2LS, UK
| | - Juan Antinao Diaz
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK
| | - Stephanie Schorge
- Clinical and Experimental Epilepsy, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Pharmacology, School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Lucy R Marshall
- Infection, Immunity, Inflammation, GOS-Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Sonja Sucic
- Institute of Pharmacology and Gaston H. Glock Laboratories for Exploratory Drug Research, Centre of Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and Gaston H. Glock Laboratories for Exploratory Drug Research, Centre of Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - John R Counsell
- Developmental Neurosciences, GOS-Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Simon J R Heales
- Genetics and Genomic Medicine, GOS-Institute of Child Health, University College London, London, WC1N 1EH, UK
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Ahad A Rahim
- Pharmacology, School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Maximilien Bencze
- Developmental Neurosciences, GOS-Institute of Child Health, University College London, London, WC1N 1EH, UK
- University Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Simon N Waddington
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, WC1E 6HX, UK.
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, 2193 Johannesburg, South Africa
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, GOS-Institute of Child Health, University College London, London, WC1N 1DZ, UK
- Department of Neurology, Great Ormond Street Hospital for Children, London, WC1N 3JH, UK
| |
Collapse
|
27
|
Abstract
Catecholamines, including dopamine, norepinephrine, and epinephrine, are modulatory transmitters released from specialized neurons throughout the brain. Collectively, catecholamines exert powerful regulation of mood, motivation, arousal, and plasticity. Transporter-mediated uptake determines the peak concentration, duration, and physical spread of released catecholamines, thus playing key roles in determining the magnitude and duration of their modulatory effects. Most studies of catecholamine clearance have focused on the presynaptic high-affinity, low-capacity dopamine (DAT), and norepinephrine (NET) transporters, which are members of the uptake1 family of monoamine transporters. However, recent studies have demonstrated that members of the uptake2 family of monoamine transporters, including organic cation transporter 2 (OCT2), OCT3, and the plasma membrane monoamine transporter (PMAT) are expressed widely throughout the brain. In contrast to DAT and NET, these transporters have higher capacity and lower affinity for catecholamines and are multi-specific, each with the capacity to transport all catecholamines. The expression of these transporters in the brain suggests that they play significant roles in regulating catecholamine homeostasis. This review summarizes studies describing the anatomical distribution of OCT2, OCT3, and PMAT, their cellular and subcellular localization, and their contribution to the regulation of the clearance of catecholamines in the brain.
Collapse
|
28
|
Ji Q, Li X. Mechanism of Dopaminergic Nerve Transmission in Different Doses of Morphine Addiction and Stress-Induced Depression. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:9987441. [PMID: 34055279 PMCID: PMC8131158 DOI: 10.1155/2021/9987441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022]
Abstract
Depression not only threatens the health and quality of life of patients but also brings a huge mental and economic burden to the patients' families. This paper mainly studies the mechanism of dopaminergic neurotransmission in different doses of morphine addiction and stress-induced depression. In the experiment, 40 male SD rats were selected. The experiment established a rat model of chronic stress depression. The rats used in this model are all raised in a single cage, and there will be various stimuli every day for 21 days, but high-intensity continuous stimuli must be avoided, and the same stimuli will not appear continuously. The experiment established a depression animal model through chronic unpredictable mild stress (CUMS), combined with the conditioned position preference (CPP) model of morphine addiction to detect the establishment of CPP in such animals, so as to explore certain stress stimuli or depression, the influence on morphine addiction, and the relationship between them. The second or third branches of pyramidal neurons were selected to analyze the PL and CA3 regions. When analyzing the density of dendrites, each animal selected at least 8 dendrites in order to count the number of dendrites and selected a length of 20 μm on each branch to record the number of dendrites. All measured values are expressed as average ± standard deviation and analyzed by SPSS17.0 statistical software, and Levene test is used in the scattered consistency test. The average NIV of PEN before injection was 11.92 ± 2.90 Hz, and the average latency was 0.16 ± 0.03 s. The results indicate that CUMS may reduce the conditioned learning and memory ability by damaging the learning loop, rather than affecting the reward loop to weaken the establishment of morphine-dependent CPP.
Collapse
Affiliation(s)
- Qing Ji
- Graduate School, Jiamusi University, Jiamusi 154000, Heilongjiang, China
| | - Xin Li
- Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi 154000, Heilongjiang, China
| |
Collapse
|
29
|
Sweet DH. Organic Cation Transporter Expression and Function in the CNS. Handb Exp Pharmacol 2021; 266:41-80. [PMID: 33963461 DOI: 10.1007/164_2021_463] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) represent major control checkpoints protecting the CNS, by exerting selective control over the movement of organic cations and anions into and out of the CNS compartment. In addition, multiple CNS cell types, e.g., astrocytes, ependymal cells, microglia, contribute to processes that maintain the status quo of the CNS milieu. To fulfill their roles, these barriers and cell types express a multitude of transporter proteins from dozens of different transporter families. Fundamental advances over the past few decades in our knowledge of transporter substrates, expression profiles, and consequences of loss of function are beginning to change basic theories regarding the contribution of various cell types and clearance networks to coordinated neuronal signaling, complex organismal behaviors, and overall CNS homeostasis. In particular, transporters belonging to the Solute Carrier (SLC) superfamily are emerging as major contributors, including the SLC22 organic cation/anion/zwitterion family of transporters (includes OCT1-3 and OCTN1-3), the SLC29 facilitative nucleoside family of transporters (includes PMAT), and the SLC47 multidrug and toxin extrusion family of transporters (includes MATE1-2). These transporters are known to interact with neurotransmitters, antidepressant and anxiolytic agents, and drugs of abuse. Clarifying their contributions to the underlying mechanisms regulating CNS permeation and clearance, as well as the health status of astrocyte, microglial and neuronal cell populations, will drive new levels of understanding as to maintenance of the CNS milieu and approaches to new therapeutics and therapeutic strategies in the treatment of CNS disorders. This chapter highlights organic cation transporters belonging to the SLC superfamily known to be expressed in the CNS, providing an overview of their identification, mechanism of action, CNS expression profile, interaction with neurotransmitters and antidepressant/antipsychotic drugs, and results from behavioral studies conducted in loss of function models (knockout/knockdown).
