1
|
Duranti E, Villa C. From Brain to Muscle: The Role of Muscle Tissue in Neurodegenerative Disorders. BIOLOGY 2024; 13:719. [PMID: 39336146 PMCID: PMC11428675 DOI: 10.3390/biology13090719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs), like amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), primarily affect the central nervous system, leading to progressive neuronal loss and motor and cognitive dysfunction. However, recent studies have revealed that muscle tissue also plays a significant role in these diseases. ALS is characterized by severe muscle wasting as a result of motor neuron degeneration, as well as alterations in gene expression, protein aggregation, and oxidative stress. Muscle atrophy and mitochondrial dysfunction are also observed in AD, which may exacerbate cognitive decline due to systemic metabolic dysregulation. PD patients exhibit muscle fiber atrophy, altered muscle composition, and α-synuclein aggregation within muscle cells, contributing to motor symptoms and disease progression. Systemic inflammation and impaired protein degradation pathways are common among these disorders, highlighting muscle tissue as a key player in disease progression. Understanding these muscle-related changes offers potential therapeutic avenues, such as targeting mitochondrial function, reducing inflammation, and promoting muscle regeneration with exercise and pharmacological interventions. This review emphasizes the importance of considering an integrative approach to neurodegenerative disease research, considering both central and peripheral pathological mechanisms, in order to develop more effective treatments and improve patient outcomes.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
2
|
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
|
3
|
Duan X, Liu H, Hu X, Yu Q, Kuang G, Liu L, Zhang S, Wang X, Li J, Yu D, Huang J, Wang T, Lin Z, Xiong N. Insomnia in Parkinson's Disease: Causes, Consequences, and Therapeutic Approaches. Mol Neurobiol 2024:10.1007/s12035-024-04400-4. [PMID: 39103716 DOI: 10.1007/s12035-024-04400-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Sleep disorders represent prevalent non-motor symptoms in Parkinson's disease (PD), affecting over 90% of the PD population. Insomnia, characterized by difficulties in initiating and maintaining sleep, emerges as the most frequently reported sleep disorder in PD, with prevalence rates reported from 27 to 80% across studies. Insomnia not only significantly impacts the quality of life of PD patients but is also associated with cognitive impairment, motor disabilities, and emotional deterioration. This comprehensive review aims to delve into the mechanisms underlying insomnia in PD, including neurodegenerative changes, basal ganglia beta oscillations, and circadian rhythms, to gain insights into the neural pathways involved. Additionally, the review explores the risk factors and comorbidities associated with insomnia in PD, providing valuable insights into its management. Special attention is given to the challenges faced by healthcare providers in delivering care to PD patients and the impact of caregiving roles on patients' quality of life. Overall, this review provides a comprehensive understanding of insomnia in PD and highlights the importance of addressing this common sleep disorder in PD patients.
Collapse
Affiliation(s)
- Xiaoyu Duan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Duke Kunshan University, No. 8 Duke Avenue, Kunshan, 215316, Jiangsu, China
| | - Hanshu Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xinyu Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qinwei Yu
- Department of Neurology, Wuhan Red Cross Hospital, 392 Hongkong Road, Wuhan, Hubei, China
| | - Guiying Kuang
- Department of Neurology, Wuhan Red Cross Hospital, 392 Hongkong Road, Wuhan, Hubei, China
| | - Long Liu
- Department of Neurology, Wuhan Red Cross Hospital, 392 Hongkong Road, Wuhan, Hubei, China
| | - Shurui Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xinyi Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Danfang Yu
- Department of Neurology, Wuhan Red Cross Hospital, 392 Hongkong Road, Wuhan, Hubei, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhicheng Lin
- Laboratory of Psychiatric Neurogenomics, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
4
|
Anwar MM, Pérez-Martínez L, Pedraza-Alva G. Exploring the Significance of Microglial Phenotypes and Morphological Diversity in Neuroinflammation and Neurodegenerative Diseases: From Mechanisms to Potential Therapeutic Targets. Immunol Invest 2024; 53:891-946. [PMID: 38836373 DOI: 10.1080/08820139.2024.2358446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Studying various microglial phenotypes and their functions in neurodegenerative diseases is crucial due to the intricate nature of their phenomics and their vital immunological role. Microglia undergo substantial phenomic changes, encompassing morphological, transcriptional, and functional aspects, resulting in distinct cell types with diverse structures, functions, properties, and implications. The traditional classification of microglia as ramified, M1 (proinflammatory), or M2 (anti-inflammatory) phenotypes is overly simplistic, failing to capture the wide range of recently identified microglial phenotypes in various brain regions affected by neurodegenerative diseases. Altered and activated microglial phenotypes deviating from the typical ramified structure are significant features of many neurodegenerative conditions. Understanding the precise role of each microglial phenotype is intricate and sometimes contradictory. This review specifically focuses on elucidating recent modifications in microglial phenotypes within neurodegenerative diseases. Recognizing the heterogeneity of microglial phenotypes in diseased states can unveil novel therapeutic strategies for targeting microglia in neurodegenerative diseases. Moreover, the exploration of the use of healthy isolated microglia to mitigate disease progression has provided an innovative perspective. In conclusion, this review discusses the dynamic landscape of mysterious microglial phenotypes, emphasizing the need for a nuanced understanding to pave the way for innovative therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Leonor Pérez-Martínez
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| | - Gustavo Pedraza-Alva
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| |
Collapse
|
5
|
Ma LM, Shi JT, Chai T, Naghavi MR, Liu HY, Yang AM, Wang J, Yang JL. Chemical constituents from Notopterygium incisum and their anti-neuroinflammatory activity. Fitoterapia 2024; 176:105976. [PMID: 38685511 DOI: 10.1016/j.fitote.2024.105976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Phytochemical research on an extract of Notopterygium incisum yielded fifteen compounds (1-15), including four previously undescribed compounds (10-13). The structures of the unreported compounds were elucidated by spectroscopic and spectrometric data analysis such as 1D and 2D NMR, IR and HR-ESI-MS. Compounds 1-5 and 10-14 were isolated from N. incisum for the first time. 7S⁎,8R⁎-Phenethyl-(7-methoxy-8-isoeugenol)-ferulate (10), 7S⁎,8R⁎-p-hydroxyphenethyl-(7-methoxy-8-isoeugenol)-ferulate (11), 7S⁎,8R⁎-benzyl-(7-methoxy-8-isoeugenol)-ferulate (12) and p-hydroxyphenethyl-(4-benzoy-3-methoxy)-cinnamate (13) are the undescribed ferulic acid derivatives. Additionly, the anti-neuroinflammatory effects of compounds were evaluated in lipopolysaccharide (LPS)-induced BV2 cells. The pharmacological results showed that 6β,10β-epoxy-4α-hydroxy-guaiane (6), teuclatriol (7) and 7S⁎,8R⁎-p-hydroxyphenethyl-(7-methoxy-8-isoeugenol)-ferulate (11) inhibited the production and expression of nitric oxide (NO) in the LPS-induced BV2 cells in a concentration-dependent manner. Acorusnol (4), teucladiol (9), 7S⁎,8R⁎-benzyl-(7-methoxy-8-isoeugenol)-ferulate (12) and p-hydroxyphenethyl-(4-benzoy-3-methoxy)-cinnamate (13) only inhibited the release of NO at concentration of 20 μM. Moreover, 7S⁎,8R⁎-p-hydroxyphenethyl-(7-methoxy-8-isoeugenol)-ferulate (11) reduced the level of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in LPS-stimulated BV2 cells. The results demonstrated 7S⁎,8R⁎-p-hydroxyphenethyl-(7-methoxy-8-isoeugenol)-ferulate (11) could be a potential anti-neuroinflammatory agent and is worthy of further study.
Collapse
Affiliation(s)
- Li-Mei Ma
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, People's Republic of China; Guangdong Key Laboratory of Natural Medicine Research and Development, College of Pharmacy, Guangdong Medical University, Dongguan 523808, People's Republic of China
| | - Jiao-Tai Shi
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, People's Republic of China
| | - Tian Chai
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, People's Republic of China
| | - Mohammad Reza Naghavi
- Department of Agronomy and Plant Breeding, Agricultural and Natural Resources College, University of Tehran, Karaj, Iran
| | - Huan-Yan Liu
- Guangdong Key Laboratory of Natural Medicine Research and Development, College of Pharmacy, Guangdong Medical University, Dongguan 523808, People's Republic of China
| | - Ai-Mei Yang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, People's Republic of China.
| | - Jun Wang
- Shandong Laboratory of Yantai Advanced Materials and Green Manufacturing, Yantai 264010, People's Republic of China.
| | - Jun-Li Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, People's Republic of China.
| |
Collapse
|
6
|
Liu T, Wu H, Sun L, Wei J. Role of Inflammation in the Development of COVID-19 to Parkinson's Disease. J Inflamm Res 2024; 17:3259-3282. [PMID: 38800597 PMCID: PMC11127656 DOI: 10.2147/jir.s460161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) can lead to neurological symptoms such as headaches, confusion, seizures, hearing loss, and loss of smell. The link between COVID-19 and Parkinson's disease (PD) is being investigated, but more research is needed for a definitive connection. Methods Datasets GSE22491 and GSE164805 were selected to screen differentially expressed gene (DEG), and immune infiltration and gene set enrichment analysis (GSEA) of the DEG were performed. WGCNA analyzed the DEG and selected the intersection genes. Potential biological functions and signaling pathways were determined, and diagnostic genes were further screened using gene expression and receiver operating characteristic (ROC) curves. Screening and molecular docking of ibuprofen as a therapeutic target. The effectiveness of ibuprofen was verified by constructing a PD model in vitro, and constructing "COVID19-PD" signaling pathway, and exploring the role of angiotensin-converting enzyme 2 (ACE2) in PD. Results A total of 13 DEG were screened from the GSE36980 and GSE5281 datasets. Kyoto encyclopedia of genes and genomes (KEGG) analysis showed that the DEG were mainly associated with the hypoxia-inducible factor (HIF-1), epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor resistance, etc. After analysis, it is found that ibuprofen alleviates PD symptoms by inhibiting the expression of nuclear factor kappa-B (NF-κB), interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α). Based on signal pathway construction, the importance of ACE2 in COVID-19-induced PD has been identified. ACE2 is found to have widespread distribution in the brain. In the 1-methyl-4-phenyl-1,2,3,6-te-trahydropyridine (MPTP)-induced ACE2-null PD mice model, more severe motor and non-motor symptoms, increased NF-κB p65 and α-synuclein (α-syn) expression with significant aggregation, decreased tyrosine hydroxylase (TH), severe neuronal loss, and neurodegenerative disorders. Conclusion Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection increases the risk of PD through an inflammatory environment and downregulation of ACE2, providing evidence for the molecular mechanism and targeted therapy associated with COVID-19 and PD.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng, 475004, People’s Republic of China
| | - Haojie Wu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng, 475004, People’s Republic of China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng, 475004, People’s Republic of China
| |
Collapse
|
7
|
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
|
8
|
Eser P, Kocabicak E, Bekar A, Temel Y. The interplay between neuroinflammatory pathways and Parkinson's disease. Exp Neurol 2024; 372:114644. [PMID: 38061555 DOI: 10.1016/j.expneurol.2023.114644] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Parkinson's disease, a progressive neurodegenerative disorder predominantly affecting elderly, is marked by the gradual degeneration of the nigrostriatal dopaminergic pathway, culminating in neuronal loss within the substantia nigra pars compacta (SNpc) and dopamine depletion. At the molecular level, neuronal loss in the SNpc has been attributed to factors including neuroinflammation, impaired protein homeostasis, as well as mitochondrial dysfunction and the resulting oxidative stress. This review focuses on the interplay between neuroinflammatory pathways and Parkinson's disease, drawing insights from current literature.
