1
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [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: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
2
|
Shan S, Cheng D, Li H, Yao W, Kou R, Ji J, Liu N, Zeng T, Zhao X. Short-term PS-NP exposure in early adulthood induces neuronal damage in middle-aged mice via microglia-mediated neuroinflammation. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137615. [PMID: 39978191 DOI: 10.1016/j.jhazmat.2025.137615] [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: 11/17/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Nanoplastics (NPs) are ubiquitous environmental pollutants that have garnered considerable attention for their potential adverse health effects. In this study, male C57BL/6 J mice were orally treated with a mixture of 50-nm and 200-nm polystyrene (PS)-NPs for one week followed by measurements of their neurobehavioral performance and neuronal damage 10 months later. Notably, PS-NPs were detected in the brains of the mice by transmission electron microscopy (TEM) and a nanoscale hyperspectral microscope imaging system 10 months after the PS-NP exposure. The mice exposed to short-term PS-NPs exhibited cognitive dysfunction and anxiety-like symptoms, neuronal damage and synapse loss, and an increase in the number of M1-polarized microglia and A1-reactive astrocytes. Interestingly, the inhibition of microglial activation by minocycline significantly mitigated the PS-NP-induced synapse loss and neuron damage. In vitro studies showed that PS-NPs could be readily internalized by three types of neurovascular unit (NVU) cells, including microglia, astrocytes, and brain microvascular endothelial cells, via multiple pathways. RNA-seq analysis confirmed that microglia-mediated neuronal injury was associated with disturbances in synapse and cell death signaling pathways. Collectively, these findings suggest that short-term PS-NP exposure-induced neuroinflammation in early adulthood may not be resolved naturally but may deteriorate under the interaction of microglia and astrocytes, leading to synapse loss, neuron degeneration, and cognitive dysfunction in middle age. The results of the present study provide important insights into the potential neurological impacts of NPs and suggest that targeting microglia to suppress inflammation might be a potential intervention strategy for neurodegeneration induced by NPs.
Collapse
Affiliation(s)
- Shan Shan
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dong Cheng
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Hui Li
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Wenhuan Yao
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Ruirui Kou
- Experimental Center, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jing Ji
- Experimental Center, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Na Liu
- Experimental Center, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
3
|
Mahnoor, Jahan S, Elahi L, Zakria M, Rabia, Ikram M, Ullah N. Alpha-Linolenic Acid for Mitigating Neuroinflammation and Dopaminergic Neuronal Loss in Parkinson's Disease: Insights From In Vivo and In Silico Studies. Clin Exp Pharmacol Physiol 2025; 52:e70043. [PMID: 40269665 DOI: 10.1111/1440-1681.70043] [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: 01/19/2025] [Revised: 03/05/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterised by dopaminergic neuronal loss and chronic neuroinflammation, leading to significant motor and non-motor deficits. This study explores the therapeutic potential of alpha-linolenic acid (ALA), a known antioxidant and anti-inflammatory agent, in a lipopolysaccharide (LPS)-induced murine model of PD. Male Balb-C mice were divided into control, LPS-treated, LPS + ALA-treated and ALA-only groups. Behavioural assessments, including the pole test, rotarod test and open field test, revealed significant motor impairments in LPS-treated mice. Co-treatment with ALA partially ameliorated motor deficits in LPS-treated mice compared to the healthy control group. However, no direct comparison was made with standard PD treatments such as levodopa. Immunohistochemistry analysis showed a 68% reduction in tyrosine hydroxylase-positive (TH+) neurons in the substantia nigra pars compacta (SNpc) of LPS-treated mice. Notably, ALA co-treatment preserved dopaminergic neurons, demonstrating its neuroprotective effects. Western blotting and ELISA revealed heightened expression of inflammatory mediators, including TNF-α, IL-1β and NF-κB, in LPS-treated mice. ALA treatment significantly reduced these markers, indicating its capacity to mitigate neuroinflammation. Molecular docking analysis revealed moderate binding affinities of ALA to NF-κB (-5.1 kcal/mol), TNF-α (-5.7 kcal/mol) and IL-1β (-3.9 kcal/mol), suggesting possible interactions with key inflammatory pathways. These interactions were comparable to known inhibitors, indicating ALA's potential for neuroprotection. This study highlights the neuroprotective and anti-inflammatory effects of ALA in reducing dopaminergic neuronal loss and mitigating neuroinflammation in an LPS-induced PD model. Although behavioural improvements were moderate, these findings underscore ALA's potential as an adjunct therapeutic candidate for PD and other neurodegenerative diseases. Further research is warranted to explore its translational applications in clinical settings.
Collapse
Affiliation(s)
- Mahnoor
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Sarwat Jahan
- Institute of Pharmaceutical Sciences (IPS), Khyber Medical University, Peshawar, Pakistan
- Department of Pharmacology, Northwest School of Medicine, Hayatabad, Pakistan
| | - Laila Elahi
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Zakria
- Institute of Pharmaceutical Sciences (IPS), Khyber Medical University, Peshawar, Pakistan
| | - Rabia
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Ikram
- Institute of Pharmaceutical Sciences (IPS), Khyber Medical University, Peshawar, Pakistan
- Division of Life Science and Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Najeeb Ullah
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| |
Collapse
|
4
|
Xie J, Wu M, Li L, Zhu L, Hu L, Li Y, Li W. Integrated bioinformatics and experimental verification to dissect the mechanisms and bioactive ingredients of Radix Rehmanniae in treating multiple sclerosis. Biochem Biophys Res Commun 2025; 763:151790. [PMID: 40233432 DOI: 10.1016/j.bbrc.2025.151790] [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: 12/15/2024] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Multiple sclerosis (MS), as a primary cause of nontraumatic disability in young adults, has no effective treatment yet. Radix Rehmanniae (RR), a typical Traditional Chinese Medicine (TCM), is commonly used in MS patients as a most frequent herbal item in TCM formulas. Our recent study demonstrated that RR alleviated neurological deficits in an experimental MS model. However, direct evidence regarding the holistic mechanisms and bioactive components of RR for MS remains unclear. In this study, we employed an integrative strategy combining bioinformatics and experimental validation to profile the holistic mechanisms of RR, identify its bioactive components, and investigate their potential targets in MS. First, a network pharmacology approach was used to construct a "compound-target-pathway" network, indicating the action of RR on MS in a multicomponent-multitarget mode, and predicting Echinacoside and Acteoside as the primary bioactive ingredients. Bioinformatics analyses of transcriptomics and single-cell RNA sequencing based on GSE datasets indicated that oxidative stress and inflammatory/immune regulation in microglia might serve as crucial mechanisms of Echinacoside and Acteoside in MS pathology. Then, in vitro assays validated that Echinacoside and Acteoside possessed anti-inflammatory and antioxidant properties by scavenging ONOO- and H2O2 directly, and suppressing microglia-derived ONOO- production through inhibition of NF-κB-mediated iNOS and NADPH oxidase. In addition, molecular docking showed strong affinities between Acteoside and inflammation-related targets TGF-β and SMAD2. These findings provide the scientific evidence for clinical application of RR and bring novel insights into MS drug development.
Collapse
Affiliation(s)
- Jing Xie
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Li Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Lixia Zhu
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Liang Hu
- School of Integrative Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Wenting Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
5
|
Braga J, Kuik EJY, Lepra M, Rusjan PM, Kish SJ, Vieira EL, Nasser Z, Verhoeff N, Vasdev N, Chao T, Bagby M, Boileau I, Kloiber S, Husain MI, Kolla N, Koshimori Y, Faiz K, Wang W, Meyer JH. Astrogliosis Marker [ 11C]SL25.1188 After COVID-19 With Ongoing Depressive and Cognitive Symptoms. Biol Psychiatry 2025; 97:816-824. [PMID: 39395470 DOI: 10.1016/j.biopsych.2024.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/30/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND After acute COVID-19, 5% of people experience persistent depressive symptoms and reduced cognitive function (COVID-DC). Theoretical models propose that astrogliosis is important in long COVID, but measures primarily indicative of astrogliosis have not been studied in the brain of long COVID or COVID-DC. The objective of the current study was to measure [11C]SL25.1188 total distribution volume ([11C]SL25.1188 VT), an index of monoamine oxidase B density and a marker of astrogliosis, with positron emission tomography in participants with COVID-DC and compare with healthy control participants. METHODS In 21 COVID-DC cases and 21 healthy control participants, [11C]SL25.1188 VT was measured in the prefrontal cortex, anterior cingulate cortex, hippocampus, dorsal putamen, and ventral striatum. Depressive symptoms were measured with the Beck Depression Inventory-II, and cognitive symptoms were measured with neuropsychological tests. RESULTS [11C]SL25.1188 VT was higher in participants with COVID-DC in the prefrontal cortex, anterior cingulate cortex, hippocampus, dorsal putamen, and ventral striatum than in healthy control participants. Depressive symptom severity negatively correlated with [11C]SL25.1188 VT across prioritized brain regions. More recent acute COVID-19 positively correlated with [11C]SL25.1188 VT, reflecting higher values since predominance of the Omicron variant. Exploratory analyses found greater [11C]SL25.1188 VT in the hippocampus, dorsal putamen, and ventral striatum of COVID-DC participants than control participants with a major depressive episode with no history of COVID-19, and there was no relationship to cognitive testing in prioritized regions. CONCLUSIONS Results strongly support the presence of monoamine oxidase B-labeled astrogliosis in COVID-DC throughout the regions assessed, although the association of greater astrogliosis with fewer symptoms raises the possibility of a protective role. The magnitude of astrogliosis in COVID-DC is greater since the emergence of the Omicron variant.
Collapse
Affiliation(s)
- Joeffre Braga
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Emily J Y Kuik
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Mariel Lepra
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Pablo M Rusjan
- Douglas Research Centre and Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Stephen J Kish
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Erica L Vieira
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Zahra Nasser
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Natasha Verhoeff
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Neil Vasdev
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Thomas Chao
- Institute of Mental Health, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Bagby
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Stefan Kloiber
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - M Ishrat Husain
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Kolla
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Waypoint Centre for Mental Health Care, Penetanguishene, Ontario, Canada
| | - Yuko Koshimori
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Khunsa Faiz
- Department of Diagnostic Radiology, Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Wei Wang
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jeffrey H Meyer
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Wen Y, Zeng X, Luo H, Cheng Y, Xing J, Zhao H, Chen H. Structural characterization and anti-neuroinflammatory activity of polysaccharides isolated from the leaves of Perilla frutescens. Int J Biol Macromol 2025:143029. [PMID: 40222528 DOI: 10.1016/j.ijbiomac.2025.143029] [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: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
In this study, the effect of crude polysaccharide (PF50) from the leaves of Perilla frutescens on the levels of inflammatory mediator proteins was evaluated through Western blotting and immunofluorescence analysis. The results showed that PF50 exerted potent anti-neuroinflammatory effects by significantly suppressing the NO release and expression of the proinflammatory cytokine mediators. Importantly, the PF50 concentration is independently protected against lipopolysaccharide (LPS)-triggered microglial activation-mediated neurotoxicity via inhibiting ROS production and improving mitochondrial function. To further investigate the active ingredients of PF50, a novel polysaccharide (PFP50-1) was purified and its anti-inflammatory activity was studied. PFP50-1 is composed of → 6) - β - D-Galp - (1 →, → 3,6) - α - D-Man p - (1 →, α - L-Araf - (1 →, → 4,6) - α-D-Glcp - (1 →, → 2,3,4) - α - L-Rhap - (1→ and β - D-Glcp - (1 →, which significantly reduced the abnormal elevation of proinflammatory cytokines in LPS-induced BV2 cells. Further experiments revealed that PFP50-1 inhibited the expression of proinflammatory mediator proteins including iNOS and COX-2. In summary, PFP50-1 has exhibited markedly anti-neuroinflammatory effects and may be one of the biologically active ingredients in PF50 for its anti-neuroinflammation and neuroprotection.
Collapse
Affiliation(s)
- Yao Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Xiang Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hongting Luo
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yu Cheng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jieyu Xing
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huan Zhao
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Haiyun Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
7
|
Hu X, Wu J, Shi L, Wang F, He K, Tan P, Hu Y, Yang Y, Wang D, Ma T, Ding S. The transcription factor MEF2C restrains microglial overactivation by inhibiting kinase CDK2. Immunity 2025; 58:946-960.e10. [PMID: 40139186 DOI: 10.1016/j.immuni.2025.02.026] [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: 11/29/2023] [Revised: 09/14/2024] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Microglial intrinsic immune checkpoints are essential safeguards to maintain immune homeostasis by preventing microglial overactivation, a process that substantially influences neurological disorders such as autism spectrum disorder (ASD). MEF2C is a crucial immune checkpoint that regulates microglial activation, but the mechanism remains unclear. We found that MEF2C-deficient (MEF2C-/-) induced microglia-like cells (iMGLs) derived from human pluripotent stem cells (hPSCs) exhibited overactivation following lipopolysaccharide stimulation, mimicking patterns observed in various neuroinflammatory disorders. High-throughput screening identified BMS265246, a cyclin-dependent kinase 2 (CDK2) inhibitor, which suppressed overactivation of MEF2C-/- iMGLs and normalized their inflammatory responses. Mechanistically, MEF2C transcriptionally upregulated p21 to inhibit CDK2 activation-mediated retinoblastoma protein (RB) degradation, thereby preventing transcription factor nuclear factor κB (NFκB) nuclear translocation and consequent microglial overactivation. BMS265246 treatment substantially ameliorated microglial overactivation and ASD-like behaviors in Mef2c-deficient mice. Our findings identify the MEF2C-p21-CDK2-RB-NFκB axis as a critical pathway to maintain microglial homeostasis and highlight CDK2 as a potential therapeutic target for neuroinflammation.