Collapse
Affiliation(s)
- Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
30
|
Meng X, Zhong J, Zeng C, Yung KKL, Zhang X, Wu X, Qu S. MiR-30a-5p Regulates GLT-1 Function via a PKCα-Mediated Ubiquitin Degradation Pathway in a Mouse Model of Parkinson's Disease. ACS Chem Neurosci 2021; 12:1578-1592. [PMID: 33882234 DOI: 10.1021/acschemneuro.1c00076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Glutamate excitotoxicity is caused by dysfunctional glutamate transporters and plays an important role in the pathogenesis of Parkinson's disease (PD); however, the mechanisms that underlie the regulation of glutamate transporters in PD are still not fully elucidated. MicroRNAs(miRNA), which are abundant in astrocytes and neurons, have been reported to play key roles in regulating the translation of glutamate-transporter mRNA. In this study, we hypothesized that the miR-30a-5p contributes to the pathogenesis of PD by regulating the ubiquitin-mediated degradation of glutamate transporter 1 (GLT-1). We demonstrated that short-hairpin RNA-mediated knockdown of miR-30a-5p ameliorated motor deficits and pathological changes like astrogliosis and reactive microgliosis in a mouse model of PD (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice). Western blotting and immunofluorescent labeling revealed that miR-30a-5p suppressed the expression and function of GLT-1 in MPTP-treated mice and specifically in astrocytes treated with 1-methyl-4-phenylpyridinium (MPP+) (cell model of PD). Both in vitro and in vivo, we found that miR-30a-5p knockdown promoted glutamate uptake and increased GLT-1 expression by hindering GLT-1 ubiquitination and subsequent degradation in a PKCα-dependent manner. Therefore, we conclude that miR-30a-5p represents a potential therapeutic target for the treatment of PD.
Collapse
Affiliation(s)
- Xingjun Meng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianping Zhong
- Department of Neurology, Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
| | - Chong Zeng
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Xiuping Zhang
- Teaching Center of Experimental Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaojuan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shaogang Qu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
31
|
Islas-Cortez M, Rios C, Rubio-Osornio M, Zamudio S, Orozco-Suarez S, Mendez-Armenta M, Nava-Ruiz C, Diaz-Ruiz A. Characterization of the antiapoptotic effect of copper sulfate on striatal and midbrain damage induced by MPP + in rats. Neurotoxicology 2020; 82:18-25. [PMID: 33127410 DOI: 10.1016/j.neuro.2020.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/18/2023]
Abstract
1-Methyl-4-phenylpyridinium ion (MPP+)-induced neurotoxicity produces cellular damage resembling that encountered in Parkinson's disease. The mechanisms of cellular death after MPP+ include the participation of oxidative stress in the loss of dopaminergic neurons. Among the mechanisms of defense against oxidative stress, several copper-dependent proteins have been implicated: Cu/Zn-SOD, ceruloplasmin, and metallothionein. Another important mechanism of damage, is MPP + interference with mitochondrial respiration. Both, oxidative stress and inhibition of mitochondrial respiration may trigger apoptosis in the neurons after MPP+. The aim of the present study was to characterize the time-course of apoptosis induced by MPP+ to determine if copper sulfate pretreatment is able to prevent the activation of caspases and decreased the neuronal apoptosis. MPP+ was microinjected into rat striatum using a stereotactic frame. The results showed increased activities of caspases 8, 9 and 3, between 72-120 hours after administration of MPP+, both in striatum and midbrain. After this study, we tested the effect of CuSO4 on MPP+ neurotoxicity, showing a diminution of the apoptotic damage induced by MPP+, decreased levels of enzymatic activity of caspases: 8 (-34 and -25 %), 9 (-25 and -42 %) and 3 (-40 and -29 %) in striatum and midbrain, respectively. Finally, we performed an immunohistochemical analysis, evidencing a decreased number of apoptotic cells in the groups pretreated with copper sulfate pretreatment compared to the control group. With these findings, it is concluded that pretreatment with copper sulfate may be a good alternative to prevent MPP+-induced apoptosis.
Collapse
Affiliation(s)
- Marcela Islas-Cortez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico; Departamento de Fisiología, Laboratorio de Neurociencia Conductual, Instituto Politécnico Nacional, Unidad Zacatenco, Ciudad de México, Mexico
| | - Camilo Rios
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico; Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, Mexico
| | - Moisés Rubio-Osornio
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Sergio Zamudio
- Departamento de Fisiología, Laboratorio de Neurociencia Conductual, Instituto Politécnico Nacional, Unidad Zacatenco, Ciudad de México, Mexico
| | - Sandra Orozco-Suarez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México, Mexico
| | - Marisela Mendez-Armenta
- Departamento de Neuropatología Experimental Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Concepción Nava-Ruiz
- Departamento de Neuropatología Experimental Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico.
| |
Collapse
|
32
|
Cai LJ, Tu L, Huang XM, Huang J, Qiu N, Xie GH, Liao JX, Du W, Zhang YY, Tian JY. LncRNA MALAT1 facilitates inflammasome activation via epigenetic suppression of Nrf2 in Parkinson's disease. Mol Brain 2020; 13:130. [PMID: 32972446 PMCID: PMC7513532 DOI: 10.1186/s13041-020-00656-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/14/2020] [Indexed: 01/01/2023] Open
Abstract
The goal of the present study was to elucidate the mechanism by which long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (lncRNA MALAT1) promotes inflammation in Parkinson’s disease (PD). 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was used to induce PD development in C57BL/6 mice, and tyrosine hydroxylase (TH) expression was analysed by immunohistochemical analysis. Western blot and qPCR analyses were conducted to assess the expression of protein and mRNA levels, respectively. Lipopolysaccharide/adenosine triphosphate (LPS/ATP) was used to activate microglia in vitro. Chromatin immunoprecipitation (ChIP), RNA pull-down and RNA immunoprecipitation chip (RIP) assays were performed to investigate the interaction among specific molecules. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to evaluate cell viability and proliferation. Flow cytometry was performed to analyse cell apoptosis after staining. The dichlorofluorescein diacetate (DCFH-DA) assay was used to measure the generation of reactive oxygen species (ROS) in cells. The results showed that MALAT1 was highly expressed in the brains of MPTP-induced PD model mice and in LPS/ATP-induced microglia cells. Knockdown of MALAT1 inhibited elevated nuclear factor (erythroid-derived 2)-like-2 factor (NRF2) expression, thereby inhibiting inflammasome activation and ROS production. MALAT1 was shown to promote neuroinflammation by recruiting enhancer of zeste homologue 2 (EZH2) to the promoter of NRF2, suppressing Nrf2 expression. In summary, MALAT1 epigenetically inhibits NRF2, thereby inducing inflammasome activation and reactive oxygen species (ROS) production in PD mouse and microglial cell models.
