1
|
Zhou S, Li T, Zhang W, Wu J, Hong H, Quan W, Qiao X, Cui C, Qiao C, Zhao W, Shen Y. The cGAS-STING-interferon regulatory factor 7 pathway regulates neuroinflammation in Parkinson's disease. Neural Regen Res 2025; 20:2361-2372. [PMID: 39359093 PMCID: PMC11759022 DOI: 10.4103/nrr.nrr-d-23-01684] [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: 10/10/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00026/figure1/v/2024-09-30T120553Z/r/image-tiff Interferon regulatory factor 7 plays a crucial role in the innate immune response. However, whether interferon regulatory factor 7-mediated signaling contributes to Parkinson's disease remains unknown. Here we report that interferon regulatory factor 7 is markedly up-regulated in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of Parkinson's disease and co-localizes with microglial cells. Both the selective cyclic guanosine monophosphate adenosine monophosphate synthase inhibitor RU.521 and the stimulator of interferon genes inhibitor H151 effectively suppressed interferon regulatory factor 7 activation in BV2 microglia exposed to 1-methyl-4-phenylpyridinium and inhibited transformation of mouse BV2 microglia into the neurotoxic M1 phenotype. In addition, siRNA-mediated knockdown of interferon regulatory factor 7 expression in BV2 microglia reduced the expression of inducible nitric oxide synthase, tumor necrosis factor α, CD16, CD32, and CD86 and increased the expression of the anti-inflammatory markers ARG1 and YM1. Taken together, our findings indicate that the cyclic guanosine monophosphate adenosine monophosphate synthase-stimulator of interferon genes-interferon regulatory factor 7 pathway plays a crucial role in the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Shengyang Zhou
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Ting Li
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Wei Zhang
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Jian Wu
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Hui Hong
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Wei Quan
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Xinyu Qiao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Chun Cui
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Chenmeng Qiao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Weijiang Zhao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Yanqin Shen
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| |
Collapse
|
2
|
Gao Y, Liu W, Shi L, Yang P, Yang L, Zhao M, Luo L. Narirutin reduces microglia-mediated neuroinflammation by inhibiting the JAK2/STAT3 pathway in MPP +/MPTP-induced Parkinson's disease models. Exp Neurol 2025; 389:115232. [PMID: 40169108 DOI: 10.1016/j.expneurol.2025.115232] [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: 10/14/2024] [Revised: 03/16/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, characterized by the loss of dopaminergic neurons in the substantia nigra compacta (SNc). Although the detailed molecular mechanisms of PD remain unknown, microglia-mediated neuroinflammation undoubtedly plays a key role in disease progression. Narirutin (Nar), a major flavonoid naturally occurring in citrus fruits, has garnered considerable research attention due to its various therapeutic applications and low toxicity. However, its effects on PD remain unclear. In this study, we explored the protective effects of Nar in 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine (MPTP)-induced PD mouse model as well as in 1-methyl-4-phenyl-pyridinium (MPP+)-induced BV2 cells. Treatment with Nar (2.0, 10.0, and 50.0 mg/kg) reduced dopaminergic neuronal loss in a dose-dependent manner and ameliorated motor impairment in PD mice. Moreover, Nar administration inhibited microglia-mediated inflammation, evidenced by decreased microglial activation in SNc and BV2 cells, and lowered levels of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) in the serum and cells. In addition, we found that Nar exerted anti-inflammatory effects by inhibiting the JAK2/STAT3 pathway. Importantly, using molecular docking and cellular thermal shift assay, we confirmed that JAK2 was a potential binding target of Nar. Overall, Nar attenuated MPTP/MPP+-induced neuroinflammation by inhibiting the JAK2/STAT3 pathway in activated microglia, thereby preventing dopaminergic neuron loss and improving motor disorders in PD mice. Our results provide new evidence supporting that Nar is promising for PD treatment and should be considered for further clinical development.
Collapse
Affiliation(s)
- Ying Gao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Wenna Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Lei Shi
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Peng Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Minggao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
| | - Li Luo
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
3
|
Zhang H, Zhang J, Huang K, Cai C, Jiang J, Su Z, Gu H, Duan Z, Shao S, Zhou M, Du Q, He F. Novel p-terphenyls with anti-neuroinflammatory activity from fruiting bodies of the Chinese edible mushroom Thelephora ganbajun Zang. Bioorg Chem 2025; 159:108414. [PMID: 40174532 DOI: 10.1016/j.bioorg.2025.108414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
The detailed mycochemical exploration of the EtOAc extract of a famous edible mushroom Thelephora ganbajun, resulted in the isolation of six new p-terphenyl derivatives, named theleganbanins A - F (1-6), together with five known ones, namely atromentin (7), fendleryl B (8), 2-O-methylatromentin (9), vialinin B (10), and ganbajunin B (11). Their structures were precisely determined through comprehensive spectroscopic analyses, especially 1D and 2D NMR data and HRMS measurement. Single crystal X-ray diffraction and comparison of calculated and experimental ECD spectra were conducted to further confirm the absolute configurations of compounds 1-6. Theleganbanins A (1) and B (2) featuring a rare α, β-unsaturated-γ-butyrolactone core were proposed to be biosynthesized through aldol condensation for the first time in naturally occurring p-terphenyl derivatives. Theleganbanin C (3) was identified as a pair of p-terphenyl enantiomers with a novel 1', 6'-dyhydro-2', 5'-pyridinedione ring. Theleganbanin D (4) was the first example of p-terphenyl derivatives with a hemiacetal furanone moiety. The anti-neuroinflammatory activities of compounds 1-2 and 4-10 were screened. As a result, these compounds showed inhibitory activity on the production of pro-inflammatory cytokines TNF-α, IL-6 and IL-1β in lipopolysaccharide (LPS)-induced BV-2 microglial cells. Further investigation showed that compound 2 could inhibit the phosphorylation of JAK2/STAT3 signaling pathway. These finding indicated that p-terphenyl derivatives from edible mushroom Thelephora ganbajun Zang would be promising drug candidates in treatment of neuroinflammatory related diseases.
Collapse
Affiliation(s)
- Hang Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jingyi Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Keyin Huang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Cheng Cai
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jinyan Jiang
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo 113-8657, Japan
| | - Zijie Su
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Haixin Gu
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zidan Duan
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shijie Shao
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Min Zhou
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qingfeng Du
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China
| | - Fei He
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China.
| |
Collapse
|
4
|
Gupta S, Jash M, Khan J, Garg S, Roy R, Arshi MU, Nayak P, Ghosh S. Discovery of potential Leonurine-based therapeutic lead MJ210 attenuates Parkinson's disease pathogenesis via NF-κB and MAPK pathways: Mechanistic insights from in vitro and in vivo rotenone models. Eur J Med Chem 2025; 289:117471. [PMID: 40090295 DOI: 10.1016/j.ejmech.2025.117471] [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: 09/27/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/18/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease affecting motor and non-motor functions, with no effective treatment yet discovered. Neuroprotective compounds, both natural and synthetic, show promise but face challenges such as crossing the blood-brain barrier, limited serum stability, and higher toxicity. To tackle these obstacles, we have devised an innovative design strategy inspired by the neuroprotective properties of Leonurine, widely utilized in managing neurological disorders. Through rigorous screening of our compound library, we have identified a potent therapeutic molecule (MJ210) that exhibited remarkable efficacy in bolstering neuroprotection against rotenone-induced PD models, both in vitro and in vivo. Our findings revealed that administering MJ210 significantly increased neuronal survival in the SH-SY5Y model of PD. This was achieved by preventing apoptosis, reducing reactive oxygen species, mitigating mitochondrial dysfunction, and dampening neuroinflammation via ERK1/2-P38-JNK and P65-NFκB signaling pathways. In addition, MJ210 demonstrated remarkable neuroprotective abilities in vivo by significantly enhancing dopamine biosynthesis, alleviating motor dysfunction, improving balance and coordination, and reversing depression in rotenone-induced PD rats, even outperforming L-DOPA, the current gold standard treatment for PD. Therefore, MJ210 emerges as a significantly promising therapeutic candidate for PD, offering the potential for managing both the severity and progression of this disease.
Collapse
Affiliation(s)
- Sanju Gupta
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Moumita Jash
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Shubham Garg
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Rajsekhar Roy
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Mohammad Umar Arshi
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Prasunpriya Nayak
- Department of Physiology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, 342005, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India.
| |
Collapse
|
5
|
Shi J, Xie J, Li Z, He X, Wei P, Sander JW, Zhao G. The Role of Neuroinflammation and Network Anomalies in Drug-Resistant Epilepsy. Neurosci Bull 2025; 41:881-905. [PMID: 39992353 PMCID: PMC12014895 DOI: 10.1007/s12264-025-01348-w] [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: 10/18/2024] [Accepted: 11/30/2024] [Indexed: 02/25/2025] Open
Abstract
Epilepsy affects over 50 million people worldwide. Drug-resistant epilepsy (DRE) accounts for up to a third of these cases, and neuro-inflammation is thought to play a role in such cases. Despite being a long-debated issue in the field of DRE, the mechanisms underlying neuroinflammation have yet to be fully elucidated. The pro-inflammatory microenvironment within the brain tissue of people with DRE has been probed using single-cell multimodal transcriptomics. Evidence suggests that inflammatory cells and pro-inflammatory cytokines in the nervous system can lead to extensive biochemical changes, such as connexin hemichannel excitability and disruption of neurotransmitter homeostasis. The presence of inflammation may give rise to neuronal network abnormalities that suppress endogenous antiepileptic systems. We focus on the role of neuroinflammation and brain network anomalies in DRE from multiple perspectives to identify critical points for clinical application. We hope to provide an insightful overview to advance the quest for better DRE treatments.
Collapse
Affiliation(s)
- Jianwei Shi
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- China International Neuroscience Institute, Beijing, 100053, China
| | - Jing Xie
- Deanery of Biomedical Sciences, Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Zesheng Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- China International Neuroscience Institute, Beijing, 100053, China
| | - Xiaosong He
- Department of Psychology, University of Science and Technology of China, Hefei, 230022, China
| | - Penghu Wei
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- China International Neuroscience Institute, Beijing, 100053, China.
| | - Josemir W Sander
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
- Chalfont Centre for Epilepsy, Chalfont St Peter, Buckinghamshire, SL9 0RJ, UK.
- Neurology Department, West China Hospital of Sichuan University, Chengdu, 61004, China.
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- China International Neuroscience Institute, Beijing, 100053, China.
| |
Collapse
|
6
|
Espejo EF, Guerra MDM, Castellano S. Association between serum myeloperoxidase enzyme activity and Parkinson's disease status. NPJ Parkinsons Dis 2025; 11:94. [PMID: 40287421 PMCID: PMC12033337 DOI: 10.1038/s41531-025-00941-0] [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: 10/25/2024] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Elevated levels of the inflammatory enzyme myeloperoxidase (MPO) in the blood are associated with the development of age-related inflammatory diseases. Given that age, inflammation, and blood MPO play a role in the pathogenesis of Parkinson´s disease (PD), we hypothesized that serum MPO could be associated with PD status. This case-control study (199 participants) was conducted using an extensive protocol, and the concentration and activity of MPO in blood serum were measured. The findings reveal that serum MPO concentration and activity are significantly increased in the patients, and that rates of PD in all individuals are associated with increasing tertiles of MPO concentration and activity. In multivariate logistic regression model adjusting for confounding factors, MPO activity (not concentration) is the factor that is most associated with PD status (OR, 6.921, P = 0.001). Mental depression is directly associated with MPO activity and with PD status (OR, 0.121, B = -2.108, P = 0.002). The use of statins or nonsteroidal anti-inflammatory drugs significantly reduces serum MPO activity, but the possible association with the odds of having PD does not survive correction for multiple testing. In summary, both serum MPO concentration and activity are increased in patients with PD, but only MPO enzyme activity is associated with PD status. These findings may have implications for the evaluation of PD.
Collapse
Affiliation(s)
| | - María-Del-Mar Guerra
- Department of Biochemistry, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Silvia Castellano
- Department of Metabolopathy, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
7
|
Lee C, Kim SS, Bae MA, Kim SH. Neuroprotective Activities of Sertraline, Tiagabine, and Bicifadine with Autophagy-Inducing Potentials in a 6-Hydroxidopamine-Treated Parkinson's Disease Cell Model. Neurochem Res 2025; 50:154. [PMID: 40278973 DOI: 10.1007/s11064-025-04404-z] [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: 10/30/2024] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
Parkinson's disease (PD) is one of neurodegenerative diseases characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The development of a neuroprotective therapy is crucial for mitigating features and progression of PD. Since autophagy induction has recently emerged as a promising neuroprotective strategy, this study aimed to identify autophagy-inducing compounds and evaluate their neuroprotective activity. Among 3,200 compounds consisting of FDA-approved drugs or are under active development, 547 compounds targeting neurological diseases were filtered in, and three compounds (sertraline, tiagabine and bicifadine) were finally identified to exhibit the autophagy-inducing activity and also demonstrated the autophagy-dependent neuroprotective action by inhibiting the mammalian target of rapamycin (mTOR) in 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in PC12 cells. Furthermore, the analysis of neurochemical changes suggested that the ability of those compounds to restore the quantity of cellular neurotransmitters such as betaine, 5-hydroxyindoleacetic acid and kynurenine might be linked to their neuroprotective function. In conclusion, compounds like sertraline, tiagabine, and bicifadine that have the ability to induce autophagy and inhibit mTOR might be repurposed as PD treatment to protect the neuronal cells.