Collapse
Affiliation(s)
- Xiaodan Hu
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Jianchen Wu
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lu Shi
- CRE Life Institute, Beijing 100000, China
| | - Folin Wang
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Kezhang He
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Pengcheng Tan
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yanyan Hu
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yuanyuan Yang
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Dan Wang
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Tianhua Ma
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| | - Sheng Ding
- New Cornerstone Science Laboratory, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
8
|
Wang H, Lu X, Ye Y, Huang C, Fang Y, Yang R, Sun M, Ren J, Song R, Xu F, Su J, Hong H, Huang C. Stimulation of microglia leads to a rapid antidepressant effect by triggering astrocytic P2Y1Rs and promoting BDNF-mediated neurogenesis in the hippocampus. Brain Behav Immun 2025; 128:134-151. [PMID: 40194747 DOI: 10.1016/j.bbi.2025.04.010] [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: 02/07/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
Reversing the decline of microglia in the dentate gyrus of stressed animals has antidepressant effects, but the molecular mechanisms are unclear. Since microglia normally interact with astrocytes and astrocytic purinergic 2Y1 receptor (P2Y1R) signaling plays an important role in regulating cellular crosstalk, we hypothesize that astrocytic P2Y1R signaling may mediate the antidepressant effects of microglia stimulation. Our results showed that a single injection of low-dose lipopolysaccharide (LPS) (100 μg/kg) elicited rapid antidepressant effects and a significant increase in adenosine triphosphate (ATP) levels in the dentate gyrus in chronically stressed mice, and that these effects of LPS were abolished by chemogenetic inhibition of microglia. Depletion of endogenous ATP, non-specific antagonization of purinergic receptors, or specific inhibition of P2Y1Rs, but not other purinergic receptors, by MRS2179 in the hippocampus abolished the antidepressant effects of low-dose LPS. Conditional gene knockout data showed that the antidepressant effect of low-dose LPS could not be observed in mice lacking P2Y1Rs in astrocytes but not in forebrain neurons. Chemogenetic inhibition of microglia in the dentate gyrus, specific deletion of P2Y1Rs in astrocytes and the absence of ATP abolished the increase in doublecortin (DCX)+ cells and brain-derived neurotrophic factor (BDNF) induced by a low dose of LPS in the dentate gyrus of stressed mice, and infusion of BDNF antibodies into the hippocampus simultaneously abolished the pro-neurogenesis and antidepressant effects of microglia stimulation in stressed mice. Taken together, these results suggest that ATP signaling mobilized by microglia stimulation has an antidepressant effect by triggering astrocytic P2Y1R-dependent synthesis of BDNF.
Collapse
Affiliation(s)
- Hanxiao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Ying Ye
- Department of Ultrasound, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001 Jiangsu, China
| | - Chen Huang
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong 226001 Jiangsu, China
| | - Yunli Fang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001 Jiangsu, China
| | - Micona Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Jie Ren
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China
| | - Rongrong Song
- Department of Emergency and Critical Care Medicine, Tongzhou People's Hospital, #999 Jianshe Road, Nantong 226300 Jiangsu, China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006 Jiangsu, China
| | - Jianbin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006 Jiangsu, China
| | - Hongxiang Hong
- Department of Spine Surgery, Affiliated Hospital 2 of Nantong University, First People's Hospital of Nantong City, #666 Shengli Road, Nantong 226006 Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001 Jiangsu, China.
| |
Collapse
|
9
|
Rambaud V, Frajerman A, Fournier M, Iftimovici A, Dwir D, Khadimallah I, Kebir O, Marzo A, Krebs MO, Chaumette B. Oxidative stress markers during the psychotic transition. J Psychiatr Res 2025; 186:137-144. [PMID: 40239390 DOI: 10.1016/j.jpsychires.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Ultra-high-risk state (UHR) concept was initially applied to promote the early detection of young help-seeking patients with higher risk of psychotic transition. However, most UHR individuals do not evolve to psychosis, stressing the need for biomarkers allowing the prediction of the transition. Substantial evidence suggest that redox dysregulation plays a major role in the pathophysiology of psychotic disorders. The aim of this study is to explore the relationship between the evolution of blood oxidative stress markers in UHR individuals. Blood samples were collected from 48 UHR individuals at their first visit and 12 months later for those who did not convert to psychosis (UHR-NC), or at the time of the transition for the converters (UHR-C). Markers for redox dysregulation, including the glutathione antioxidant system, superoxide dismutase, thioredoxin, TBARS, macrophage migration inhibitory factor, peroxiredoxin-4, MMP9 and sRAGE, were assessed in erythrocytes, serum and plasma. Statistical analyses revealed a combination of peripheral redox markers associated with the risk of transition to psychosis. These markers were able to discriminate between UHR-C and UHR-NC subjects at baseline. A decrease in blood levels of peroxiredoxin-4, an antioxidant enzyme, was associated with a lower risk of transition. GPx activity and TBARS levels were associated with the later severity of symptoms during the course of psychosis. These findings suggest the interest of peripheral biomarkers of oxidative stress to monitor the risk of psychosis. Overall, these findings hold promises for early detection and argue for the development of treatments targeting redox pathways in psychosis.
Collapse
Affiliation(s)
- Victoria Rambaud
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Ariel Frajerman
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France; GHU Paris Psychiatrie et Neurosciences, Paris, France
| | - Margot Fournier
- Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Department of Psychiatry, Lausanne, Switzerland
| | - Anton Iftimovici
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France; GHU Paris Psychiatrie et Neurosciences, Paris, France
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Department of Psychiatry, Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Department of Psychiatry, Lausanne, Switzerland
| | - Oussama Kebir
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France; GHU Paris Psychiatrie et Neurosciences, Paris, France
| | - Aude Marzo
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Marie-Odile Krebs
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France; GHU Paris Psychiatrie et Neurosciences, Paris, France
| | - Boris Chaumette
- Université Paris Cité, INSERM U1266, Institute of Psychiatry and Neuroscience of Paris, Paris, France; GHU Paris Psychiatrie et Neurosciences, Paris, France; Department of Psychiatry, McGill University, Montreal, France.
| |
Collapse
|
10
|
Alonaizan R, K Alotaibi W, Alsulami A, M Alkhulaifi F, Alomar S. Sex-Differences Influence Depressive-Like Behaviour via Alterations in Microglial Expression of GIF-1, TREM2, and IL-1β in an Acute Lipopolysaccharide-Induced Murine Neuroinflammation Model. Immunol Invest 2025; 54:317-333. [PMID: 39701694 DOI: 10.1080/08820139.2024.2440006] [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: 12/21/2024]
Abstract
BACKGROUND Neurodegenerative diseases (NDs) have caused serious health issues worldwide. A growing body of evidence suggests a correlation between neuroinflammation and abnormal microglial activity with ND symptoms. Microglia survey play crucial roles in CNS during health and the injury. It is proposed that sex affects microglial roles during inflammation, resulting in mouse behavioural changes and expression alterations in key markers related to microglia functions. METHODS Male and female C57BL/6 mice were injected with a single dose of LPS (5 mg/kg, i.p.) or saline. After 48 h, an open field test was conducted, followed by brain tissues collection for measuring the expression of IGF-1, IL-1β and TREM2 and Immunohistochemistry (IHC) analysis for NLRP3 level. RESULTS Males displayed greater depressive-like behaviour in the OFT, with lower levels of IGF-1, IL-1β, and NLRP3 and high TREM2 expression. Female mice did not exhibit this behaviour, in contrast to male mice, they exhibited increased IL-1β and NLRP3 expression. DISCUSSION This study revealed that LPS-induced sex-specific changes in genes involved in neuronal cell survival caused behavioural alterations in male mice. Moreover, females had observed inflammatory responses that had no impact on behavioural alterations. Overall, both sexes exhibited sex-specific microglial activation states.
Collapse
Affiliation(s)
- Rasha Alonaizan
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Wafa K Alotaibi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Asma Alsulami
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Fadwa M Alkhulaifi
- Biology Department, College of Science, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Suliman Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Song J, Wang T, Hong JS, Wang Y, Feng J. TFEB-dependent autophagy-lysosomal pathway is required for NRF2-driven antioxidative action in obstructive sleep apnea-induced neuronal injury. Cell Signal 2025; 128:111630. [PMID: 39875050 PMCID: PMC11913475 DOI: 10.1016/j.cellsig.2025.111630] [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: 10/24/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Nearly one billion individuals worldwide suffer from obstructive sleep apnea (OSA) and are potentially impacted by related neurodegeneration. TFEB is considered a master regulator of autophagy and lysosomal biogenesis, but little is known about its role in neuronal oxidative stress and resultant injury induced by OSA. This study aimed to investigate these issues. Here, we demonstrated that neuronal TFEB induction is repressed in OSA mouse models. Activation of a TFEB-dependent autophagy-lysosomal pathway (ALP) reduces hippocampal neuronal cell death and mitigates OSA-related cognitive impairment. Neuronal NRF2 induction was also found to be defective in OSA mouse models. A series of staining assays for HO1, SOD3, ROS, GSH, 8-OHdG, MDA and PI revealed that enhancement of NRF2 expression restores neuronal redox balance and protects hippocampal neurons. We then identified a novel interplay between TFEB-dependent ALP and NRF2-mediated relief of oxidative stress. Inhibition of NRF2 hinders TFEB expression and lysosomal biogenesis. Conversely, knockdown of TFEB or blocking autophagy dampens the antioxidative effect of NRF2. Our findings highlight the unexpected and crucial role of TFEB-dependent ALP as a downstream event of NRF2 in NRF2-promoted redox balance. This study provides novel insights into the mechanism behind NRF2-driven antioxidative action and the regulation of TFEB-dependent ALP.
Collapse
Affiliation(s)
- Junxiu Song
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China
| | - Tian Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China; Respiratory Department, Cangzhou People's Hospital, 061000, Hebei, China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC 27709, USA
| | - Yubao Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China.
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin Medical University, 300052 Tianjin, China.
| |
Collapse
|
12
|
Costa-Ferro ZSM, Cunha RS, Rossi EA, Loiola EC, Cipriano BP, Figueiredo JCQ, da Silva EA, de Lima AVR, de Jesus Ribeiro AM, Moitinho Junior VS, Adanho CSA, Nonaka CKV, Silva AMDS, da Silva KN, Rocha GV, De Felice FG, do Prado-Lima PAS, Souza BSDF. Extracellular vesicles derived from mesenchymal stem cells alleviate depressive-like behavior in a rat model of chronic stress. Life Sci 2025; 366-367:123479. [PMID: 39983828 DOI: 10.1016/j.lfs.2025.123479] [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: 09/12/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Depression is a prevalent chronic psychiatric disorder with a growing impact on global health. Current treatments often fail to achieve full remission, highlighting the need for alternative therapeutic strategies. Mesenchymal stem cells (MSCs) have attracted significant interest for their therapeutic potential in neuropsychiatric disorders, primarily due to their capacity to target neuroinflammation. This study aimed to investigate if extracellular vesicles derived from human umbilical MSCs (hucMSCs) promote behavioral beneficial actions in a rat model of chronic unpredictable mild stress (CUMS). We show that a single dose of hucMSCs or their derived EVs (hucMSC-EVs) via the tail vein alleviated depressive-like behavior in rats, reduced markers of neuroinflammation, reduced pro-inflammatory cytokines (IL-1β and TNF-α), and increased the number and dendritic complexity of DCX-positive cells in the dentate gyrus. Proteomic analysis of EVs revealed the presence of proteins involved in modulation of inflammatory processes and cell activation. Our study demonstrates EVs derived from hucMSCs can effectively mitigate depressive symptoms by modulating neuroinflammatory pathways and enhancing neurogenesis. These findings support further exploration of MSC-derived EVs as a novel therapeutic option for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zaquer Suzana Munhoz Costa-Ferro
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil
| | - Rachel Santana Cunha
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil
| | - Erik Aranha Rossi
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil; Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Erick Correia Loiola
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil
| | - Barbara Porto Cipriano
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil
| | - Júlio César Queiroz Figueiredo
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil
| | - Elisama Araújo da Silva
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil
| | - Adne Vitória Rocha de Lima
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil
| | - Adlas Michel de Jesus Ribeiro
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil
| | | | - Corynne Stephanie Ahouefa Adanho
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil
| | - Carolina Kymie Vasques Nonaka
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil
| | | | - Kátia Nunes da Silva
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil
| | - Gisele Vieira Rocha
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil
| | - Fernanda Guarino De Felice
- D'OR Institute for Research and Education, Rio de Janeiro, Brazil; Centre for Neuroscience Studies, Departments of Biomedical and Molecular Sciences & Psychiatry, Queen's University, Kingston, ON, Canada; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil; Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil; D'Or Institute for Research and Education, Salvador, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, Brazil; Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Xiao Y, He Y, Zhong D, Liu B, Tang Z, Lan X, Dong Y, Du H, Liu Y, Luo J. Effect of Engineered Cyanobacterial Capsules on a Neurogenic Bladder after Spinal Cord Injury. ACS NANO 2025; 19:11841-11860. [PMID: 40116782 DOI: 10.1021/acsnano.4c14140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
The presence of a neurogenic bladder is a severe but common complication of spinal cord injury (SCI). Multiple pathological factors, such as hypoxia, ischemia, and oxidative stress caused by SCI, promote M1 microglial polarization and the release of proinflammatory factors to amplify inflammation. An excessive inflammatory response stimulates the generation of reactive oxygen species (ROS) and induces oxidative stress to promote neuronal ferroptosis, thus leading to bladder dysfunction after SCI. Therefore, promoting the recovery of neural function by regulating the interaction between microglia and neurons is important. For this purpose, we developed an engineered immunoregulatory cyanobacterial capsule named siRNA@Cyanzyme, which consists of MnO2@zeolitic-imidazolate framework@cyanobacteria (Cyanzyme) and a small-interfering RNA targeting ACSL4 (siRNA-ACSL4). Cyanzyme reversed M1 microglial polarization via photosynthetic oxygen to promote anti-inflammatory factor release. MnO2 nanoenzymes grown on the surface of ZIF-8 eliminated excessive ROS to reduce oxidative stress. Moreover, Cyanzyme increased the delivery efficiency of siRNA-ACSL4, which is a key regulator of ferroptosis. Both treatments alleviated GABAergic neuron damage to mitigate bladder dysfunction. Our data demonstrated that siRNA@Cyanzyme effectively reversed M1 microglial polarization, reduced neuronal ferroptosis, and ultimately restored neurogenic bladder function.