Collapse
Affiliation(s)
- Li-Jun Cai
- Department of Neurology, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Li Tu
- Department of General Medical, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, P.R. China
| | - Xiao-Mo Huang
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Jia Huang
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Nan Qiu
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Guang-Hong Xie
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Jian-Xiong Liao
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Wei Du
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Ying-Yue Zhang
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China
| | - Jin-Yong Tian
- Department of Emergency, Guizhou Provincial People's Hospital, No.83 Zhongshan East Road, Guiyang, 550002, Guizhou Province, P.R. China.
| |
Collapse
|
33
|
Zhai K, Shi XY, Yi FS, Huang ZY, Wu XZ, Dong SF, Wang W, Wu MT, Shi HZ. IL-10 promotes malignant pleural effusion by regulating T H 1 response via an miR-7116-5p/GPR55/ERK pathway in mice. Eur J Immunol 2020; 50:1798-1809. [PMID: 32506440 DOI: 10.1002/eji.202048574] [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: 01/30/2020] [Revised: 05/02/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
IL-10, produced by a wide variety of cells, is a highly pleiotropic cytokine that plays a critical role in the control of immune responses. However, its regulatory activity in tumor immunity remains poorly understood. In this study, we report that IL-10 deficiency robustly suppressed the formation of malignant pleural effusion (MPE) and significantly enhanced miR-7116-5p expression in pleural CD4+ T cells. We demonstrated that miR-7116-5p suppressed IL-10-mediated MPE formation by inhibiting pleural vascular permeability as well as tumor angiogenesis and tumor growth. IL-10 promoted MPE formation by suppressing miR-7116-5p that enhances TH 1 response. We identified G protein-coupled receptor 55 (GPR55) as a potential target of miR-7116-5p, and miR-7116-5p promoted TH 1 cell function by downregulating GPR55. Moreover, GPR55 promoted MPE formation by inhibiting TH 1 cell expansion through the ERK phosphorylation pathway. These results uncover an IL-10-mediated pathway controlling TH 1 cells and demonstrate a central role for miR-7116-5p/GPR55/ERK signaling in the physiological regulation of IL-10-driven pro-malignant responses.
Collapse
Affiliation(s)
- Kan Zhai
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xin-Yu Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Feng-Shuang Yi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhong-Yin Huang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiu-Zhi Wu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shu-Feng Dong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Min-Ting Wu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huan-Zhong Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Jiang W, Wang X, Wang W, Hua F, Zhang Z, Zhang Z, Xiang J, Yang X. Inhibition of NK1R attenuates LPS-induced microglial inflammation and consequent death of PC12 cells. Brain Res Bull 2020; 162:115-124. [PMID: 32540418 DOI: 10.1016/j.brainresbull.2020.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 10/24/2022]
Abstract
Microglia, the resident immune cells in the central nervous system, play a critical role under physiological conditions, but they may be activated and exaggerate the pathological development of Parkinson's disease (PD). Recent reports have suggested that neurokinin 1 receptor (NK1R) is involved in various inflammatory diseases, including PD. However, whether neurokinin 1 (NK1) is involved in the activation of microglial cells remains unclear. In the present study, we found that (1) NK1R is located in microglial cells and upregulated in lipopolysaccharide (LPS)-activated BV2 microglia. Application of CP-99994, a selective antagonist of NK1R, inhibited the production of inflammatory mediators such as tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), IL-6, inducible macrophage-type nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in activated BV2 cells. (2) NK1R antagonist suppressed the morphological changes in LPS-stimulated BV2. (3) Microglial inactivation by NK1R antagonist resulted in decreased microglial migration. (4) NK1R antagonist reduced nuclear translocation of nuclear factor kappa-B (NF-κB) and attenuated phosphorylation of mitogen-activated protein kinases (MAPKs) in LPS-stimulated BV2. (5) The cell death of PC12 induced by microglia-mediated neuroinflammation was reversed in a Transwell co-culture system by NK1R antagonist. Collectively, these results showed that inhibition of NK1R attenuates LPS-induced microglial inflammatory response and dopaminergic neurotoxicity, which may be due to the decreased MAPK/NF-κB signal pathway. Thus, NK1R may be a therapeutic target in neuroinflammation, especially in PD.
Collapse
Affiliation(s)
- Weifeng Jiang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaoying Wang
- Department of Ultrasound, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wei Wang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zuohui Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jie Xiang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Xinxin Yang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| |
Collapse
|
35
|
Liu WW, Wei SZ, Huang GD, Liu LB, Gu C, Shen Y, Wang XH, Xia ST, Xie AM, Hu LF, Wang F, Liu CF. BMAL1 regulation of microglia-mediated neuroinflammation in MPTP-induced Parkinson's disease mouse model. FASEB J 2020; 34:6570-6581. [PMID: 32246801 DOI: 10.1096/fj.201901565rr] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/21/2019] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
Dysfunction of the circadian rhythm is one of most common nonmotor symptoms in Parkinson's disease (PD), but the molecular role of the circadian rhythm in PD is unclear. We here showed that inactivation of brain and muscle ARNT-like 1 (BMAL1) in 1-methyl-4-phenyl-1,2,4,5-tetrahydropyridine (MPTP)-treated mice resulted in obvious motor functional deficit, loss of dopaminergic neurons (DANs) in the substantia nigra pars compacta (SNpc), decrease of dopamine (DA) transmitter, and increased activation of microglia and astrocytes in the striatum. Time on the rotarod or calorie consumption, and food and water intake were reduced in the Bmal1-/- mice after MPTP treatment, suggesting that absence of Bmal1 may exacerbate circadian and PD motor function. We observed a significant reduction of DANs (~35%) in the SNpc, the tyrosine hydroxylase protein level in the striatum (~60%), the DA (~22%), and 3,4-dihydroxyphenylacetic acid content (~29%), respectively, in MPTP-treated Bmal1-/- mice. Loss of Bmal1 aggravated the inflammatory reaction both in vivo and in vitro. These findings suggest that BMAL1 may play an essential role in the survival of DANs and maintain normal function of the DA signaling pathway via regulating microglia-mediated neuroinflammation in the brain.