Collapse
Affiliation(s)
- Pinar Eser
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey.
| | - Ersoy Kocabicak
- Ondokuz Mayis University, Health Practise and Research Hospital, Neuromodulation Center, Samsun, Turkey
| | - Ahmet Bekar
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
9
|
Lee MY, Kim M. Effects of Red ginseng on neuroinflammation in neurodegenerative diseases. J Ginseng Res 2024; 48:20-30. [PMID: 38223824 PMCID: PMC10785270 DOI: 10.1016/j.jgr.2023.08.003] [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: 03/29/2023] [Revised: 07/17/2023] [Accepted: 08/25/2023] [Indexed: 01/16/2024] Open
Abstract
Red ginseng (RG) is widely used as a herbal medicine. As the human lifespan has increased, numerous diseases have developed, and RG has also been used to treat various diseases. Neurodegenerative diseases are major problems that modern people face through their lives. Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are featured by progressive nerve system damage. Recently, neuroinflammation has emerged as a degenerative factor and is an immune response in which cytokines with nerve cells that constitute the nervous system. RG, a natural herbal medicine with fewer side effects than chemically synthesized drugs, is currently in the spotlight. Therefore, we reviewed studies reporting the roles of RG in treating neuroinflammation and neurodegenerative diseases and found that RG might help alleviate neurodegenerative diseases by regulating neuroinflammation.
Collapse
Affiliation(s)
- Min Yeong Lee
- Department of Chemistry & Life Science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Hwarangro 815, Nowongu, Seoul, Republic of Korea
| |
Collapse
|
10
|
Hu Z, Sun P, George A, Zeng X, Li M, Lin TH, Ye Z, Wei X, Jiang X, Song SK, Yang R. Diffusion basis spectrum imaging detects pathological alterations in substantia nigra and white matter tracts with early-stage Parkinson's disease. Eur Radiol 2023; 33:9109-9119. [PMID: 37438642 DOI: 10.1007/s00330-023-09780-0] [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: 08/31/2022] [Revised: 03/13/2023] [Accepted: 03/30/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVES Using diffusion basis spectrum imaging (DBSI) to examine the microstructural changes in the substantia nigra (SN) and global white matter (WM) tracts of patients with early-stage PD. METHODS Thirty-seven age- and sex-matched patients with early-stage PD and 22 healthy controls (HCs) were enrolled in this study. All participants underwent clinical assessments and diffusion-weighted MRI scans, analyzed by diffusion tensor imaging (DTI) and DBSI to assess the pathologies of PD in SN and global WM tracts. RESULTS The lower DTI fraction anisotropy (FA) was seen in SN of PD patients (PD: 0.316 ± 0.034 vs HCs: 0.331 ± 0.019, p = 0.015). The putative cells marker-DBSI-restricted fraction (PD: 0.132 ± 0.051 vs HCs: 0.105 ± 0.039, p = 0.031) and the edema/extracellular space marker-DBSI non-restricted-fraction (PD: 0.150 ± 0.052 vs HCs: 0.122 ± 0.052, p = 0.020) were both significantly higher and the density of axons/dendrites marker-DBSI fiber-fraction (PD: 0.718 ± 0.073 vs HCs: 0.773 ± 0.071, p = 0.003) was significantly lower in SN of PD patients. DBSI-restricted fraction in SN was negatively correlated with HAMA scores (r = - 0.501, p = 0.005), whereas DTI-FA was not correlated with any clinical scales. In WM tracts, only higher DTI axial diffusivity (AD) among DTI metrics was found in multiple WM regions in PD, while lower DBSI fiber-fraction and higher DBSI non-restricted-fraction were detected in multiple WM regions. DBSI non-restricted-fraction in both left fornix (cres)/stria terminalis (r = -0.472, p = 0.004) and right posterior thalamic radiation (r = - 0.467, p = 0.005) was negatively correlated with MMSE scores. CONCLUSION DBSI could potentially detect and quantify the extent of inflammatory cell infiltration, fiber/dendrite loss, and edema in both SN and WM tracts in patients with early-stage PD, a finding remains to be further investigated through more extensive longitudinal DBSI analysis. CLINICAL RELEVANCE STATEMENT Our study shows that DBSI indexes can potentially detect early-stage PD's pathological changes, with a notable ability to distinguish between inflammation and edema. This implies that DBSI has the potential to be an imaging biomarker for early PD diagnosis. KEY POINTS • Diffusion basis spectrum imaging detected higher restricted-fraction in Parkinson's disease, potentially reflecting inflammatory cell infiltration. • Diffusion basis spectrum imaging detected higher non-restricted-fraction and lower fiber-fraction in Parkinson's disease, indicating the presence of edema and/or dopaminergic neuronal/dendritic loss. • Diffusion basis spectrum imaging metrics correlated with non-motor symptoms, suggesting its potential diagnostic role to detect early-stage PD dysfunctions.
Collapse
Affiliation(s)
- Zexuan Hu
- Department of Radiology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangdong, 510310, Guangzhou, China
| | - Peng Sun
- Biomedical MR Laboratory, Mallinckrodt Institute of Radiology, Washington University School of Medicine, Room 2313, 4525 Scott Ave, Campus Box 8227, St. Louis, MO, 63110-1093, USA
| | - Ajit George
- Biomedical MR Laboratory, Mallinckrodt Institute of Radiology, Washington University School of Medicine, Room 2313, 4525 Scott Ave, Campus Box 8227, St. Louis, MO, 63110-1093, USA
| | - Xiangling Zeng
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, #1 Panfu Road, Yuexiu District, Guangdong, 510180, Guangzhou, China
| | - Mengyan Li
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, #1 Panfu Road, Yuexiu District, Guangdong, 510180, Guangzhou, China
| | - Tsen-Hsuan Lin
- Biomedical MR Laboratory, Mallinckrodt Institute of Radiology, Washington University School of Medicine, Room 2313, 4525 Scott Ave, Campus Box 8227, St. Louis, MO, 63110-1093, USA
| | - Zezhong Ye
- Biomedical MR Laboratory, Mallinckrodt Institute of Radiology, Washington University School of Medicine, Room 2313, 4525 Scott Ave, Campus Box 8227, St. Louis, MO, 63110-1093, USA
| | - Xinhua Wei
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, #1 Panfu Road, Yuexiu District, Guangdong, 510180, Guangzhou, China
| | - Xinqing Jiang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, #1 Panfu Road, Yuexiu District, Guangdong, 510180, Guangzhou, China
| | - Sheng-Kwei Song
- Biomedical MR Laboratory, Mallinckrodt Institute of Radiology, Washington University School of Medicine, Room 2313, 4525 Scott Ave, Campus Box 8227, St. Louis, MO, 63110-1093, USA.
| | - Ruimeng Yang
- Department of Radiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, #1 Panfu Road, Yuexiu District, Guangdong, 510180, Guangzhou, China.
| |
Collapse
|
11
|
Ning H, Zhou H, Yang N, Ren J, Wang H, Liu W, Zhao Y. Effect of Zishen pingchan granules combined with pramipexole on serum BDNF, IL-1β, IL-6, CRP, TNF-α levels in depressed patients with Parkinson's disease: Results of a randomized, double-blind, controlled study. Exp Gerontol 2023; 182:112295. [PMID: 37734668 DOI: 10.1016/j.exger.2023.112295] [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: 08/05/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
INTRODUCTION Depression is a common comorbidity in Parkinson's Disease (PD) and treatment of depression can significantly support PD management. Zishen pingchan granules (ZPG), a traditional Chinese herbal formula, may help ameliorate depressive symptoms in PD patients. However, the molecular mechanisms underlying the effects of ZPG remain unclear. This study aimed to investigate the impact of ZPG on serum levels of brain-derived neurotrophic factor (BDNF), interleukin-1β (IL-1β), interleukin-6 (IL-6), C-reactive protein (CRP), and tumor necrosis factor-α (TNF-α) in PD patients with depression. METHODS Eighty PD patients treated with pramipexole but still experiencing mild to moderate depression symptoms were randomly allocated to a group receiving 12-week ZPG treatment (n = 40) or placebo (n = 40). The Hamilton Depression Scale 17 items (HAM-D-17) was utilized to evaluate changes in depressive symptoms from baseline over 12 weeks, while the Unified Parkinson's Disease Rating Scales (UPDRS) part 3 was employed to assess changes in motor symptoms over the same duration. Serum levels of BDNF, IL-1β, IL-6, CRP, and TNF-α were measured at baseline and post-treatment. RESULTS Seventy-one participants completed the study. Following treatment, both groups showed significantly reduced HAMD scores. The placebo group demonstrated a decrease in BDNF levels, while the ZPG group showed an increase in IL-6 levels post-treatment. In the examination of the group-time interaction, the ZPG group exhibited a greater decrease in HAMD scores and increase in IL-6 levels compared to the placebo group. Conversely, the placebo group showed a greater decrease in BDNF levels compared to the ZPG group. However, no significant group differences were observed in UPDRS part 3 change scores or serum levels of IL-1β, CRP, or TNF-α change from baseline. CONCLUSION ZPG may potentially ameliorate depressive symptoms in PD patients, with the potential mechanism involving mitigation of reductions in serum BDNF level and an increase in IL-6 level.
Collapse
Affiliation(s)
- Houxu Ning
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China; Department of Chinese Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Zhou
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ning Yang
- Department of Chinese Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jingru Ren
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Haidong Wang
- Department of Chinese Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
12
|
De Marchi F, Munitic I, Vidatic L, Papić E, Rački V, Nimac J, Jurak I, Novotni G, Rogelj B, Vuletic V, Liscic RM, Cannon JR, Buratti E, Mazzini L, Hecimovic S. Overlapping Neuroimmune Mechanisms and Therapeutic Targets in Neurodegenerative Disorders. Biomedicines 2023; 11:2793. [PMID: 37893165 PMCID: PMC10604382 DOI: 10.3390/biomedicines11102793] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Many potential immune therapeutic targets are similarly affected in adult-onset neurodegenerative diseases, such as Alzheimer's (AD) disease, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and frontotemporal dementia (FTD), as well as in a seemingly distinct Niemann-Pick type C disease with primarily juvenile onset. This strongly argues for an overlap in pathogenic mechanisms. The commonly researched immune targets include various immune cell subsets, such as microglia, peripheral macrophages, and regulatory T cells (Tregs); the complement system; and other soluble factors. In this review, we compare these neurodegenerative diseases from a clinical point of view and highlight common pathways and mechanisms of protein aggregation, neurodegeneration, and/or neuroinflammation that could potentially lead to shared treatment strategies for overlapping immune dysfunctions in these diseases. These approaches include but are not limited to immunisation, complement cascade blockade, microbiome regulation, inhibition of signal transduction, Treg boosting, and stem cell transplantation.
Collapse
Affiliation(s)
- Fabiola De Marchi
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy;
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia;
| | - Lea Vidatic
- Laboratory for Neurodegenerative Disease Research, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia;
| | - Eliša Papić
- Department of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (E.P.); (V.R.); (V.V.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (E.P.); (V.R.); (V.V.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Jerneja Nimac
- Department of Biotechnology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; (J.N.); (B.R.)
- Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Igor Jurak
- Molecular Virology Laboratory, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia;
| | - Gabriela Novotni
- Department of Cognitive Neurology and Neurodegenerative Diseases, University Clinic of Neurology, Medical Faculty, University Ss. Cyril and Methodius, 91701 Skoplje, North Macedonia;
| | - Boris Rogelj
- Department of Biotechnology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; (J.N.); (B.R.)
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Vladimira Vuletic
- Department of Neurology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (E.P.); (V.R.); (V.V.)