Collapse
Affiliation(s)
- Chaemi Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, Korea
| | - Seong Soon Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Myung Ae Bae
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Seong Hwan Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea.
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, Korea.
| |
Collapse
|
8
|
Belviranlı M, Okudan N, Sezer T. Potential therapeutic effects of curcumin, with or without L-DOPA, on motor and cognitive functions and hippocampal changes in rotenone-treated rats. Metab Brain Dis 2025; 40:174. [PMID: 40208367 PMCID: PMC11985604 DOI: 10.1007/s11011-025-01602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
The neurodegenerative condition known as Parkinson's disease (PD) is a long-term condition that causes both motor and non-motor symptoms. It is known that curcumin has a strong neuroprotective potential. This experimental study was designed to examine the anti-inflammatory, anti-apoptotic and neuroprotective effects of curcumin administered alone and in combination with L-DOPA in the hippocampus as well as behavioral symptoms in rotenone-induced PD model. Forty-two 4-month-old adult male Wistar rats were randomly divided into six groups as follows: Control, Curcumin, Rotenone, Rotenone plus curcumin, Rotenone plus L-DOPA and Rotenone plus curcumin plus L-DOPA. Control group received vehicles, curcumin group received curcumin (200 mg kg-1, daily for 35 days), rotenone group received rotenone (2 mg kg-1, daily for 35 days), and test groups received curcumin or L-DOPA (10 mg kg-1, daily for the last 15 days) or their combination in addition the rotenone. Pole, sucrose preference, open field, elevated plus maze, and Morris water maze tests were performed after treatment. Molecular and biochemical analyses were performed in the hippocampus tissue and serum samples. Rotenone injection caused impairments in motor activity, depressive-like behavior, and learning and memory functions. Rotenone also increased the expressions of α-synuclein, caspase 3, NF-κB, and decreased the expressions of parkin and BDNF in the hippocampus. However, especially curcumin and L-DOPA combined treatment normalized all these impaired molecular and behavioral variables. In conclusion, curcumin may exert beneficial effects in treatment strategies for PD-related hippocampal effects, especially when added to L-DOPA therapy.
Collapse
Affiliation(s)
- Muaz Belviranlı
- School of Medicine, Department of Physiology, Selçuk University, Konya, 42131, Turkey.
| | - Nilsel Okudan
- School of Medicine, Department of Physiology, Selçuk University, Konya, 42131, Turkey
| | - Tuğba Sezer
- School of Medicine, Department of Physiology, Selçuk University, Konya, 42131, Turkey
| |
Collapse
|
9
|
Zhao FL, Zhang JR, Liu MH, Liu HY, Mao CJ, Wang F, Chen JP, Liu CF. Tan I modulates astrocyte inflammatory responses through enhanced NAD +-Sirt1 pathway: Insights from metabolomics studies. Int Immunopharmacol 2025; 151:114364. [PMID: 40024217 DOI: 10.1016/j.intimp.2025.114364] [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: 01/09/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Over the past decade, research has increasingly demonstrated that oligomeric α-synuclein (O-αS) plays a pivotal role in the pathogenesis of Parkinson's disease (PD), particularly in mediating dopaminergic neuron injury and neuroinflammation. In this study, we investigated the anti-inflammatory effects of tanshinone I (Tan I), an active component of the traditional Chinese medicine Danshen, on O-αS-induced inflammation in primary mouse astrocytes. Using metabolomics analysis, we identified key pathways regulated by Tan I. Our results showed that Tan I significantly suppressed O-αS-induced mRNA expression of pro-inflammatory cytokines, including interleukin-1β, IL-6, tumor necrosis factor-α and cyclooxygenase-2. Metabolomic profiling revealed that Tan I enhanced NAD+ metabolism, leading to activation of the NAD+-Sirt1 pathway and subsequent inhibition of nuclear factor-κB activity. Together, these findings suggest that Tan I attenuates neuroinflammatory response in astrocytes by modulating NAD+-dependent signaling mechanisms.
Collapse
Affiliation(s)
- Feng-Lun Zhao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Jia-Rui Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Man-Hua Liu
- Department of Neurology, Changshu Hospital affiliated to Nanjing University of Chinese Medicine, Changshu 215500, China
| | - Hui-Yi Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Ju-Ping Chen
- Department of Neurology, Changshu Hospital affiliated to Nanjing University of Chinese Medicine, Changshu 215500, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| |
Collapse
|
10
|
Fredlund F, Fryklund C, Trujeque-Ramos O, Staley HA, Pardo J, Luk KC, Tansey MG, Swanberg M. Lack of neuroprotection after systemic administration of the soluble TNF inhibitor XPro1595 in an rAAV6-α-Syn + PFFs-induced rat model for Parkinson's disease. Neurobiol Dis 2025; 207:106841. [PMID: 39954745 DOI: 10.1016/j.nbd.2025.106841] [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: 10/07/2024] [Revised: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
Parkinson's disease (PD) is characterized by dopaminergic neurodegeneration, α-Synuclein (α-Syn) pathology, and inflammation. Microglia in the substantia nigra pars compacta (SNpc) upregulate major histocompatibility complex class II (MHCII), and variants in genes encoding MHCII affect PD risk. Additionally, elevated TNF levels and α-Syn-reactive T cells in circulation suggest a strong link between innate and adaptive immune responses in PD. We have previously reported that reduced levels of the class II transactivator, the master regulator of MHCII expression, increases susceptibility to α-Syn-induced PD-like pathology in rats and are associated with higher serum levels of soluble TNF (sTNF). Here, we demonstrate that inhibiting sTNF with a dominant-negative TNF variant, XPro1595, known to be neuroprotective in endotoxin- and toxin-induced neurodegeneration models, fails to protect against robust α-Syn-induced PD-like pathology in rats. We used a model combining rAAV-mediated α-Syn overexpression in SNpc with striatal injection of α-Syn preformed fibrils two weeks later. Systemic XPro1595 treatment was initiated one-week post-rAAV-α-Syn. We observed up to 70 % loss of striatal dopaminergic fibers without treatment, and no protective effects on dopaminergic neurodegeneration after XPro1595 administration. Pathological α-Syn levels as well as microglial and astrocytic activation were not reduced in SNpc or striatum following XPro1595 treatment. An increase in IL-6 and IL-1β levels in CSF was observed in rats treated with XPro1595, possibly explaining a lack of protective effects following treatment. Our results highlight the need to determine the importance of timing of treatment initiation, which is crucial for future applications of sTNF therapies in PD patients.
Collapse
Affiliation(s)
- Filip Fredlund
- Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Claes Fryklund
- Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Olivia Trujeque-Ramos
- Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hannah A Staley
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Joaquin Pardo
- Instituto de Investigaciones Bioquímicas de La Plata "Profesor Doctor Rodolfo R. Brenner", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Buenos Aires, Argentina; Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Malú G Tansey
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Maria Swanberg
- Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
11
|
Wang H, Cheng Z, Xu Z, Wang M, Sun X, Liu W, Wang J, Yang Q, Zhang T, Song J, Du Y, Zhang X. Systemic Inflammatory Factors and Neuropsychiatric Disorders: A Bidirectional Mendelian Randomization Study. Brain Behav 2025; 15:e70478. [PMID: 40200869 PMCID: PMC11979492 DOI: 10.1002/brb3.70478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND The present study employed Mendelian randomization to scrutinize the causal connections that may exist between 91 distinct inflammatory markers and six neuropsychiatric disorders, namely Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), anxiety disorders (ANX), depressive disorders (DEP), and unexplained encephalopathy (UE). DISCUSSION The methodology utilized involved the standard inverse variance weighting method within a two-sample, two-way Mendelian randomization framework and integrated statistics from genome-wide association studies. To ascertain the robustness of the identified causal associations, sensitivity analyses were performed with the aid of the MR-Egger method and the weighted median test. CONCLUSION The results revealed that 14 distinct systemic inflammatory modulators are potentially causally linked to the risk of developing various neuropsychiatric disorders. Specifically, five were associated with AD, eight with ANX, six with DEP, and one with UE. However, the causal associations involving systemic inflammatory markers with PD and MS require further investigation, particularly with the identification of additional significant genetic variants. Furthermore, the concentration levels of 33 systemic inflammatory factors could be modulated by the occurrence of neuropsychiatric conditions, indicated by this study. These include five affected by AD, eight by PD, six by MS, 12 by ANX, five by DEP, and five by UE.
Collapse
Affiliation(s)
- Hao Wang
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Ziji Cheng
- Department of Anatomy, College of Chinese Integrative MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zixuan Xu
- School of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Min Wang
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Xingyang Sun
- School of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Wen Liu
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Jingtian Wang
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Qian Yang
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Tuo Zhang
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Jie Song
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Yanjun Du
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| | - Xiaoming Zhang
- College of Acupuncture‐Moxibustion and OrthopedicsHubei University of Chinese MedicineWuhanChina
| |
Collapse
|
12
|
Vujosevic S, Limoli C, Kozak I. Hallmarks of aging in age-related macular degeneration and age-related neurological disorders: novel insights into common mechanisms and clinical relevance. Eye (Lond) 2025; 39:845-859. [PMID: 39289517 PMCID: PMC11933422 DOI: 10.1038/s41433-024-03341-5] [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: 04/14/2024] [Revised: 08/13/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Age-related macular degeneration (AMD) and age-related neurological diseases (ANDs), such as Alzheimer's and Parkinson's Diseases, are increasingly prevalent conditions that significantly contribute to global morbidity, disability, and mortality. The retina, as an accessible part of the central nervous system (CNS), provides a unique window to study brain aging and neurodegeneration. By examining the associations between AMD and ANDs, this review aims to highlight novel insights into fundamental mechanisms of aging and their role in neurodegenerative disease progression. This review integrates knowledge from the emerging field of aging research, which identifies common denominators of biological aging, specifically loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, and inflammation. Finally, we emphasize the clinical relevance of these pathways and the potential for cross-disease therapies that target common aging hallmarks. Identifying these shared pathways could open avenues to develop therapeutic strategies targeting mechanisms common to multiple degenerative diseases, potentially attenuating disease progression and promoting the healthspan.
Collapse
Affiliation(s)
- Stela Vujosevic
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
- Eye Clinic, IRCCS MultiMedica, Milan, Italy.
| | - Celeste Limoli
- Eye Clinic, IRCCS MultiMedica, Milan, Italy
- University of Milan, Milan, Italy
| | - Igor Kozak
- Moorfields Eye Hospital Centre, Abu Dhabi, UAE
- Ophthalmology and Vision Science, University of Arizona, Tucson, USA
| |
Collapse
|
13
|
Seo MH, Yeo S. Acupuncture regulates α-synuclein expression via serping1 in an MPTP-induced mouse model of Parkinsonism. Acupunct Med 2025; 43:85-94. [PMID: 40130647 DOI: 10.1177/09645284251327195] [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: 03/26/2025]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by a substantial loss of dopaminergic cells in the substantia nigra (SN) and the formation of intracellular Lewy bodies, which are mainly composed of α-synuclein (α-syn). Acupuncture has been used to improve the symptoms of PD in humans and exhibits a neuroprotective effect against Parkinsonism in animal models. We further investigated the neuroprotective effect of acupuncture via its effect on α-syn levels, dopaminergic cell death and serping1 expression in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of chronic PD. METHODS Mice were divided into a control group receiving phosphate-buffered saline (CTL) and three model groups receiving MPTP that either remained untreated (MPTP) received verum acupuncture at GB34 and LR3 (MPTP_A) or received control acupuncture at sites not corresponding to any traditional acupuncture point location (MPTP_NA). The signal intensity of serping1 gene expression was assessed using microarray, and α-syn level and dopaminergic cell death were measured by Western blotting, immunohistochemistry and immunofluorescence. To further investigate the relationship between expression of serping1 and α-syn, a cell culture experiment was carried out in a 1-methyl-4-phenylpyridinium (MPP+) treated neuroblastoma cell line (SH-SY5Y) with and without serping1 knockdown using short interfering (si)RNA. RESULTS Acupuncture at GB34 and LR3 attenuated the MPTP-induced decrease in tyrosine hydroxylase (TH) levels and increase in α-syn levels in the SN. Furthermore, verum acupuncture prevented the increase in serping1 level induced by MPTP. In SH-SY5Y cells, MPP+ treatment increased α-syn and decreased both TH expression and cell viability; however, these effects were mitigated by serping1 knockdown. CONCLUSIONS Our results suggest that an MPTP-induced reduction in serping1 level leads to decreased TH and increased α-syn expression, and that these effects can be attenuated/blocked by acupuncture at GB34 and LR3. Our findings provide new evidence for the neuroprotective effects of acupuncture on dopaminergic cells, which may be mediated by control of serping1 expression.