Collapse
Affiliation(s)
- Wen-Wen Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Shi-Zhuang Wei
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Guo-Dong Huang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Lu-Bing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chao Gu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yun Shen
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xian-Hui Wang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, China
| | - Shu-Ting Xia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - An-Mu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li-Fang Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
36
|
Titze-de-Almeida SS, Soto-Sánchez C, Fernandez E, Koprich JB, Brotchie JM, Titze-de-Almeida R. The Promise and Challenges of Developing miRNA-Based Therapeutics for Parkinson's Disease. Cells 2020; 9:cells9040841. [PMID: 32244357 PMCID: PMC7226753 DOI: 10.3390/cells9040841] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are small double-stranded RNAs that exert a fine-tuning sequence-specific regulation of cell transcriptome. While one unique miRNA regulates hundreds of mRNAs, each mRNA molecule is commonly regulated by various miRNAs that bind to complementary sequences at 3’-untranslated regions for triggering the mechanism of RNA interference. Unfortunately, dysregulated miRNAs play critical roles in many disorders, including Parkinson’s disease (PD), the second most prevalent neurodegenerative disease in the world. Treatment of this slowly, progressive, and yet incurable pathology challenges neurologists. In addition to L-DOPA that restores dopaminergic transmission and ameliorate motor signs (i.e., bradykinesia, rigidity, tremors), patients commonly receive medication for mood disorders and autonomic dysfunctions. However, the effectiveness of L-DOPA declines over time, and the L-DOPA-induced dyskinesias commonly appear and become highly disabling. The discovery of more effective therapies capable of slowing disease progression –a neuroprotective agent–remains a critical need in PD. The present review focus on miRNAs as promising drug targets for PD, examining their role in underlying mechanisms of the disease, the strategies for controlling aberrant expressions, and, finally, the current technologies for translating these small molecules from bench to clinics.
Collapse
Affiliation(s)
- Simoneide S. Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, FAV, University of Brasilia, Brasília 70910-900, Brazil;
| | - Cristina Soto-Sánchez
- Neuroprosthetics and Visual Rehabilitation Research Unit, Bioengineering Institute, Miguel Hernández University, 03202 Alicante, Spain; (C.S.-S.); (E.F.)
| | - Eduardo Fernandez
- Neuroprosthetics and Visual Rehabilitation Research Unit, Bioengineering Institute, Miguel Hernández University, 03202 Alicante, Spain; (C.S.-S.); (E.F.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine—CIBER-BBN, 28029 Madrid, Spain
| | - James B. Koprich
- Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada; (J.B.K.); (J.M.B.)
| | - Jonathan M. Brotchie
- Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada; (J.B.K.); (J.M.B.)
| | - Ricardo Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, FAV, University of Brasilia, Brasília 70910-900, Brazil;
- Correspondence: ; Tel.: +55-61-3107-7222
| |
Collapse
|
37
|
Lu J, Dou F, Yu Z. The potassium channel KCa3.1 represents a valid pharmacological target for microgliosis-induced neuronal impairment in a mouse model of Parkinson's disease. J Neuroinflammation 2019; 16:273. [PMID: 31878950 PMCID: PMC6931251 DOI: 10.1186/s12974-019-1682-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Background Recent studies described a critical role for microglia in Parkinson’s disease (PD), where these central nerve system resident immune cells participate in the neuroinflammatory microenvironment that contributes to dopaminergic neurons loss in the substantia nigra. Understanding the phenotype switch of microgliosis in PD could help to identify the molecular mechanism which could attenuate or delay the progressive decline in motor function. KCa3.1 has been reported to regulate the “pro-inflammatory” phenotype switch of microglia in neurodegenerative pathological conditions. Methods We here investigated the effects of gene deletion or pharmacological blockade of KCa3.1 activity in wild-type or KCa3.1−/− mice after treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a mouse model of PD. MPTP-induced PD mouse model was subjected to the rotarod test to evaluate the locomotor ability. Glia activation and neuron loss were measured by immunostaining. Fluo-4 AM was used to measure cytosolic Ca2+ level in 1-methyl-4-phenylpyridinium (MPP+)-induced microgliosis in vitro. Results We report that treatment of MPTP-induced PD mouse model with gene deletion or pharmacological blockade of KCa3.1 with senicapoc improves the locomotor ability and the tyrosine hydroxylase (TH)-positive neuron number and attenuates the microgliosis and neuroinflammation in the substantia nigra pars compacta (SNpc). KCa3.1 involves in store-operated Ca2+ entry-induced Ca2+ overload and endoplasmic reticulum stress via the protein kinase B (AKT) signaling pathway during microgliosis. Gene deletion or blockade of KCa3.1 restored AKT/mammalian target of rapamycin (mTOR) signaling both in vivo and in vitro. Conclusions Taken together, these results demonstrate a key role for KCa3.1 in driving a pro-inflammatory microglia phenotype in PD.