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Rajka M. Liscic
- Department of Neurology, Sachsenklinik GmbH, Muldentalweg 1, 04828 Bennewitz, Germany;
| | - Jason R. Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy;
| | - Letizia Mazzini
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy;
| | - Silva Hecimovic
- Laboratory for Neurodegenerative Disease Research, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia;
| |
Collapse
|
13
|
Wang L, Bai Y, Tao Y, Shen W, Zhou H, He Y, Wu H, Huang F, Shi H, Wu X. Bear bile powder alleviates Parkinson's disease-like behavior in mice by inhibiting astrocyte-mediated neuroinflammation. Chin J Nat Med 2023; 21:710-720. [PMID: 37777320 DOI: 10.1016/s1875-5364(23)60449-2] [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: 03/21/2023] [Indexed: 10/02/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in middle-aged and elderly people. In particular, increasing evidence has showed that astrocyte-mediated neuroinflammation is involved in the pathogenesis of PD. As a precious traditional Chinese medicine, bear bile powder (BBP) has a long history of use in clinical practice. It has numerous activities, such as clearing heat, calming the liver wind and anti-inflammation, and also exhibits good therapeutic effect on convulsive epilepsy. However, whether BBP can prevent the development of PD has not been elucidated. Hence, this study was designed to explore the effect and mechanism of BBP on suppressing astrocyte-mediated neuroinflammation in a mouse model of PD. PD-like behavior was induced in the mice by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (30 mg·kg-1) for five days, followed by BBP (50, 100, and 200 mg·kg-1) treatment daily for ten days. LPS stimulated rat C6 astrocytic cells were used as a cell model of neuroinflammation. THe results indicated that BBP treatment significantly ameliorated dyskinesia, increased the levels of tyrosine hydroxylase (TH) and inhibited astrocyte hyperactivation in the substantia nigra (SN) of PD mice. Furthermore, BBP decreased the protein levels of glial fibrillary acidic protein (GFAP), cyclooxygenase 2 (COX2) and inducible nitric oxide synthase (iNOS), and up-regulated the protein levels of takeda G protein-coupled receptor 5 (TGR5) in the SN. Moreover, BBP significantly activated TGR5 in a dose-dependent manner, and decreased the protein levels of GFAP, iNOS and COX2, as well as the mRNA levels of GFAP, iNOS, COX2, interleukin (IL) -1β, IL-6 and tumor necrosis factor-α (TNF-α) in LPS-stimulated C6 cells. Notably, BBP suppressed the phosphorylation of protein kinase B (AKT), inhibitor of NF-κB (IκBα) and nuclear factor-κB (NF-κB) proteins in vivo and in vitro. We also observed that TGR5 inhibitor triamterene attenuated the anti-neuroinflammatory effect of BBP on LPS-stimulated C6 cells. Taken together, BBP alleviates the progression of PD mice by suppressing astrocyte-mediated inflammation via TGR5.
Collapse
Affiliation(s)
- Lupeng Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuyan Bai
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanlin Tao
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei Shen
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Houyuan Zhou
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yixin He
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
14
|
Gebrie A. The melanocortin receptor signaling system and its role in neuroprotection against neurodegeneration: Therapeutic insights. Ann N Y Acad Sci 2023; 1527:30-41. [PMID: 37526975 DOI: 10.1111/nyas.15048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
The melanocortin signaling system consists of the melanocortin peptides, their distinctive receptors, accessory proteins, and endogenous antagonists. Melanocortin peptides are small peptide hormones that have been studied in a variety of physiological and pathological conditions. There are five types of melanocortin receptors, and they are distributed within the central nervous system and in several tissues of the periphery. The G protein-coupled melanocortin receptors typically signal through adenylyl cyclase and other downstream signaling pathways. Depending on the ligand, surface expression of melanocortin receptor, receptor occupancy period, related proteins, the type of cell, and other parameters, the signaling pathways are complicated and pleiotropic. While it is known that all five melanocortin receptors are coupled to Gs, they can also occasionally couple to Gq or Gi. Both direct and indirect neuroprotection are induced by the melanocortin signaling system. Targeting several of the components of the melanocortin signaling system (ligands, receptors, accessory proteins, signaling effectors, and regulators) may provide therapeutic opportunities. Activation of the melanocortin system improves different functional traits in neurodegenerative diseases. There is a potential for additional melanocortin system interventions by interfering with dimerization or dissociation. This review aims to discuss the melanocortin receptor signaling system and its role in neuroprotection, as well as its therapeutic potential.
Collapse
Affiliation(s)
- Alemu Gebrie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
15
|
Yang PN, Chen WL, Lee JW, Lin CH, Chen YR, Lin CY, Lin W, Yao CF, Wu YR, Chang KH, Chen CM, Lee-Chen GJ. Coumarin-chalcone hybrid LM-021 and indole derivative NC009-1 targeting inflammation and oxidative stress to protect BE(2)-M17 cells against α-synuclein toxicity. Aging (Albany NY) 2023; 15:8061-8089. [PMID: 37578928 PMCID: PMC10497001 DOI: 10.18632/aging.204954] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/17/2023] [Indexed: 08/16/2023]
Abstract
Parkinson's disease (PD) is featured mainly by the loss of dopaminergic neurons and the presence of α-synuclein-containing aggregates in the substantia nigra of brain. The α-synuclein fibrils and aggregates lead to increased oxidative stress and neural toxicity in PD. Chronic inflammation mediated by microglia is one of the hallmarks of PD pathophysiology. In this report, we showed that coumarin-chalcone hybrid LM-021 and indole derivative NC009-1 reduced the expression of major histocompatibility complex-II, NLR family pyrin domain containing (NLRP) 3, caspase-1, inducible nitric oxide synthase, interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in α-synuclein-activated mouse BV-2 microglia. Release of pro-inflammatory mediators including nitric oxide, IL-1β, IL-6 and TNF-α was also mitigated. In BE(2)-M17 cells expressing A53T α-synuclein aggregates, LM-021 and NC009-1 reduced α-synuclein aggregation, neuroinflammation, oxidative stress and apoptosis, and promoted neurite outgrowth. These protective effects were mediated by downregulating NLRP1, IL-1β and IL-6, and their downstream pathways including nuclear factor (NF)-κB inhibitor alpha (IκBα)/NF-κB P65 subunit (P65), c-Jun N-terminal kinase (JNK)/proto-oncogene c-Jun (JUN), mitogen-activated protein kinase 14 (P38)/signal transducer and activator of transcription (STAT) 1, and Janus kinase 2 (JAK2)/STAT3. The study results indicate LM-021 and NC009-1 as potential new drug candidates for PD.
Collapse
Affiliation(s)
- Pei-Ning Yang
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Wan-Ling Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Jun-Wei Lee
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Chih-Hsin Lin
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Yi-Ru Chen
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
16
|
Huang H, Li S, Zhang Y, He C, Hua Z. Microglial Priming in Bilirubin-Induced Neurotoxicity. Neurotox Res 2023; 41:338-348. [PMID: 37058197 DOI: 10.1007/s12640-023-00643-6] [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: 11/04/2022] [Revised: 03/05/2023] [Accepted: 04/02/2023] [Indexed: 04/15/2023]
Abstract
Neuroinflammation is a major contributor to bilirubin-induced neurotoxicity, which results in severe neurological deficits. Microglia are the primary immune cells in the brain, with M1 microglia promoting inflammatory injury and M2 microglia inhibiting neuroinflammation. Controlling microglial inflammation could be a promising therapeutic strategy for reducing bilirubin-induced neurotoxicity. Primary microglial cultures were prepared from 1-3-day-old rats. In the early stages of bilirubin treatment, pro-/anti-inflammatory (M1/M2) microglia mixed polarization was observed. In the late stages, bilirubin persistence induced dominant proinflammatory microglia, forming an inflammatory microenvironment and inducing iNOS expression as well as the release of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β. Simultaneously, nuclear factor-kappa B (NF-κB) was activated and translocated into the nucleus, upregulating inflammatory target genes. As well known, neuroinflammation can have an effect on N-methyl-D-aspartate receptor (NMDAR) expression or function, which is linked to cognition. Treatment with bilirubin-treated microglia-conditioned medium did affect the expression of IL-1β, NMDA receptor subunit 2A (NR2A), and NMDA receptor subunit 2B (NR2B) in neurons. However, VX-765 effectively reduces the levels of proinflammatory cytokines TNF-α, IL-6, and IL-1β, as well as the expressions of CD86, and increases the expressions of anti-inflammatory related Arg-1. A timely reduction in proinflammatory microglia could protect against bilirubin-induced neurotoxicity.
Collapse
Affiliation(s)
- Hongmei Huang
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Siyu Li
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Yan Zhang
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Chunmei He
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Ziyu Hua
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China.
| |
Collapse
|
17
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
18
|
Lv Y, Wu M, Wang Z, Wang J. Ferroptosis: From regulation of lipid peroxidation to the treatment of diseases. Cell Biol Toxicol 2023; 39:827-851. [PMID: 36459356 DOI: 10.1007/s10565-022-09778-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022]
Abstract
Ferroptosis is a regulated cell death mainly manifested by iron-dependent lipid peroxide accumulation. The leading cause of ferroptosis is the imbalance of intracellular oxidative systems (e.g., LOXs, POR, ROS) and antioxidant systems (e.g., GSH/GPx4, CoQ10/FSP1, BH4/GCH1), which is regulated by a complex network. In the past decade, this metabolic network has been continuously refined, and the links with various pathophysiological processes have been gradually established. Apoptosis has been regarded as the only form of regulated cell death for a long time, and the application of chemotherapeutic drugs to induce apoptosis of cancer cells is the mainstream method. However, studies have reported that cancer cells' key features are resistance to apoptosis and chemotherapeutics. For high proliferation, cancer cells often have very active lipid metabolism and iron metabolism, which pave the way for ferroptosis. Interestingly, researchers found that drug-resistant or highly aggressive cancer cells are more prone to ferroptosis. Therefore, ferroptosis may be a potential strategy to eliminate cancer cells. In addition, links between ferroptosis and other diseases, such as neurological disorders and ischemia-reperfusion injury, have also been found. Understanding these diseases from the perspective of ferroptosis may provide new insights into clinical treatment. Herein, the metabolic processes in ferroptosis are reviewed, and the potential mechanisms and targets of ferroptosis in different diseases are summarized.
Collapse
Affiliation(s)
- Yonghui Lv
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Meiying Wu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
19
|
Anwer DM, Gubinelli F, Kurt YA, Sarauskyte L, Jacobs F, Venuti C, Sandoval IM, Yang Y, Stancati J, Mazzocchi M, Brandi E, O’Keeffe G, Steece-Collier K, Li JY, Deierborg T, Manfredsson FP, Davidsson M, Heuer A. A comparison of machine learning approaches for the quantification of microglial cells in the brain of mice, rats and non-human primates. PLoS One 2023; 18:e0284480. [PMID: 37126506 PMCID: PMC10150977 DOI: 10.1371/journal.pone.0284480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Microglial cells are brain-specific macrophages that swiftly react to disruptive events in the brain. Microglial activation leads to specific modifications, including proliferation, morphological changes, migration to the site of insult, and changes in gene expression profiles. A change in inflammatory status has been linked to many neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease. For this reason, the investigation and quantification of microglial cells is essential for better understanding their role in disease progression as well as for evaluating the cytocompatibility of novel therapeutic approaches for such conditions. In the following study we implemented a machine learning-based approach for the fast and automatized quantification of microglial cells; this tool was compared with manual quantification (ground truth), and with alternative free-ware such as the threshold-based ImageJ and the machine learning-based Ilastik. We first trained the algorithms on brain tissue obtained from rats and non-human primate immunohistochemically labelled for microglia. Subsequently we validated the accuracy of the trained algorithms in a preclinical rodent model of Parkinson's disease and demonstrated the robustness of the algorithms on tissue obtained from mice, as well as from images provided by three collaborating laboratories. Our results indicate that machine learning algorithms can detect and quantify microglial cells in all the three mammalian species in a precise manner, equipotent to the one observed following manual counting. Using this tool, we were able to detect and quantify small changes between the hemispheres, suggesting the power and reliability of the algorithm. Such a tool will be very useful for investigation of microglial response in disease development, as well as in the investigation of compatible novel therapeutics targeting the brain. As all network weights and labelled training data are made available, together with our step-by-step user guide, we anticipate that many laboratories will implement machine learning-based quantification of microglial cells in their research.