Collapse
Affiliation(s)
- Min Hyung Seo
- College of Korean Medicine, Sangji University, Wonju, Republic of Korea
| | - Sujung Yeo
- College of Korean Medicine, Sangji University, Wonju, Republic of Korea
- Research Institute of Korean Medicine, Sangji University, Wonju, Republic of Korea
| |
Collapse
|
14
|
Lewis SJG, Sue CM, Cooper A, Halliday GM. Future is now: an Australasian perspective on disease-modifying trials in Parkinson's and prodromal disease. BMJ Neurol Open 2025; 7:e001070. [PMID: 40129903 PMCID: PMC11931937 DOI: 10.1136/bmjno-2025-001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
There is increasing interest in the role of platform trials, where several investigational products targeting disease modification in Parkinson's Disease can be assessed in parallel. Indeed, several initiatives are currently gearing up across North America and Europe to conduct such studies. However, to date, little attention has been paid to ongoing efforts that already exist in Australia and look soon to expand across to New Zealand as part of greater collaboration. This viewpoint will highlight some of these ongoing efforts addressing the challenges and potential solutions for delivering successful studies.
Collapse
Affiliation(s)
- Simon JG Lewis
- Macquarie Medical School, Macquarie University, Sydney, New South Wales, Australia
| | - Carolyn M Sue
- Kinghorn Chair, Neurodegeneration, Neuroscience Research Australia, Randwick, New South Wales, Australia
- Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Antony Cooper
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, Faculty of Medicine and Health, Central Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Sefah B, Ashie Y, Osafo N, Mante PK. Hydroethanolic Extract of Salvia officinalis L. Leaves Improves Memory and Alleviates Neuroinflammation in ICR Mice. ScientificWorldJournal 2025; 2025:2198542. [PMID: 40151497 PMCID: PMC11949616 DOI: 10.1155/tswj/2198542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Neurodegenerative disorders are known to be commonly associated with neuroinflammation. Plants with antioxidant and anti-inflammatory properties hold prospect in alleviating neuroinflammation. One such plant with documented anti-inflammatory and antioxidant potential is Salvia officinalis L. This study looked at effects of the hydroethanolic leaf extract of S. officinalis L. on lipopolysaccharide (LPS)-induced neuroinflammation and associated memory impairment using an ICR mouse model. Assessment of the phytochemical constituents in S. officinalis L. and its acute toxicity was conducted. Mice were treated with S. officinalis L. extract (30, 100, and 300 mg/kg) after LPS administration. Object recognition and elevated plus maze tests were employed to assess neuroinflammation-induced behavioral changes. Brain samples were taken to determine the levels of TNF-α and conduct histopathological analysis. The hydroethanolic extract of S. officinalis L. was found to contain alkaloids, glycoside, tannins, flavonoids, and coumarins and exhibited no observable acute toxicity. The extract showed the presence of eicosatrienoic acid, methyl ester, and phenanthrene derivatives. The extract improved memory and cognitive performance but had no significant effect on brain tissue TNF-α expression. S. officinalis L. treatment in mice with neuroinflammation also resulted in reduced mononuclear infiltration and gliosis and reduced apoptotic and necrotic neurons as well as no observable brain lesions. S. officinalis L. holds promising pharmacological activity at reducing neuroinflammation and its associated cognitive impairment.
Collapse
Affiliation(s)
- Bernard Sefah
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Yolanda Ashie
- Department of Pathology, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Newman Osafo
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Priscilla Kolibea Mante
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
16
|
d'Errico P, Früholz I, Meyer-Luehmann M, Vlachos A. Neuroprotective and plasticity promoting effects of repetitive transcranial magnetic stimulation (rTMS): A role for microglia. Brain Stimul 2025; 18:810-821. [PMID: 40118248 DOI: 10.1016/j.brs.2025.03.012] [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: 01/06/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique used to modulate neocortical excitability, with expanding applications in neurological and psychiatric disorders. However, the cellular and molecular mechanisms underlying its effects, particularly the role of microglia -the resident immune cells of the central nervous system- remain poorly understood. This review synthesizes recent findings on how different rTMS protocols influence microglial function under physiological conditions and in disease models. Emerging evidence indicates that rTMS modulates microglial activation, promoting neuroprotective and plasticity-enhancing processes not only in models of brain disorders, such as Alzheimer's and Parkinson's disease, but also in healthy neural circuits. While much of the current research has focused on the inflammatory profile of microglia, critical aspects such as activity-dependent synaptic remodeling, phagocytic activity, and process motility remain underexplored. Given the substantial heterogeneity of microglial responses across brain regions, age, and sex, as well as their differential roles in health and disease, a deeper understanding of their involvement in rTMS-induced plasticity is essential. Future studies should integrate selective microglial manipulation and advanced structural, functional, and molecular profiling techniques to clarify their causal involvement. Addressing these gaps will be pivotal in optimizing rTMS protocols and maximizing its therapeutic potential across a spectrum of neurological and neuropsychiatric conditions.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Iris Früholz
- Department of Neurology, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
Salarvandian S, Digaleh H, Khodagholi F, Javadpour P, Asadi S, Zaman AAO, Dargahi L. Harmonic activity of glutamate dehydrogenase and neuroplasticity: The impact on aging, cognitive dysfunction, and neurodegeneration. Behav Brain Res 2025; 480:115399. [PMID: 39675635 DOI: 10.1016/j.bbr.2024.115399] [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: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
In recent years, glutamate has attracted significant attention for its roles in various brain processes. However, one of its key regulators, glutamate dehydrogenase (GDH), remains understudied despite its pivotal role in several biochemical pathways. Dysfunction or dysregulation of GDH has been implicated in aging and various neurological disorders, such as Alzheimer's disease and Parkinson's disease. In this review, the impact of GDH on aging, cognitive impairment, and neurodegenerative conditions, as exemplars of the phenomena that may affected by neuroplasticity, has been reviewed. Despite extensive research on synaptic plasticity, the precise influence of GDH on brain structure and function remains undiscovered. This review of existing literature on GDH and neuroplasticity reveals diverse and occasionally conflicting effects. Future research endeavors should aim to describe the precise mechanisms by which GDH influences neuroplasticity (eg. synaptic plasticity and neurogenesis), particularly in the context of human aging and disease progression. Studies on GDH activity have been limited by factors such as insufficient sample sizes and varying experimental conditions. Researchers should focus on investigating the molecular mechanisms by which GDH modulates neuroplasticity, utilizing various animal strains and species, ages, sexes, GDH isoforms, brain regions, and cell types. Understanding GDH's role in neuroplasticity may offer innovative therapeutic strategies for neurodegenerative and psychiatric diseases, potentially slowing the aging process and promoting brain regeneration.
Collapse
Affiliation(s)
- Shakiba Salarvandian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Digaleh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sareh Asadi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Orang Zaman
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Kim BR, Lee KH, Paik K, Kim M, Bae JM, Choi CW, Youn SW. Comorbid diseases in bullous pemphigoid: A population-based case-control study. J Dermatol 2025; 52:460-471. [PMID: 39670559 DOI: 10.1111/1346-8138.17577] [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: 08/22/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Bullous pemphigoid (BP) is the most prevalent autoimmune blistering disorder, triggered by autoantibodies targeting hemidesmosome components. It is associated with substantial morbidity and increased mortality. No studies comprehensively evaluate all comorbidities before and after diagnosing patients with BP. We aimed to investigate all BP-associated comorbid diseases and their patterns of associations. This nationwide population-based study included 5066 patients with BP and 10 132 controls between 2011 and 2021. We performed an automated mass screening of 546 diagnostic codes to identify BP-associated comorbidities 5 years before and after BP diagnosis, and analyzed associations patterns of comorbidities. Patients with BP had increased odds of having pressure ulcers, intracerebral hemorrhage, scabies, neuropsychiatric disorders, psoriasis, drug eruption, and acute renal failure before BP diagnosis. After BP diagnosis, they had increased odds pneumonia, sepsis, chronic renal disease, and cardiac arrest. Strong interrelationships were observed between five neuropsychiatric conditions before BP diagnosis and a strong bidirectional association between Alzheimer's dementia and pneumonia after BP diagnosis. This large case-control study of patients with BP thoroughly identified all relevant comorbidities before and after BP diagnosis, highlighting their clinical significance as predisposing and prognostic factors in patients with BP.
Collapse
Affiliation(s)
- Bo Ri Kim
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Kun Hee Lee
- Department of Applied Statistics, Yonsei University, Seoul, Republic of Korea
| | - Kyungho Paik
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Minjae Kim
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Jung Min Bae
- Department of Dermatology, St Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chong Won Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Sang Woong Youn
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| |
Collapse
|
19
|
Ma Q, Tian JL, Lou Y, Guo R, Ma XR, Wu JB, Yang J, Tang BJ, Li S, Qiu M, Duan S, Zhao JW, Zhang J, Xu ZZ. Oligodendrocytes drive neuroinflammation and neurodegeneration in Parkinson's disease via the prosaposin-GPR37-IL-6 axis. Cell Rep 2025; 44:115266. [PMID: 39913287 DOI: 10.1016/j.celrep.2025.115266] [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: 11/15/2023] [Revised: 10/28/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease and is difficult to treat due to its elusive mechanisms. Recent studies have identified a striking association between oligodendrocytes and PD progression, yet how oligodendrocytes regulate the pathogenesis of PD is still unknown. Here, we show that G-protein-coupled receptor 37 (GPR37) is upregulated in oligodendrocytes of the substantia nigra and that prosaposin (PSAP) secretion is increased in parkinsonian mice. The released PSAP can induce interleukin (IL)-6 upregulation and secretion from oligodendrocytes via a GPR37-dependent pathway, resulting in enhanced neuroinflammation, dopamine neuron degeneration, and behavioral deficits. GPR37 deficiency in oligodendrocytes prevents neurodegeneration in multiple PD models. Finally, the hallmarks of the PSAP-GPR37-IL-6 axis are observed in patients with PD. Thus, our results reveal that dopaminergic neurons interact with oligodendrocytes via secreted PSAP, and our findings identify the PSAP-GPR37-IL-6 axis as a driver of PD pathogenesis and a potential therapeutic target that might alleviate PD progression in patients.
Collapse
Affiliation(s)
- Qiang Ma
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China.
| | - Jin-Lan Tian
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Yao Lou
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Ran Guo
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiao-Ru Ma
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jian-Bin Wu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Yang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Bing-Jie Tang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Shumin Duan
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Jing-Wei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Zhang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Department of Pathology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China; National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou 310002, China
| | - Zhen-Zhong Xu
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China.
| |
Collapse
|
20
|
Cheng WY, Lee XZ, Lai MSL, Ho YS, Chang RCC. PKR modulates sterile systemic inflammation-triggered neuroinflammation and brain glucose metabolism disturbances. Front Immunol 2025; 16:1469737. [PMID: 40070845 PMCID: PMC11893411 DOI: 10.3389/fimmu.2025.1469737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Sterile systemic inflammation may contribute to neuroinflammation and accelerate the progression of neurodegenerative diseases. The double-stranded RNA-dependent protein kinase (PKR) is a key signaling molecule that regulates immune responses by regulating macrophage activation, various inflammatory pathways, and inflammasome formation. This study aims to study the role of PKR in regulating sterile systemic inflammation-triggered neuroinflammation and cognitive dysfunctions. Here, the laparotomy mouse model was used to study neuroimmune responses triggered by sterile systemic inflammation. Our study revealed that genetic deletion of PKR in mice potently attenuated the laparotomy-induced peripheral and neural inflammation and cognitive deficits. Furthermore, intracerebroventricular injection of rAAV-DIO-PKR-K296R to inhibit PKR in cholinergic neurons of ChAT-IRES-Cre-eGFP mice rescued the laparotomy-induced changes in key metabolites of brain glucose metabolism, particularly the changes in phosphoenolpyruvate and succinate levels, and cognitive impairment in short-term and spatial working memory. Our results demonstrated the critical role of PKR in regulating neuroinflammation, brain glucose metabolism and cognitive dysfunctions in a peripheral inflammation model. PKR could be a novel pharmacological target for treating systemic inflammation-induced neuroinflammation and cognitive dysfunctions.
Collapse
Affiliation(s)
- Wai-Yin Cheng
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Xin-Zin Lee
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Michael Siu-Lun Lai
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yuen-Shan Ho
- School of Nursing, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Lim JS, Li X, Lee DY, Yao L, Yoo G, Kim Y, Eum SM, Cho YC, Yoon S, Park SJ. Antioxidant and Anti-Inflammatory Activities of Methanol Extract of Senna septemtrionalis (Viv.) H.S. Irwin & Barneby Through Nrf2/HO-1-Mediated Inhibition of NF-κB Signaling in LPS-Stimulated Mouse Microglial Cells. Int J Mol Sci 2025; 26:1932. [PMID: 40076558 PMCID: PMC11900505 DOI: 10.3390/ijms26051932] [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: 12/24/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Botanical extracts are recognized in traditional medicine for their therapeutic potential and safety standards. Botanical extracts are viable and sustainable alternatives to synthetic drugs, being essential in drug discovery for various diseases. Senna septemtrionalis (Viv.) H.S. Irwin & Barneby is a medical plant traditionally used to treat inflammation. However, its antioxidant and anti-inflammatory properties and the molecular pathways activated in microglial cells require further investigation. Therefore, this study examines the antioxidant and anti-inflammatory properties of Senna septemtrionalis (Viv.) H.S. Irwin & Barneby methanol extracts (SMEs) in lipopolysaccharide (LPS)-stimulated mouse microglial cells. SMEs significantly inhibit LPS-induced nitric oxide (NO) and proinflammatory cytokine production, which are mediated through the dephosphorylation of mitogen-activated protein kinases and inhibition of nuclear factor kappa B (NF-κB) translocation into the nucleus. Additionally, SME treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase (HO)-1, reducing oxidative stress, indicated by a decrease in reactive oxygen species and restoration of the total glutathione content in LPS-stimulated BV2 cells. The inhibitory effects of SMEs on inflammatory mediator production and NF-κB nuclear translocation were significantly reversed by Sn-protoporphyrin, a specific HO-1 inhibitor. These findings demonstrate that SME protects microglial cells by activating the Nrf2/HO-1 pathway and inhibiting NF-κB translocation.