Collapse
Affiliation(s)
- Yuhong Xiao
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yizhe He
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Da Zhong
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330006, P.R. China
| | - Bo Liu
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - ZhiBo Tang
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaoyong Lan
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - YiYang Dong
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huixian Du
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yu Liu
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Luo
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Ors H, Alimogullari E, Aslan Erdem S, Elmazoglu Z, Ceylan AF. Rosmarinus officinalis Ethanolic Extracts Rescues BV-2 Cells via Modulating Inflammation and Redox Balance: Comparative Study With Carnosol and Carnosic Acid. Cell Biochem Funct 2025; 43:e70073. [PMID: 40219627 PMCID: PMC11992469 DOI: 10.1002/cbf.70073] [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: 06/06/2024] [Revised: 03/03/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Neuroinflammation generally refers to an inflammatory response within the central nervous system caused by various pathological insults, including infection, trauma, ischemia, and toxins. As the brain's sentinel immune cell, microglia are tasked as the first responders to infection or tissue injury and initiating an inflammatory response. The perennial shrub plant Rosmarinus officinalis L. was reported to possess anti-inflammatory, anticancer, anti-nociceptive, antidiabetic, neuroprotective, and antioxidative properties. The present study aimed to investigate the effects of Rosmarinus officinalis ethanolic extracts on the lipopolysaccharide (LPS)-induced neuroinflammation model of BV-2 cells in comparison to carnosol and carnosic acid, phenolic diterpenes of the plant. Ultrasound-assisted extraction was used to have ethanolic extract of the plant. LPS was used to induce inflammation in BV-2 cells. Tumor necrosis alpha (TNF-α), interleukin 1 beta (IL-1β) secretion, reactive oxygen species (ROS) production, GSH/GSSG ratio, protein carbonyl level, and caspase-3 activity were evaluated. Inflammation induced by LPS was reduced by the ethanolic extract. Both carnosol and carnosic acid decreased the TNF-α and IL-1β levels as well. The ethanolic extract reduced ROS production and protein carbonylation, and increased GSH/GSSG ratio more effectively compared to the effects of carnosol and carnosic acid. Results depicted that caspase-3 activity was reduced by the ethanolic extract and this effect was more pronounced compared to carnosol and carnosic acid. The present study indicates the ethanolic extract of Rosmarinus officinalis rescues BV-2 cells from apoptosis via alleviating inflammation and oxidative stress.
Collapse
Affiliation(s)
- Hatice Ors
- Faculty of Medicine, Department of Medical PharmacologyAnkara Yildirim Beyazit UniversityAnkaraTurkey
| | - Ebru Alimogullari
- Faculty of Medicine, Department of Histology and EmbryologyAnkara Yildirim Beyazit UniversityAnkaraTurkey
| | - Sinem Aslan Erdem
- Faculty of Pharmacy, Department of PharmacognosyAnkara UniversityAnkaraTurkey
| | - Zubeyir Elmazoglu
- Faculty of Pharmacy, Department of PharmacologyAnkara Medipol UniversityAnkaraTurkey
| | - Asli F. Ceylan
- Faculty of Medicine, Department of Medical PharmacologyAnkara Yildirim Beyazit UniversityAnkaraTurkey
| |
Collapse
|
15
|
Zhao H, Huang L, Liu J, Feng M, Liu Y, Li H, Gong S, Chen C, Zeng S, Ren W. A vascular endothelial cell, neuron, and microglia tri-culture model to study hypertension-related depression. Front Cell Neurosci 2025; 19:1553309. [PMID: 40230380 PMCID: PMC11994666 DOI: 10.3389/fncel.2025.1553309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Hypertension-related Depression (HD) is a complex mental disorder that exerts a significant negative impact on patients' quality of life. Previous studies have demonstrated that damages to vascular endothelial and hippocampus are the primary pathological features in HD rats. Under hypertensive conditions, inflammatory cytokines in peripheral blood vessels can induce central nervous system inflammation through penetration of a damaged blood-brain barrier, peripheral immune cells, and neural pathways, damaging the brain and triggering HD. Therefore, interactions between vascular endothelial cells, neurons, and glial cells are critical for the understanding of HD. However, in vivo animal models are often limited by the complexity of intrinsic systems, high inter-individual variability, and stringent ethical regulations. A reliable model that could be easily manipulated is needed for investigating the mechanisms involved in communication between vascular endothelial cells, neurons, and glial cells in HD. We therefore aimed to create a composite tri-culture model consisting of rat aortic endothelial cells (RAECs), neurons, and microglia to study HD. First, RAECs were stimulated with lipopolysaccharide to mimic endothelial injury under hypertensive conditions. Vascular endothelial function and inflammatory levels were assessed using fluorescent probes and enzyme-linked immunosorbent assays. RAECs treated with 1 μg/ml LPS for 24 h had reduced levels of nitric oxide, increased levels of endothelin-1 and inflammatory mediators. These findings are consistent with the endothelial dysfunction and inflammatory responses observed in spontaneously hypertensive rats, which suggests that the lipopolysaccharide-induced RAECs model effectively mimics key pathological features of hypertension-related endothelial injury. Subsequently, the supernatants from lipopolysaccharide-induced RAECs were combined with 200 μM corticosterone and transferred to neuron-microglia co-cultures to simulate damages to hippocampal neuron under HD conditions. To evaluate the features of cells, neuronal viability was measured by CCK-8 and live-dead assays. Nissl staining was used to assess neuronal Nissl bodies, while the levels of inflammatory factors and monoamine neurotransmitters in the culture supernatants were evaluated by enzyme-linked immunosorbent assays. Reactive oxygen species in neurons were visualized by a fluorescent probe, apoptosis was detected using TUNEL assays, and immunofluorescence was used to assess microglial phenotypes and the levels of TLR4 and NF-κB. It was found that neurons in the tri-culture model had reduced viability, higher levels of apoptosis, fewer Nissl bodies, increased inflammation, and reduced levels of monoamine neurotransmitters. Additionally, the number of M1 microglia was increased, along with elevated levels of TLR4 and NF-κB proteins. These findings were similar to damages of hippocampal neuron, abnormal levels of monoamine neurotransmitters, microglia polarization, and hippocampal inflammatory response observed in the HD rat model. In conclusion, our findings indicate that the tri-culture model can effectively simulate the pathological characteristics of HD, especially in vascular endothelial damage, neuroinflammation, monoamine neurotransmitters disorders. Therefore, the tri-culture model would provides a reliable and invaluable experimental tool for further research on the pathogenesis and treatment of HD.
Collapse
Affiliation(s)
- Hongxia Zhao
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lingge Huang
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Jian Liu
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Min Feng
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Yeqian Liu
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Hong Li
- Department of Pharmacy, The Second People’s Hospital of Anhui, Hefei, Anhui, China
| | - Shan Gong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chunming Chen
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Shuiqing Zeng
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Weiqiong Ren
- Department of Pharmacy, The First Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
16
|
Massaro A, Calvi P, Restivo I, Giardina M, Mulè F, Tesoriere L, Amato A, Nuzzo D, Picone P, Terzo S, Allegra M. Kumquat Fruit Administration Counteracts Dysmetabolism-Related Neurodegeneration and the Associated Brain Insulin Resistance in the High-Fat Diet-Fed Mice. Int J Mol Sci 2025; 26:3077. [PMID: 40243721 PMCID: PMC11988715 DOI: 10.3390/ijms26073077] [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: 02/20/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic disorders and brain insulin resistance (IR) are major risk factors for the development of neurodegenerative conditions. Kumquat fruit (KF) administration has demonstrated significant anti-dysmetabolic effects, improving peripheral IR in murine models of metabolic syndrome. Along these lines, this study evaluated the neuroprotective effects of KF supplementation in a model of dysmetabolism-induced neuronal damage and its ability to counteract the disruption of brain insulin signalling. To this end, biochemical and histological analysis assessed neuroapoptosis, disruption of brain insulin signalling and neuroinflammation in a model of high-fat diet (HFD)-induced neuronal damage. Our findings demonstrate, for the first time, that KF supplementation significantly counteracts HFD-induced neuroapoptosis downregulating pro-apoptotic genes (FAS-L, BIM and P27) and upregulating the anti-apoptotic ones (BDNF and BCL-2). Coherently, KF positively influenced the expression of selected genes related to Alzheimer's Disease. Relevantly, these effects were associated to KF ability to restore brain insulin signalling by increasing insulin receptor expression, reducing IRS-1 serine phosphorylation, enhancing both AKT activation and GSK-3β inactivation. Accordingly, KF suppressed HFD-neuroinflammation, counteracting the overexpression of NF-κB and its downstream enzymatic products, iNOS and COX-2. Collectively, these findings demonstrate the neuroprotective benefits of KF administration, supporting its potential as a dietary intervention for dysmetabolic-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Alessandro Massaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Pasquale Calvi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Ignazio Restivo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Marta Giardina
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Flavia Mulè
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Luisa Tesoriere
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Antonella Amato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Domenico Nuzzo
- Institute for Biomedical Research and Innovation—IRIB, 90146 Palermo, Italy; (D.N.); (P.P.)
| | - Pasquale Picone
- Institute for Biomedical Research and Innovation—IRIB, 90146 Palermo, Italy; (D.N.); (P.P.)
| | - Simona Terzo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| | - Mario Allegra
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (P.C.); (I.R.); (M.G.); (F.M.); (L.T.); (A.A.); (M.A.)
| |
Collapse
|
17
|
Zhuang C, Yan H, Lu J, Zhou Y, Liu Y, Shi G, Li Y. Compensatory enhancement of orexinergic system functionality induced by amyloid-β protein: a neuroprotective response in Alzheimer's disease. Front Physiol 2025; 16:1529981. [PMID: 40196718 PMCID: PMC11973307 DOI: 10.3389/fphys.2025.1529981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Background Amyloid-β protein (Aβ) accumulation is a defining characteristic of Alzheimer's disease (AD), resulting in neurodegeneration and a decline in cognitive function. Given orexin's well-documented role in enhancing memory and cognition, this study investigates its potential to regulate Aβ-induced neurotoxicity, offering new perspectives into AD management. Methods This paper simulated Aβ accumulation in the hippocampus of AD patients by administering Aβ1-42 oligomers into the bilateral hippocampal dentate gyrus of ICR mice. Inflammatory cytokines (IL-6, TNF-α) and orexin-A levels were measured by ELISA. Additionally, the excitability of orexinergic neurons was assessed by IHC targeting c-Fos expression. These methodologies evaluated the Aβ-induced neuroinflammation, orexinergic system functionality, and dexamethasone's (Dex) effects on these processes. Results Injection of Aβ1-42 oligomer resulted in elevated levels of IL-6, TNF-α, and orexin-A in the hippocampus, as well as increased excitability of orexinergic neurons in the lateral hypothalamus (LH). Dex treatment reduced neuroinflammation, causing a reduction in orexin-A levels and the excitability of orexinergic neurons. Conclusion Aβ-induced neuroinflammation is accompanied by enhanced levels of orexin-A and orexinergic neuron excitability. These findings suggest that the enhanced functionality of the orexinergic system may become a compensatory neuroprotective mechanism to counteract neuroinflammation and enhance cognitive function.
Collapse
Affiliation(s)
- Chenyu Zhuang
- Medical College, Yangzhou University, Yangzhou, China
| | - Hengyu Yan
- Medical College, Yangzhou University, Yangzhou, China
| | - Jiayu Lu
- Medical College, Yangzhou University, Yangzhou, China
| | - Yifan Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Yanqing Liu
- Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Guoshan Shi
- Department of Basic Medical Sciences, Guizhou University of Chinese Medicine, Guiyang, China
| | - Yan Li
- Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Department of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
18
|
Gong J, Li J, Li J, He A, Ren B, Zhao M, Li K, Zhang Y, He M, Liu Y, Wang Z. Impact of Microglia-Derived Extracellular Vesicles on Resident Central Nervous System Cell Populations After Acute Brain Injury Under Various External Stimuli Conditions. Mol Neurobiol 2025:10.1007/s12035-025-04858-w. [PMID: 40126599 DOI: 10.1007/s12035-025-04858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Acute brain injuries (ABI) caused by various emergencies can lead to structural and functional damage to brain tissue. Common causes include traumatic brain injury, cerebral hemorrhage, ischemic stroke, and heat stroke. Globally, ABI represent a significant portion of neurosurgical cases. Previous studies have emphasized the significant therapeutic potential of stem cell-derived extracellular vesicles (EVs). Recent research indicates that EVs extracted from resident cells in the central nervous system (CNS) also show therapeutic potential following brain injury. Microglia, as innate immune cells of the CNS, respond to changes in the internal environment by altering their phenotype and secreting EVs that impact various CNS cells, including neurons, astrocytes, oligodendrocytes, endothelial cells, neural stem cells (NSCs), and microglia themselves. Notably, under different external stimuli, microglia can either promote neuronal survival, angiogenesis, and myelin regeneration while reducing glial scarring and inflammation, or they can exert opposite effects. This review summarizes and evaluates the current research findings on how microglia-derived EVs influence various CNS cells after ABI under different external stimuli. It analyzes the interaction mechanisms between EVs and resident CNS cells and discusses potential future research directions and clinical applications.