Collapse
Affiliation(s)
- Danish M. Anwer
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| | - Francesco Gubinelli
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| | - Yunus A. Kurt
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| | - Livija Sarauskyte
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| | - Febe Jacobs
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| | - Chiara Venuti
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| | - Ivette M. Sandoval
- Barrow Neurological Institute, Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Phoenix, Arizona, United States of America
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Jennifer Stancati
- Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Martina Mazzocchi
- Brain Development and Repair Group, Department of Anatomy and Neuroscience University College Cork, Cork, Ireland
| | - Edoardo Brandi
- Neural Plasticity and Repair, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Gerard O’Keeffe
- Brain Development and Repair Group, Department of Anatomy and Neuroscience University College Cork, Cork, Ireland
| | - Kathy Steece-Collier
- Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States of America
| | - Jia-Yi Li
- Neural Plasticity and Repair, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Fredric P. Manfredsson
- Barrow Neurological Institute, Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Phoenix, Arizona, United States of America
| | - Marcus Davidsson
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
- Barrow Neurological Institute, Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Phoenix, Arizona, United States of America
| | - Andreas Heuer
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University Lund, Sweden
| |
Collapse
|
20
|
Cheataini F, Ballout N, Al Sagheer T. The effect of neuroinflammation on the cerebral metabolism at baseline and after neural stimulation in neurodegenerative diseases. J Neurosci Res 2023. [PMID: 37186320 DOI: 10.1002/jnr.25198] [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: 11/06/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
Neuroinflammation is a reaction of nervous tissue to an attack caused by an infection, a toxin, or a neurodegenerative disease. It involves brain metabolism adaptation in order to meet the increased energy needs of glial cell activation, but the nature of these adaptations is still unknown. Increasing interest concerning neuroinflammation leads to the identification of its role in neurodegenerative diseases. Few reports studied the effect of metabolic alteration on neuroinflammation. Metabolic damage initiates a pro-inflammatory response by microglial activation. Moreover, the exact neuroinflammation effect on cerebral cell metabolism remains unknown. In this study, we reviewed systematically the neuroinflammation effect in animal models' brains. All articles showing the relationship of neuroinflammation with brain metabolism, or with neuronal stimulation in neurodegenerative diseases were considered. Moreover, this review examines also the mitochondrial damage effect in neurodegeneration diseases. Then, different biosensors are classified regarding their importance in the determination of metabolite change. Finally, some therapeutic drugs inhibiting neuroinflammation are cited. Neuroinflammation increases lymphocyte infiltration and cytokines' overproduction, altering cellular energy homeostasis. This review demonstrates the importance of neuroinflammation as a mediator of disease progression. Further, the spread of depolarization effects pro-inflammatory genes expression and microglial activation, which contribute to the degeneration of neurons, paving the road to better management and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Fatima Cheataini
- Neuroscience Research Center (NRC), Faculty of Medical Science, Lebanese University, Hadath, Beirut, Lebanon
| | - Nissrine Ballout
- Neuroscience Research Center (NRC), Faculty of Medical Science, Lebanese University, Hadath, Beirut, Lebanon
| | - Tareq Al Sagheer
- Neuroscience Research Center (NRC), Faculty of Medical Science, Lebanese University, Hadath, Beirut, Lebanon
| |
Collapse
|
21
|
Liu Z, Shen C, Li H, Tong J, Wu Y, Ma Y, Wang J, Wang Z, Li Q, Zhang X, Dong H, Yang Y, Yu M, Wang J, Zhou R, Fei J, Huang F. NOD-like receptor NLRC5 promotes neuroinflammation and inhibits neuronal survival in Parkinson's disease models. J Neuroinflammation 2023; 20:96. [PMID: 37072793 PMCID: PMC10111753 DOI: 10.1186/s12974-023-02755-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 03/02/2023] [Indexed: 04/20/2023] Open
Abstract
Parkinson's disease (PD) is mainly characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and neuroinflammation mediated by overactivated microglia and astrocytes. NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) has been reported to participate in various immune disorders, but its role in neurodegenerative diseases remains unclear. In the current study, we found that the expression of NLRC5 was increased in the nigrostriatal axis of mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced PD, as well as in primary astrocytes, microglia and neurons exposed to different neurotoxic stimuli. In an acute MPTP-induced PD model, NLRC5 deficiency significantly reduced dopaminergic system degeneration and ameliorated motor deficits and striatal inflammation. Furthermore, we found that NLRC5 deficiency decreased the expression of the proinflammatory genes IL-1β, IL-6, TNF-α and COX2 in primary microglia and primary astrocytes treated with neuroinflammatory stimuli and reduced the inflammatory response in mixed glial cells in response to LPS treatment. Moreover, NLRC5 deficiency suppressed activation of the NF-κB and MAPK signaling pathways and enhanced the activation of AKT-GSK-3β and AMPK signaling in mixed glial cells. Furthermore, NLRC5 deficiency increased the survival of primary neurons treated with MPP+ or conditioned medium from LPS-stimulated mixed glial cells and promoted activation of the NF-κB and AKT signaling pathways. Moreover, the mRNA expression of NLRC5 was decreased in the blood of PD patients compared to healthy subjects. Therefore, we suggest that NLRC5 promotes neuroinflammation and dopaminergic degeneration in PD and may serve as a marker of glial activation.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Heng 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
| | - Jiabin Tong
- 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 Wu
- 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
| | - 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
| | - Zishan 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
| | - 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
| | - 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
| | - 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
| | - Yufang Yang
- 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 Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Renyuan Zhou
- 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., Shanghai, 201203, 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
|
22
|
Ma C, Liu Y, Li S, Ma C, Huang J, Wen S, Yang S, Wang B. Microglial cGAS drives neuroinflammation in the MPTP mouse models of Parkinson's disease. CNS Neurosci Ther 2023. [PMID: 36914567 DOI: 10.1111/cns.14157] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Neuroinflammation has been widely accepted as a cause of the degenerative process. Increasing interest has been devoted to developing intervening therapeutics for preventing neuroinflammation in Parkinson's disease (PD). It is well known that virus infections, including DNA viruses, are associated with an increased risk of PD. In addition, damaged or dying dopaminergic neurons can release dsDNA during PD progression. However, the role of cGAS, a cytosolic dsDNA sensor, in PD progression remains unclear. METHODS Adult male wild-type mice and age-matched male cGAS knockout (cGas-/- ) mice were treated with MPTP to induce neurotoxic PD model, and then behavioral tests, immunohistochemistry, and ELISA were conducted to compare disease phenotype. Chimeric mice were reconstituted to explore the effects of cGAS deficiency in peripheral immune cells or CNS resident cells on MPTP-induced toxicity. RNA sequencing was used to dissect the mechanistic role of microglial cGAS in MPTP-induced toxicity. cGAS inhibitor administration was conducted to study whether GAS may serve as a therapeutic target. RESULTS We observed that the cGAS-STING pathway was activated during neuroinflammation in MPTP mouse models of PD. cGAS deficiency in microglia, but not peripheral immune cells, controlled neuroinflammation and neurotoxicity induced by MPTP. Mechanistically, microglial cGAS ablation alleviated the neuronal dysfunction and inflammatory response in astrocytes and microglia by inhibiting antiviral inflammatory signaling. Additionally, the administration of cGAS inhibitors conferred the mice neuroprotection during MPTP exposure. CONCLUSIONS Collectively, these findings demonstrate microglial cGAS promote neuroinflammation and neurodegeneration during the progression of MPTP-induced PD mouse models and suggest cGAS may serve as a therapeutic target for PD patients. LIMITATIONS OF THE STUDY Although we demonstrated that cGAS promotes the progression of MPTP-induced PD, this study has limitations. We identified that cGAS in microglia accelerate disease progression of PD by using bone marrow chimeric experiments and analyzing cGAS expression in CNS cells, but evidence would be more straightforward if conditional knockout mice were used. This study contributed to the knowledge of the role of the cGAS pathway in PD pathogenesis; nevertheless, trying more PD animal models in the future will help us to understand the disease progression deeper and explore possible treatments.
Collapse
Affiliation(s)
- Chunmei Ma
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, Nanjing Medical University, Nanjing, China
| | - Ying Liu
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sheng Li
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, Nanjing Medical University, Nanjing, China.,Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chanyuan Ma
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jiajia Huang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuang Wen
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, Nanjing Medical University, Nanjing, China
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, Nanjing Medical University, Nanjing, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Mobahat M, Sadroddiny E, Nooshabadi VT, Ebrahimi-Barough S, Goodarzi A, Malekshahi ZV, Ai J. Curcumin-loaded human endometrial stem cells derived exosomes as an effective carrier to suppress alpha-synuclein aggregates in 6OHDA-induced Parkinson's disease mouse model. Cell Tissue Bank 2023; 24:75-91. [PMID: 35641803 DOI: 10.1007/s10561-022-10008-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/22/2022] [Indexed: 11/25/2022]
Abstract
Parkinson disease (PD) is considered as one of the most worldwide neurodegenerative disorders. The major reasons associated to neurodegeneration process of PD pathogenesis are oxidative stress. Many studies reported that natural antioxidant molecules, especially, curcumin can suppress inflammatory pathways and preserve dopaminergic neurons damage in PD. Further, the poor pharmacokinetics, instability of chemical structure because of fast hydrolytic degradation at physiologic condition and especially, the presence of the blood brain barrier (BBB) has regarded as a considerable restriction factor for transfer of neurotherapeutic molecules to the brain tissue. The present research aims to the fabrication of nanoformulated curcumin loaded human endometrial stem cells derived exosomes (hEnSCs EXOs-Cur) to study on enhancing curcumin penetration to the brain across BBB and to improve anti- Parkinsonism effects of curcumin against neural death and alpha-synuclein aggregation. hEnSCs EXOs-Cur characterization results demonstrated the accurate size and morphology of formulated curcumin loaded exosomes with a proper stability and sustained release profile. In vivo studies including behavioral, Immunohistochemical and molecular evaluations displayed that novel formulation of hEnSCs EXO-Cur is able to cross BBB, enhance motor uncoordinated movements, suppress the aggregation of αS protein and rescue neuronal cell death through elevation of BCL2 expression level as an anti-apoptotic protein and the expression level reduction of BAX and Caspase 3 as apoptotic markers.
Collapse
Affiliation(s)
- Mahsa Mobahat
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, , School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Goodarzi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Shiraz, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, , School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Zong Q, Pan Y, Liu Y, Wu Z, Huang Z, Zhang Y, Ma K. pNaktide mitigates inflammation-induced neuronal damage and behavioral deficits through the oxidative stress pathway. Int Immunopharmacol 2023; 116:109727. [PMID: 36689848 DOI: 10.1016/j.intimp.2023.109727] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023]
Abstract
Neuroinflammation is closely related to the etiology and progression of neurodegenerative diseases such as Parkinson disease and Alzheimer disease. pNaktide, an Src inhibitor, exerts antioxidant effects by mimicking Na/K-ATPase. It has been verified that its anti-inflammation and anti-oxidation ability could be embodied in obesity, steatohepatitis, uremic cardiomyopathy, aging, and prostate cancer. This study aimed to investigate the effects and mechanisms of pNaktide in lipopolysaccharide (LPS)-induced behavioral damage, neuroinflammation, and neuronal damage. We found that pNaktide improved anxiety, memory, and motor deficits. pNaktide inhibited MAPK and NF-κB pathways induced by TLR4 activation, inhibited the NLRP3 inflammasome complex, and reduced the expression of inflammatory factors, complement factors, and chemokines. pNaktide inhibited the activation of Nrf2 and HO-1 antioxidant stress pathways by LPS and reduced the level of oxidative stress. Inhibition of autophagy and enhancement of apoptosis induced by LPS were also alleviated by pNaktide, which restored LPS-induced injury to newborn neurons in the hippocampus region. In summary, pNaktide attenuates neuroinflammation, reduces the level of oxidative stress, has neuroprotective effects, and may be used for the treatment of neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Qinglan Zong
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Yongfang Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Zhengcun Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Zhangqiong Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Kaili Ma
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| |
Collapse
|
25
|
ElGamal RZ, Tadros MG, Menze ET. Linagliptin counteracts rotenone's toxicity in non-diabetic rat model of Parkinson's disease: Insights into the neuroprotective roles of DJ-1, SIRT-1/Nrf-2 and implications of HIF1-α. Eur J Pharmacol 2023; 941:175498. [PMID: 36623635 DOI: 10.1016/j.ejphar.2023.175498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/04/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
While all current therapies' main focus is enhancing dopaminergic effects and remission of symptoms, delaying Parkinson's disease (PD) progression remains a challenging mission. Linagliptin, a Dipeptidyl Peptidase-4 (DPP-4) Inhibitor, exhibited neuroprotection in various neurodegenerative diseases. This study aims to evaluate the neuroprotective effects of Linagliptin in a rotenone-induced rat model of PD and investigate the possible underlying mechanisms of Linagliptin's actions. The effects of two doses of Linagliptin (5 and 10 mg/kg) on spontaneous locomotion, catalepsy, coordination and balance, and histology were assessed. Then, after Linagliptin showed promising results, it was further tested for its potential anti-inflammatory, antiapoptotic effects, and different pathways for oxidative stress. Linagliptin prevented rotenone-induced motor deficits and histological damage. Besides, it significantly inhibited the rotenone-induced increase in pro-inflammatory cytokines: Tumor Necrosis Factor-α (TNF-α) and Interleukin-6 (IL-6) and decrease in caspase 3 levels. These effects were associated with induction in the levels of Protein deglycase also known as DJ-1, Hypoxia-inducible factor 1-alpha (HIF-1α), potentiation in the Sirtuin 1 (SIRT-1)/Nuclear factor erythroid-2-related factor 2 (Nrf-2)/Heme oxygenase-1 (HO-1) pathway, and an increase in the antioxidant activity of catalase which provided neuroprotection to the neurons from rotenone-induced PD. Collectively, these results suggest that Linagliptin might be a suitable candidate for the management of PD.