Collapse
Affiliation(s)
- Jae Sung Lim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Xiangying Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Da Young Lee
- R&D Center, CUOME BIO Co., Ltd., Sandan-gil, Hwasun-eup, Hwasun-gun 58141, Jeollanam-do, Republic of Korea;
| | - Lulu Yao
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Guijae Yoo
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Jeollabuk-do, Republic of Korea;
| | - Yunyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Sang Mi Eum
- International Biological Material Research Center, Korea Research Institute of Bioscience & Biotechnology, 125 Gwahak-ro, Daejeon 34141, Republic of Korea;
| | - Young-Chang Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Somy Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea
| |
Collapse
|
22
|
Liu Y, Qin K, Jiang C, Gao J, Hou B, Xie A. TMEM106B Knockdown Exhibits a Neuroprotective Effect in Parkinson's Disease via Decreasing Inflammation and Iron Deposition. Mol Neurobiol 2025; 62:1813-1825. [PMID: 39044012 PMCID: PMC11772555 DOI: 10.1007/s12035-024-04373-4] [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: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
Parkinson's disease (PD) is closely related to iron accumulation and inflammation. Emerging evidence indicates that TMEM106B plays an essential role in PD. But whether TMEM106B could act on neuroinflammation and iron metabolism in PD has not yet been investigated. The aim of this study was to investigate the pathological mechanisms of inflammation and iron metabolism of TMEM106B in PD. 1-methyl-4-phenylpyridinium (MPP+)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced SH-SY5Y cells and mice were treated with LV-shTMEM106B and AAV-shTMEM106B to construct PD cellular and mouse models. Pole tests and open-field test (OFT) were performed to evaluate the locomotion of the mice. Immunohistochemistry and iron staining were used to detect TH expression and iron deposition in the SN. Iron staining was used to measure the levels of iron. Western blotting was used to detect the expression of inflammatory factors (tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6)), NOD-like receptor protein 3 (NLRP3) inflammasome, divalent metal transporter 1 (DMT1), and Ferroportin1 (FPN1)). Knockdown of TMEM106B improved motor ability and rescued dopaminergic (DA) neuron loss. TMEM106B knockdown attenuated the increases of TNF-α, IL-6, NLRP3 inflammasome, and DMT1 expression in the MPP+ and MPTP-induced PD models. Furthermore, TMEM106B knockdown also increases the expression of FPN1. This study provides the first evidence that knockdown of TMEM106B prevents dopaminergic neurodegeneration by modulating neuroinflammation and iron metabolism.
Collapse
Affiliation(s)
- Yumei Liu
- Department of Neurology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Kunpeng Qin
- Department of Neurology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Chunyan Jiang
- Department of Neurology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Jinzhao Gao
- Department of Neurology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Binghui Hou
- Department of Neurology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, Shandong, China.
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, Shandong, China.
| |
Collapse
|
23
|
Nassar M, Nassar O, Abosheaishaa H, Misra A. Comparative outcomes of systemic diseases in people with type 2 diabetes, or obesity alone treated with and without GLP-1 receptor agonists: a retrospective cohort study from the Global Collaborative Network : Author list. J Endocrinol Invest 2025; 48:483-497. [PMID: 39302577 DOI: 10.1007/s40618-024-02466-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are increasingly used to manage type 2 diabetes (T2D) and obesity. Despite their recognized benefits in glycemic control and weight management, their impact on broader systemic has been less explored. OBJECTIVE This study aimed to evaluate the impact of GLP-1RAs on a variety of systemic diseases in people with T2D or obesity. METHODS We conducted a retrospective cohort study using data from the Global Collaborative Network, accessed through the TriNetX analytics platform. The study comprised two primary groups: individuals with T2D and those with obesity. Each group was further divided into subgroups based on whether they received GLP-1RA treatment or not. Data were analyzed over more than a 5-year follow-up period, comparing incidences of systemic diseases; systemic lupus erythematosus (SLE), systemic sclerosis (SS), rheumatoid arthritis (RA), ulcerative colitis (UC), crohn's disease (CD), alzheimer's disease (AD), parkinson's disease (PD), dementia, bronchial asthma (BA), osteoporosis, and several cancers. RESULTS In the T2D cohorts, GLP-1RA treatment was associated with significantly lower incidences of several systemic and metabolic conditions as compared to those without GLP-1RA, specifically, dementia (Risk Difference (RD): -0.010, p < 0.001), AD (RD: -0.003, p < 0.001), PD (RD: -0.002, p < 0.001), and pancreatic cancer (RD: -0.003, p < 0.001). SLE and SS also saw statistically significant reductions, though the differences were minor in magnitude (RD: -0.001 and - 0.000 respectively, p < 0.001 for both). Conversely, BA a showed a slight increase in risk (RD: 0.002, p < 0.001). CONCLUSIONS GLP-1RAs demonstrate potential benefits in reducing the risk of several systemic conditions in people with T2D or obesity. Further prospective studies are needed to confirm these effects fully and understand the mechanisms.
Collapse
Affiliation(s)
- Mahmoud Nassar
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Omar Nassar
- Williamsville East High School, Buffalo, NY, USA
| | - Hazem Abosheaishaa
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anoop Misra
- Fortis-C-DOC Centre of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India
- National Diabetes, Obesity and Cholesterol Foundation (N-DOC), New Delhi, India
- Diabetes Foundation (India) (DFI) India, New Delhi, India
| |
Collapse
|
24
|
Clarke JR, Bacelar TS, Fernandes GG, Silva RCD, Antonio LS, Queiroz M, de Souza RV, Valadão LF, Ribeiro GS, De Lima EV, Colodeti LC, Mangeth LC, Wiecikowski A, da Silva TN, Paula-Neto HA, da Costa R, Cordeiro Y, Passos GF, Figueiredo CP. Abatacept inhibits Th17 differentiation and mitigates α-synuclein-induced dopaminergic dysfunction in mice. Mol Psychiatry 2025; 30:547-555. [PMID: 39152331 DOI: 10.1038/s41380-024-02618-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is a multifaceted disease characterized by degeneration of nigrostriatal dopaminergic neurons, which results in motor and non-motor dysfunctions. Accumulation of α-synuclein (αSYN) in Lewy bodies is a key pathological feature of PD. Although the exact cause of PD remains unknown, accumulating evidence suggests that brain infiltration of T cells plays a critical role in the pathogenesis of disease, contributing to neuroinflammation and dopaminergic neurodegeneration. Here, we used a mouse model of brain-infused aggregated αSYN, which recapitulates motor and non-motor dysfunctions seen in PD patients. We found that αSYN-induced motor dysfunction in mice is accompanied by an increased number of brain-residing Th17 (IL17+ CD4+) cells, but not CD8+ T cells. To evaluate whether the modulation of T cell response could rescue αSYN-induced damage, we chronically treated animals with abatacept (8 mg/kg, sc, 3x per week), a selective T-cell co-stimulation modulator. We found that abatacept treatment decreased Th1 (IFNƔ+ CD4+) and Th17 (IL17+ CD4+) cells in the brain, rescued motor function and prevented dopaminergic neuronal loss in αSYN-infused mice. These results highlight the significance of effector CD4+ T cells, especially Th17, in the progression of PD and introduce novel possibilities for repurposing immunomodulatory drugs used for arthritis as PD-modifying therapies.
Collapse
Affiliation(s)
- Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Thiago Sa Bacelar
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Raquel Costa da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia S Antonio
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana Queiroz
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Renata V de Souza
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia F Valadão
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriel S Ribeiro
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Emanuelle V De Lima
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944-590, Brazil
| | - Lilian C Colodeti
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luana C Mangeth
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Adalgisa Wiecikowski
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Talita N da Silva
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Heitor A Paula-Neto
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Robson da Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Yraima Cordeiro
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Giselle F Passos
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Claudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
25
|
Giannakis A, Tsamis K, Konitsiotis S. Increased incidence of akinetic crises in Parkinson's disease patients treated with Levodopa-Carbidopa Intestinal Gel: A case series and review of the literature. Clin Neurol Neurosurg 2025; 249:108777. [PMID: 39914276 DOI: 10.1016/j.clineuro.2025.108777] [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: 11/27/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025]
Abstract
Akinetic crisis (AC) is a severe, potentially life-threatening condition affecting individuals with Parkinson's Disease (PD). Similar to neuroleptic malignant syndrome, AC is characterized by a rapid worsening of motor symptoms, including bradykinesia, rigidity, and dysphagia. Additional symptoms may include hyperthermia and autonomic dysfunction. Large-scale prospective studies indicate that the incidence of AC among PD patients ranges from 3.1 % to 3.9 %. In our department, we recorded nine episodes of AC in seven out of 44 patients treated with Levodopa-Carbidopa Intestinal Gel (LCIG) since 2011. This incidence rate is notably higher than the figures reported in larger studies. No significant differences were observed between our patients and those in previous studies, except for mean disease duration. Therefore, LCIG therapy may be linked to a higher incidence of AC. Notably, one of our patients with a parkin gene mutation experienced AC three times. This aligns with previous findings that genetic PD, particularly with parkin mutations, is associated with a higher risk of AC, especially recurrent episodes. Further research is necessary to definitively establish a link between LCIG treatment and AC. Large-scale prospective studies are needed to assess the potential increased risk of this serious complication, which can significantly impact morbidity and mortality in advanced PD patients.
Collapse
Affiliation(s)
- Alexandros Giannakis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece.
| | - Konstantinos Tsamis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece
| | - Spiridon Konitsiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Stavrou Niarchou Av., University Campus, Ioannina, Greece
| |
Collapse
|
26
|
Breßer M, Siemens KD, Schneider L, Lunnebach JE, Leven P, Glowka TR, Oberländer K, De Domenico E, Schultze JL, Schmidt J, Kalff JC, Schneider A, Wehner S, Schneider R. Macrophage-induced enteric neurodegeneration leads to motility impairment during gut inflammation. EMBO Mol Med 2025; 17:301-335. [PMID: 39762650 PMCID: PMC11822118 DOI: 10.1038/s44321-024-00189-w] [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: 05/14/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
Current studies pictured the enteric nervous system and macrophages as modulators of neuroimmune processes in the inflamed gut. Expanding this view, we investigated the impact of enteric neuron-macrophage interactions on postoperative trauma and subsequent motility disturbances, i.e., postoperative ileus. In the early postsurgical phase, we detected strong neuronal activation, followed by transcriptional and translational signatures indicating neuronal death and synaptic damage. Simultaneously, our study revealed neurodegenerative profiles in macrophage-specific transcriptomes after postoperative trauma. Validating the role of resident and monocyte-derived macrophages, we depleted macrophages by CSF-1R-antibodies and used CCR2-/- mice, known for reduced monocyte infiltration, in POI studies. Only CSF-1R-antibody-treated animals showed decreased neuronal death and lessened synaptic decay, emphasizing the significance of resident macrophages. In human gut samples taken early and late during abdominal surgery, we substantiated the mouse model data and found reactive and apoptotic neurons and dysregulation in synaptic genes, indicating a species' overarching mechanism. Our study demonstrates that surgical trauma activates enteric neurons and induces neurodegeneration, mediated by resident macrophages, introducing neuroprotection as an option for faster recovery after surgery.