Collapse
Affiliation(s)
- Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Anqi He
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingcheng Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Kexin Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuchi Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mengyao He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| |
Collapse
|
19
|
Li Z, Zhang J, Zhang X, Jin Q, Zheng X, Mo L, Da Z. Oxygen metabolism abnormalities and high-altitude cerebral edema. Front Immunol 2025; 16:1555910. [PMID: 40176814 PMCID: PMC11961428 DOI: 10.3389/fimmu.2025.1555910] [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: 01/05/2025] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
Hypobaric hypoxia is widely recognized as a prominent risk factor for high-altitude cerebral edema (HACE), which contributes to the exacerbation of multiple pathological mechanisms, including oxidative stress, mitochondrial dysfunction, disruption of blood-;brain barrier integrity, neuroinflammation, and neuronal apoptosis. Among these mechanisms, abnormalities in oxygen metabolism, including hypoxia, oxidative stress, and mitochondrial dysfunction, play pivotal roles in the pathophysiology of HACE. In this review, our objective is to enhance our comprehension of the underlying molecular mechanisms implicated in HACE by investigating the potential involvement of oxygen metabolism. Addressing aberrations in oxygen metabolism holds promise for providing innovative therapeutic strategies for managing HACE.
Collapse
Affiliation(s)
- Zhi Li
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Jianping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaoxia Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Qiaoying Jin
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Xingxing Zheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Li Mo
- Department of Ophthalmology, Minxian People’s Hospital, Minxian, Gansu, China
| | - Zejiao Da
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
20
|
Saad HM, Atef E, Elsayed AE. New Insights on the Potential Role of Pyroptosis in Parkinson's Neuropathology and Therapeutic Targeting of NLRP3 Inflammasome with Recent Advances in Nanoparticle-Based miRNA Therapeutics. Mol Neurobiol 2025:10.1007/s12035-025-04818-4. [PMID: 40100493 DOI: 10.1007/s12035-025-04818-4] [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: 09/25/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Parkinson's disease (PD) is a widespread neurodegenerative disorder characterized by the gradual degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc). This review aims to summarize the recent advancements in the pathophysiological mechanisms of pyroptosis, mediated by NLRP3 inflammasome, in advancing PD and the anti-pyroptotic agents that target NLRP3 inflammatory pathways and miRNA. PD pathophysiology is primarily linked to the aggregation of α-synuclein, the overproduction of reactive oxygen species (ROS), and the development of neuroinflammation due to microglial activation. Prior research indicated that a significant quantity of microglia is activated in both PD patients and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse models, triggering neuroinflammation and resulting in a cascade of cellular death. Microglia possess an inflammatory complex pathway termed the nucleotide-binding oligomerization domain-, leucine-rich repeat, and pyrin domain-containing 3 (NLRP3) inflammasome. Activation of the NLRP-3 inflammasome results in innate cytokines maturation, including IL-18 and IL-1β, which initiates the neuroinflammatory signal and induces a type of inflammatory cell death known as pyroptosis. Upon neuronal damage, intracellular levels of damage-associated molecular patterns (DAMPs), including reactive oxygen species (ROS), would build. DAMPs induce unregulated cell death and subsequent release of oxidative intermediates and pro-inflammatory cytokines, leading to the progression of PD. Thus, targeting of neuroinflammation using antipyroptotic medications can be efficiently achieved by blocking NLRP3 and obstructing IL-1β signaling and release. Furthermore, many research studies showed that miRNAs have been identified as regulators of the NLRP3 inflammasome and Nrf2 signal, which subsequently modulate the NLRP3-Nrf2 axis in PD. Nanotechnology promises potential for the advancement of miRNA-based therapies. Nanoparticles that ensure miRNA stability, traverse the blood-brain barrier (BBB) and distribute miRNA targeting regions needed to be created. In conclusion, targeting the pyroptosis pathway via NLRP3 or miRNA may serve as a prospective therapeutic strategy for PD in the future.
Collapse
Affiliation(s)
- Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Esraa Atef
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom, 32511, Egypt
| | - Abeer E Elsayed
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt
| |
Collapse
|
21
|
Dalrymple AN, Jones ST, Fallon JB, Shepherd RK, Weber DJ. Overcoming failure: improving acceptance and success of implanted neural interfaces. Bioelectron Med 2025; 11:6. [PMID: 40083033 PMCID: PMC11907899 DOI: 10.1186/s42234-025-00168-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Implanted neural interfaces are electronic devices that stimulate or record from neurons with the purpose of improving the quality of life of people who suffer from neural injury or disease. Devices have been designed to interact with neurons throughout the body to treat a growing variety of conditions. The development and use of implanted neural interfaces is increasing steadily and has shown great success, with implants lasting for years to decades and improving the health and quality of life of many patient populations. Despite these successes, implanted neural interfaces face a multitude of challenges to remain effective for the lifetime of their users. The devices are comprised of several electronic and mechanical components that each may be susceptible to failure. Furthermore, implanted neural interfaces, like any foreign body, will evoke an immune response. The immune response will differ for implants in the central nervous system and peripheral nervous system, as well as over time, ultimately resulting in encapsulation of the device. This review describes the challenges faced by developers of neural interface systems, particularly devices already in use in humans. The mechanical and technological failure modes of each component of an implant system is described. The acute and chronic reactions to devices in the peripheral and central nervous system and how they affect system performance are depicted. Further, physical challenges such as micro and macro movements are reviewed. The clinical implications of device failures are summarized and a guide for determining the severity of complication was developed and provided. Common methods to diagnose and examine mechanical, technological, and biological failure modes at various stages of development and testing are outlined, with an emphasis on chronic in vivo characterization of implant systems. Finally, this review concludes with an overview of some of the innovative solutions developed to reduce or resolve the challenges faced by implanted neural interface systems.
Collapse
Affiliation(s)
- Ashley N Dalrymple
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
- Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, USA.
- NERVES Lab, University of Utah, Salt Lake City, UT, USA.
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Sonny T Jones
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- NERVES Lab, University of Utah, Salt Lake City, UT, USA
| | - James B Fallon
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
- Medical Bionics Department, University of Melbourne, Melbourne, VIC, Australia
| | - Robert K Shepherd
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Mu J, Zhang Z, Jiang C, Geng H, Duan J. Role of Tau Protein Hyperphosphorylation in Diabetic Retinal Neurodegeneration. J Ophthalmol 2025; 2025:3278794. [PMID: 40109357 PMCID: PMC11922625 DOI: 10.1155/joph/3278794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 12/25/2024] [Accepted: 02/22/2025] [Indexed: 03/22/2025] Open
Abstract
Diabetic retinal neurodegeneration (DRN) is an early manifestation of diabetic retinopathy (DR) characterized by neurodegeneration that precedes microvascular abnormalities in the retina. DRN is characterized by apoptosis of retinal ganglion cells (involves alterations in retinal ganglion cells [RGCs], photoreceptors, amacrine cells and bipolar cells and so on), reactive gliosis, and reduced retinal neuronal function. Tau, a microtubule-associated protein, is a key mediator of neurotoxicity in neurodegenerative diseases, with functions in phosphorylation-dependent microtubule assembly and stabilization, axonal transport, and neurite outgrowth. The hyperphosphorylated tau (p-tau) loses its ability to bind to microtubules and aggregates to form paired helical filaments (PHFs), which further form neurofibrillary tangles (NFTs), leading to abnormal cell scaffolding and cell death. Studies have shown that p-tau can cause degeneration of RGCs in DR, making tau pathology a new pathophysiological model for DR. Here, we review the mechanisms by which p-tau contribute to DRN, including insulin resistance or lack of insulin, mitochondrial damage such as mitophagy impairment, mitochondrial axonal transport defects, mitochondrial bioenergetics dysfunction, and impaired mitochondrial dynamics, Abeta toxicity, and inflammation. Therefore, this article proposes that tau protein hyperphosphorylation plays a crucial role in the pathogenesis of DRN and may serve as a novel therapeutic target for combating DRN.
Collapse
Affiliation(s)
- Jingyu Mu
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Zengrui Zhang
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Chao Jiang
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Haoming Geng
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Junguo Duan
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
- Ineye Hospital of Chengdu University of TCM, Chengdu, Sichuan, China
| |
Collapse
|
23
|
Fu X, Zhang Y, Chen G, Mao G, Tang J, Xu J, Han Y, Chen H, Ding L. Responsive nanoparticles synergize with Curcumin to break the "reactive oxygen Species-Neuroinflammation" vicious cycle, enhancing traumatic brain injury outcomes. J Nanobiotechnology 2025; 23:172. [PMID: 40045354 PMCID: PMC11881390 DOI: 10.1186/s12951-025-03251-y] [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: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Traumatic brain injury (TBI) disrupts oxygen homeostasis in the brain, leading to excessive reactive oxygen species (ROS) production and dysregulated antioxidant mechanisms, which fail to clear excess ROS. This ROS overload promotes the expression of pro-inflammatory genes, releasing cytokines and chemokines and creating a vicious "ROS-neuroinflammation" cycle, making it essential to break this cycle for effective TBI treatment. In this study, we developed cysteine-alanine-glutamine-lysine (CAQK) peptide-modified antioxidant nanoparticles (C-PPS/C) for co-delivery of curcumin (Cur) to modulate oxidative and neuroinflammatory disturbances after TBI. In TBI mice, C-PPS/C nanoparticles accumulated in injured brain regions, where poly (propylene sulfide)120 scavenged ROS, reducing oxidative stress, while Cur release further suppressed ROS and inflammation. C-PPS/C nanoparticles broke the "ROS-neuroinflammation" cycle, protecting the blood-brain barrier (BBB), reducing acute brain edema, and promoting long-term neurological recovery. Further investigation showed that C-PPS/C nanoparticles inhibited the NF-κB pathway, reducing pro-inflammatory gene expression and mitigating inflammation, suggesting a promising approach for TBI treatment.
Collapse
Affiliation(s)
- Xianhua Fu
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Yongkang Zhang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guojie Chen
- Clinical Laboratory, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Guangyao Mao
- Clinical Laboratory, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jiajia Tang
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Jin Xu
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Yuhan Han
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Honglin Chen
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China.
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China.
| |
Collapse
|
24
|
Li J, Liu S, Chen C, Deng J, Du Z, Yang S, Deng H, Zhang Z, Huang Y, Fu J, Zhang W, Poon WS, Hou H, Wang J. The Epileptiogenic Modified Therapy: Regulating the Dynamic of Microglia via ROS-Responsive Cascade Nano-Formulation. Adv Healthc Mater 2025; 14:e2403700. [PMID: 39713896 DOI: 10.1002/adhm.202403700] [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: 09/27/2024] [Revised: 12/13/2024] [Indexed: 12/24/2024]
Abstract
Pharmacological treatment of epilepsy presents several challenges, particularly the ineffectiveness of antiseizure medicines (ASMs) in modifying disease. In fact, the removal of reactive oxygen species (ROS) and preconditioning with tolerable dose of nitric oxide (NO) can activate neuroprotective mechanisms during latency and enhance tolerance to oxidative stress during seizures. To address this, a ROS-responsive cascade Nano-formulation (RRCN) is developed, which will transform ROS into NO. Remarkably, RRCN significantly reduces seizure severity, prolongs seizure latency, and extends inter-seizure intervals, though it does not increase the latency of generalized seizures. Microglia, the primary immune cells of the brain, play a crucial role in the initiation and progression of epilepsy. In the kainic acid (KA) epilepsy model, microglial processes elongate, branching increases, and interactions between microglia and neurons are strengthened in the CA1 and CA3 regions of the hippocampus compared to the Vehicle group. RRCN reverses these dynamic changes in microglia and their interactions with neurons, which are mediated by the NO/HIF/ErBb2 pathway. Thus, RRCN can inhibit seizures generalization by preconditioning the dynamic changes of microglia.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chenghan Chen
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Jiahong Deng
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Zibo Du
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Simin Yang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Hongying Deng
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Zhixia Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Yiyu Huang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Jingwen Fu
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Wangming Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Wai Sang Poon
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jun Wang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
25
|
Li D, Yong Y, Qiao C, Jiang H, Lin J, Wei J, Zhou Y, Li F. Low-Intensity Pulsed Ultrasound Dynamically Modulates the Migration of BV2 Microglia. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:494-507. [PMID: 39632209 DOI: 10.1016/j.ultrasmedbio.2024.11.010] [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: 08/06/2024] [Revised: 10/18/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Low-intensity pulsed ultrasound (LIPUS) is a promising modality for neuromodulation. Microglia are the resident immune cells in the brain and their mobility is critical for maintaining brain homeostasis and alleviating neuroimmune pathologies. However, it is unclear whether and how LIPUS modulates microglial migration in physiological conditions. METHODS Here we examined the in vitro effects of LIPUS on the mobility of BV2 microglia by live cell imaging. Single-cell tracing of BV2 microglia migration was analyzed using ImageJ and Chemotaxis and Migration Tool software. Pharmacological manipulation was performed to determine the key molecular players involved in regulating ultrasound-dependent microglia migration. RESULTS We found that the distance of microglial migration was enhanced by LIPUS with increasing acoustic pressure. Removing the extracellular Ca2+ influx or depletion of intracellular Ca2+ stores suppressed ultrasound-enhanced BV2 migration. Furthermore, we found that blocking the reorganization of actin, or suppressing purinergic signaling by application of apyrase or hemi-channel inhibitors, both diminished ultrasound-induced BV2 migration. LIPUS stimulation also enhanced microglial migration in a lipopolysaccharide (LPS)-induced inflammatory environment. CONCLUSION LIPUS promoted microglia migration in both physiological and inflammatory environments. Calcium, cytoskeleton, and purinergic signaling were involved in regulating ultrasound-dependent microglial mobility. Our study reveals the biomechanical impact of ultrasound on microglial migration and highlights the potential of using ultrasound-based tools for modulation of microglial function.