Collapse
Affiliation(s)
- Rania Z ElGamal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University - Kantara Branch, Ismailia, 41636, Egypt.
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
26
|
Huang B, Hu G, Zong X, Yang S, He D, Gao X, Liu D. α-Cyperone protects dopaminergic neurons and inhibits neuroinflammation in LPS-induced Parkinson's disease rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Int Immunopharmacol 2023; 115:109698. [PMID: 36634417 DOI: 10.1016/j.intimp.2023.109698] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Our previous study showed that α-Cyperone inhibited the inflammatory response triggered by activated microglia and protected dopaminergic neuron in in vitro cell model of Parkinson's disease (PD). It is unclear the effect of α-Cyperone in animal models of PD. In this study, our results indicated that α-Cyperone ameliorated motor dysfunction, protected dopaminergic neurons, and inhibited the reduction of dopamine and its metabolites in lipopolysaccharide (LPS)-induced PD rat model. Moreover, α-Cyperone suppressed the activation of microglia and the expression of neuroinflammatory factor (TNF-α, IL-6, IL-1β, iNOS, COX-2 and ROS). Furthermore, the molecular mechanism research revealed that α-Cyperone inhibited neuroinflammation and oxidative stress to exert protective effect in microglia by activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Moreover, α-Cyperone upregulated the expression of antioxidant enzymes (GCLC, GCLM and NQO1) in microglia. In conclusion, our study demonstrates α-Cyperone alleviates dopaminergic neurodegeneration by inhibiting neuroinflammation and oxidative stress in LPS-induced PD rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bingxu Huang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Guiqiu Hu
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaofeng Zong
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuo Yang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dewei He
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Xiyu Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dianfeng Liu
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China.
| |
Collapse
|
27
|
Investigating Therapeutic Effects of Indole Derivatives Targeting Inflammation and Oxidative Stress in Neurotoxin-Induced Cell and Mouse Models of Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24032642. [PMID: 36768965 PMCID: PMC9917106 DOI: 10.3390/ijms24032642] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Neuroinflammation and oxidative stress have been emerging as important pathways contributing to Parkinson's disease (PD) pathogenesis. In PD brains, the activated microglia release inflammatory factors such as interleukin (IL)-β, IL-6, tumor necrosis factor (TNF)-α, and nitric oxide (NO), which increase oxidative stress and mediate neurodegeneration. Using 1-methyl-4-phenylpyridinium (MPP+)-activated human microglial HMC3 cells and the sub-chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD, we found the potential of indole derivative NC009-1 against neuroinflammation, oxidative stress, and neurodegeneration for PD. In vitro, NC009-1 alleviated MPP+-induced cytotoxicity, reduced NO, IL-1β, IL-6, and TNF-α production, and suppressed NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in MPP+-activated HMC3 cells. In vivo, NC009-1 ameliorated motor deficits and non-motor depression, increased dopamine and dopamine transporter levels in the striatum, and reduced oxidative stress as well as microglia and astrocyte reactivity in the ventral midbrain of MPTP-treated mice. These protective effects were achieved by down-regulating NLRP3, CASP1, iNOS, IL-1β, IL-6, and TNF-α, and up-regulating SOD2, NRF2, and NQO1. These results strengthen the involvement of neuroinflammation and oxidative stress in PD pathogenic mechanism, and indicate NC009-1 as a potential drug candidate for PD treatment.
Collapse
|
28
|
Gubinelli F, Sarauskyte L, Venuti C, Kulacz I, Cazzolla G, Negrini M, Anwer D, Vecchio I, Jakobs F, Manfredsson F, Davidsson M, Heuer A. Characterisation of functional deficits induced by AAV overexpression of alpha-synuclein in rats. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 4:100065. [PMID: 36632447 PMCID: PMC9827042 DOI: 10.1016/j.crneur.2022.100065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Background In the last decades different preclinical animal models of Parkinson's disease (PD) have been generated, aiming to mimic the progressive neuronal loss of midbrain dopaminergic (DA) cells as well as motor and non-motor impairment. Among all the available models, AAV-based models of human alpha-synuclein (h-aSYN) overexpression are promising tools for investigation of disease progression and therapeutic interventions. Objectives The goal with this work was to characterise the impairment in motor and non-motor domains following nigrostriatal overexpression of h-aSYN and correlate the behavioural deficits with histological assessment of associated pathology. Methods Intranigral injection of an AAV9 expressing h-aSYN was compared with untreated animals, 6-OHDA and AAV9 expressing either no transgene or GFP. The animals were assessed on a series of simple and complex behavioural tasks probing motor and non-motor domains. Post-mortem neuropathology was analysed using immunohistochemical methods. Results Overexpression of h-aSYN led to progressive degeneration of DA neurons of the SN and axonal terminals in the striatum (STR). We observed extensive nigral and striatal pathology, resembling that of human PD brain, as well as the development of stable progressive deficit in simple motor tasks and in non-motor domains such as deficits in motivation and lateralised neglect. Conclusions In the present work we characterized a rat model of PD that closely resembles human PD pathology at the histological and behavioural level. The correlation of cell loss with behavioural performance enables the selection of rats which can be used in neuroprotective or neurorestorative therapies.
Collapse
Affiliation(s)
- F. Gubinelli
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - L. Sarauskyte
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - C. Venuti
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - I. Kulacz
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - G. Cazzolla
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - M. Negrini
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - D. Anwer
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - I. Vecchio
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - F. Jakobs
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - F.P. Manfredsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - M. Davidsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA,Molecular Neuromodulation, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - A. Heuer
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden,Corresponding author. Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Sölvegatan 19, 22 184, Lund, Sweden.
| |
Collapse
|
29
|
Rahman MM, Wang X, Islam MR, Akash S, Supti FA, Mitu MI, Harun-Or-Rashid M, Aktar MN, Khatun Kali MS, Jahan FI, Singla RK, Shen B, Rauf A, Sharma R. Multifunctional role of natural products for the treatment of Parkinson's disease: At a glance. Front Pharmacol 2022; 13:976385. [PMID: 36299886 PMCID: PMC9590378 DOI: 10.3389/fphar.2022.976385] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
Natural substances originating from plants have long been used to treat neurodegenerative disorders (NDs). Parkinson's disease (PD) is a ND. The deterioration and subsequent cognitive impairments of the midbrain nigral dopaminergic neurons distinguish by this characteristic. Various pathogenic mechanisms and critical components have been reported, despite the fact that the origin is unknown, such as protein aggregation, iron buildup, mitochondrial dysfunction, neuroinflammation and oxidative stress. Anti-Parkinson drugs like dopamine (DA) agonists, levodopa, carbidopa, monoamine oxidase type B inhibitors and anticholinergics are used to replace DA in the current treatment model. Surgery is advised in cases where drug therapy is ineffective. Unfortunately, the current conventional treatments for PD have a number of harmful side effects and are expensive. As a result, new therapeutic strategies that control the mechanisms that contribute to neuronal death and dysfunction must be addressed. Natural resources have long been a useful source of possible treatments. PD can be treated with a variety of natural therapies made from medicinal herbs, fruits, and vegetables. In addition to their well-known anti-oxidative and anti-inflammatory capabilities, these natural products also play inhibitory roles in iron buildup, protein misfolding, the maintenance of proteasomal breakdown, mitochondrial homeostasis, and other neuroprotective processes. The goal of this research is to systematically characterize the currently available medications for Parkinson's and their therapeutic effects, which target diverse pathways. Overall, this analysis looks at the kinds of natural things that could be used in the future to treat PD in new ways or as supplements to existing treatments. We looked at the medicinal plants that can be used to treat PD. The use of natural remedies, especially those derived from plants, to treat PD has been on the rise. This article examines the fundamental characteristics of medicinal plants and the bioactive substances found in them that may be utilized to treat PD.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Xiaoyan Wang
- Department of Pathology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Fatema Akter Supti
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mohona Islam Mitu
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Harun-Or-Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Most. Nazmin Aktar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Most. Sumaiya Khatun Kali
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Farhana Israt Jahan
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Pakistan
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
30
|
Zeng J, Wang X, Pan F, Mao Z. The relationship between Parkinson's disease and gastrointestinal diseases. Front Aging Neurosci 2022; 14:955919. [PMID: 36034146 PMCID: PMC9399652 DOI: 10.3389/fnagi.2022.955919] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/25/2022] [Indexed: 01/03/2023] Open
Abstract
An increasing number of studies have provided evidence for the hypothesis that the pathogenesis of Parkinson's disease (PD) may derive from the gut. Firstly, Lewy pathology can be induced in the enteric nervous system (ENS) and be transported to the central nervous system (CNS) via the vagal nerve. Secondly, the altered composition of gut microbiota causes an imbalance between beneficial and deleterious microbial metabolites which interacts with the increased gut permeability and the gut inflammation as well as the systemic inflammation. The activated inflammatory status then affects the CNS and promotes the pathology of PD. Given the above-mentioned findings, researchers start to pay attention to the connection between PD and gastrointestinal diseases including irritable bowel syndrome, inflammatory bowel disease (IBD), microscopic colitis (MC), gastrointestinal infections, gastrointestinal neoplasms, and colonic diverticular disease (CDD). This review focuses on the association between PD and gastrointestinal diseases as well as the pathogenesis of PD from the gut.