Collapse
Affiliation(s)
- Mona Breßer
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kevin D Siemens
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Patrick Leven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Tim R Glowka
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristin Oberländer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Elena De Domenico
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim Schmidt
- Department of General, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
27
|
de Deus JL, Maia JM, Soriano RN, Amorim MR, Branco LGS. Psychedelics in neuroinflammation: Mechanisms and therapeutic potential. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111278. [PMID: 39892847 DOI: 10.1016/j.pnpbp.2025.111278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Neuroinflammation is a critical factor in the pathogenesis of various neurodegenerative and psychiatric disorders, including Alzheimer's disease, Parkinson's disease, and major depressive disorder. Psychedelics, such as psilocybin, lysergic acid diethylamide (LSD), and dimethyltryptamine (DMT), have demonstrated promising therapeutic effects on neuroinflammation, primarily through interactions with serotonin (5-HT) receptors, particularly the 5-HT2A receptor. Activation of these receptors by psychedelics modulates the production of pro-inflammatory cytokines, regulates microglial activity, and shifts the balance between neurotoxic and neuroprotective metabolites. Additionally, psychedelics affect critical signaling pathways, including the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt), and mechanistic target of rapamycin (mTOR) pathways, promoting neuroplasticity and exerting anti-inflammatory effects. Beyond the serotonergic system, other neurotransmitter systems-including the glutamatergic, dopaminergic, noradrenergic, gamma-aminobutyric acid (GABAergic), and cholinergic systems-also play significant roles in mediating the effects of psychedelics. This review examines the intricate mechanisms by which psychedelics modulate neuroinflammation and underscores their potential as innovative therapeutic agents for treating neuroinflammatory and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Junia Lara de Deus
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC, USA; Department of Oral and Basic Biology Ribeirão Preto, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Juliana Marino Maia
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares,MG, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Mateus R Amorim
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC, USA; Program of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz G S Branco
- Department of Oral and Basic Biology Ribeirão Preto, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Program of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
28
|
Tripodi G, Lombardo M, Kerav S, Aiello G, Baldelli S. Nitric Oxide in Parkinson's Disease: The Potential Role of Dietary Nitrate in Enhancing Cognitive and Motor Health via the Nitrate-Nitrite-Nitric Oxide Pathway. Nutrients 2025; 17:393. [PMID: 39940251 PMCID: PMC11819985 DOI: 10.3390/nu17030393] [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: 12/11/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, leading to motor symptoms such as tremor, rigidity, and bradykinesia. The pathological hallmarks of PD include Lewy bodies and mechanisms like oxidative/nitrosative stress, chronic inflammation, and mitochondrial dysfunction. Nitric oxide (NO), produced by nitric oxide synthase (NOS) isoforms, plays a dual role in neuroprotection and neurodegeneration. Excessive NO production exacerbates neuroinflammation and oxidative/nitrosative damage, contributing to dopaminergic cell death. This review explores NO's role in PD pathogenesis and investigates dietary nitrate as a therapeutic strategy to regulate NO levels. METHODS A literature review of studies addressing the role of NO in PD was conducted using major scientific databases, including PubMed, Scopus, and Web of Science, using keywords such as "nitric oxide", "NOSs", "Parkinson's disease", and "nitrate neuroprotection in PD". Studies on nitrate metabolism via the nitrate-nitrite-NO pathway and its effects on PD hallmarks were analyzed. Studies regarding the role of nitrosamine formation in PD, which are mainly formed during the nitrification process of amines (nitrogen-containing compounds), often due to chemical reactions in the presence of nitrite or nitrate, were also examined. In particular, nitrate has been shown to induce oxidative stress, affect the mitochondrial function, and contribute to inflammatory phenomena in the brain, another factor closely related to the pathogenesis of PD. RESULTS Excessive NO production, particularly from iNOS and nNOS, was strongly associated with neuroinflammation and oxidative/nitrosative stress, amplifying neuronal damage in PD. Dietary nitrate was shown to enhance NO bioavailability through the nitrate-nitrite-NO pathway, mitigating inflammation and oxidative/nitrosative damage. CONCLUSIONS Dysregulated NO production contributes significantly to PD progression via inflammatory and oxidative/nitrosative pathways. Dietary nitrate, by modulating NO levels, offers a promising therapeutic strategy to counteract these pathological mechanisms. Further clinical trials are warranted to establish its efficacy and optimize its use in PD management.
Collapse
Affiliation(s)
- Gianluca Tripodi
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (G.T.); (M.L.); (G.A.)
| | - Mauro Lombardo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (G.T.); (M.L.); (G.A.)
| | - Sercan Kerav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, 17100 Çanakkale, Türkiye;
| | - Gilda Aiello
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (G.T.); (M.L.); (G.A.)
| | - Sara Baldelli
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (G.T.); (M.L.); (G.A.)
- IRCCS San Raffaele Roma, 00166 Rome, Italy
| |
Collapse
|
29
|
Abhilash PL, Bharti U, Rashmi SK, Philip M, Raju TR, Kutty BM, Sagar BKC, Alladi PA. Aging and MPTP Sensitivity Depend on Molecular and Ultrastructural Signatures of Astroglia and Microglia in Mice Substantia Nigra. Cell Mol Neurobiol 2025; 45:13. [PMID: 39833644 PMCID: PMC11753320 DOI: 10.1007/s10571-024-01528-8] [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: 10/27/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Both astroglia and microglia show region-specific distribution in CNS and often maladapt to age-associated alterations within their niche. Studies on autopsied substantia nigra (SN) of Parkinson's disease (PD) patients and experimental models propose gliosis as a trigger for neuronal loss. Epidemiological studies propose an ethnic bias in PD prevalence, since Caucasians are more susceptible than non-whites. Similarly, different mice strains are variably sensitive to MPTP. We had earlier likened divergent MPTP sensitivity of C57BL/6 J and CD-1 mice with differential susceptibility to PD, based on the numbers of SN neurons. We examined whether the variability was incumbent to inter-strain differences in glial features of male C57BL/6 J and CD-1 mice. Stereological counts showed relatively more microglia and fewer astrocytes in the SN of normal C57BL/6 J mice, suggesting persistence of an immune-vigilant state. MPTP-induced microgliosis and astrogliosis in both strains suggest their involvement in pathogenesis. ELISA of pro-inflammatory cytokines in the ventral-midbrain revealed augmentation of TNF-α and IL-6 at middle age in both strains that reduced at old age, suggesting middle age as a critical, inflamm-aging-associated time point. TNF-α levels were high in C57BL/6 J, through aging and post-MPTP, while IL-6 and IL-1β were upregulated at old age. CD-1 had higher levels of anti-inflammatory cytokine TGF-β. MPTP challenge caused upregulation of enzymes MAO-A, MAO-B, and iNOS in both strains. Post-MPTP enhancement in fractalkine and hemeoxygenase-1 may be neuron-associated compensatory signals. Ultrastructural observations of elongated astroglial/microglial mitochondria vis-à-vis the shrunken ones in neurons suggest a scale-up of their functions with neurotoxic consequences. Thus, astroglia and microglia may modulate aging and PD susceptibility.
Collapse
Affiliation(s)
- P L Abhilash
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Upasna Bharti
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Santhosh Kumar Rashmi
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Mariamma Philip
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - T R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Bindu M Kutty
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - B K Chandrasekhar Sagar
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
30
|
Torrente D, Su EJ, Citalán-Madrid AF, Schielke GP, Magaoay D, Warnock M, Stevenson T, Mann K, Lesept F, Delétage N, Blanc M, Norris EH, Vivien D, Lawrence DA. The interaction of tPA with NMDAR1 drives neuroinflammation and neurodegeneration in α-synuclein-mediated neurotoxicity. J Neuroinflammation 2025; 22:8. [PMID: 39810216 PMCID: PMC11731172 DOI: 10.1186/s12974-025-03336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The thrombolytic protease tissue plasminogen activator (tPA) is expressed in the CNS, where it regulates diverse functions including neuronal plasticity, neuroinflammation, and blood-brain-barrier integrity. However, its role in different brain regions such as the substantia nigra (SN) is largely unexplored. In this study, we characterize tPA expression, activity, and localization in the SN using a combination of retrograde tracing and β-galactosidase tPA reporter mice. We further investigate tPA's potential role in SN pathology in an α-synuclein mouse model of Parkinson's disease (PD). To characterize the mechanism of tPA action in α-synuclein-mediated pathology in the SN and to identify possible therapeutic pathways, we performed RNA-seq analysis of the SN and used multiple transgenic mouse models. These included tPA deficient mice and two newly developed transgenic mice, a knock-in model expressing endogenous levels of proteolytically inactive tPA (tPA Ala-KI) and a second model overexpressing proteolytically inactive tPA (tPA Ala-BAC). Our findings show that striatal GABAergic neurons send tPA+ projections to dopaminergic (DA)-neurons in the SN and that tPA is released from SN-derived synaptosomes upon stimulation. We also found that tPA levels in the SN increased following α-synuclein overexpression. Importantly, tPA deficiency protects DA-neurons from degeneration, prevents behavioral deficits, and reduces microglia activation and T-cell infiltration induced by α-synuclein overexpression. RNA-seq analysis indicates that tPA in the SN is required for the upregulation of genes involved in the innate and adaptive immune responses induced by α-synuclein overexpression. Overexpression of α-synuclein in tPA Ala-KI mice, expressing only proteolytically inactive tPA, confirms that tPA-mediated neuroinflammation and neurodegeneration is independent of its proteolytic activity. Moreover, overexpression of proteolytically inactive tPA in tPA Ala-BAC mice leads to increased neuroinflammation and neurodegeneration compared to mice expressing normal levels of tPA, suggesting a tPA dose response. Finally, treatment of mice with glunomab, a neutralizing antibody that selectively blocks tPA binding to the N-methyl-D-aspartate receptor-1 (NMDAR1) without affecting NMDAR1 ion channel function, identifies the tPA interaction with NMDAR1 as necessary for tPA-mediated neuroinflammation and neurodegeneration in response to α-synuclein-mediated neurotoxicity. Thus, our data identifies a novel pathway that promotes DA-neuron degeneration and suggests a potential therapeutic intervention for PD targeting the tPA-NMDAR1 interaction.
Collapse
Affiliation(s)
- Daniel Torrente
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, USA
| | - Enming J Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Alí Francisco Citalán-Madrid
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Gerald P Schielke
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Daniel Magaoay
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark Warnock
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Tamara Stevenson
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Kris Mann
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Flavie Lesept
- Lys Therapeutics, Main offices: 56 rue Saint Jean de Dieu, Lyon, 69007, France
- Lys Therapeutics, HQ: Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Nathalie Delétage
- Lys Therapeutics, Main offices: 56 rue Saint Jean de Dieu, Lyon, 69007, France
- Lys Therapeutics, HQ: Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Manuel Blanc
- Lys Therapeutics, Main offices: 56 rue Saint Jean de Dieu, Lyon, 69007, France
- Lys Therapeutics, HQ: Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, USA
| | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders (PhIND), UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Normandie Univ, Caen, France
- Department of Clinical Research, Caen University Hospital, CHU, Caen, France
| | - Daniel A Lawrence
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA.
| |
Collapse
|
31
|
Ghaith WZ, Wadie W, El-Yamany MF. Crosstalk between SIRT1/Nrf2 signaling and NLRP3 inflammasome/pyroptosis as a mechanistic approach for the neuroprotective effect of linagliptin in Parkinson's disease. Int Immunopharmacol 2025; 145:113716. [PMID: 39642562 DOI: 10.1016/j.intimp.2024.113716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
In recent years, special attention has been paid to highlighting the antiparkinsonian effect of linagliptin. However, the mechanism of its action has not yet been well investigated. The present study aimed to verify the neuroprotective effect of linagliptin in the rotenone model of Parkinson's disease (PD) and further explore its potential molecular mechanisms. Rats were intoxicated with rotenone (2 mg/kg/day; sc) and treated with linagliptin (10 mg/kg/day; po) for 14 consecutive days. The present finding showed that linagliptin ameliorated the histopathological changes of rotenone on substantia nigra and striata. Linagliptin decreased α-synuclein immunoreactivity along with an increase in tyrosine hydroxylase immunoreactivity and striatal dopamine content. This was reflected in the marked improvement of the behavior and motor deficits in rotenone-intoxicated rats. On the molecular level, linagliptin upregulated sirtuin 1 (SIRT1)/ nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, reduced ionized calcium-binding adaptor molecule 1 (Iba1) protein expression, restored glutathione (GSH) content, and elevated heme oxygenase-1 (HO-1) level in rats with rotenone intoxication. Moreover, linagliptin inhibited NOD-like receptor protein 3 (NLRP3)/caspase-1/interleukin-1β (IL-1β) cascade with subsequent reduction in gasdermin D (GSDMD) expression. Therefore, the present study reveals the ability of linagliptin, through the activation of SIRT1/Nrf2 signaling, to suppress NLRP3 inflammasome-mediated pyroptosis and protect against rotenone-induced parkinsonism.
Collapse
Affiliation(s)
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt.
| |
Collapse
|
32
|
Sakiyama H, Baba K, Kimura Y, Ogawa K, Nishiike U, Hayakawa H, Yoshida M, Aguirre C, Ikenaka K, Nagano S, Mochizuki H. Accelerated senescence exacerbates α-synucleinopathy in senescence-accelerated prone 8 mice via persistent neuroinflammation. Neurochem Int 2025; 182:105906. [PMID: 39586378 DOI: 10.1016/j.neuint.2024.105906] [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: 09/29/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Parkinson's disease (PD) is characterized by the formation of α-synuclein (α-syn) aggregates, which lead to dopaminergic neuronal degeneration. The incidence of PD increases with age, and senescence is considered to be a major risk factor for PD. In this study, we evaluated the effect of senescence on PD pathology using α-synuclein preformed fibrils (PFF) injection model in senescence-accelerated mice. We injected PFF into the substantia nigra (SN) of senescence-accelerated prone 8 (SAMP8) mice and senescence-accelerated resistant 1 (SAMR1) mice. At 24 weeks after injection of saline or PFF, we found that SAMP8 mice injected with PFF exhibited robust Lewy pathology and exacerbated degeneration of dopaminergic neurons in the SN compared to PFF-injected SAMR1 mice. We further observed an increase in the number of Iba1-positive cells in the brains of PFF-injected SAMP8 mice. RNA sequencing revealed that several genes related to neuroinflammation were upregulated in the brains of PFF-injected SAMP8 mice compared to SAMR1 mice. Inflammatory chemokine CC-chemokine ligand 21 (CCL21) was upregulated in PFF-injected SAMP8 mice and expressed in the glial cells of these mice. Our research indicates that accelerated senescence leads to persistent neuroinflammation, which plays an important role in the exacerbation of α-synucleinopathy.