Collapse
Affiliation(s)
- Dandan Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yu Yong
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Chaofeng Qiao
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China; School of Basic Medical Sciences, Beihua University, Jilin City, China
| | - Hao Jiang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jiawei Lin
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianpeng Wei
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yufeng Zhou
- Chongqing Medical University, State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing City, China
| | - Fenfang Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
26
|
Wang J, Du H, Li M, Yan T, Jia Y. Schisandra chinensis lignans exerts endocannabinoids-like antidepressive effect: The phagocytotic relationship of activated CB2R-mediated M2 microglia and "stressed-but-viable" neuron. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119385. [PMID: 39832627 DOI: 10.1016/j.jep.2025.119385] [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: 10/23/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Schisandrachinensis, a traditional functional Chinese medicine, is known for its ability to tonify the kidneys, calm the heart, and tranquilize the mind. Recent pharmacological research has demonstrated its anti-inflammatory and neuroprotective effects. AIM OF THE STUDY We had previously demonstrated that Schisandra chinensis lignans (SCL) promote microglia polarization to M2 phenotype via targeting cannabinoid receptor type-2 (CB2R) to exert antidepressant effects. Considering the pathological features of abnormal microglial phagocytosis in chronic unpredictable mild stress (CUMS)-induced depression model, the current study aimed to build relationship between microglial phagocytosis and phenotype, further to explore whether SCL exerts antidepression by ameliorating abnormal phagocytotic "stressed-but-viable" neuron by targeting microglial CB2R. MATERIALS AND METHODS Endocannabinoid levels were analyzed using Triple Quadrupole LC/MS. In vivo immunofluorescence assay was employed to evaluate the microglial abnormal phagocytosis. Then, we build models which one was microglia BV2 phagocytized FITC-IgG conjugated latex beads, the other was BV2 co-cultured with stressed-but-viable neurons. Phagocytosis was quantified using flow cytometry. The expression of calreticulin (CRT) in total, intracellular, and surface fractions was validated by Western blot, flow cytometry, and immunofluorescence. The other proteins, such as LRP1, microglial phenotype markers and the PERK-eIF2α pathway, were assessed by Western blot. The qRT-PCR was used to evaluate microglial phenotype markers. Based on the interaction between endocannabinoids and SCL with CB2R, we conducted a CB2R pharmacological antagonist in the CUMS model and used siRNA against CB2R in BV2 cells to verify the findings. RESULTS SCL improved the disrupted levels of endocannabinoids induced by CUMS. In vivo studies revealed that the CB2R antagonist AM630 reversed the SCL-reduced efficiency of microglial mistakenly phagocytosed stressed-but-viable neurons and the up-regulated level of M2 phenotype. In the in vitro studies, we identified SCL activated M2 microglia via CB2R targeting, leading to a reduction in the neuronal cell-surface CRT, inhibition of the "eat-me" signaling, and alleviation abnormal phagocytosis. In-depth investigation performed in the co-culture model revealed that this mechanism involved the inactivation of the PERK-eIF2α pathway in neuronal cells by M2 microglia to exert the above-mentioned effects. CONCLUSION Overall, the improved abnormal phagocytotic process appears to be influenced by SCL and endocannabinoids, promoting microglial polarization toward the M2 phenotype in a CB2R-dependent manner. Specifically, this mechanism involves M2 microglia inactivation of the PERK-eIF2α pathway in stressed-but-viable neurons, thereby reducing CRT translocation to the cell surface and enhancing the regulation of abnormal phagocytosis, ultimately contributing to an antidepressant effect.
Collapse
Affiliation(s)
- Jinyu Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Haoyu Du
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Mengru Li
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
27
|
Wei X, Wang S, Zhang M, Yan Y, Wang Z, Wei W, Tuo H, Wang Z. Alterations of diffusion kurtosis measures in gait-related white matter in the "ON-OFF state" of Parkinson's disease. Chin Med J (Engl) 2025:00029330-990000000-01448. [PMID: 40012092 DOI: 10.1097/cm9.0000000000003486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Gait impairment is closely related to quality of life in patients with Parkinson's disease (PD). This study aimed to explore alterations in brain microstructure in PD patients and healthy controls (HCs) and to identify the correlation of gait impairment in the ON and OFF states of patients with PD, respectively. METHODS We enrolled 24 PD patients and 29 healthy controls (HCs) from the Movement Disorders Program at Beijing Friendship Hospital Capital Medical University between 2019 and 2020. We acquired magnetic resonance imaging (MRI) scans and processed the diffusion kurtosis imaging (DKI) images. Preprocessing of diffusion-weighted data was performed with Mrtrix3 software, using a directional distribution function to track participants' main white matter fiber bundles. Demographic and clinical characteristics were recorded. Quantitative gait and clinical scales were used to assess the status of medication ON and OFF in PD patients. RESULTS The axial kurtosis (AK), mean kurtosis (MK), and radial kurtosis (RK) of five specific white matter fiber tracts, the bilateral corticospinal tract, left superior longitudinal fasciculus, left anterior thalamic radiation, forceps minor, and forceps major were significantly higher in PD patients compared to HCs. Additionally, the MK values were negatively correlated with Timed Up and Go Test (TUG) scores in both the ON and OFF in PD patients. Within the PD group, higher AK, MK, and RK values, whether the patients were ON or OFF, were associated with better gait performance (i.e., higher velocity and stride length). CONCLUSIONS PD exhibits characteristic regional patterns of white matter microstructural degradation. Correlations between objective gait parameters and DKI values suggest that dopamine-responsive gait function depends on preserved white matter microstructure. DKI-based Tract-Based Spatial Statistics (TBSS) analysis may serve as a tool for evaluating PD-related motor impairments (e.g., gait abnormalities) and could yield potential neuroimaging biomarkers.
Collapse
Affiliation(s)
- Xuan Wei
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shiya Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Mingkai Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Yan
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zheng Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wei Wei
- Division of Science and Technology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Houzhen Tuo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
28
|
Sivalingam AM, Sureshkumar DD, Pandurangan V. Cerebellar pathology in forensic and clinical neuroscience. Ageing Res Rev 2025; 106:102697. [PMID: 39988260 DOI: 10.1016/j.arr.2025.102697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/30/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Recent research underscores the cerebellum's growing importance in forensic science and neurology, showing its functions extend beyond motor control, especially in identifying causes of death. Critical neuropathological markers including alpha-synuclein and tau protein aggregates, cellular degeneration, inflammation, and vascular changes are vital for identifying neurodegenerative diseases, injuries, and toxic exposures. Advanced forensic methods, such as Magnetic resonance imaging (MRI), immunohistochemistry, and molecular analysis, have greatly improved the accuracy of diagnoses. Promising new therapies, including neuroprotective agents like resveratrol and transcranial magnetic stimulation (TMS), offer potential in treating cerebellar disorders. The cerebellum's vulnerability to toxins, drugs, and traumatic brain injuries (TBIs) highlights its forensic relevance. Moreover, advancements in genetic diagnostics, such as next-generation sequencing and CRISPR-Cas9, are enhancing the understanding and treatment of genetic conditions like Joubert syndrome and Dandy-Walker malformation. These findings emphasize the need for further research into cerebellar function and its broader significance in both forensic science and neurology.
Collapse
Affiliation(s)
- Azhagu Madhavan Sivalingam
- Natural Products & Nanobiotechnology Research Lab, Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), (Saveetha University), Thandalam, Chennai, Tamil Nadu 602 105, India.
| | - Darshitha D Sureshkumar
- Department of Forensic Science, NIMS Institute of Allied Medical Science and Technology, (NIMS University), Jaipur, Rajasthan 303121, India
| | - Vijayalakshmi Pandurangan
- Department of Radiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), (Saveetha University), Thandalam, Chennai-602 105, Tamil Nadu, India
| |
Collapse
|
29
|
Guo J, Li CG, Mai FY, Liang JR, Chen ZH, Luo J, Zhou MC, Wang YL, Yang WT. Lithospermic acid targeting heat shock protein 90 attenuates LPS-induced inflammatory response via NF-кB signalling pathway in BV2 microglial cells. Immunol Res 2025; 73:54. [PMID: 39969702 PMCID: PMC11839720 DOI: 10.1007/s12026-025-09600-1] [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: 11/20/2024] [Accepted: 01/19/2025] [Indexed: 02/20/2025]
Abstract
Microglia function as a vital constituent in the maintenance of brain homeostasis. Aberrant microglial activation, however, may contribute to neurodegenerative diseases. Lithospermic acid (LA) is a plant-derived polycyclic phenolic carboxylic acid isolated from Salvia miltiorrhiza. The present study investigated the potential effects of lithospermic acid on LPS-induced neuroinflammation in BV2 microglial cells and determined the mechanism of action of this compound. Cells were pre-treated with lithospermic acid for 1 h and incubated with LPS for 24 h. qPCR, immunofluorescence, and immunoblot assays were used to determine the expression of iNOS, COX2, NF-κB p65, and HSP90 expression. ELISA was employed to measure the production of pro-inflammatory cytokines. Lithospermic acid dramatically reduced LPS-stimulated cell migration and decreased NF-κB p65 nuclear translocation. Furthermore, lithospermic acid also markedly decreased the production of pro-inflammatory cytokines, including IL-6, IL-1β, and TNF-α in a dose-dependent manner. Additionally, lithospermic acid inhibited NO and PGE2 production in response to LPS, and it also inhibited the expression of iNOS and COX2 in a dose-dependent manner. Molecular docking and experimental verification have demonstrated that lithospermic acid inhibits the activity and expression of HSP90. Small interfering RNA knockdown of HSP90 expression, which abrogated LPS-induced inflammation. These findings suggest that the lithospermic acid targeting HSP90 attenuates LPS-induced inflammatory response via the NF-κB signalling pathway in BV2 microglial cells. Collectively, lithospermic acid may offer therapeutic benefits for neurodegenerative disorders associated with microglial activation and could serve as a potential inhibitor/agent for the treatment of neuroinflammation.
Collapse
Affiliation(s)
- Jie Guo
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Chen-Guang Li
- Pain Department, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, China
| | - Feng-Yi Mai
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, 518055, China
| | - Jing-Rong Liang
- Pain Department, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, China
| | - Ze-Hao Chen
- Pain Department, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, China
| | - Jiao Luo
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Ming-Chao Zhou
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Yu-Long Wang
- Department of Rehabilitation Medicine, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Wen-Tao Yang
- Pain Department, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, China.
| |
Collapse
|
30
|
Arıcı Sağlıyan G, Çetin FH, Akyürek F, Tok O, Çiçek Zekey Ö, Tezcan ME, Sağlıyan B, Türkoğlu S, Uçar HN, Öztürk B, Altınbaş K. Arginine metabolism and neurocognitive impairment in offspring of bipolar parents: a high-risk case-control study. Front Psychiatry 2025; 16:1511397. [PMID: 40034185 PMCID: PMC11872899 DOI: 10.3389/fpsyt.2025.1511397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction The aim of this study is to investigate whether arginine and its metabolites can be an endophenotype for bipolar disorder (BD) and to evaluate the role of arginine metabolites and neurocognitive function levels in unaffected healthy children of parents diagnosed with BD in cognitive impairment. Methods The study included 37 healthy children of parents diagnosed with BD Type I as the high-risk group and 36 healthy children of parents without any psychiatric disorders as the control group. The arginine, n-monomethyl-l-arginine acetate (L-NMMA), asymmetric dimethylarginine (ADMA), symmetric dimethylarginine (SDMA), citrulline, homoarginine, ornithine serum levels, and nitric oxide synthase (NOS) activity level of both groups were compared. Results The study found that in the high-risk group, ADMA, SDMA, and ornithine levels were significantly higher compared to the controls, while citrulline and NOS activity level were significantly lower in the high-risk group compared to the controls. All neurocognitive performances of the high-risk group were considered statistically significantly worse compared to controls. The impairment in neurocognitive functions in the high-risk group was found to be correlated with ADMA, L-NMMA, citrulline, homoarginine, ornithine levels, and NOS activity level. Discussion These findings highlight a potential link between arginine metabolism and executive dysfunction in individuals at high risk for BD. Further longitudinal studies are essential to fully understand the complex interactions between these factors.