Collapse
Affiliation(s)
- Jiaqi Zeng
- Department of Gastroenterology and Hepatology, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xinchan Wang
- Department of Gastroenterology and Hepatology, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Nankai University, Tianjin, China
| | - Fei Pan
- Department of Gastroenterology and Hepatology, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhiqi Mao
- Department of Neurosurgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
31
|
Marino F, Pinoli M, Rasini E, Martini S, Luini A, Pulze L, Dalla Gasperina D, Grossi P, Legnaro M, Ferrari M, Congiu T, Pacheco R, Osorio-Barrios F, de Eguileor M, Cosentino M. Dopaminergic Inhibition of Human Neutrophils is Exerted Through D1-Like Receptors and Affected By Bacterial Infection. Immunol Suppl 2022; 167:508-527. [PMID: 35897164 DOI: 10.1111/imm.13550] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/11/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Dopamine (DA) affects immune functions in healthy subjects and during disease by acting on D1-like (D1 and D5) and D2-like (D2, D3 and D4) dopaminergic receptors (DR), however its effects on human polymorphonuclear leukocytes (PMN) are still poorly defined. METHODS We investigated DR expression in human PMN and the ability of DA to affect cell migration and reactive oxygen species (ROS) production. Experiments were performed on cells from healthy subjects (HS) and from patients (Pts) with bacterial infections as well, during the acute phase and after recovery. Some experiments were also performed in mice KO for the DRD5 gene. RESULTS PMN from HS express both D1-like and D2-like DR, and exposure to DA results in inhibition of activation-induced morphological changes, migration and ROS production which depend on the activation of D1-like DR. In agreement with these findings, DA inhibited migration of PMN obtained from wild-type mice, but not from DR D5 KO mice. In Pts with bacterial infections, during the febrile phase D1-like DR D5 on PMN were downregulated and DA failed to affect PMN migration. Both D1-like DR D5 expression and DA-induced inhibition of PMN migration were however restored after recovery. CONCLUSION Dopaminergic inhibition of human PMN is a novel mechanism which is likely to play a key role in the regulation of innate immunity. Evidence obtained in Pts with bacterial infections provides novel clues for the therapeutic modulation of PMN during infectious disease.
Collapse
Affiliation(s)
- Franca Marino
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| | - Monica Pinoli
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| | - Emanuela Rasini
- Center for Research in Medical Pharmacology University of Insubria, Varese, Italy
| | - Stefano Martini
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| | - Alessandra Luini
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| | - Laura Pulze
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | | | - Paolo Grossi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Massimiliano Legnaro
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| | - Marco Ferrari
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| | - Terenzio Congiu
- Department of Surgical Sciences, University of Cagliari, Italy
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Ñuñoa, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile
| | | | - Magda de Eguileor
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Marco Cosentino
- Center for Research in Medical Pharmacology; University of Insubria, Varese, Italy
| |
Collapse
|
32
|
Joshi P, Bisht A, Joshi S, Semwal D, Nema NK, Dwivedi J, Sharma S. Ameliorating potential of curcumin and its analogue in central nervous system disorders and related conditions: A review of molecular pathways. Phytother Res 2022; 36:3143-3180. [PMID: 35790042 DOI: 10.1002/ptr.7522] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/26/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Curcumin, isolated from turmeric (Curcuma longa L.) is one of the broadly studied phytomolecule owing to its strong antioxidant and anti-inflammatory potential and has been considered a promising therapeutic candidate in a wide range of disorders. Considering, its low bioavailability, different curcumin analogs have been developed to afford desired pharmacokinetic profile and therapeutic outcome in varied pathological states. Several preclinical and clinical studies have indicated that curcumin ameliorates mitochondrial dysfunction, inflammation, oxidative stress apoptosis-mediated neural cell degeneration and could effectively be utilized in the treatment of different neurodegenerative diseases. Hence, in this review, we have summarized key findings of experimental and clinical studies conducted on curcumin and its analogues with special emphasis on molecular pathways, viz. NF-kB, Nrf2-ARE, glial activation, apoptosis, angiogenesis, SOCS/JAK/STAT, PI3K/Akt, ERK1/2 /MyD88 /p38 MAPK, JNK, iNOS/NO, and MMP pathways involved in imparting ameliorative effects in the therapy of neurodegenerative disorders and associated conditions.
Collapse
Affiliation(s)
- Priyanka Joshi
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.,R & D, Patanjali Ayurved Ltd, Patanjali Food and Herbal Park, Haridwar, Uttarakhand, India
| | - Akansha Bisht
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| | - Sushil Joshi
- R & D, Patanjali Ayurved Ltd, Patanjali Food and Herbal Park, Haridwar, Uttarakhand, India
| | - Deepak Semwal
- Faculty of Biomedical Sciences, Uttarakhand Ayurved University, Dehradun, Uttarakhand, India
| | - Neelesh Kumar Nema
- Paramount Kumkum Private Limited, Prestige Meridian-1, Bangalore, Karnataka, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| |
Collapse
|
33
|
Bian LH, Yao ZW, Wang ZY, Wang XM, Li QY, Yang X, Li JY, Wei XJ, Wan GH, Wang YQ, Shi JL, Guo JY. Nardosinone regulates the slc38a2 gene to alleviate Parkinson's symptoms in rats through the GABAergic synaptic and cAMP pathways. Biomed Pharmacother 2022; 153:113269. [PMID: 35728354 DOI: 10.1016/j.biopha.2022.113269] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 11/02/2022] Open
Abstract
In a rotenone-induced Parkinson's disease (PD) rat model, behavioral investigation, pathological examination, inflammatory factor analysis, and mitochondrial structure and function investigation verified the anti-PD efficacy of nardosinone. A combined transcriptome and proteome analysis proposed that the anti-PD target of nardosinone is the slc38a2 gene and may involve the GABAergic synaptic pathway and cAMP-signaling pathway. Analysis of targeted slc38a2 knockout cells and expression of key enzyme-encoding genes in both pathways verified the target and pathways proposed by the 'omics analysis. This further confirms that nardosinone can regulate the slc38a2 gene, a potential new target for the treatment of Parkinson's disease, and plays an anti-PD role through the GABAergic synaptic and cAMP pathways.
Collapse
Affiliation(s)
- Li-Hua Bian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zi-Wei Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Zhe-Yi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China; Qilu Hospital, Shandong University, Jinan 250012, Shandong, China.
| | - Xiao-Mei Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Qiu-Yu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Xue Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Jia-Yuan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Xiao-Jia Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Guo-Hui Wan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Yu-Qing Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Jin-Li Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Jian-You Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China.
| |
Collapse
|
34
|
Dietary fat intake and risk of Parkinson disease: results from the Swedish National March Cohort. Eur J Epidemiol 2022; 37:603-613. [PMID: 35416636 PMCID: PMC9288363 DOI: 10.1007/s10654-022-00863-8] [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: 10/15/2021] [Accepted: 03/15/2022] [Indexed: 11/28/2022]
Abstract
Background Following progressive aging of the population worldwide, the prevalence of Parkinson disease is expected to increase in the next decades. Primary prevention of the disease is hampered by limited knowledge of preventable causes. Recent evidence regarding diet and Parkinson disease is inconsistent and suggests that dietary habits such as fat intake may have a role in the etiology. Objective To investigate the association between intake of total and specific types of fat with the incidence of Parkinson disease. Methods Participants from the Swedish National March Cohort were prospectively followed-up from 1997 to 2016. Dietary intake was assessed at baseline using a validated food frequency questionnaire. Food items intake was used to estimate fat intake, i.e. the exposure variable, using the Swedish Food Composition Database. Total, saturated, monounsaturated and polyunsaturated fat intake were categorized into quartiles. Parkinson disease incidence was ascertained through linkages to Swedish population-based registers. Cox proportional hazards regression models were used to estimate hazard ratios (HR) with 95% confidence intervals (CI) of the association between fat intake from total or specific types of fats and the incidence of Parkinson disease. The lowest intake category was used as reference. Isocaloric substitution models were also fitted to investigate substitution effects by replacing energy from fat intake with other macronutrients or specific types of fat. Results 41,597 participants were followed up for an average of 17.6 years. Among them, 465 developed Parkinson disease. After adjusting for potential confounders, the highest quartile of saturated fat intake was associated with a 41% increased risk of Parkinson disease compared to the lowest quartile (HR Q4 vs. Q1: 1.41; 95% CI: 1.04–1.90; p for trend: 0.03). Total, monounsaturated or polyunsaturated fat intake were not significantly associated with Parkinson disease. The isocaloric substitution models did not show any effect. Conclusions We found that a higher consumption of large amounts of saturated fat might be associated with an increased risk of Parkinson disease. A diet low in saturated fat might be beneficial for disease prevention. Supplementary Information The online version contains supplementary material available at 10.1007/s10654-022-00863-8.
Collapse
|
35
|
Chakkittukandiyil A, Sajini DV, Karuppaiah A, Selvaraj D. The principal molecular mechanisms behind the activation of Keap1/Nrf2/ARE pathway leading to neuroprotective action in Parkinson's disease. Neurochem Int 2022; 156:105325. [PMID: 35278519 DOI: 10.1016/j.neuint.2022.105325] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder. PD is associated with the loss of dopaminergic neurons in the substantia nigra pars compacta region of the midbrain. Present therapies for PD provide only symptomatic relief by restoring the dopamine (DA) level. However, they are not disease modifying agents and so they do not delay the disease progression. Alpha-synuclein aggregation, oxidative stress, mitochondrial dysfunction and chronic inflammation are considered to be the major pathological mechanisms mediating neurodegeneration in PD. To resist oxidative stress, the human body has an antioxidant defence mechanism consisting of many antioxidants and cytoprotective genes. The expression of those genes are largely controlled by the Kelch-like ECH-associated protein 1/Nuclear factor - erythroid - 2 - related factor 2/Antioxidant response element (Keap1/Nrf2/ARE) signalling pathway. The transcription factor Nrf2 is activated in response to oxidative or electrophilic stress and protects the cells from oxidative stress and inflammation. Nrf2 has been widely considered as a therapeutic target for neurodegeneration and several drugs are now being tested in clinical trials. Regulation of the Keap1/Nrf2/ARE pathway by small molecules which can act as Nrf2 activators could be effective for treating oxidative stress and neuroinflammation in PD. In this review, we had discussed the principal molecular mechanisms behind the neuroprotective effects of Keap1/Nrf2/ARE pathway in PD. Additionally, we also discussed the small molecules and phytochemicals that could activate the Nrf2 mediated anti-oxidant pathway for neuroprotection in PD.
Collapse
Affiliation(s)
- Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Deepak Vasudevan Sajini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Arjunan Karuppaiah
- Department of Pharmaceutics, PSG College of Pharmacy, Peelamedu, Coimbatore, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| |
Collapse
|
36
|
Ponce J, Ulu A, Hanson C, Cameron-Smith E, Bertoni J, Wuebker J, Fisher A, Siu KC, Marmelat V, Adamec J, Bhatti D. Role of Specialized Pro-resolving Mediators in Reducing Neuroinflammation in Neurodegenerative Disorders. Front Aging Neurosci 2022; 14:780811. [PMID: 35250536 PMCID: PMC8891627 DOI: 10.3389/fnagi.2022.780811] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are neurodegenerative disorders that affect millions of individuals worldwide. As incidence of these conditions increases with age, there will undoubtedly be an increased prevalence of cases in the near future. Neuroinflammation is a hallmark in the development and progression of neurodegenerative diseases and prevention or resolution of chronic neuroinflammation may represent a novel approach to treatment. The present review highlights the potential of the anti-inflammatory and pro-resolving effects of polyunsaturated fatty acid (PUFA)-derived mediators (Specialized Pro-resolving Mediators—SPM) in neurodegenerative disorders. PUFA-derived SPM are biosynthesized in response to chemicals produced from acute inflammatory responses. Preclinical studies from both AD and PD models suggest a dysregulation of SPM and their receptors in neurological disorders. Decreased SPM may be due to inadequate substrate, an imbalance between SPM and pro-inflammatory mediators or a disruption in SPM synthesis. SPMs hold great promise for neuroprotection in AD by altering expression of pro-inflammatory genes, modulating macrophage function, serving as a biomarker for AD status, and promoting resolution of neuroinflammation. In PD, data suggest SPM are able to cross the blood-brain barrier, inhibit microglial activation and decrease induced markers of inflammation, possibly as a result of their ability to downregulate NFκB signaling pathways. Several in vivo and in vitro studies suggest a benefit from administration of SPMs in both neurodegenerative disorders. However, extrapolation of these outcomes to humans is difficult as no models are able to replicate all features of AD or PD. Minimal data evaluating these PUFA-derived metabolites in humans with neurodegenerative disorders are available and a gap in knowledge exists regarding behavior of SPM and their receptors in patients with these conditions. There is also large gap in our knowledge regarding which lipid mediator would be most effective in which model of AD or PD and how dietary intake or supplementation can impact SPM levels. Future direction should include focused, translational efforts to investigate SPM as an add-on (in addition to standard treatment) or as standalone agents in patients with neurodegenerative disorders.