Collapse
Affiliation(s)
- Jia Lu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Fangfang Dou
- Basic Research Department, Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
38
|
Liu Z, Qiu AW, Huang Y, Yang Y, Chen JN, Gu TT, Cao BB, Qiu YH, Peng YP. IL-17A exacerbates neuroinflammation and neurodegeneration by activating microglia in rodent models of Parkinson's disease. Brain Behav Immun 2019; 81:630-645. [PMID: 31351185 DOI: 10.1016/j.bbi.2019.07.026] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 12/26/2022] Open
Abstract
Neuroinflammation has been involved in pathogenesis of Parkinson's disease (PD), a chronic neurodegenerative disease characterized neuropathologically by progressive dopaminergic neuronal loss in the substantia nigra (SN). We recently have shown that helper T (Th)17 cells facilitate dopaminergic neuronal loss in vitro. Herein, we demonstrated that interleukin (IL)-17A, a proinflammatory cytokine produced mainly by Th17 cells, contributed to PD pathogenesis depending on microglia. Mouse and rat models for PD were prepared by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or striatal injection of 1-methyl-4-phenylpyridinium (MPP+), respectively. Both in MPTP-treated mice and MPP+-treated rats, blood-brain barrier (BBB) was disrupted and IL-17A level increased in the SN but not in cortex. Effector T (Teff) cells that were adoptively transferred via tail veins infiltrated into the brain of PD mice but not into that of normal mice. The Teff cell transfer aggravated nigrostriatal dopaminergic neurodegeneration, microglial activation and motor impairment. Contrarily, IL-17A deficiency alleviated BBB disruption, dopaminergic neurodegeneration, microglial activation and motor impairment. Anti-IL-17A-neutralizing antibody that was injected into lateral cerebral ventricle in PD rats ameliorated the manifestations mentioned above. IL-17A activated microglia but did not directly affect dopaminergic neuronal survival in vitro. IL-17A exacerbated dopaminergic neuronal loss only in the presence of microglia, and silencing IL-17A receptor gene in microglia abolished the IL-17A effect. IL-17A-treated microglial medium that contained higher concentration of tumor necrosis factor (TNF)-α facilitated dopaminergic neuronal death. Further, TNF-α-neutralizing antibody attenuated MPP+-induced neurotoxicity. The findings suggest that IL-17A accelerates neurodegeneration in PD depending on microglial activation and at least partly TNF-α release.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ao-Wang Qiu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ya Yang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jin-Na Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ting-Ting Gu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Bei-Bei Cao
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
39
|
Porro C, Panaro MA, Lofrumento DD, Hasalla E, Trotta T. The multiple roles of exosomes in Parkinson's disease: an overview. Immunopharmacol Immunotoxicol 2019; 41:469-476. [PMID: 31405314 DOI: 10.1080/08923973.2019.1650371] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The extracellular vesicles (EVs) represent a relatively new field of research in neurodegenerative disease and they are thought to be one of the ways that neurodegenerative pathologies, such as Parkinson's Disease (PD), spread in the brain. EVs are membrane vesicles released from cells into the extracellular space and they are produced by all cells of the nervous tissue. The classification of the vesicle subtypes comprises exosomes, microvesicles/microparticles, apoptotic bodies. EVs change in number and content in response to environmental conditions and may function as shuttles for the delivery of cargo between cells. Recent data suggest that exosomes secreted by both activated microglia and neurons play an important role in α-synuclein (α-syn) spreading and increase of neuroinflammation, thus exacerbating neuronal dysfunction and disease progression. α-syn is a presynaptic protein secreted by neurons in small amounts, and it is the main component of Lewy bodies, one of the histopathological features of PD. Several factors have shown to induce and/or modulate α-syn structure and oligomerization in vitro. Under pathological conditions, progressive accumulation of α-syn and the formation of oligomers have been proposed to play a critical role in the pathogenesis of PD. This review gives an overview about the multiple roles of exosomes in PD, despite their role in the progression of neurodegeneration, exosomes could represent a specific drug delivery tool for a difficult target such as the brain, which poses an obstacle to most drugs and they could also represent new biomarkers to track the progression of PD.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari , Italy
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento , Lecce , Italy
| | - Elona Hasalla
- Department of Pre-Clinic Subjects, Faculty of Medical Sciences, University of Elbasan "Aleksander Xhuvani" , Elbasan , Albania
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia , Foggia , Italy
| |
Collapse
|
40
|
Hinkle JT, Dawson VL, Dawson TM. The A1 astrocyte paradigm: New avenues for pharmacological intervention in neurodegeneration. Mov Disord 2019; 34:959-969. [PMID: 31136698 PMCID: PMC6642014 DOI: 10.1002/mds.27718] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/27/2019] [Accepted: 04/24/2019] [Indexed: 12/25/2022] Open
Abstract
We recently demonstrated that NLY01, a novel glucagon-like peptide-1 receptor agonist, exerts neuroprotective effects in two mouse models of PD in a glia-dependent manner. NLY01 prevented microglia from releasing inflammatory mediators known to convert astrocytes into a neurotoxic A1 reactive subtype. Importantly, we provided evidence that this neuroprotection was not mediated by a direct action of NLY01 on neurons or astrocytes (e.g., by activating neurotrophic pathways or modulating astrocyte reactivity per se). In the present article, we provide a generalist review of microglia and astrocytes in neurodegeneration and discuss the emerging paradigm of A1 astrocyte neurotoxicity in more detail. We comment on specific inferences that are naturally suggested by our work in this area and the differential level of support it offers to each. Finally, we discuss implications for the overall goal of creating disease-modifying therapies for PD, survey emerging methodologies for accelerating translational research on glia in neurodegeneration, and describe expected challenges for developing glia-directed therapies that do not impede essential physiological functions carried out by glia in the CNS. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jared T. Hinkle
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L. Dawson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology,Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Ted M. Dawson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology,Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
41
|