Collapse
Affiliation(s)
| | - Fatih Hilmi Çetin
- Department of Child and Adolescent Psychiatry, Diamind Mental Academy, Konya, Türkiye
| | - Fikret Akyürek
- Department of Medical Biochemistry, Selcuk University Faculty of Medicine, Konya, Türkiye
| | - Oğuzhan Tok
- Department of Medical Biochemistry, Selcuk University Faculty of Medicine, Konya, Türkiye
| | - Özlem Çiçek Zekey
- Department of Child and Adolescent Psychiatry, Sivas Numune Hospital, Sivas, Türkiye
| | - Mustafa Esad Tezcan
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine, Konya, Türkiye
| | - Bilal Sağlıyan
- Department of Psychiatry, Faculty of Medicine, Selcuk University, Konya, Türkiye
| | - Serhat Türkoğlu
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine, Konya, Türkiye
| | - Halit Necmi Uçar
- Department of Child and Adolescent Psychiatry, Diamind Mental Academy, Konya, Türkiye
| | - Bahadır Öztürk
- Department of Medical Biochemistry, Selcuk University Faculty of Medicine, Konya, Türkiye
| | | |
Collapse
|
31
|
Sun J, Lu L, Lian Y, Xu S, Zhu Y, Wu Y, Lin Q, Hou J, Li Y, Yu Z. Sodium butyrate attenuates microglia-mediated neuroinflammation by modulating the TLR4/MyD88/NF-κB pathway and microbiome-gut-brain axis in cardiac arrest mice. Mol Brain 2025; 18:13. [PMID: 39962509 PMCID: PMC11834616 DOI: 10.1186/s13041-025-01179-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: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Cardiac arrest (CA) is one of the most common illnesses worldwide. Post-CA brain injury (PCABI) is a major cause of death and poor recovery in CA patients and the current CA treatments are not very effective. The microbiome-gut-brain axis has been found to significantly affect brain ischemia injury. Furthermore, in ischemic stroke patients, short-chain fatty acids (SCFA), especially sodium butyrate (SB), have been observed to promote neuroprotective effects by modulating inflammatory response and microglial polarization in the cortex. However, the precise mechanism of SB on CA-induced injury remains elusive. Therefore, this research study established an oxygen-glucose deprivation and reoxygenation (OGD/R) model using BV-2 microglial and HT22 cells to simulate cerebral ischemia/reperfusion injury in vitro and a potassium chloride-induced CA mouse model to mimic CA in vivo. The data revealed that SB markedly improved neurological scores and reduced neuronal death and apoptosis. Moreover, it reduced M1 microglia and neuroinflammation in CA mice. In addition, SB increased intestinal integrity and alleviated systemic inflammation. The 16S rDNA sequencing analysis indicated that SB intervention mitigated CA-induced gut microbiota dysbiosis and SCFA depletion. It was also observed that CA mice's brain and OGD/R-exposed BV2 cells had substantially increased levels of MyD88, phosphorylated NF-κB p65, and TLR4 proteins, which were reduced after SB treatment. In summary, this study revealed that SB can protect against cerebral ischemia-reperfusion injury by controlling microglia polarization and microbiome-gut-brain axis to inhibit brain inflammation via the TLR4/MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Jianfei Sun
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liping Lu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
| | - Yingtao Lian
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
| | - Ying Zhu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yanping Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianhui Lin
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Hou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yinping Li
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430060, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, No. 99 ZhangZhidong Road, Wuhan, 430060, Hubei, China.
| |
Collapse
|
32
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [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: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
33
|
Cao ZL, Zhu LX, Wang HM, Zhu LJ. Microglial Regulation of Neural Networks in Neuropsychiatric Disorders. Neuroscientist 2025:10738584251316558. [PMID: 39932233 DOI: 10.1177/10738584251316558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Microglia serve as vital innate immune cells in the central nervous system, playing crucial roles in the generation and development of brain neurons, as well as mediating a series of immune and inflammatory responses. The morphologic transitions of microglia are closely linked to their function. With the advent of single-cell sequencing technology, the diversity of microglial subtypes is increasingly recognized. The intricate interactions between microglia and neuronal networks have significant implications for psychiatric disorders and neurodegenerative diseases. A deeper investigation of microglia in neurologic diseases such as Alzheimer disease, depression, and epilepsy can provide valuable insights in understanding the pathogenesis of diseases and exploring novel therapeutic strategies, thereby addressing issues related to central nervous system disorders.
Collapse
Affiliation(s)
- Zi-Lin Cao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Xia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, China
| | - Hong-Mei Wang
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
34
|
Naderi R, Salimi R, Jafari A, Mehranfard N. Age-dependent increase in apoptosis is associated with dysregulation of miR-92a/Akt/mTOR and NF-κB signaling pathways in male rats. Neurosci Lett 2025; 848:138115. [PMID: 39800254 DOI: 10.1016/j.neulet.2025.138115] [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: 07/20/2024] [Revised: 10/02/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Brain aging is the leading risk factor for most neurodegenerative diseases and has been linked with high rates of neuron loss. Thus, identifying molecular mechanisms underlying neuron loss and pharmacological modulation may be of great importance for slowing or preventing age-related diseases. Herein, we investigated the roles of miR-92a, Akt, mTOR, and NF-κB in age-associated apoptosis in the hippocampus (a critical structure involved in brain aging) of male rats alone and in combination with prazosin. Twenty-four male Wistar rats were grouped into young control (3-month-old), aged (18-month-old), and aged + prazosin groups (n = 8 for each). Prazosin (1 mg/kg; i.p.) was administered for 4 weeks to aged rats. Apoptosis was detected by TUNEL staining. Western blot for Akt, mTOR, and NF-κB was conducted. miR-92a gene expression was performed by using RT-PCR. The results indicated a marked enhancement of apoptosis in the aging hippocampus. We also detected substantial up-regulation of NF-κB as well as substantial down-regulation of phosphorylated-Akt and mTOR in the aging hippocampus. Moreover, miR-92a gene expression was markedly reduced in the aging hippocampus. Treatment with prazosin significantly suppressed apoptosis and reversed miR-92a gene expression, as well as Akt, mTOR, and NF-κB protein expressions in the aging hippocampus. Considering the NF-κB regulatory role on miRNAs, our results suggest that NF-κB may be a negative transcriptional regulator of miR-92a, which in turn could regulate the Akt/mTOR signaling. In this regard, NF-κB upregulation may mediate the downregulation of miR-92a/Akt/mTOR axis, and thereby contribute to age-related neurodegeneration. This may provide a novel treatment target for delaying or preventing age-related problems.
Collapse
Affiliation(s)
- Roya Naderi
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Rahil Salimi
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
35
|
Kong G, Liu J, Wang J, Yu X, Li C, Deng M, Liu M, Wang S, Tang C, Xiong W, Fan J. Engineered Extracellular Vesicles Modified by Angiopep-2 Peptide Promote Targeted Repair of Spinal Cord Injury and Brain Inflammation. ACS NANO 2025; 19:4582-4600. [PMID: 39853366 PMCID: PMC11803916 DOI: 10.1021/acsnano.4c14675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Engineered extracellular vesicles play an increasingly important role in the treatment of spinal cord injury. In order to prepare more effective engineered extracellular vesicles, we biologically modified M2 microglia. Angiopep-2 (Ang2) is an oligopeptide that can target the blood-brain barrier. Through single-cell sequencing and immunofluorescence experiments, we confirmed that the expression of LRP-1, the targeted receptor of Ang2, was elevated after spinal cord injury. Subsequently, we integrated the Ang2 peptide segment into M2 microglia to obtain Ang2-EVs, which could successfully target the site of spinal cord injury. However, in order to improve the function of Ang2-EVs, we pretreated M2 microglia with melatonin, which has anti-inflammatory effects, to obtain M-Ang2-EVs. The results of single-nucleus sequencing of the mouse spinal cord verified that neurons and OPCs gradually transformed into subtypes related to nerve repair functions after treatment with M-Ang2-EVs. This is consistent with the sequencing and enrichment analysis of miRNAs contained in M-Ang2-EVs. We further verified through experiments that M-Ang2-EVs can promote microglia/macrophages to phagocytose sphingomyelin, promote axon remyelination and axon elongation, and maintain the integrity of the blood-spinal barrier. Since Ang2 can also target the blood-brain barrier, we found that M-Ang2-EVs can also reduce brain inflammation that results from spinal cord injury. Our study applied the Angiopep-2 peptide to spinal cord injury to enhance the targeting of injured cells, and successfully construct engineered extracellular vesicles that can target the spinal cord injury site and the brain.
Collapse
Affiliation(s)
- Guang Kong
- Department
of Orthopedics, Xijing Hospital, Fourth
Military Medical University, Xi’an 710000 Shaanxi, China
| | - Jie Liu
- Department
of Orthopedics, The Affiliated Taizhou People’s
Hospital of Nanjing Medical University, Taizhou School of Clinical
Medicine, Nanjing Medical University, 366 Taihu Road, Taizhou 225300 Jiangsu, China
| | - Juan Wang
- Department
of Human Anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing 210000 Jiangsu, China
| | - Xiaohu Yu
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Cong Li
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Mingyang Deng
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Minhao Liu
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Siming Wang
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Chunming Tang
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 300
Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Wu Xiong
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| | - Jin Fan
- Department
of Orthopedics, The First Affiliated Hospital
of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210000 Jiangsu, China
| |
Collapse
|
36
|
Sun Q, Zhu J, Zhao X, Huang X, Qu W, Tang X, Ma D, Shu Q, Li X. Mettl3-m 6A-NPY axis governing neuron-microglia interaction regulates sleep amount of mice. Cell Discov 2025; 11:10. [PMID: 39905012 PMCID: PMC11794856 DOI: 10.1038/s41421-024-00756-y] [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: 05/14/2024] [Accepted: 11/13/2024] [Indexed: 02/06/2025] Open
Abstract
Sleep behavior is regulated by diverse mechanisms including genetics, neuromodulation and environmental signals. However, it remains completely unknown regarding the roles of epitranscriptomics in regulating sleep behavior. In the present study, we showed that the deficiency of RNA m6A methyltransferase Mettl3 in excitatory neurons specifically induces microglia activation, neuroinflammation and neuronal loss in thalamus of mice. Mettl3 deficiency remarkably disrupts sleep rhythm and reduces the amount of non-rapid eye movement sleep. We also showed that Mettl3 regulates neuropeptide Y (NPY) via m6A modification and Mettl3 conditional knockout (cKO) mice displayed significantly decreased expression of NPY in thalamus. In addition, the dynamic distribution pattern of NPY is observed during wake-sleep cycle in cKO mice. Ectopic expression of Mettl3 and NPY significantly inhibits microglia activation and neuronal loss in thalamus, and restores the disrupted sleep behavior of cKO mice. Collectively, our study has revealed the critical function of Mettl3-m6A-NPY axis in regulating sleep behavior.
Collapse
Affiliation(s)
- Qihang Sun
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinpiao Zhu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xingsen Zhao
- Institute of Biotechnology, Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Xiaoli Huang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Wenzheng Qu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xia Tang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Daqing Ma
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Division of Anesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| | - Qiang Shu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xuekun Li
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
37
|
Chen L, Shen Q, Liu Y, Zhang Y, Sun L, Ma X, Song N, Xie J. Homeostasis and metabolism of iron and other metal ions in neurodegenerative diseases. Signal Transduct Target Ther 2025; 10:31. [PMID: 39894843 PMCID: PMC11788444 DOI: 10.1038/s41392-024-02071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
As essential micronutrients, metal ions such as iron, manganese, copper, and zinc, are required for a wide range of physiological processes in the brain. However, an imbalance in metal ions, whether excessive or insufficient, is detrimental and can contribute to neuronal death through oxidative stress, ferroptosis, cuproptosis, cell senescence, or neuroinflammation. These processes have been found to be involved in the pathological mechanisms of neurodegenerative diseases. In this review, the research history and milestone events of studying metal ions, including iron, manganese, copper, and zinc in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), will be introduced. Then, the upstream regulators, downstream effector, and crosstalk of mental ions under both physiologic and pathologic conditions will be summarized. Finally, the therapeutic effects of metal ion chelators, such as clioquinol, quercetin, curcumin, coumarin, and their derivatives for the treatment of neurodegenerative diseases will be discussed. Additionally, the promising results and limitations observed in clinical trials of these metal ion chelators will also be addressed. This review will not only provide a comprehensive understanding of the role of metal ions in disease development but also offer perspectives on their modulation for the prevention or treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yingjuan Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yunqi Zhang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Liping Sun
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
38
|
Soylu KO, Yemisci M, Karatas H. The link between spreading depolarization and innate immunity in the central nervous system. J Headache Pain 2025; 26:25. [PMID: 39901107 PMCID: PMC11792447 DOI: 10.1186/s10194-024-01938-5] [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: 11/07/2024] [Accepted: 12/12/2024] [Indexed: 02/05/2025] Open
Abstract
Spreading depolarization (SD) is a complex event that induces significant cellular stress in the central nervous system, leading to a robust inflammatory response without causing cell death in healthy tissues which may be called as neuro-parainflammation. Research has established a clear link between SD and the activation of pro-inflammatory pathways, particularly through the release of cytokines like interleukin-1β and tumor necrosis factor-α, and the involvement of inflammatory mediators such as cyclooxygenase-2 and high mobility group box 1 (HMGB1). Mechanistically, the opening of pannexin-1 (Panx1) channels and the activation of the (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome play critical roles in this process, facilitating the release of inflammatory signals that can exacerbate conditions like migraine. Furthermore, the interplay between neurons and glial cells, particularly astrocytes and microglia, underscores the intricate nature of neuroinflammation triggered by SD. Importantly, these findings indicate that these inflammatory processes may also have systemic implications, affecting immune responses beyond the central nervous system. Overall, this body of work highlights the need for further exploration of the mechanisms underlying SD-induced inflammation and potential therapeutic targets to mitigate neuroinflammatory disorders. Inflammation extends beyond the central nervous system to peripheral structures, including the meninges and trigeminovascular system, which are critical for headache initiation. Genetic factors, particularly familial hemiplegic migraine (FHM), exacerbate neuroinflammatory responses to SD, leading to increased susceptibility and prolonged headache behaviors. Collectively, these findings underscore the complex cellular interactions and innate inflammatory processes underlying SD and their relevance to migraine mechanisms, suggesting potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Kadir Oguzhan Soylu
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Türkiye
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Faculty of Medicine, Department of Neurology, Hacettepe University, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Türkiye
| | - Hulya Karatas
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Türkiye.
| |
Collapse
|
39
|
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
|
40
|
Li Y, Zhang W, Zhang Q, Li Y, Xin C, Tu R, Yan H. Oxidative stress of mitophagy in neurodegenerative diseases: Mechanism and potential therapeutic targets. Arch Biochem Biophys 2025; 764:110283. [PMID: 39743032 DOI: 10.1016/j.abb.2024.110283] [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: 08/05/2024] [Revised: 10/28/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Neurodegenerative diseases are now significant chronic progressive neurological conditions that affect individuals' physical health. Oxidative stress is crucial in the development of these diseases. Among the various neurodegenerative diseases, mitochondrial damage has become a major factor in oxidative stress and disease advancement. During this process, oxidative stress and mitophagy plays an important role. In this paper, we introduced the role of mitophagy and oxidative stress in detail, and expounded the relationship between them. In addition, we summarized the pathogenesis of some neurodegenerative diseases and the mechanism of three antioxidants. The former includes AD, PD, HD and ALS, while the latter includes carnosine, adiponectin and resveratrol. Provide goals and directions for further research and treatment of neurodegenerative diseases. This review summarizes the impact of oxidative stress on neurodegenerative diseases by regulating mitophagy, provides a deeper understanding of their pathological mechanisms, and suggests potential new therapeutic targets.