Collapse
Affiliation(s)
- Hiroshi Sakiyama
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Kousuke Baba
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan; Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Yasuyoshi Kimura
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Kotaro Ogawa
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Ujiakira Nishiike
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Hideki Hayakawa
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Miki Yoshida
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Cesar Aguirre
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Seiichi Nagano
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan; Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University, Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
33
|
Khan S, Haider MF. A Comprehensive Review on Repurposing the Nanocarriers for the Treatment of Parkinson's Disease: An Updated Patent and Clinical Trials. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:181-195. [PMID: 39400019 DOI: 10.2174/0118715273323074241001071645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024]
Abstract
Parkinson's Disease (PD) is a progressive neurodegenerative disorder marked by the deterioration of dopamine-producing neurons, resulting in motor impairments like tremors and rigidity. While the precise cause remains elusive, genetic and environmental factors are implicated. Mitochondrial dysfunction, oxidative stress, and protein misfolding contribute to the disease's pathology. Current therapeutics primarily aim at symptom alleviation, employing dopamine replacement and deep brain stimulation. However, the quest for disease-modifying treatments persists. Ongoing clinical trials explore novel approaches, such as neuroprotective agents and gene therapies, reflecting the evolving PD research landscape. This review provides a comprehensive overview of PD, covering its basics, causal factors, major pathways, existing treatments, and a nuanced exploration of ongoing clinical trials. As the scientific community strives to unravel PD's complexities, this review offers insights into the multifaceted strategies pursued for a better understanding and enhanced management of this debilitating condition.
Collapse
Affiliation(s)
- Sara Khan
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Md Faheem Haider
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
34
|
Zambom-Ferraresi F, Zambom-Ferraresi F, Izco M, Martínez-Velilla N, Extramiana L, Fernández-Irigoyen J, Santamaría E, Llamas-Urbano A, Hanaee Y, Martinez-Moreno JM, Barbarroja N, Pérez-Sánchez C. Monitoring Cerebrospinal Fluid Inflammatory Mediators by Olink Target 48 Cytokine Panel. Methods Mol Biol 2025; 2914:51-64. [PMID: 40167910 DOI: 10.1007/978-1-0716-4462-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The diagnosis and monitoring of neurological disorders are challenging due to their complexity and the difficulty of obtaining samples of damaged tissue while the patient is alive. Cerebrospinal fluid (CSF) is an excellent biofluid for conducting diagnostic and monitoring studies because it is in direct contact with the brain, allowing for the analysis of secreted proteins and peptides associated with various neurological disorders.The Proximity Extension Assay, which combines antibodies with oligonucleotides to detect proteins via qPCR, is revolutionizing the field of biomarker identification. The Olink Target 48 Cytokine Panel is an optimal tool for evaluating the inflammatory profile of CSF samples, requiring only one microliter. The workflow involves incubation with antibody-linked oligonucleotides, extension, amplification through a PCR assay, and detection using microfluidic qPCR, enabling precise and accurate quantification of cytokine levels in pg/mL. This approach offers valuable insights into inflammatory processes within the CSF, serving as a robust tool for studying and understanding inflammation in neurological contexts.
Collapse
Affiliation(s)
- Fabricio Zambom-Ferraresi
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Fabiola Zambom-Ferraresi
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Maite Izco
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Nicolás Martínez-Velilla
- Department of Geriatric Medicine, Hospital Universitario de Navarra (HUN), Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Leire Extramiana
- Clinical Neuroproteomics Laboratory, Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Laboratory, Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Laboratory, Proteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | | | - Yasin Hanaee
- Cobiomic Bioscience. EBT University of Cordoba/IMIBIC, Cordoba, Spain
| | | | - Nuria Barbarroja
- Cobiomic Bioscience. EBT University of Cordoba/IMIBIC, Cordoba, Spain
- Rheumatology Unit/Reina Sofia Hospital/University of Cordoba/IMIBIC, Cordoba, Spain
| | - Carlos Pérez-Sánchez
- Cobiomic Bioscience. EBT University of Cordoba/IMIBIC, Cordoba, Spain.
- Rheumatology Unit/Reina Sofia Hospital/University of Cordoba/IMIBIC, Cordoba, Spain.
| |
Collapse
|
35
|
Asfour AAR, Evren AE, Sağlık Özkan BN, Yurttaş L. Investigating the potential of novel thiazole derivatives in treating Alzheimer's and Parkinson's diseases. J Biomol Struct Dyn 2024:1-17. [PMID: 39672098 DOI: 10.1080/07391102.2024.2437521] [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: 01/22/2024] [Accepted: 05/17/2024] [Indexed: 12/15/2024]
Abstract
The study aimed to investigate 12 novel thiazole compounds in the treatment of neurodegenerative disorders. The compounds produced were evaluated for their inhibitory efficacy against acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and monoamine oxidases (MAOs). Among the compounds, 5d, 5e, and 5j showed the highest AChE inhibitory activity. The IC50 values for compounds are 0.223 ± 0.010 µM, 0.092 ± 0.003 µM, and 0.054 ± 0.002 µM, respectively. In addition, molecular docking analyses and molecular dynamic simulation were used to examine the interactions of these compounds with protein sites. The results suggest that thiazole-ring compounds could serve as a promising basis for the development of drugs aimed at treating neurodegenerative diseases (NDD), caused by Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Abd Al Rahman Asfour
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
- Institute of Graduate Education, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
| | - Asaf Evrim Evren
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
- Department of Pharmacy Services, Bilecik Seyh Edebali University, Vocational School of Health Services, Bilecik
| | | | - Leyla Yurttaş
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
36
|
Tian H, Yao J, Ba Q, Meng Y, Cui Y, Quan L, Gong W, Wang Y, Yang Y, Yang M, Gao C. Cerebral biomimetic nano-drug delivery systems: A frontier strategy for immunotherapy. J Control Release 2024; 376:1039-1067. [PMID: 39505218 DOI: 10.1016/j.jconrel.2024.10.058] [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: 04/29/2024] [Revised: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Brain diseases are a significant threat to human health, especially in the elderly, and this problem is growing as the aging population increases. Efficient brain-targeted drug delivery has been the greatest challenge in treating brain disorders due to the unique immune environment of the brain, including the blood-brain barrier (BBB). Recently, cerebral biomimetic nano-drug delivery systems (CBNDSs) have provided a promising strategy for brain targeting by mimicking natural biological materials. Herein, this review explores the latest understanding of the immune microenvironment of the brain, emphasizing the immune mechanisms of the occurrence and progression of brain disease. Several brain targeting systems are summarized, including cell-based, exosome-based, protein-based, and microbe-based CBNDSs, and their immunological mechanisms are highlighted. Moreover, given the rise of immunotherapy, the latest applications of CBNDSs in immunotherapy are also discussed. This review provides a comprehensive understanding of CBNDSs and serves as a guideline for immunotherapy in treating brain diseases. In addition, it provides inspiration for the future of CBNDSs.
Collapse
Affiliation(s)
- Hao Tian
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Jiaxin Yao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Qi Ba
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Yuanyuan Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanan Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liangzhu Quan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
37
|
Du J, Zhong Y, Fan B, Yang X, Ye R, Huang Y, Li Z, Liang B, Xian H, Deng Y, Huang X, Chen X, Shi C, Yu X, Wu B, Yang X, Huang Z. Human umbilical cord mesenchymal stem cells mitigate A1 astrocyte neuroinflammation induced by 1,2-dichloroethane via ERBB pathway inhibition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117365. [PMID: 39571258 DOI: 10.1016/j.ecoenv.2024.117365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
1,2-Dichloroethane (1,2-DCE), a prevalent industrial and environmental contaminant, induces toxic encephalopathy through inhalation, leading to neurotoxic effects and inflammation-driven brain edema. Human umbilical cord mesenchymal stem cells (HUCMSCs) secrete bioactive factors, including miRNAs, proteins, and lipids via exosomes, exhibiting anti-inflammatory and immune-regulatory properties. However, their potential in treating 1,2-DCE-induced neuroinflammation and the underlying mechanisms remain unclear. This study investigates how HUCMSCs mitigate 1,2-DCE-induced neuroinflammation. We exposed SVG p12 cells to 1,2-DCE and assessed inflammatory markers and A1 astrocyte activation. Co-culturing these cells with HUCMSCs, we used RNA sequencing to analyze inflammatory modulation. Additionally, HUCMSCs were administered to CD-1 male mice post-1,2-DCE exposure, evaluating the reduction in A1 astrocyte inflammation via behavioral tests, molecular analyses, and tissue staining. Pre-treating HUCMSCs with exosome inhibitors and co-culturing them with 1,2-DCE-treated SVG p12 cells investigated miRNA transfer. Results showed that 1,2-DCE activated A1 astrocytes, leading to increases in interleukin-1β (IL-1β, 4.9-fold), tumor necrosis factor-α (TNF-α, 2.5-fold), complement 3 (C3, 2.1-fold), and glial fibrillary acidic protein (GFAP, 1.4-fold). HUCMSCs effectively reversed 1,2-DCE-induced A1 astrocyte inflammation, attenuating IL-1β, TNF-α, and A1 astrocyte activation. RNA-seq highlighted modulation of the erb-b2 receptor tyrosine kinase (ERBB) pathway via Ral-binding protein 1-associated Eps domain-containing 2 (REPS2). In vivo confirmation underscored these findings. Importantly, HUCMSC-derived exosomes, particularly miR-3064-5p, reversed 1,2-DCE-activated A1 astrocyte inflammation, suggesting therapeutic potential. Collectively, HUCMSCs alleviate 1,2-DCE-induced neuroinflammation via exosome-mediated miR-3064-5p secretion, targeting REPS2 to mitigate neuroinflammation. This study advances the understanding of their therapeutic roles and highlights HUCMSC exosomal miRNA transfer for treating 1,2-DCE-induced neuroinflammatory conditions.
Collapse
Affiliation(s)
- Jiaxin Du
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yizhou Zhong
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bingchi Fan
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiaohong Yang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Rongyi Ye
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yuji Huang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhiming Li
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Boxuan Liang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hongyi Xian
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yanhong Deng
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiyun Huang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiaoqing Chen
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Congying Shi
- Institution of Guangdong Cord Blood Bank, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510663, China; Department of Experimental Center, Guangzhou Municipality Tianhe Nuoya Bio-engineering Co., Ltd, Guangzhou, Guangdong 510663, China
| | - Xibao Yu
- Institution of Guangdong Cord Blood Bank, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510663, China; Department of Experimental Center, Guangzhou Municipality Tianhe Nuoya Bio-engineering Co., Ltd, Guangzhou, Guangdong 510663, China
| | - Banghua Wu
- Institute of Chemical Surveillance, Guangdong Provincial Hospital for Occupational Disease Prevention and Treatment, Guangzhou 510300, China
| | - Xingfen Yang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zhenlie Huang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
38
|
Navale GR, Ahmed I, Lim MH, Ghosh K. Transition Metal Complexes as Therapeutics: A New Frontier in Combatting Neurodegenerative Disorders through Protein Aggregation Modulation. Adv Healthc Mater 2024; 13:e2401991. [PMID: 39221545 DOI: 10.1002/adhm.202401991] [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: 05/30/2024] [Revised: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Neurodegenerative disorders (NDDs) are a class of debilitating diseases that progressively impair the protein structure and result in neurological dysfunction in the nervous system. Among these disorders, Alzheimer's disease (AD), prion diseases such as Creutzfeldt-Jakob disease (CJD), and Parkinson's disease (PD) are caused by protein misfolding and aggregation at the cellular level. In recent years, transition metal complexes have gained significant attention for their potential applications in diagnosing, imaging, and curing these NDDs. These complexes have intriguing possibilities as therapeutics due to their diverse ligand systems and chemical properties and can interact with biological systems with minimal detrimental effects. This review focuses on the recent progress in transition metal therapeutics as a new era of hope in the battle against AD, CJD, and PD by modulating protein aggregation in vitro and in vivo. It may shed revolutionary insights into unlocking new opportunities for researchers to develop metal-based drugs to combat NDDs.