Yao L, Zhu Z, Wu J, Zhang Y, Zhang H, Sun X, Qian C, Wang B, Xie L, Zhang S, Lu G. MicroRNA-124 regulates the expression of p62/p38 and promotes autophagy in the inflammatory pathogenesis of Parkinson's disease. FASEB J 2019; 33:8648-8665. [PMID: 30995872 DOI: 10.1096/fj.201900363r] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor and nonmotor symptoms due to the selective loss of midbrain dopaminergic neurons. The evidence for a chronic inflammatory reaction mediated by microglial cells in the brain is particularly strong in PD. In our previous study, we have shown that brain-specific microRNA-124 (miR-124) is significantly down-regulated in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD and that it can also inhibit neuroinflammation during the development of PD. However, further investigation is required to understand whether the abnormal expression of miR-124 regulates microglial activation. In this study, we found that the expression of sequestosome 1 (p62) and phospho-p38 mitogen-activated protein kinases (p-p38) showed a significant increase in LPS-treated immortalized murine microglial cell line BV2 cells in an MPTP-induced mouse model of PD. Knockdown of p62 could suppress the secretion of proinflammatory cytokines and p-p38 of microglia. Besides, inhibition of p38 suppressed the secretion of proinflammatory cytokines and promoted autophagy in BV2 cells. Moreover, our study is the first to identify a unique role of miR-124 in mediating the microglial inflammatory response by targeting p62 and p38 in PD. In the microglial culture supernatant transfer model, the knockdown of p62 in BV2 cells prevented apoptosis and death of human neuroblastoma cell lines SH-SY5Y (SH-SY5Y) cells following microglia activation. In addition, the exogenous delivery of miR-124 could suppress p62 and p-p38 expression and could also attenuate the activation of microglia in the substantia nigra par compacta of MPTP-treated mice. Taken together, our data suggest that miR-124 could inhibit neuroinflammation during the development of PD by targeting p62, p38, and autophagy, indicating that miR-124 could be a potential therapeutic target for regulating the inflammatory response in PD.-Yao, L., Zhu, Z., Wu, J., Zhang, Y., Zhang, H., Sun, X., Qian, C., Wang, B., Xie, L., Zhang, S., Lu, G. MicroRNA-124 regulates the expression of p62/p38 and promotes autophagy in the inflammatory pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Longping Yao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Zhiyuan Zhu
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Jiayu Wu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yizhou Zhang
- Tarbut V'Torah Community Day School, Irvine, California, USA
| | - Hongbo Zhang
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Xiang Sun
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Chen Qian
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Baoyan Wang
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Linghai Xie
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Shizhong Zhang
- Department of Neurosurgery Southern Medical University, Guangzhou, China.,The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
42
|
Jiao Z, Zhang W, Chen C, Zhu X, Chen X, Zhou M, Peng G, Liu H, Qiu J, Lin Y, Huang S, Mo M, Yang X, Qu S, Xu P. Gene Dysfunction Mediates Immune Response to Dopaminergic Degeneration in Parkinson's Disease. ACS Chem Neurosci 2019; 10:803-811. [PMID: 30289236 DOI: 10.1021/acschemneuro.8b00373] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many publications reported that genetic dysfunction mediates abnormal immune responses in the brain, which is important for the development of neurodegenerative diseases, especially for Parkinson's disease (PD). This immune disorder results in subsequent inflammatory reaction, which stimulates microglia or other immune cells to secrete cytokines and chemokines and disturbs the proportion of peripheral blood lymphocyte subsets contributing to dopaminergic (DA) neuron apoptosis. Furthermore, the abnormal immune related signal pathways caused by genetic variants promote chronic inflammation destroying the blood-brain barrier, which allows infiltration of different molecules and blood cells into the central nervous system (CNS) exerting toxicity on DA neurons. As a result, the inflammatory reaction in the CNS accelerates the progression of Parkinson's disease and promotes α-synuclein aggregation and diffusion among DA neurons in the procession of Parkinson's disease. Thus, for disease evaluation, the genetic mediated abnormal immune response in PD may be assessed based on the multiple immune molecules and inflammatory factors, as well as the ratio of lymphocyte subsets from PD patient's peripheral blood as potential biomarkers.
Collapse
Affiliation(s)
- Zhigang Jiao
- Central Laboratory, Shunde Hospital, Southern Medical University, Foshan 528300, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Foshan 528300, China
| | - Wenlong Zhang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Chaojun Chen
- Department of Neurology, Guangzhou Chinese Medical Integrated Hospital (Huadu), Guangdong 510800, China
| | - Xiaoqin Zhu
- Guangzhou Medical University, Guangzhou 511436, China
| | - Xiang Chen
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Miaomiao Zhou
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Guoyou Peng
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Hanqun Liu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jiewen Qiu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuwan Lin
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Shuxuan Huang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Mingshu Mo
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Xinling Yang
- Department of Neurology, the Third Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Shaogang Qu
- Central Laboratory, Shunde Hospital, Southern Medical University, Foshan 528300, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Foshan 528300, China
| | - Pingyi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
43
|
Activation of microglia synergistically enhances neurodegeneration caused by MPP + in human SH-SY5Y cells. Eur J Pharmacol 2019; 850:64-74. [PMID: 30684467 DOI: 10.1016/j.ejphar.2019.01.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 01/21/2023]
Abstract
While MPP+ may not directly activate microglia, the initial neuronal damage inflicted by the toxin may trigger microglia, possibly leading to synergistic pro-apoptotic interaction between neuro-inflammation and toxin-induced neurotoxicity, which may further aggravate neurodegeneration. However, what molecular targets are synergistically up or downregulated during this interaction is not well understood. Here, we addressed this by co-culturing fully differentiated human SH-SY5Y cells treated with parkinsonian toxin 1-Methyl-4-phenylpyridinium (MPP+), with endotoxin-activated microglial cell line EOC 20 to determine how this interaction affects pro-apoptotic (p38, JNK, and bax:bcl2 ratios) and pro-survival (NF-κB, MEK1) signaling at both mRNA and protein levels. Concurrent MPP+ and endotoxin-treatment aggravated a decrease in SH-SY5Y cell viability and caused strong synergistic increases in the bax:bcl2 ratio, but also NF-κB and JNK signaling. These effects were attenuated by microglia inhibitor minocycline. Altogether, these data provide further molecular insights into the important role or even conditional requirement of microglia activation in the progressive neurodegenerative nature of PD.