Collapse
Affiliation(s)
- Yixin Li
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Wanying Zhang
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Qihang Zhang
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yunzhe Li
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Chonghui Xin
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Rongze Tu
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Haijing Yan
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China.
| |
Collapse
|
41
|
Kharkongor R, Stephen J, Khan U, Radhakrishnan R. Exposure to an enriched environment and fucoidan supplementation ameliorate learning and memory function in rats subjected to global cerebral ischemia. Neurosci Lett 2025; 847:138094. [PMID: 39736397 DOI: 10.1016/j.neulet.2024.138094] [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: 09/06/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025]
Abstract
An enriched environment (EE) constitutes a proficient strategy that instigates social, cognitive, and motor faculties, fostering healing and heightening learning and memory function after ischemia, while fucoidan derived from brown seaweed encompasses a diverse array of bioactivities and is known to possess neuroprotective properties. This study aims to investigate the effectiveness of combining fucoidan and EE in a rat model of vascular dementia to overcome cognitive challenges. The rats were randomly assigned as Sham, Lesion - 4-vessel occlusion (4VO) i.e., transient global cerebral ischemia (tGCI), 4VO + F50mg/kg, 4VO + EE, and 4VO + F50mg/kg + EE. At the end of the study periods, the rats were exposed to the Novel object task, T-maze, and the Morris water maze. The profile of hippocampal pyramidal neurons and their dendrites was assessed through the CFV, and Golgi cox stained brain sections. Neuroinflammatory markers (IL-1β, IL-6, NF-κB, TNF-α) and synaptogenic markers (BDNF, SYP, PSD-95) were evaluated through western blot analysis. The levels of oxidative stress marker (LPO) and antioxidants (SOD, CAT, GSH, GST, GPX) in the hippocampus were quantified through biochemical assay. The findings revealed that the cognitive deficits were significantly reduced in both the 4VO + F50mg/kg and 4VO + F50mg/kg + EE treatment groups and inflammatory markers were reduced with increased antioxidant levels and synaptogenic markers when compared with the lesion group. However, through this study, the combination therapy involving fucoidan and exposure to an EE was proven effective in preserving neural integrity and restoring cognitive function against the damage caused by oxidative stress and inflammation following tGCI.
Collapse
Affiliation(s)
- Ronyson Kharkongor
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - JenishaChris Stephen
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - UlfathTasneem Khan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Rameshkumar Radhakrishnan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, India.
| |
Collapse
|
42
|
Tiong SQ, Mohgan RN, Quek JY, Liew JYS, Wong GYS, Thang ZQ, Chan ZL, Gan SY, Chan EWL. Inhibition of the Transforming Growth Factor-β Signaling Pathway Confers Neuroprotective Effects on Beta-Amyloid-Induced Direct Neurotoxicity and Microglia-Mediated Neuroinflammation. Neurol Res Int 2025; 2025:8948290. [PMID: 39949498 PMCID: PMC11824711 DOI: 10.1155/nri/8948290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025] Open
Abstract
Background: Abnormal elevation of transforming growth factor-beta (TGF-β) has been observed among Alzheimer's disease (AD) patients. This may be due to microglia-mediated release of proinflammatory cytokines, which promote neuroinflammation and neuronal apoptosis. Silencing of TGFBR1, a gene encoding TGF-β receptor type I (TGF-βR1), has resulted in neuronal survival from amyloid-beta (Aβ)-induced neurotoxicity. Therefore, the present study investigated the neuroprotective effect of TGF-βR1 inhibitors (RepSox, Galunisertib, and Vactosertib) against Aβ-induced direct neurotoxicity and microglia-mediated neuroinflammation. Methods: The neuroprotective effect of TGF-βR1 inhibitors against Aβ-induced direct neurotoxicity and microglia-mediated neuroinflammation were investigated using the RealTime-Glo™ MT Cell Viability Assay. The inhibitory effect of TGF-βR1 inhibitors on Aβ-induced microglia-mediated production of proinflammatory cytokines (TNF-α and IL-1β) was determined using enzyme-linked immunosorbent assay (ELISA). Results: TGF-βR1 inhibitors (RepSox, Galunisertib, and Vactosertib) at the tested concentrations (6.25-150 nM) showed no significant cytotoxicity effects on SH-SY5Y and BV-2 cells. Moreover, treatments with these inhibitors exhibited neuroprotection on SH-SY5Y cells against Aβ-induced direct neurotoxicity. The trend of cell viability after 24 h treatment also supports the microscopic images of the cells' morphology. Furthermore, pretreatment with these inhibitors conferred indirect neuroprotective effect against Aβ-induced microglia-mediated neuroinflammation by attenuating the production of proinflammatory cytokines (TNF-α and IL-1β). Conclusion: The inhibition of the TGF-β signaling pathway in neuronal and microglia cells by TGF-βR1 inhibitors resulted in neuroprotection against Aβ-induced direct neurotoxicity and microglia-mediated neuroinflammation. Hence, targeting the TGF-β signaling pathway in both neuronal and microglia cells could provide a promising therapeutic strategy in AD.
Collapse
Affiliation(s)
- Shao Qin Tiong
- School of Pharmacy, IMU University, Bukit Jalil, Kuala Lumpur, Malaysia
| | | | - Jia Yee Quek
- School of Pharmacy, IMU University, Bukit Jalil, Kuala Lumpur, Malaysia
| | | | | | - Zi Qing Thang
- School of Pharmacy, IMU University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Zhi Ling Chan
- School of Pharmacy, IMU University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Sook Yee Gan
- School of Pharmacy, IMU University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Elaine Wan Ling Chan
- Institute for Research, Development and Innovation, IMU University, Bukit Jalil, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Gill AJ, Smith MD, Galleguillos D, Garton T, Mace JW, Gadani SP, Kumar S, Pokharel A, Solem K, Potluri S, Hussein O, Rogines GS, Singh A, Clark A, Calabresi PA, Gharagozloo M. NLRX1 limits inflammatory neurodegeneration in the anterior visual pathway. J Neuroinflammation 2025; 22:21. [PMID: 39875919 PMCID: PMC11773851 DOI: 10.1186/s12974-025-03339-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Chronic innate immune activation in the central nervous system (CNS) significantly contributes to neurodegeneration in progressive multiple sclerosis (MS). Using multiple experimental autoimmune encephalomyelitis (EAE) models, we discovered that NLRX1 protects neurons in the anterior visual pathway from inflammatory neurodegeneration. We quantified retinal ganglion cell (RGC) density and optic nerve axonal degeneration, gliosis, and T-cell infiltration in Nlrx1-/- and wild-type (WT) EAE mice and found increased RGC loss and axonal injury in Nlrx1-/- mice compared to WT mice in both active immunization EAE and spontaneous opticospinal encephalomyelitis (OSE) models. To minimize the effects of Nlrx1-/- on peripheral lymphocyte priming during EAE, we performed adoptive transfer experiments, in which activated myelin-specific T cells were transferred into lymphocyte-deficient Rag-/- or Nlrx1-/-Rag-/- mice. In this model, we found more severe microgliosis and astrogliosis in the optic nerve of Nlrx1-/-Rag-/- mice compared to Rag-/- mice, suggesting a regulatory role of NLRX1 in innate immune cells. Transcriptome analysis in primary astrocytes activated with LPS and IFNγ demonstrated that NLRX1 suppresses NF-κB activation and regulates mitochondrial oxidative phosphorylation in inflammatory reactive astrocytes. The novel pharmacologic NLRX1 activators NX-13 and LABP-66 decreased LPS-mediated gene expression of inflammatory cytokines and chemokines in mixed glial cultures. Moreover, treating EAE mice with oral LABP-66, compared to vehicle, after the onset of paralysis resulted in less anterior visual pathway neurodegeneration. These data suggest that pharmacologic NLRX1 activators have the potential to limit inflammatory neurodegeneration. This study highlights that NLRX1 could serve as a promising target for neuroprotection in progressive MS and other neurodegenerative diseases. Further studies are needed to better understand the cell-specific mechanisms underlying the neuroprotective role of NLRX1 in response to inflammation in the CNS.
Collapse
Affiliation(s)
- Alexander J Gill
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Matthew D Smith
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Danny Galleguillos
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Thomas Garton
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Jackson W Mace
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Sachin P Gadani
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Swati Kumar
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Aayush Pokharel
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Krista Solem
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Saahith Potluri
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Omar Hussein
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Giuliana Sardi Rogines
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Arihant Singh
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Annatje Clark
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Marjan Gharagozloo
- Department of Neurology, Division of Neuroimmunology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
44
|
Cai Z, Liang C, Huang K, Luo J, Lu R, Lai Y, Zheng D, Lin Z, Zhong J, Dai J, Huang J, Zhang H, Chen J. Curcumin prevents neurodegeneration by blocking HDAC6-NLRP3 pathway-dependent neuroinflammation in Parkinson's disease. Int Immunopharmacol 2025; 146:113928. [PMID: 39724731 DOI: 10.1016/j.intimp.2024.113928] [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: 09/16/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Curcumin is a hydrophobic polyphenolic compound with potent anti-inflammatory properties. However, whether it can achieve therapeutic effects by alleviating neuroinflammation in patients with Parkinson's disease (PD) and its potential mechanism are still unknown. This study explored the effects of curcumin on neuroinflammation in dopaminergic neurons and deciphered its direct target in the histone deacetylase 6 (HDAC6)-Nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) pathway, revealing the potential role of curcumin in the treatment of Parkinson's disease. Here, we show that curcumin alleviated the degeneration of neurons in a PD model by mitigating the activation of the NLRP3-mediated inflammatory response both in vivo and in vitro. Furthermore, we discovered that curcumin prevented neuroinflammation by blocking the HDAC6-NLRP3 pathway in a PD model. Moreover, overexpression of HDAC6 could eliminate the effect of curcumin on the neuroinflammatory response mediated by NLRP3. Curcumin and the HDAC6 inhibitor WT161 could alleviate neurodegeneration. In addition, activated HDAC6 directly deacetylated NLRP3 at lysine 84 to maintain its stability, which increased the inflammatory response and promoted neurodegeneration. These findings show that curcumin, a neuroinflammation inhibitor, blocks neurodegeneration via the HDAC6-NLRP3 pathway and represents a potentially practical pharmacological approach for treating neuroinflammation-driven neurodegenerative diseases. For the first time, HDAC6 was shown to directly regulate the acetylation of NLRP3.
Collapse
Affiliation(s)
- Ziwei Cai
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Congmin Liang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Kailun Huang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Jiankun Luo
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Renjian Lu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Yixi Lai
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Dongyan Zheng
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Zhuomiao Lin
- Department of Clinical Pharmacy, Meizhou People's Hospital (Huangtang Hospital), Huangtang Road 63#, Meijiang District, Meizhou 514031, China
| | - Jiahong Zhong
- Department of Clinical Pharmacy, Meizhou People's Hospital (Huangtang Hospital), Huangtang Road 63#, Meijiang District, Meizhou 514031, China
| | - Juanxiu Dai
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China
| | - Jiewen Huang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan 523710, Guangdong, China
| | - He Zhang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China.
| | - Jialong Chen
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, Guangdong, PR China.
| |
Collapse
|
45
|
Tang P, Sun Y, Yang C, Zhang N. Early functional and structural hippocampal impairment in a bilateral common carotid artery stenosis mouse model. Animal Model Exp Med 2025. [PMID: 39853719 DOI: 10.1002/ame2.12549] [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: 06/24/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Subcortical ischemic vascular dementia (SIVD) is a common subtype of vascular dementia. Currently, the bilateral common carotid artery stenosis (BCAS) mouse model is the most suitable SIVD rodent model. In this study, we investigated the functional and structural impairments in the hippocampus 1 month after BCAS. METHODS We used behavioral tests, laser speckle flowmetry, long-term potentiation, histochemical staining, molecular experiments, and voxel-based morphometry to evaluate the hippocampal impairments. RESULTS Behavioral studies revealed that BCAS mice exhibited worse performance. Laser speckle flowmetry detected an obvious decrease in cerebral blood flow. The synaptic plasticity of the perforant path-dentate gyrus pathway was inhibited. Decreased fractional anisotropy and increased mean diffusivity were detected in the hippocampus via diffusion tensor imaging data. A reduction in gray matter volume, which was most prominent in the hippocampus and its surrounding areas, was detected via voxel-based morphometry analysis. Impairments in cell morphology and myelin integrity were validated using histochemical staining and molecular biology techniques. In addition, the numbers of GFAP+ astrocytes and Iba1+ microglia increased in the hippocampus. CONCLUSIONS Overall, our study demonstrates early functional and structural impairments in the hippocampus contributing to learning and memory deficits after 1 month of BCAS, indicating that the hippocampus is vulnerable to chronic cerebral ischemia.