Collapse
Affiliation(s)
- Govinda R Navale
- Department of Chemistry, Indian Institute of Technology, Roorkee, 247667, India
| | - Imtiaz Ahmed
- Department of Chemistry, Indian Institute of Technology, Roorkee, 247667, India
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kaushik Ghosh
- Department of Chemistry, Indian Institute of Technology, Roorkee, 247667, India
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, 247667, India
| |
Collapse
|
39
|
Safreena N, Nair IC, Chandra G. Therapeutic potential of Parkin and its regulation in Parkinson's disease. Biochem Pharmacol 2024; 230:116600. [PMID: 39500382 DOI: 10.1016/j.bcp.2024.116600] [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: 07/25/2024] [Revised: 10/05/2024] [Accepted: 10/28/2024] [Indexed: 11/14/2024]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the midbrain substantia nigra, resulting in motor and non-motor symptoms. While the exact etiology of PD remains elusive, a growing body of evidence suggests that dysfunction in the parkin protein plays a pivotal role in the pathogenesis of the disease. Parkin is an E3 ubiquitin ligase that ubiquitinates substrate proteins to control a number of crucial cellular processes including protein catabolism, immune response, and cellular apoptosis.While autosomal recessive mutations in the PARK2 gene, which codes for parkin, are linked to an inherited form of early-onset PD, heterozygous mutations in PARK2 have also been reported in the more commonly occurring sporadic PD cases. Impairment of parkin's E3 ligase activity is believed to play a pathogenic role in both familial and sporadic forms of PD.This article provides an overview of the current understanding of the mechanistic basis of parkin's E3 ligase activity, its major physiological role in controlling cellular functions, and how these are disrupted in familial and sporadic PD. The second half of the manuscript explores the currently available and potential therapeutic strategies targeting parkin structure and/or function in order to slow down or mitigate the progressive neurodegeneration in PD.
Collapse
Affiliation(s)
- Narukkottil Safreena
- Cell Biology Laboratory, Center for Development and Aging Research, Inter University Center for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam 686009, Kerala, India
| | - Indu C Nair
- SAS SNDP Yogam College, Konni, Pathanamthitta 689691, Kerala, India
| | - Goutam Chandra
- Cell Biology Laboratory, Center for Development and Aging Research, Inter University Center for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam 686009, Kerala, India.
| |
Collapse
|
40
|
Sood R, Sanjay, Kang SU, Yoon NY, Lee HJ. Malvidin-3-O-Glucoside Mitigates α-Syn and MPTP Co-Induced Oxidative Stress and Apoptosis in Human Microglial HMC3 Cells. Int J Mol Sci 2024; 25:12733. [PMID: 39684444 DOI: 10.3390/ijms252312733] [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: 09/11/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease (PD) is a widespread age-related neurodegenerative disorder characterized by the presence of an aggregated protein, α-synuclein (α-syn), which is encoded by the SNCA gene and localized to presynaptic terminals in a normal human brain. The α-syn aggregation is induced by the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mitochondrial neurotoxin and is therefore used to mimic PD-like pathology in various in vitro and in vivo models. However, in vitro PD-like pathology using α-syn and MPTP in human microglial cells has not yet been reported. Malvidin-3-O-glucoside (M3G) is a major anthocyanin primarily responsible for pigmentation in various fruits and beverages and has been reported to possess various bioactivities. However, the neuroprotective effects of M3G in humanized in vitro PD-like pathologies have not been reported. Therefore, individual and co-treatments of α-syn and MPTP in a human microglial (HMC3) cell line were used to establish a humanized PD-like pathology model in vitro. The individual treatments were significantly less cytotoxic when compared to the α-syn and MPTP co-treatment. This study examined the neuroprotective effects of M3G by treating HMC3 cells with α-syn (8 μg/mL) and MPTP (2 mM) individually or in a co-treatment in the presence or absence of M3G (50 μM). M3G demonstrated anti-apoptotic, anti-inflammatory, and antioxidative properties against the α-syn- and MPTP-generated humanized in vitro PD-like pathology. This study determined that the cytoprotective effects of M3G are mediated by nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase (HO)-1 signaling.
Collapse
Affiliation(s)
- Rachit Sood
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
| | - Sanjay
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Na Young Yoon
- Food Safety and Processing Research Division, National Institute of Fisheries Science, Busan 46083, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Science and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
41
|
Ben Shaul T, Frenkel D, Gurevich T. The Interplay of Stress, Inflammation, and Metabolic Factors in the Course of Parkinson's Disease. Int J Mol Sci 2024; 25:12409. [PMID: 39596474 PMCID: PMC11594997 DOI: 10.3390/ijms252212409] [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: 09/29/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative condition for which there are symptomatic treatments but no disease-modifying therapies (DMTs). Extensive research over the years has highlighted the need for a multi-target DMT approach in PD that recognizes the various risk factors and their intricate interplay in contributing to PD-related neurodegeneration. Widespread risk factors, such as emotional stress and metabolic factors, have increasingly become focal points of exploration. Our review aims to summarize interactions between emotional stress and selected key players in metabolism, such as insulin, as potential mechanisms underlying neurodegeneration in PD.
Collapse
Affiliation(s)
- Tal Ben Shaul
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tanya Gurevich
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
42
|
Abu-Baih DH, Abd El-Mordy FM, Saber EA, Ali SFES, Hisham M, Alanazi MA, Altemani FH, Algehainy NA, Lehmann L, Abdelmohsen UR. Unlocking the potential of edible Ulva sp. seaweeds: Metabolomic profiling, neuroprotective mechanisms, and implications for Parkinson's disease management. Arch Pharm (Weinheim) 2024; 357:e2400418. [PMID: 39086040 DOI: 10.1002/ardp.202400418] [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: 05/25/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
Green seaweed (Ulva sp.) is frequently used as a food component and nutraceutical agent because of its high polysaccharide and natural fiber content in Asian countries. This study investigates both metabolomic profiling of Ulva sp. and the neuroprotective efficacy of its ethanol extract and its underlying mechanisms in a rotenone-induced rat model of neurodegeneration, mimicking Parkinson's disease (PD) in humans. Metabolomic profiling of Ulva sp. extract was done using liquid chromatography high resolution electrospray ionization mass spectrometry and led to the identification of 22 compounds belonging to different chemical classes.Catenin Beta Additionally, this study demonstrated the neuroprotective properties against rotenone-induced PD, which was achieved through the suppression of elevated levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 together with the inhibition of reactive oxygen species (ROS) generation, apoptosis, inflammatory mediators, and the phosphoinositide 3-kinases/serine/threonine protein kinase (PI3K/AKT) pathway. Using a protein-protein interaction network, AKT1, GAPDH, TNF-α, IL-6, caspase 3, signal transducer and activator of transcription 3, Catenin Beta 1, epidermal growth factor receptor, B-cell lymphoma -2, and HSP90AA1 were identified as the top 10 most significant genes. Finally, molecular docking results showed that compounds 1, 3, and 7 might possess a promising anti-parkinsonism effect by binding to active sites of selected hub genes. Therefore, it is hypothesized that the Ulva sp. extract has the potential to be further developed as a potential therapeutic agent for the treatment of PD.
Collapse
Affiliation(s)
- Dalia H Abu-Baih
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Deraya University, Minya, Egypt
- Deraya Center for Scientific Research, Deraya University, New Minia City, Minia, Egypt
| | - Fatma Mohamed Abd El-Mordy
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | | | | | - Mohamed Hisham
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New-Minia, Egypt
| | - Mohammad A Alanazi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Faisal H Altemani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh A Algehainy
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Leane Lehmann
- Chair of Food Chemistry, Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Usama Ramadan Abdelmohsen
- Deraya Center for Scientific Research, Deraya University, New Minia City, Minia, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
43
|
Muhammad AJ, Al-Baqami FF, Alanazi FE, Alattar A, Alshaman R, Rehman NU, Riadi Y, Shah FA. The Interplay of Carveol and All-Trans Retinoic Acid (ATRA) in Experimental Parkinson's Disease: Role of Inflammasome-Mediated Pyroptosis and Nrf2. Neurochem Res 2024; 49:3118-3130. [PMID: 39190122 DOI: 10.1007/s11064-024-04226-5] [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: 04/25/2024] [Revised: 07/21/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
Parkinson's disease (PD) is a debilitating and the second most common neurodegenerative disorder with a high prevalence. PD has a multifaceted etiology characterized by an altered redox state and an excessive inflammatory response. Extensive research has consistently demonstrated the role of the nuclear factor E2-related factor (Nrf2) and inflammasomes, notably NLRP3 in neurodegenerative diseases. In this study, our focus was on exploring the potential neuroprotective properties of carveol in Parkinson's disease. Our findings suggest that carveol may exhibit these effects through Nrf2 and by suppressing pyroptosis. Male albino mice were treated with carveol, and the animal PD model was induced through a single intranigral dose of 2 µg/2µl lipopolysaccharide (LPS). To further demonstrate the essential role of the Nrf2 pathway, we utilized all-trans retinoic acid (ATRA) to inhibit the Nrf2. Our finding showed the induction of pyroptosis as evidenced by increased levels of NLRP3 and other inflammatory mediators, including IL-1β, iNOS, p-NFKB, and apoptotic cell death indicated by positive fluoro Jade B (FJB) staining. Moreover, increased levels of lipid peroxides and reactive oxygen species indicated a significant rise in oxidative stress due to LPS. The administration of carveol mitigates oxidative stress and suppresses inflammatory pathways through the augmentation of intrinsic antioxidant defenses, primarily via the activation of the Nrf2. Conversely, ATRA reversed carveol protective effects by increasing FJB-positive cells, inflammatory and oxidative biomarkers. Taken together, our findings suggest that carveol mitigated LPS-induced Parkinson-like symptoms, partially through the activation of the Nrf2 and downregulation of pyroptosis notably NLRP3.
Collapse
Affiliation(s)
- Asmaa Jan Muhammad
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Faisal F Al-Baqami
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia
| | - Fawaz E Alanazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Najeeb Ur Rehman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia
| | - Yassine Riadi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia
- Department of Pharmaceutical Chemistry, College of Pharmacy Prince Sattam Bin Abdul Aziz University, Al-Kharj, Saudi Arabia
| | - Fawad Ali Shah
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj, 16242, Saudi Arabia.
| |
Collapse
|
44
|
Poniatowski ŁA, Joniec-Maciejak I, Wawer A, Sznejder-Pachołek A, Machaj E, Ziętal K, Mirowska-Guzel D. Dose-Ranging Effects of the Intracerebral Administration of Atsttrin in Experimental Model of Parkinson's Disease Induced by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in Mice. Mol Neurobiol 2024; 61:9432-9458. [PMID: 38642286 PMCID: PMC11496375 DOI: 10.1007/s12035-024-04161-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024]
Abstract
Parkinson's disease is one of the most common neurodegenerative disorders characterized by a multitude of motor and non-motor clinical symptoms resulting from the progressive and long-lasting abnormal loss of nigrostriatal dopaminergic neurons. Currently, the available treatments for patients with Parkinson's disease are limited and exert only symptomatic effects, without adequate signs of delaying or stopping the progression of the disease. Atsttrin constitutes the bioengineered protein which ultrastructure is based on the polypeptide chain frame of the progranulin (PGRN), which exerts anti-inflammatory effects through the inhibition of TNFα. The conducted preclinical studies suggest that the therapeutic implementation of Atsttrin may be potentially effective in the treatment of neurodegenerative diseases that are associated with the occurrence of neuroinflammatory processes. The aim of the proposed study was to investigate the effect of direct bilateral intracerebral administration of Atsttrin using stereotactic methods in the preclinical C57BL/6 mouse model of Parkinson's disease inducted by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication. The analysis of the dose dependency effects of the increasing doses of Atsttrin has covered a number of parameters and markers regarding neurodegenerative processes and inflammatory responses including IL-1α, TNFα, IL-6, TH, and TG2 mRNA expressions. Accordingly, the evaluation of the changes in the neurochemical profile included DA, DOPAC, 3-MT, HVA, NA, MHPG, 5-HT, and 5-HIAA concentration levels. The intracerebral administration of Atsttrin into the striatum effectively attenuated the neuroinflammatory reaction in evaluated neuroanatomical structures. Furthermore, the partial restoration of monoamine content and its metabolic turnover were observed. In this case, taking into account the previously described pharmacokinetic profile and extrapolated bioavailability as well as the stability characteristics of Atsttrin, an attempt was made to describe as precisely as possible the quantitative and qualitative effects of increasing doses of the compound within the brain tissue microenvironment in the presented preclinical model of the disease. Collectively, this findings demonstrated that the intracerebral administration of Atsttrin may represent a potential novel therapeutic method for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Łukasz A Poniatowski
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
- Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Salvador-Allende-Straße 30, 17036, Neubrandenburg, Germany
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| | - Adriana Wawer
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Anna Sznejder-Pachołek
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Ewa Machaj
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Katarzyna Ziętal
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| |
Collapse
|
45
|
Cheng W, Fang K, Ouyang X, Jin L, Song Y, Yu B. Vagus nerve stimulation with a small total charge transfer improves motor behavior and reduces neuroinflammation in a mouse model of Parkinson's disease. Neurochem Int 2024; 180:105871. [PMID: 39362497 DOI: 10.1016/j.neuint.2024.105871] [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: 06/27/2024] [Revised: 09/21/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Conventional treatments are ineffective in reversing disease progression. Recently, the therapeutic and rehabilitation potential of vagus nerve stimulation (VNS) in PD has been explored. However, the underlying mechanisms remain largely unknown. In this study, we investigated the neuroprotective effects of VNS in a lateral lesioned mice model of PD. Excluding controls, experimental mice received cuff electrode implantation on the left vagus nerve and 6-hydroxydopamine administration into the bilateral striatum. After ten days, electrical stimulation was delivered for 11 consecutive days onto PD animals. Behavioral tests were performed after stimulation. The expression of TH, Iba-1, GFAP, adrenergic receptors and cytokines in the SN and striatum was detected by immunofluorescence or western blotting. The activity of noradrenergic neurons in the locus coeruleus (LC) was also measured. Our results suggest that VNS improved behavioral performance in rod rotation, open field tests and pole-climbing tests in PD mice, accompanied by a decrease in the loss of dopaminergic neurons in the SN and increased TH expression in the striatum. Neuroinflammation-related factors, such as GFAP, Iba-1, TNF-α and IL-1β were also suppressed in PD mice after VNS compared to those without treatment. Furthermore, the proportion of c-Fos-positive noradrenergic neurons in the LC increased when animals received VNS. Additionally, the expression of the adrenergic receptor of α1BR was also upregulated after VNS compared to PD mice. In conclusion, VNS has potential as a novel PD therapy for neuroprotective effects, and indicate that activation of norepinephric neurons in LC may plays an important role in VNS treatment for PD.