Collapse
Affiliation(s)
- Jana Ponce
- Division of Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Jana Ponce,
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Corrine Hanson
- Division of Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
| | - Erin Cameron-Smith
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - John Bertoni
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jenna Wuebker
- Department of Pharmaceutical and Nutrition Care, Nebraska Medicine, Omaha, NE, United States
| | - Alfred Fisher
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Ka-Chun Siu
- Division of Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
| | - Vivien Marmelat
- Department of Biomechanics, College of Education, Health, and Human Sciences, University of Nebraska - Omaha, Omaha, NE, United States
| | - Jiri Adamec
- Department of Biochemistry, College of Arts and Sciences, University of Nebraska - Lincoln, Lincoln, NE, United States
| | - Danish Bhatti
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
37
|
Kasen A, Houck C, Burmeister AR, Sha Q, Brundin L, Brundin P. Upregulation of α-synuclein following immune activation: Possible trigger of Parkinson's disease. Neurobiol Dis 2022; 166:105654. [DOI: 10.1016/j.nbd.2022.105654] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/20/2022] Open
|
38
|
Lackova A, Beetz C, Oppermann S, Bauer P, Pavelekova P, Lorincova T, Ostrozovicova M, Kulcsarova K, Cobejova J, Cobej M, Levicka P, Liesenerova S, Sendekova D, Sukovska V, Gdovinova Z, Han V, Rizig M, Houlden H, Skorvanek M. Prevalence of Fabry Disease among Patients with Parkinson's Disease. PARKINSON'S DISEASE 2022; 2022:1014950. [PMID: 35111290 PMCID: PMC8803460 DOI: 10.1155/2022/1014950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND An increased prevalence of Parkinson's disease (PD) disease has been previously reported in subjects with Fabry disease (FD) carrying alpha-galactosidase (GLA) mutations and their first-line relatives. Moreover, decreased alpha-galactosidase A (AGLA) enzymatic activity has been reported among cases with PD compared to controls. OBJECTIVE The aim of our study was to determine the prevalence of FD among patients with PD. METHODS We recruited 236 consecutive patients with PD from February 2018 to December 2020. Clinical and sociodemographic data, including the MDS-UPDRS-III scores and HY stage (the Hoehn and Yahr scale), were collected, and in-depth phenotyping was performed in subjects with identified GLA variants. A multistep approach, including standard determination of AGLA activity and LysoGb3 in males, and next-generation based GLA sequencing in all females and males with abnormal AGLA levels was performed in a routine diagnostic setting. RESULTS The mean age of our patients was 68.9 ± 8.9 years, 130 were men (55.1%), and the mean disease duration was 7.77 ± 5.35 years. Among 130 men, AGLA levels were low in 20 patients (15%), and subsequent Lyso-Gb3 testing showed values within the reference range for all tested subjects. In 126 subsequently genetically tested patients, four heterozygous p.(Asp313Tyr) GLA variants (3.2%, MAF 0.016) were identified; all were females. None of the 4 GLA variant carriers identified had any clinical manifestation suggestive of FD. CONCLUSIONS The results of this study suggest a possible relationship between FD and PD in a small proportion of cases. Nevertheless, the GLA variant found in our cohort is classified as a variant of unknown significance. Therefore, its pathogenic causative role in the context of PD needs further elucidation, and these findings should be interpreted with caution.
Collapse
Affiliation(s)
- Alexandra Lackova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| | | | | | | | - Petra Pavelekova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| | - Tatiana Lorincova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Miriam Ostrozovicova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| | - Kristina Kulcsarova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| | - Jana Cobejova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Martin Cobej
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Petra Levicka
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Simona Liesenerova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Daniela Sendekova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Viktoria Sukovska
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
| | - Zuzana Gdovinova
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| | - Vladimir Han
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| | - Mie Rizig
- University College London, Institute of Neurology, Department of Neuromuscular Disorders, Queen Square, WC1N 3BG London, UK
| | - Henry Houlden
- University College London, Institute of Neurology, Department of Neuromuscular Disorders, Queen Square, WC1N 3BG London, UK
| | - Matej Skorvanek
- Department of Neurology, University of Pavol Jozef Šafárik, Košice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Košice, Slovakia
| |
Collapse
|
39
|
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: 18] [Impact Index Per Article: 9.0] [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
|
40
|
Kermanian F, Seghatoleslam M, Mahakizadeh S. MDMA related neuro-inflammation and adenosine receptors. Neurochem Int 2022; 153:105275. [PMID: 34990730 DOI: 10.1016/j.neuint.2021.105275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/25/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022]
Abstract
3,4-methylenedioxymethamphetamine (MDMA) is a world-wide abused psychostimulant, which has the neurotoxic effects on dopaminergic and serotonergic neurons in both rodents and non-human primates. Adenosine acts as a neurotransmitter in the brain through the activation of four specific G-protein-coupled receptors and it acts as a neuromodulator of dopamine neurotransmission. Recent studies suggest that stimulation of adenosine receptors oppose many behavioral effects of methamphetamines. This review summarizes the specific cellular mechanisms involved in MDMA neuroinflammatory effects, along with the protective effects of adenosine receptors.
Collapse
Affiliation(s)
- Fatemeh Kermanian
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Masoumeh Seghatoleslam
- Evaluative Clinical Sciences, Sunnybrook Research Institute, University of Toronto, ON, Canada
| | - Simin Mahakizadeh
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
41
|
Safar MM, Abdelkader NF, Ramadan E, Kortam MA, Mohamed AF. Novel mechanistic insights towards the repositioning of alogliptin in Parkinson's disease. Life Sci 2021; 287:120132. [PMID: 34774622 DOI: 10.1016/j.lfs.2021.120132] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/27/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease that impairs people's lives tremendously. The development of innovative treatment modalities for PD is a significant unmet medical need. The critical function of glucagon-like peptide-1 (GLP-1) in neurodegenerative diseases has raised impetus in investigating the repositioning of a dipeptidyl peptidase IV inhibitor, alogliptin (ALO), as an effective treatment for PD. As a result, the focus of this research was to assess the effect of ALO in a rat rotenone (ROT) model of PD. For 21 days, ROT (1.5 mg/kg) was delivered subcutaneously every other day. ALO (30 mg/kg/day), delivered by gavage for 21 days, recovered motor performance and improved motor coordination in the open-field and rotarod testing. These impacts were highlighted by restoring striatal dopamine content and correcting histological changes that occurred concurrently. The ALO molecular signaling was determined by increasing the quantity of GLP-1 and the protein expression of its downstream signaling pathway, pT172-AMPK/SIRT1/PGC-1α. Furthermore, it curbed neuroinflammation via hampering HMGB1/TLR4/NLRP3 inflammasome activation and conquered striatal microglia activation. Pre-administration of dorsomorphin reversed the neuroprotective effects. In conclusion, the promising neuroprotective effect of ALO highlights the repositioning of ALO as a prospective revolutionary candidate for combating PD.
Collapse
Affiliation(s)
- Marwa M Safar
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, the British University in Egypt, Cairo, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Noha F Abdelkader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Eman Ramadan
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, the British University in Egypt, Cairo, Egypt
| | - Mona A Kortam
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
42
|
Schilder BM, Navarro E, Raj T. Multi-omic insights into Parkinson's Disease: From genetic associations to functional mechanisms. Neurobiol Dis 2021; 163:105580. [PMID: 34871738 PMCID: PMC10101343 DOI: 10.1016/j.nbd.2021.105580] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/17/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Genome-Wide Association Studies (GWAS) have elucidated the genetic components of Parkinson's Disease (PD). However, because the vast majority of GWAS association signals fall within non-coding regions, translating these results into an interpretable, mechanistic understanding of the disease etiology remains a major challenge in the field. In this review, we provide an overview of the approaches to prioritize putative causal variants and genes as well as summarise the primary findings of previous studies. We then discuss recent efforts to integrate multi-omics data to identify likely pathogenic cell types and biological pathways implicated in PD pathogenesis. We have compiled full summary statistics of cell-type, tissue, and phentoype enrichment analyses from multiple studies of PD GWAS and provided them in a standardized format as a resource for the research community (https://github.com/RajLabMSSM/PD_omics_review). Finally, we discuss the experimental, computational, and conceptual advances that will be necessary to fully elucidate the effects of functional variants and genes on cellular dysregulation and disease risk.
Collapse
Affiliation(s)
- Brian M Schilder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; UK Dementia Research Institute at Imperial College London, London, United Kingdom.
| | - Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Sección Departamental de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
43
|
Su WM, Gu XJ, Hou YB, Zhang LY, Cao B, Ou RW, Wu Y, Chen XP, Song W, Zhao B, Shang HF, Chen YP. Association Analysis of WNT3, HLA-DRB5 and IL1R2 Polymorphisms in Chinese Patients With Parkinson's Disease and Multiple System Atrophy. Front Genet 2021; 12:765833. [PMID: 34868249 PMCID: PMC8636743 DOI: 10.3389/fgene.2021.765833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/29/2021] [Indexed: 02/05/2023] Open
Abstract
Background: The association between inflammation and neurodegeneration has long been observed in parkinson's disease (PD) and multiple system atrophy (MSA). Previous genome-wide association studies (GWAS) and meta-analyses have identified several risk loci in inflammation-associated genes associated with PD. Objective: To investigate whether polymorphisms in some inflammation-associated genes could modulate the risk of developing PD and MSA in a Southwest Chinese population. Methods: A total of 2,706 Chinese subjects comprising 1340 PD, 483 MSA and 883 healthy controls were recruited in the study. Three polymorphisms (rs2074404 GG/GT/TT, rs17425622 CC/CT/TT, rs34043159 CC/CT/TT) in genes linked to inflammation in all the subjects were genotyped by using the Sequenom iPLEX Assay. Results: The allele G of WNT3 rs2074404 can increase risk on PD (OR: 1.048, 95% CI: 1.182-1.333, p = 0.006), exclusively in the LOPD subgroup (OR: 1.166, 95% CI:1.025-1.327, p = 0.019), but not in EOPD or MSA. And the recessive model analysis also demonstrated an increased PD risk in GG genotype of this locus (OR = 1.331, p = 0.007). However, no significant differences were observed in the genotype distributions and alleles of HLA-DRB5 rs17425622 and IL1R2 rs34043159 between the PD patients and controls, between the MSA patients and controls, or between subgroups of PD or MSA and controls. Conclusion: Our results suggested the allele G of WNT3 rs2074404 have an adverse effect on PD and particularly, on the LOPD subgroup among a Chinese population.
Collapse
Affiliation(s)
- Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao-Jing Gu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Bing Hou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ling-Yu Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ru-Wei Ou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Wu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Song
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bi Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
- Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Mirzaei S, Kulkarni K, Zhou K, Crack PJ, Aguilar MI, Finkelstein DI, Forsythe JS. Biomaterial Strategies for Restorative Therapies in Parkinson's Disease. ACS Chem Neurosci 2021; 12:4224-4235. [PMID: 34634903 DOI: 10.1021/acschemneuro.1c00484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological disorder, in which dopaminergic midbrain neurons degenerate, leading to dopamine depletion that is associated with neuronal death. In this Review, we initially describe the pathogenesis of PD and established therapies that unfortunately only delay progression of the disease. With a rapidly escalating incidence in PD, there is an urgent need to develop new therapies that not only halt progression but even reverse degeneration. Biomaterials are playing critical roles in these new therapies which include controlled and site-specific delivery of neurotrophins, increased engraftment of implanted neural stem cells, and redirection of endogenous stem cell populations away from their niche to encourage reparative mechanisms. This Review will therefore cover important design features of biomaterials used in regenerative medicine and tissue engineering strategies targeted at PD.