Collapse
Affiliation(s)
- Ping Tang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi Sun
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunsheng Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| |
Collapse
|
46
|
Li X, Zhu B, Dong N, Zhao Z, Cao J, Zhou L, Gao Z, Su B. Early Detection of High-Altitude Hypoxic Brain Injury by In Vivo Electrochemistry. Angew Chem Int Ed Engl 2025; 64:e202416395. [PMID: 39497570 DOI: 10.1002/anie.202416395] [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: 08/26/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
High-altitude hypoxic brain injury (HHBI) is a kind of acute mountain sickness and the survival rate of patients with HHBI can be improved only if it is detected and treated at the early stage. However, limited by speediness and accuracy, it is still very difficult for most of current approaches to realize the early detection of HHBI. We propose herein a novel strategy for this goal based on spatiotemporal changes in the brain oxygen level. As revealed by in vivo electrochemistry, the characteristic changes of brain oxygen level under the high-altitude exposure are directly associated with the brain hypoxia status. Given brain hypoxia is the main pathogenesis of HHBI, the degree of HHBI can be diagnosed by the variation of brain oxygen, making the early detection of HHBI feasible. In addition, the risk of HHBI for mouse exposed to high-altitude hypoxia environments can be also prognosed days in advance.
Collapse
Affiliation(s)
- Xinru Li
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
| | - Boyu Zhu
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
| | - Nuo Dong
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
| | - Ziyi Zhao
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
| | - Jiayi Cao
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
| | - Lin Zhou
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
| | - Zhigang Gao
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 310052, Hangzhou, China
| | - Bin Su
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, 310058, Hangzhou, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 310052, Hangzhou, China
| |
Collapse
|
47
|
Jansen R, Pawlitzki M, Gliem M, Meuth SG, Schreiber S, Görtler MW, Neumann J. LFA-1: A potential key player in microglia-mediated neuroprotection against oxygen-glucose deprivation in vitro. PLoS One 2025; 20:e0314020. [PMID: 39787147 PMCID: PMC11717251 DOI: 10.1371/journal.pone.0314020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/04/2024] [Indexed: 01/12/2025] Open
Abstract
For the last 38 years, all neuroprotective agents for patients with ischemic stroke have failed in clinical trials. The innate immune system, particularly microglia, is a much-discussed target for neuroprotective agents. Promising results for neuroprotection by inhibition of integrins with drugs such as natalizumab in animal stroke models have not been translated into clinical practice. Our present study reveals the relevance of a β2 integrin, lymphocyte function-associated antigen-1 (LFA-1), as a potential key player in protecting neuronal cell death after oxygen-glucose deprivation in organotypic hippocampal cell cultures. In addition, we identified microglial cells as effector cells for LFA-1-mediated neuroprotection. The counterpart of LFA-1 on microglia is unclear, but we show strong expression of ICAM-5 in hippocampal neurons, suggesting a critical role for direct crosstalk between microglia and neurons for neuronal survival under oxygen-glucose deprivation. The enigma of neuroprotection after ischemic stroke remains to be solved, and our findings highlight the continuing importance and lack of understanding of integrin-mediated pathways after ischemic stroke and the need for further intensive research.
Collapse
Affiliation(s)
- Robin Jansen
- Medical Faculty and University Hospital Düsseldorf, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Marc Pawlitzki
- Medical Faculty and University Hospital Düsseldorf, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Michael Gliem
- Medical Faculty and University Hospital Düsseldorf, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven G. Meuth
- Medical Faculty and University Hospital Düsseldorf, Department of Neurology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Stefanie Schreiber
- Medical Faculty, Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Michael-W. Görtler
- Medical Faculty, Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Jens Neumann
- Medical Faculty, Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
48
|
Zhou L, Shi H, Xiao M, Liu W, Wang L, Zhou S, Chen S, Wang Y, Liu C. Remimazolam attenuates lipopolysaccharide-induced neuroinflammation and cognitive dysfunction. Behav Brain Res 2025; 476:115268. [PMID: 39322063 DOI: 10.1016/j.bbr.2024.115268] [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: 05/16/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE Remimazolam, a novel benzodiazepine, is widely used as an anesthetic in endoscopic procedures; however, its effects on cognitive function remain unclear, limiting its broader application in general anaesthesia. Neuroinflammation is a well-established key factor in the etiology and progression of cognitive dysfunction, including conditions such as Alzheimer's disease, Parkinson's disease, postoperative delirium, and postoperative cognitive dysfunction. Preclinical studies have demonstrated that remimazolam exerts anti-inflammatory and neuroprotective effects, and clinical reports indicate a reduced incidence of postoperative delirium in patients treated with remimazolam. Nevertheless, whether remimazolam improves cognitive function through its anti-inflammatory properties remains uncertain. This study aimed to investigate the neuroprotective effects of remimazolam and its underlying mechanism in a lipopolysaccharide (LPS)-induced model of neuroinflammation, neuronal injury, and cognitive dysfunction METHODS: C57BL/6 J male mice were administered LPS intraperitoneally to establish a model of neuroinflammation-induced cognitive impairment. A subset of mice received remimazolam via intraperitoneal injection 30 minutes prior to LPS administration. Cognitive performance was evaluated using behavioural tests, including the Morris Water Maze (MWM), Novel Object Recognition (NOR) test, and Open Field Test (OFT). Hippocampal tissues were analyzed by haematoxylin-eosin (HE) staining to assess structural changes. Inflammatory markers, including Interleukin (IL)-6, IL-1β, and tumor necrosis factor-α, were quantified using enzyme-linked immunosorbent assay (ELISA) and real-time quantitative PCR. Immunofluorescence was used to detect translocator protein (TSPO) and markers of microglia activation (IBA-1, CD16/32, and CD206). RESULTS (1) Remimazolam reversed LPS-induced cognitive deficits, as evidenced by shorter spatial exploration latency and increased platform crossings in the MWM, and an elevated recognition index in the NOR test. (2) Remimazolam improved hippocampal morphology, reducing LPS-induced neuronal damage. (3) Remimazolam significantly decreased levels of hippocampal inflammatory cytokines, inhibited microglial activation, promoted M2-type microglia polarization, and increased TSPO expression. CONCLUSION Remimazolam demonstrated neuroprotective and anti-neuroinflammatory effects in a mouse model of LPS-induced cognitive impairment. These effects are likely mediated through the regulation of TSPO, which inhibits microglial activation and promotes the polarization of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype.
Collapse
Affiliation(s)
- Leguang Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China; University of South China Hengyang Medical School Clinical Anatomy & Reproductive Medicine Application Institute, China
| | - Hongzhao Shi
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Mengzhe Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Wenjie Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Lijuan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Shangtao Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Shenghua Chen
- University of South China Hengyang Medical School Clinical Anatomy & Reproductive Medicine Application Institute, China
| | - Yan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China.
| | - Chengxi Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China.
| |
Collapse
|
49
|
Godeanu S, Mușat MI, Scheller A, Osiac E, Cătălin B. Minimal differences observed when comparing the morphological profiling of microglia obtained by confocal laser scanning and optical sectioning microscopy. Front Neuroanat 2025; 18:1507140. [PMID: 39829733 PMCID: PMC11739110 DOI: 10.3389/fnana.2024.1507140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Background While widefield microscopy has long been constrained by out-of-focus scattering, advancements have generated a solution in the form of confocal laser scanning microscopy (cLSM) and optical sectioning microscopy using structured illumination (OSM). In this study, we aim to investigate, using microglia branching, if cLSM and OSM can produce images with comparable morphological characteristics. Results By imaging the somatosensory microglia from a tissue slice of a 3-week-old mouse and establishing morphological parameters that characterizes the microglial branching pattern, we were able to show that there is no difference in total length of the branch tree, number of branches, mean branch length and number of primary to terminal branches. We did find that area-based parameters such as mean occupied area and mean surveillance area were bigger in cLSM isolated microglia compared to OSM ones. Additionally, by investigating the difference in acquisition time between techniques and personal costs we were able to establish that the amortization could be made in 6.11 ± 2.93 years in the case of countries with a Human Development Index (HDI) = 7-9 and 7.06 ± 3.13 years, respectably, for countries with HDI < 7. As such, OSM systems seem a valid option if one just wants basic histological evaluation, and cLSM should be considered for groups that demand higher resolution or volumetric images.
Collapse
Affiliation(s)
- Sânziana Godeanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Saarbrücken, Germany
| | - Mădălina Iuliana Mușat
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Saarbrücken, Germany
- Center for Gender-Specific Biology and Medicine (CGBM), University of Saarland, Saarbrücken, Germany
| | - Eugen Osiac
- Department of Biophysics, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Bogdan Cătălin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
50
|
Elser H, Frankland TB, Chen C, Tartof SY, Mayeda ER, Lee GS, Northrop AJ, Torres JM, Benmarhnia T, Casey JA. Wildfire Smoke Exposure and Incident Dementia. JAMA Neurol 2025; 82:40-48. [PMID: 39585704 PMCID: PMC11589856 DOI: 10.1001/jamaneurol.2024.4058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/03/2024] [Indexed: 11/26/2024]
Abstract
Importance Long-term exposure to total fine particulate matter (PM2.5) is a recognized dementia risk factor, but less is known about wildfire-generated PM2.5, an increasingly common PM2.5 source. Objective To assess the association between long-term wildfire and nonwildfire PM2.5 exposure and risk of incident dementia. Design, Setting, and Participants This open cohort study was conducted using January 2008 to December 2019 electronic health record (EHR) data among members of Kaiser Permanente Southern California (KPSC), which serves 4.7 million people across 10 California counties. KPSC members aged 60 years or older were eligible for inclusion. Members were excluded if they did not meet eligibility criteria, if they had a dementia diagnosis before cohort entry, or if EHR data lacked address information. Data analysis was conducted from May 2023 to May 2024. Exposures Three-year rolling mean wildfire and nonwildfire PM2.5 in member census tracts from January 2006 to December 2019, updated quarterly and estimated via monitoring and remote-sensing data and statistical techniques. Main Outcome and Measures The primary outcome was incident dementia, identified using diagnostic codes in the EHR. Odds of dementia diagnoses associated with 3-year mean wildfire and nonwildfire PM2.5 exposure were estimated using a discrete-time approach with pooled logistic regression. Models adjusted for age, sex, race and ethnicity (considered as a social construct rather than as a biological determinant), marital status, smoking status, calendar year, and census tract-level poverty and population density. Stratified models assessed effect measure modification by age, sex, race and ethnicity, and census tract-level poverty. Results Among 1.64 million KPSC members aged 60 years or older during the study period, 1 223 107 members were eligible for inclusion in this study. The study population consisted of 644 766 female members (53.0%). In total, 319 521 members identified as Hispanic (26.0%), 601 334 members identified as non-Hispanic White (49.0%), and 80 993 members received a dementia diagnosis during follow-up (6.6%). In adjusted models, a 1-μg/m3 increase in the 3-year mean of wildfire PM2.5 exposure was associated with an 18% increase in the odds of dementia diagnosis (odds ratio [OR], 1.18; 95% CI, 1.03-1.34). In comparison, a 1-μg/m3 increase in nonwildfire PM2.5 exposure was associated with a 1% increase (OR, 1.01; 95% CI, 1.01-1.02). For wildfire PM2.5 exposure, associations were stronger among members less than 75 years old upon cohort entry, members from racially minoritized subgroups, and those living in high-poverty vs low-poverty census tracts. Conclusions and Relevance In this cohort study, after adjusting for measured confounders, long-term exposure to wildfire and nonwildfire PM2.5 over a 3-year period was associated with dementia diagnoses. As the climate changes, interventions focused on reducing wildfire PM2.5 exposure may reduce dementia diagnoses and related inequities.
Collapse
Affiliation(s)
- Holly Elser
- Department of Neurology, University of Pennsylvania, Philadelphia
- Editorial Fellow, JAMA Neurology
| | - Timothy B. Frankland
- Kaiser Permanente Hawaii Center for Integrated Health Care Research, Honolulu, Hawaii
| | - Chen Chen
- Scripps Institution of Oceanography, University of California, San Diego
| | - Sara Y. Tartof
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
| | - Elizabeth Rose Mayeda
- Department of Epidemiology, UCLA Fielding School of Public Health, University of California, Los Angeles
| | - Gina S. Lee
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, California
| | | | - Jacqueline M. Torres
- Department of Epidemiology & Biostatistics, University of California, San Francisco
| | - Tarik Benmarhnia
- Scripps Institution of Oceanography, University of California, San Diego
- Irset Institut de Recherche en Santé, Environnement et Travail, UMR-S 1085, Inserm, University of Rennes, EHESP, Rennes, France
| | - Joan A. Casey
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle
- Department of Epidemiology, University of Washington School of Public Health, Seattle
| |
Collapse
|