Collapse
|
44
|
Yang R, Wang H, Wen J, Ma K, Chen D, Chen Z, Huang C. Regulation of microglial process elongation, a featured characteristic of microglial plasticity. Pharmacol Res 2018; 139:286-297. [PMID: 30476531 DOI: 10.1016/j.phrs.2018.11.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/08/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
Abstract
Microglia, a type of glia within the brain characterized by a ramified morphology, are essential for removing neuronal debris and restricting the expansion of a lesion site. Upon moderate activation, they undergo a transformation in morphology inducing beneficial responses. However, upon strong stimulation, they mediate neuronal damage via production of pro-inflammatory cytokines. The inhibition of this cascade is considered an effective strategy for neuroinflammation-associated disorder therapy. During this pathological activation microglia also undergo a shortening of process length which contributes to the pathogenesis of such disorders. Thus, microglial plasticity should be considered to have two components: one is the production of inflammatory mediators, and the other is the dynamic changes in their processes. The former role has been well-documented in previous studies, while the latter one remains largely unknown. Recently, we and others have reported that the elongation of microglial process is associated with the transformation of microglia from a pro-inflammatory to an anti-inflammatory state, suggesting that the shortening of process length would make the microglia lose their ability to restrict pathological injury, while the elongation of microglial process would help attenuate neuroinflammation. Compared with the traditional anti-neuroinflammatory strategy, stimulating elongation of microglial process not only reduces the production of pro-inflammatory cytokines, but restores the ability of microglia to scan their surrounding environments, thus rendering their homeostasis regulation more effective. In this review, we provide a discussion of the factors that regulate microglial process elongation in vitro and in vivo, aiming to further drive the understanding of microglial process plasticity.
Collapse
Affiliation(s)
- Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| | - Hui Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Department of Neuroscience & Cell Biology, Rutgers-Robert Wood Johnson Medical School, 675 Hoes lane, Piscataway, 08854, NJ, United States
| | - Jie Wen
- Beijing Allwegene Health, B-607 Wanlin Technology Mansion, 8 Malianwa North Road, Beijing 100094, China
| | - Kai Ma
- Probiotics Australia, 24-30 Blanck Street, Ormeau, QLD, 4208, Australia
| | - Dongjian Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
45
|
Zhou Y, Wu J, Sheng R, Li M, Wang Y, Han R, Han F, Chen Z, Qin ZH. Reduced Nicotinamide Adenine Dinucleotide Phosphate Inhibits MPTP-Induced Neuroinflammation and Neurotoxicity. Neuroscience 2018; 391:140-153. [PMID: 30195055 DOI: 10.1016/j.neuroscience.2018.08.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/03/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022]
Abstract
It is generally believed that oxidative stress and neuroinflammation are implicated in the pathogenesis of Parkinson's disease (PD). Reduced nicotinamide adenine dinucleotide phosphate (NADPH) has been demonstrated to have potent neuroprotective effects against oxidative stress. In the present research, we investigated if NADPH could offer neuroprotection by inhibiting glia-mediated neuroinflammation induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a mechanism contributing to PD pathogenesis. The current data demonstrated that MPTP/MPP+ increased levels of reactive oxygen species (ROS), activated glial cells, and inflammasome proteins in the substantia nigra (SNpc), in addition to inducing the nuclear translocation of nuclear factor-κB (NF-κB) and phosphorylation of p38 MAPK. These responses were inhibited by supplementation of exogenous NADPH. Moreover, NADPH effectively decreased MPP+-induced excessive production of ROS, p38 phosphorylation and inflammatory protein of Cyclooxygenase2 (COX2) in cultured microglial BV-2 cells in vitro studies. Similarly, the p38 MAPK inhibitor SB203580 suppressed the upregulation of MPP+-induced p38 phosphorylation and COX2 protein levels. Co-culture of neuronal cells with MPP+-primed BV-2 cells increased the levels of tumor necrosis factor-alpha (TNF-α) and induced cell death of neuronal cells. These effects were diminished by TNF-α neutralizing antibody and NADPH. NADPH reduced motor dysfunction and the loss of dopaminergic (DA) cells induced by MPTP. Therefore, the present study demonstrates that NADPH protects DA neurons by inhibiting oxidative stress and glia-mediated neuroinflammation both in vitro and in vivo, thus suggesting a potential of clinical application for PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Junchao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Yan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Feng Han
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
46
|
Ju C, Li Y, Shen Y, Liu Y, Cai J, Liu N, Ma G, Tang Y. Transplantation of Cardiac Mesenchymal Stem Cell-Derived Exosomes for Angiogenesis. J Cardiovasc Transl Res 2018; 11:429-437. [PMID: 30276617 DOI: 10.1007/s12265-018-9824-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/31/2018] [Indexed: 12/25/2022]
Abstract
We demonstrated the effects of exosomes secreted by cardiac mesenchymal stem cells (C-MSC-Exo) in protecting acute ischemic myocardium from reperfusion injury. To investigate the effect of exosomes from C-MSC on angiogenesis, we injected C-MSC-Exo or PBS intramuscularly into ischemic hind limb. Blood perfusion of limb was evaluated by laser Doppler Imaging. We observed that ischemic limb treated with C-MSC-Exo exhibits improved blood perfusion compared to ischemic limb treated with PBS at 2 weeks and 1 month after induction of limb ischemia. To explore the potential mechanisms underlying C-MSC-Exo's angiogenetic effect, we performed microRNA array analysis and identify mmu-miR-7116-5p as the most abundant enriched miRNA detected in C-MSC-Exo. Bioinformatics' analysis shows that miR-7116-5p negatively regulates protein polyubiquitination. In conclusion, our study demonstrated that intramuscular delivery of C-MSC-Exo after limb ischemia improves blood perfusion, and we identified the most abundant miRNAs that are preferentially enriched in C-MSC-Exo.