Collapse
Affiliation(s)
- Samaneh Mirzaei
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Kun Zhou
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Peter J. Crack
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - David I. Finkelstein
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - John S. Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
45
|
Phosphorylated α-synuclein aggregated in Schwann cells exacerbates peripheral neuroinflammation and nerve dysfunction in Parkinson's disease through TLR2/NF-κB pathway. Cell Death Discov 2021; 7:289. [PMID: 34642321 PMCID: PMC8511120 DOI: 10.1038/s41420-021-00676-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/11/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
To investigate the mechanism of peripheral neuropathy in Parkinson's disease (PD), we prepared a PD mice model by long-term exposure of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to mimic PD pathology in humans and the sciatic nerves were taken for further research. It turned out that phosphorylated α-synuclein (p-α-syn) was significantly deposited in Schwann cells (SCs) of sciatic nerves possibly contributing to degenerated myelin SCs and atrophied axons in MPTP group. Further analysis confirmed that toll-like receptors (TLRs) were implicated with PD peripheral neuropathy, in which TLR2 exhibits the predominant expression. Increased expression of inflammatory factors about TLR2/nuclear factor kappa-B (NF-κB) pathway was noted in MPTP group compared to saline group, with proteins on other pathways showing no changes. Moreover, MPTP-challenged mice exhibited worse motor ability and damaged nerve conduction, implicating that p-α-syn neurotoxicity might be relevant to impairments of motor and sensory nerves. After the treatment of CU-CPT22, a TLR2 antagonist, p-α-syn accumulation, motor and sensory function were ameliorated in CU-CPT22 combined with MPTP group. Thus, we demonstrated that pathological p-α-syn might combine TLR2 to affect SCs activation, inflammatory response as well as motor and sensory function through TLR2/nuclear factor kappa-B (NF-κB) signaling pathway. This study firstly demonstrates a novel mechanism of p-α-syn accumulated in SCs of peripheral nerves, which extends our understanding on SCs-mediated peripheral neuroinflammation related to TLR2/NF-κB signaling pathway and sheds light on potential new therapeutic avenues for PD.
Collapse
|
46
|
Ishijima T, Nakajima K. Inflammatory cytokines TNFα, IL-1β, and IL-6 are induced in endotoxin- stimulated microglia through different signaling cascades. Sci Prog 2021; 104:368504211054985. [PMID: 34821182 PMCID: PMC10450609 DOI: 10.1177/00368504211054985] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
By using an animal model in which inflammatory cytokines are induced in lipopolysaccharide (LPS)-injected rat brain, we investigated the induction of tumor necrosis factor alpha (TNFα), interleukin-1beta (IL-1β), and IL-6. Immunoblotting and immunohistochemistry revealed that all three cytokines were transiently induced in the cerebral cortex at about 12 h after LPS injection. To clarify which glial cell type induced the cytokines, we examined the respective abilities of astrocytes and microglia in vitro. Primary microglia largely induced TNFα, IL-1β and IL-6 in response to LPS, but primary astrocytes induced only limited levels of TNFα. Thus, we used specific inhibitors to focus on microglia in surveying signaling molecules involved in the induction of TNFα, IL-1β, and IL-6. The experiments using mitogen-activated protein kinases (MAPK) inhibitors revealed that c-Jun N-terminal kinase (JNK)/p38, external signal regulated kinase (ERK)/JNK, and ERK/JNK/p38 are necessary for the induction of TNFα, IL-1β, and IL-6, respectively. The experiments using protein kinase C (PKC) inhibitor clarified that PKCα is required for the induction of all these cytokines in LPS-stimulated microglia. Furthermore, LPS-dependent IL-1β/IL-6 induction was suppressed by pretreatment with a nitric oxide (NO) scavenger, suggesting that NO is involved in the signaling cascade of IL-1β/IL-6 induction. Thus, an inducible NO synthase induced in the LPS-injected cerebral cortex might be related to the induction of IL-1β/IL-6 through the production of NO in vivo. Taken together, these results demonstrated that microglia induce different kinds of inflammatory cytokine through specific combinations of MAPKs and by the presence or absence of NO.
Collapse
Affiliation(s)
- Takashi Ishijima
- Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| | - Kazuyuki Nakajima
- Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| |
Collapse
|
47
|
Paul KC, Binder AM, Horvath S, Kusters C, Yan Q, Rosario ID, Yu Y, Bronstein J, Ritz B. Accelerated hematopoietic mitotic aging measured by DNA methylation, blood cell lineage, and Parkinson's disease. BMC Genomics 2021; 22:696. [PMID: 34565328 PMCID: PMC8474781 DOI: 10.1186/s12864-021-08009-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Aging and inflammation are important components of Parkinson's disease (PD) pathogenesis and both are associated with changes in hematopoiesis and blood cell composition. DNA methylation (DNAm) presents a mechanism to investigate inflammation, aging, and hematopoiesis in PD, using epigenetic mitotic aging and aging clocks. Here, we aimed to define the influence of blood cell lineage on epigenetic mitotic age and then investigate mitotic age acceleration with PD, while considering epigenetic age acceleration biomarkers. RESULTS We estimated epigenetic mitotic age using the "epiTOC" epigenetic mitotic clock in 10 different blood cell populations and in a population-based study of PD with whole-blood. Within subject analysis of the flow-sorted purified blood cell types DNAm showed a clear separation of epigenetic mitotic age by cell lineage, with the mitotic age significantly lower in myeloid versus lymphoid cells (p = 2.1e-11). PD status was strongly associated with accelerated epigenetic mitotic aging (AccelEpiTOC) after controlling for cell composition (OR = 2.11, 95 % CI = 1.56, 2.86, p = 1.6e-6). AccelEpiTOC was also positively correlated with extrinsic epigenetic age acceleration, a DNAm aging biomarker related to immune system aging (with cell composition adjustment: R = 0.27, p = 6.5e-14), and both were independently associated with PD. Among PD patients, AccelEpiTOC measured at baseline was also associated with longitudinal motor and cognitive symptom decline. CONCLUSIONS The current study presents a first look at epigenetic mitotic aging in PD and our findings suggest accelerated hematopoietic cell mitosis, possibly reflecting immune pathway imbalances, in early PD that may also be related to motor and cognitive progression.
Collapse
Affiliation(s)
- Kimberly C Paul
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| | - Alexandra M Binder
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Cynthia Kusters
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Irish Del Rosario
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Yu Yu
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Jeff Bronstein
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Beate Ritz
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, California, USA
| |
Collapse
|
48
|
Ghazanfari N, van Waarde A, Dierckx RAJO, Doorduin J, de Vries EFJ. Is cyclooxygenase-1 involved in neuroinflammation? J Neurosci Res 2021; 99:2976-2998. [PMID: 34346520 PMCID: PMC9542093 DOI: 10.1002/jnr.24934] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/08/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022]
Abstract
Purpose: Reactive microglia are an important hallmark of neuroinflammation. Reactive microglia release various inflammatory mediators, such as cytokines, chemokines, and prostaglandins, which are produced by enzymes like cyclooxygenases (COX). The inducible COX‐2 subtype has been associated with inflammation, whereas the constitutively expressed COX‐1 subtype is generally considered as a housekeeping enzyme. However, recent evidence suggests that COX‐1 can also be upregulated and may play a prominent role in the brain during neuroinflammation. In this review, we summarize the evidence that supports this involvement of COX‐1. Methods: Five databases were used to retrieve relevant studies that addressed COX‐1 in the context of neuroinflammation. The search resulted in 32 articles, describing in vitro, in vivo, post mortem, and in vivo imaging studies that specifically investigated the COX‐1 isoform under such conditions. Results: Reviewed literature generally indicated that the overexpression of COX‐1 was induced by an inflammatory stimulus, which resulted in an increased production of prostaglandin E2. The pharmacological inhibition of COX‐1 was shown to suppress the induction of inflammatory mediators like prostaglandin E2. Positron emission tomography (PET) imaging studies in animal models confirmed the overexpression of COX‐1 during neuroinflammation. The same imaging method, however, could not detect any upregulation of COX‐1 in patients with Alzheimer's disease. Conclusion: Taken together, studies in cultured cells and living rodents suggest that COX‐1 is involved in neuroinflammation. Most postmortem studies on human brains indicate that the concentration of COX‐1‐expressing microglial cells is increased near sites of inflammation. However, evidence for the involvement of COX‐1 in neuroinflammation in the living human brain is still largely lacking.
Collapse
Affiliation(s)
- Nafiseh Ghazanfari
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
49
|
Cerebral Organoids Derived from a Parkinson's Patient Exhibit Unique Pathogenesis from Chikungunya Virus Infection When Compared to a Non-Parkinson's Patient. Pathogens 2021; 10:pathogens10070913. [PMID: 34358063 PMCID: PMC8308834 DOI: 10.3390/pathogens10070913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/05/2021] [Accepted: 07/15/2021] [Indexed: 12/25/2022] Open
Abstract
(1) Background: Arboviruses of medical and veterinary significance have been identified on all seven continents, with every human and animal population at risk for exposure. Like arboviruses, chronic neurodegenerative diseases, like Alzheimer’s and Parkinson’s disease, are found wherever there are humans. Significant differences in baseline gene and protein expression have been determined between human-induced pluripotent stem cell lines derived from non-Parkinson’s disease individuals and from individuals with Parkinson’s disease. It was hypothesized that these inherent differences could impact cerebral organoid responses to viral infection. (2) Methods: In this study, cerebral organoids from a non-Parkinson’s and Parkinson’s patient were infected with Chikungunya virus and observed for two weeks. (3) Results: Parkinson’s organoids lost mass and exhibited a differential antiviral response different from non-Parkinson’s organoids. Neurotransmission data from both infected non-Parkinson’s and Parkinson’s organoids had dysregulation of IL-1, IL-10, and IL-6. These cytokines are associated with mood and could be contributing to persistent depression seen in patients following CHIKV infection. Both organoid types had increased expression of CXCL10, which is linked to demyelination. (4) Conclusions: The differential antiviral response of Parkinson’s organoids compared with non-Parkinson’s organoids highlights the need for more research in neurotropic infections in a neurologically compromised host.
Collapse
|
50
|
Yao L, Wu J, Koc S, Lu G. Genetic Imaging of Neuroinflammation in Parkinson's Disease: Recent Advancements. Front Cell Dev Biol 2021; 9:655819. [PMID: 34336822 PMCID: PMC8320775 DOI: 10.3389/fcell.2021.655819] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative aging disorders characterized by motor and non-motor symptoms due to the selective loss of midbrain dopaminergic (DA) neurons. The decreased viability of DA neurons slowly results in the appearance of motor symptoms such as rigidity, bradykinesia, resting tremor, and postural instability. These symptoms largely depend on DA nigrostriatal denervation. Pharmacological and surgical interventions are the main treatment for improving clinical symptoms, but it has not been possible to cure PD. Furthermore, the cause of neurodegeneration remains unclear. One of the possible neurodegeneration mechanisms is a chronic inflammation of the central nervous system, which is mediated by microglial cells. Impaired or dead DA neurons can directly lead to microglia activation, producing a large number of reactive oxygen species and pro-inflammatory cytokines. These cytotoxic factors contribute to the apoptosis and death of DA neurons, and the pathological process of neuroinflammation aggravates the primary morbid process and exacerbates ongoing neurodegeneration. Therefore, anti-inflammatory treatment exerts a robust neuroprotective effect in a mouse model of PD. Since discovering the first mutation in the α-synuclein gene (SNCA), which can cause disease-causing, PD has involved many genes and loci such as LRRK2, Parkin, SNCA, and PINK1. In this article, we summarize the critical descriptions of the genetic factors involved in PD's occurrence and development (such as LRRK2, SNCA, Parkin, PINK1, and inflammasome), and these factors play a crucial role in neuroinflammation. Regulation of these signaling pathways and molecular factors related to these genetic factors can vastly improve the neuroinflammation of PD.
Collapse
Affiliation(s)
- Longping Yao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiayu Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sumeyye Koc
- Department of Neuroscience, Institute of Health Sciences, Ondokuz Mayıs University, Samsun, Turkey
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|