Collapse
Affiliation(s)
- Wen Cheng
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Kexin Fang
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Xiaorong Ouyang
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Lingjing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Yunping Song
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China.
| | - Bin Yu
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
46
|
Cantero-Fortiz Y, Boada M. The role of inflammation in neurological disorders: a brief overview of multiple sclerosis, Alzheimer's, and Parkinson's disease'. Front Neurol 2024; 15:1439125. [PMID: 39539666 PMCID: PMC11558529 DOI: 10.3389/fneur.2024.1439125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroinflammation is a central feature in the pathophysiology of several neurodegenerative diseases, including MS, AD, and PD. This review aims to synthesize current research on the role of inflammation in these conditions, emphasizing the potential of inflammatory biomarkers for diagnosis and treatment. We highlight recent findings on the mechanisms of neuroinflammation, the utility of biomarkers in disease differentiation, and the implications for therapeutic strategies. Advances in understanding inflammatory pathways offer promising avenues for developing targeted interventions to improve patient outcomes. Future research should focus on validating these biomarkers in larger cohorts and integrating them into clinical practice to enhance diagnostic accuracy and therapeutic efficacy.
Collapse
Affiliation(s)
- Yahveth Cantero-Fortiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
47
|
Xie F, Shen B, Luo Y, Zhou H, Xie Z, Zhu S, Wei X, Chang Z, Zhu Z, Ding C, Jin K, Yang C, Batzu L, Chaudhuri KR, Chan LL, Tan EK, Wang Q. Repetitive transcranial magnetic stimulation alleviates motor impairment in Parkinson's disease: association with peripheral inflammatory regulatory T-cells and SYT6. Mol Neurodegener 2024; 19:80. [PMID: 39456006 PMCID: PMC11515224 DOI: 10.1186/s13024-024-00770-4] [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: 08/24/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) has been used to treat various neurological disorders. However, the molecular mechanism underlying the therapeutic effect of rTMS on Parkinson's disease (PD) has not been fully elucidated. Neuroinflammation like regulatory T-cells (Tregs) appears to be a key modulator of disease progression in PD. If rTMS affects the peripheral Tregs in PD remains unknown. METHODS Here, we conducted a prospective clinical study (Chinese ClinicalTrials. gov: ChiCTR 2100051140) involving 54 PD patients who received 10-day rTMS (10 Hz) stimulation on the primary motor cortex (M1) region or sham treatment. Clinical and function assessment as well as flow cytology study were undertaken in 54 PD patients who were consecutively recruited from the department of neurology at Zhujiang hospital between September 2021 and January 2022. Subsequently, we implemented flow cytometry analysis to examine the Tregs population in spleen of MPTP-induced PD mice that received rTMS or sham treatment, along with quantitative proteomic approach reveal novel molecular targets for Parkinson's disease, and finally, the RNA interference method verifies the role of these new molecular targets in the treatment of PD. RESULTS We demonstrated that a 10-day rTMS treatment on the M1 motor cortex significantly improved motor dysfunction in PD patients. The beneficial effects persisted for up to 40 days, and were associated with an increase in peripheral Tregs. There was a positive correlation between Tregs and motor improvements in PD cases. Similarly, a 10-day rTMS treatment on the brains of MPTP-induced PD mice significantly ameliorated motor symptoms. rTMS reversed the downregulation of circulating Tregs and tyrosine hydroxylase neurons in these mice. It also increased anti-inflammatory mediators, deactivated microglia, and decreased inflammatory cytokines. These effects were blocked by administration of a Treg inhibitor anti-CD25 antibody in MPTP-induced PD mice. Quantitative proteomic analysis identified TLR4, TH, Slc6a3 and especially Syt6 as the hub node proteins related to Tregs and rTMS therapy. Lastly, we validated the role of Treg and rTMS-related protein syt6 in MPTP mice using the virus interference method. CONCLUSIONS Our clinical and experimental studies suggest that rTMS improves motor function by modulating the function of Tregs and suppressing toxic neuroinflammation. Hub node proteins (especially Syt6) may be potential therapeutic targets. TRIAL REGISTRATION Chinese ClinicalTrials, ChiCTR2100051140. Registered 15 December 2021, https://www.chictr.org.cn/bin/project/edit?pid=133691.
Collapse
Affiliation(s)
- Fen Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Bibiao Shen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Hang Zhou
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhenchao Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Xiaobo Wei
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhaohua Zhu
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Changhai Ding
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Chengwu Yang
- Division of Biostatistics and Health Services Research, Department of Population and Quantitative Health Sciences, T. H. Chan School of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Lucia Batzu
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS, UK
| | - K Ray Chaudhuri
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS, UK
| | - Ling-Ling Chan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- 7Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.
- 7Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
48
|
Laguna A, Peñuelas N, Gonzalez-Sepulveda M, Nicolau A, Arthaud S, Guillard-Sirieix C, Lorente-Picón M, Compte J, Miquel-Rio L, Xicoy H, Liu J, Parent A, Cuadros T, Romero-Giménez J, Pujol G, Giménez-Llort L, Fort P, Bortolozzi A, Carballo-Carbajal I, Vila M. Modelling human neuronal catecholaminergic pigmentation in rodents recapitulates age-related neurodegenerative deficits. Nat Commun 2024; 15:8819. [PMID: 39394193 PMCID: PMC11470033 DOI: 10.1038/s41467-024-53168-7] [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: 08/14/2023] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
One key limitation in developing effective treatments for neurodegenerative diseases is the lack of models accurately mimicking the complex physiopathology of the human disease. Humans accumulate with age the pigment neuromelanin inside neurons that synthesize catecholamines. Neurons reaching the highest neuromelanin levels preferentially degenerate in Parkinson's, Alzheimer's and apparently healthy aging individuals. However, this brain pigment is not taken into consideration in current animal models because common laboratory species, such as rodents, do not produce neuromelanin. Here we generate a tissue-specific transgenic mouse, termed tgNM, that mimics the human age-dependent brain-wide distribution of neuromelanin within catecholaminergic regions, based on the constitutive catecholamine-specific expression of human melanin-producing enzyme tyrosinase. We show that, in parallel to progressive human-like neuromelanin pigmentation, these animals display age-related neuronal dysfunction and degeneration affecting numerous brain circuits and body tissues, linked to motor and non-motor deficits, reminiscent of early neurodegenerative stages. This model could help explore new research avenues in brain aging and neurodegeneration.
Collapse
Affiliation(s)
- Ariadna Laguna
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Institut de Neurociències-Autonomous University of Barcelona (INc-UAB), 08193, Cerdanyola del Vallès, Spain
| | - Núria Peñuelas
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Marta Gonzalez-Sepulveda
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alba Nicolau
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Sébastien Arthaud
- CNRS UMR5292, INSERM U1028, Lyon Neuroscience Research Centre (CRNL), SLEEP team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Claude Bernard, Lyon 1, Lyon, France
| | - Camille Guillard-Sirieix
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Marina Lorente-Picón
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Joan Compte
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Lluís Miquel-Rio
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC); Center for Networked Biomedical Research on Mental Health (CIBERSAM), 08036, Barcelona, Spain
- Systems Neuropharmacology Research Group, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036, Barcelona, Spain
| | - Helena Xicoy
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
| | - Jiong Liu
- CNRS UMR5292, INSERM U1028, Lyon Neuroscience Research Centre (CRNL), SLEEP team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Claude Bernard, Lyon 1, Lyon, France
| | - Annabelle Parent
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Thais Cuadros
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Jordi Romero-Giménez
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
| | - Gemma Pujol
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
| | - Lydia Giménez-Llort
- Institut de Neurociències-Autonomous University of Barcelona (INc-UAB), 08193, Cerdanyola del Vallès, Spain
- Department of Psychiatry and Forensic Medicine-Autonomous University of Barcelona (INc-UAB), 08193, Cerdanyola del Vallès, Spain
| | - Patrice Fort
- CNRS UMR5292, INSERM U1028, Lyon Neuroscience Research Centre (CRNL), SLEEP team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Claude Bernard, Lyon 1, Lyon, France
| | - Analia Bortolozzi
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC); Center for Networked Biomedical Research on Mental Health (CIBERSAM), 08036, Barcelona, Spain
- Systems Neuropharmacology Research Group, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036, Barcelona, Spain
| | - Iria Carballo-Carbajal
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Institut de Neurociències-Autonomous University of Barcelona (INc-UAB), 08193, Cerdanyola del Vallès, Spain.
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, 08193, Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain.
| |
Collapse
|
49
|
Moon S, Sarmento CVM, Smirnova IV, Colgrove Y, Lai SM, Lyons KE, Liu W. A pilot randomized clinical trial examining the effects of Qigong on inflammatory status and sleep quality in people with Parkinson's disease. J Bodyw Mov Ther 2024; 40:1002-1007. [PMID: 39593404 PMCID: PMC11602653 DOI: 10.1016/j.jbmt.2024.07.025] [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: 11/28/2022] [Revised: 05/31/2024] [Accepted: 07/07/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Neuroinflammation contributes to degeneration of neurons in people with Parkinson's disease (PD). The concentrations of inflammatory cytokines, IL-1β, IL-6, and TNF-α, are often increased in people with PD and are associated with various non-motor symptoms. Qigong exercise is a mind-body practice which has been used as a rehabilitation intervention for people with PD. A previous study showed a strong association between sleep quality and inflammatory status. This study investigated the effect of Qigong on inflammatory status in people with PD and explored potential relationships between inflammatory status and sleep quality in this population. METHODS The study was a randomized controlled trial. A total of 17 participants completed the 12-week intervention (Qigong (n = 8), sham Qigong (n = 9)). Questionnaires were utilized to assess non-motor symptoms, including sleep quality. Inflammatory cytokines were measured by a high sensitivity antibody-based multiplex assay. RESULTS After the 12-week intervention, a decreasing trend in the concentrations of IL-1β and IL-6 was found in the Qigong group. Moderate to strong correlations were found between changes in IL-1β concentrations and sleep quality. CONCLUSION Inflammation is an important aspect of PD. This study explored the inflammatory status after a mind-body exercise. Further studies need to extend our findings to confirm the effect of Qigong in people with PD.
Collapse
Affiliation(s)
- Sanghee Moon
- Department of Kinesiology, University of New Hampshire, Durham, NH, USA; Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Caio V M Sarmento
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA; Department of Physical Therapy, California State University, Fresno, CA, USA
| | - Irina V Smirnova
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yvonne Colgrove
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sue-Min Lai
- Department of Population Health, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kelly E Lyons
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Wen Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
50
|
Qin X, Li B, Hu B, Huang J, Tian X, Zhang X, Wang Y, Huang W. Rhein inhibits M1 polarization of BV2 microglia through MAPK/IκB signalling pathway and reduces neurotoxicity caused by neuroinflammation. Int J Dev Neurosci 2024; 84:533-545. [PMID: 38858813 DOI: 10.1002/jdn.10352] [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: 11/27/2023] [Revised: 01/29/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Rhein is an anthraquinone compound with anti-inflammatory pharmacological activity. It has been found to play a neuroprotective role in neurological diseases, but the neuroprotective mechanism of rhein remains unclear. METHODS SH-SY5Y cells serving as neuron-like cells and BV2 microglia were used. The toxicity of rhein on BV2 microglia and the viability of SH-SY5Y cells were measured by CCK-8 assay. The mRNA expression and secretion of pro-inflammatory cytokines were detected by qPCR and ELISA. Iba1, CD86 and pathway signalling protein in BV2 microglia were assessed by Western blot and immunofluorescence. Apoptosis of SH-SY5Y cells exposed to neuroinflammation was analysed through flow cytometry. RESULTS Rhein inhibited MAPK/IκB signalling pathways. Further studies revealed that rhein inhibited the production of pro-inflammatory cytokines TNF-α, IL-6, IL-1β and iNOS in BV2 cells and also inhibited the expression of M1 polarization markers Iba1 and CD86 in BV2 cells. Furthermore, rhein reduced the apoptotic rate and restored cell viability of SH-SY5Y cells exposed to neuroinflammation. CONCLUSIONS Our study demonstrated that rhein inhibited microglia M1 polarization via MAPK/IκB signalling pathway and protected nerve cells through suppressing neuroinflammation.
Collapse
Affiliation(s)
- Xin Qin
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Yichang Central People's Hospital, Yichang, China
| | - Bowen Li
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Binbin Hu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juan Huang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xingfu Tian
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinyue Zhang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ye Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Huang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|