Collapse
Affiliation(s)
- Chengwei Ju
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Youngjun Li
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yan Shen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yutao Liu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jingwen Cai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gengshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
47
|
Cheray M, Joseph B. Epigenetics Control Microglia Plasticity. Front Cell Neurosci 2018; 12:243. [PMID: 30123114 PMCID: PMC6085560 DOI: 10.3389/fncel.2018.00243] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 01/31/2023] Open
Abstract
Microglia, resident immune cells of the central nervous system, fulfill multiple functions in the brain throughout life. These microglial functions range from participation in innate and adaptive immune responses, involvement in the development of the brain and its homeostasis maintenance, to contribution to degenerative, traumatic, and proliferative diseases; and take place in the developing, the aging, the healthy, or the diseased brain. Thus, an impressive level of cellular plasticity, appears as a requirement for the pleiotropic biological functions of microglia. Epigenetic changes, including histone modifications or DNA methylation as well as microRNA expression, are important modifiers of gene expression, and have been involved in cell phenotype regulation and reprogramming and are therefore part of the mechanisms regulating cellular plasticity. Here, we review and discuss the epigenetic mechanisms, which are emerging as contributors to this microglial cellular plasticity and thereby can constitute interesting targets to modulate microglia associated brain diseases, including developmental diseases, neurodegenerative diseases as well as cancer.
Collapse
Affiliation(s)
- Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
48
|
Gasser PJ, Lowry CA. Organic cation transporter 3: A cellular mechanism underlying rapid, non-genomic glucocorticoid regulation of monoaminergic neurotransmission, physiology, and behavior. Horm Behav 2018; 104:173-182. [PMID: 29738736 PMCID: PMC7137088 DOI: 10.1016/j.yhbeh.2018.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/11/2023]
Abstract
Contribution to Special Issue on Fast effects of steroids. Corticosteroid hormones act at intracellular glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) to alter gene expression, leading to diverse physiological and behavioral responses. In addition to these classical genomic effects, corticosteroid hormones also exert rapid actions on physiology and behavior through a variety of non-genomic mechanisms, some of which involve GR or MR, and others of which are independent of these receptors. One such GR-independent mechanism involves corticosteroid-induced inhibition of monoamine transport mediated by "uptake2" transporters, including organic cation transporter 3 (OCT3), a low-affinity, high-capacity transporter for norepinephrine, epinephrine, dopamine, serotonin and histamine. Corticosterone directly and acutely inhibits OCT3-mediated transport. This review describes the studies that initially characterized uptake2 processes in peripheral tissues, and outlines studies that demonstrated OCT3 expression and corticosterone-sensitive monoamine transport in the brain. Evidence is presented supporting the hypothesis that corticosterone can exert rapid, GR-independent actions on neuronal physiology and behavior by inhibiting OCT3-mediated monoamine clearance. Implications of this mechanism for glucocorticoid-monoamine interactions in the context-dependent regulation of behavior are discussed.
Collapse
Affiliation(s)
- Paul J Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver Veterans Affairs Medical Center (VAMC), Denver, CO 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO 80220, USA.
| |
Collapse
|
49
|
Gasser PJ. Roles for the uptake 2 transporter OCT3 in regulation of dopaminergic neurotransmission and behavior. Neurochem Int 2018; 123:46-49. [PMID: 30055194 DOI: 10.1016/j.neuint.2018.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/30/2022]
Abstract
Transporter-mediated uptake determines the peak concentration, duration, and physical spread of released monoamines. Most studies of monoamine clearance focus on the presynaptic uptake1 transporters SERT, NET and DAT. However, recent studies have demonstrated the expression of the uptake2 transporter OCT3 (organic cation transporter 3), throughout the rodent brain. In contrast to NET, DAT and SERT, OCT3 has higher capacity and lower affinity for substrates, is sodium-independent, and is multi-specific, with the capacity to transport norepinephrine, dopamine, serotonin and histamine. OCT3 is insensitive to inhibition by cocaine and antidepressant drugs but is inhibited directly by the glucocorticoid hormone corticosterone. Thus, OCT3 represents a novel, stress hormone-sensitive, monoamine transport mechanism. Incorporating this transporter into current models of monoaminergic neurotransmission requires information on: A) the cellular and subcellular localization of the transporter; B) the effects of OCT3 inhibitors on monoamine clearance; and C) the consequences of decreased OCT3-mediated transport on physiology and/or behavior. This review summarizes studies describing the anatomical distribution of OCT3, its cellular and subcellular localization, its contribution to the regulation of dopaminergic signaling, and its roles in the regulation of behavior. Together, these and other studies suggest that both Uptake1 and Uptake2 transporters play key roles in regulating monoaminergic neurotransmission and the effects of monoamines on behavior.
Collapse
Affiliation(s)
- Paul J Gasser
- Department of Biomedical Sciences, Marquette University, 561 N 15th Street, Milwaukee, WI, 53233, USA.
| |
Collapse
|
50
|
Wang Q, He Q, Chen Y, Shao W, Yuan C, Wang Y. JNK-mediated microglial DICER degradation potentiates inflammatory responses to induce dopaminergic neuron loss. J Neuroinflammation 2018; 15:184. [PMID: 29907159 PMCID: PMC6003208 DOI: 10.1186/s12974-018-1218-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Background Amplified inflammation is important for the progression of Parkinson’s disease (PD). However, how this enhanced inflammation is regulated remains largely unknown. Deletion of DICER leads to progressive dopamine neuronal loss and induces gliosis. We hypothesized that the homeostasis of microglial DICER would be responsible for the amplified inflammation in the mouse model of PD. Methods The microglia or C57BL/6 mice were treated or injected with l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium (MPP+), respectively, for the model establishment. Microglia and astrocytes sorted by fluorescence-activated cell sorter (FACS) were assayed by quantitative real-time PCR, Western blotting, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), immunohistofluorescence, and mass spectrometry. Results Microglial DICER was phosphorylated at serine 1456 by c-jun N-terminal kinase (JNK) and downregulated in response to 1-methyl-4-phenylpyridinium (MPP+), a causative agent in PD. Inhibition of JNK phosphorylation of DICER at serine 1456 rescued the MPP+-induced DICER degradation, suppressed microglial inflammatory process, and prevented the loss of tyrosine hydroxylase-expressing neurons in the mouse MPTP model. Conclusions JNK-mediated microglial DICER degradation potentiates inflammation to induce dopaminergic neuronal loss. Thus, preventing microglial DICER degradation could be a novel strategy for controlling neuroinflammation in PD. Electronic supplementary material The online version of this article (10.1186/s12974-018-1218-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Wang
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Qian He
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Yifei Chen
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Wei Shao
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Chao Yuan
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Yizheng Wang
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|