1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Jin J, Zhang H, Lu Q, Tian L, Yao S, Lai F, Liang Y, Liu C, Lu Y, Tian S, Zhao Y, Ren W. Nanocarrier-mediated siRNA delivery: a new approach for the treatment of traumatic brain injury-related Alzheimer's disease. Neural Regen Res 2025; 20:2538-2555. [PMID: 39314170 PMCID: PMC11801294 DOI: 10.4103/nrr.nrr-d-24-00303] [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: 03/16/2024] [Revised: 07/06/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
Traumatic brain injury and Alzheimer's disease share pathological similarities, including neuronal loss, amyloid-β deposition, tau hyperphosphorylation, blood-brain barrier dysfunction, neuroinflammation, and cognitive deficits. Furthermore, traumatic brain injury can exacerbate Alzheimer's disease-like pathologies, potentially leading to the development of Alzheimer's disease. Nanocarriers offer a potential solution by facilitating the delivery of small interfering RNAs across the blood-brain barrier for the targeted silencing of key pathological genes implicated in traumatic brain injury and Alzheimer's disease. Unlike traditional approaches to neuroregeneration, this is a molecular-targeted strategy, thus avoiding non-specific drug actions. This review focuses on the use of nanocarrier systems for the efficient and precise delivery of siRNAs, discussing the advantages, challenges, and future directions. In principle, siRNAs have the potential to target all genes and non-targetable proteins, holding significant promise for treating various diseases. Among the various therapeutic approaches currently available for neurological diseases, siRNA gene silencing can precisely "turn off" the expression of any gene at the genetic level, thus radically inhibiting disease progression; however, a significant challenge lies in delivering siRNAs across the blood-brain barrier. Nanoparticles have received increasing attention as an innovative drug delivery tool for the treatment of brain diseases. They are considered a potential therapeutic strategy with the advantages of being able to cross the blood-brain barrier, targeted drug delivery, enhanced drug stability, and multifunctional therapy. The use of nanoparticles to deliver specific modified siRNAs to the injured brain is gradually being recognized as a feasible and effective approach. Although this strategy is still in the preclinical exploration stage, it is expected to achieve clinical translation in the future, creating a new field of molecular targeted therapy and precision medicine for the treatment of Alzheimer's disease associated with traumatic brain injury.
Collapse
Affiliation(s)
- Jie Jin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Huajing Zhang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Chuanchuan Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yujia Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sijia Tian
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
3
|
Xiong H, Zheng S, Qi X, Liu J. μGlia-Flow, an automatic workflow for microglia segmentation and classification. J Neurosci Methods 2025; 419:110446. [PMID: 40220906 DOI: 10.1016/j.jneumeth.2025.110446] [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: 02/05/2025] [Revised: 03/19/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Microglia are important immune cells in the central nervous system, playing a key role in various pathological processes. The morphological diversity of microglia is closely linked to the development of brain diseases, yet accurate segmentation and automatic classification of microglia remain challenging. NEW METHOD We proposed a workflow, μGlia-Flow, which integrates both segmentation and classification for microglia analysis. The Frangi filtering algorithm was employed for branch segmentation, and an edge-guided attention TransUNet (EGA-Net) was used for soma segmentation. A Vision Transformer (ViT) network was applied to classify different morphologies. RESULTS The Frangi filtering algorithm produces more complete branches with smoother edges and clearer structures. The EGA-Net improves Dice and IoU scores by 4.02 % and 6.75 %, respectively. ViT achieves over 99 % precision in classification. Post-processing reveals decreasing complexity during activation, validating the accuracy of μGlia-Flow. COMPARISON WITH EXISTING METHODS μGlia-Flow introduces deep learning, significantly improving segmentation accuracy and addressing the parameter dependency of existing classification methods. CONCLUSION we present an automatic workflow for segmenting and classifying microglia, providing a powerful tool for different morphology analysis.
Collapse
Affiliation(s)
- Huangrui Xiong
- School of Information Science and Technology, MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China; Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Siling Zheng
- School of Information Science and Technology, MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Xiuhong Qi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Ji Liu
- School of Information Science and Technology, MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China; Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
4
|
Yan K, He Q, Tang J, Peng W, Dou B, Chen H, Bei W. Actinobacillus pleuropneumoniae infection activates IL-1β expression in porcine alveolar macrophages via β-amyloid production. Microb Pathog 2025; 204:107559. [PMID: 40220800 DOI: 10.1016/j.micpath.2025.107559] [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: 12/21/2024] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Actinobacillus pleuropneumoniae (A. pleuropneumoniae), a porcine respiratory tract pathogen, causes porcine pleuropneumonia. Porcine alveolar macrophages (PAMs) play a crucial role during A. pleuropneumoniae infection. Amyloid precursor protein (APP) can be cleaved by β- and γ-secretase to produce β-amyloid (Aβ). APP and Aβ are associated with the inflammatory response. They activate microglia and astrocytes to secrete IL-1β, IL-6, and other cytokines. In this study, we found that during the interaction between A. pleuropneumoniae and PAMs, the two-component system CpxAR upregulates wecA expression, increasing lipopolysaccharide (LPS) production. LPS promotes APP production and cleavage to generate Aβ. The Aβ activates NF-κB, leading to increased IL-1β expression. We hypothesize that A. pleuropneumoniae infection of PAMs regulates APP production and cleavage to control Aβ levels. Different quantities of Aβ induce PAMs to produce varying amounts of cytokines, leading to different pathological processes in porcine pleuropneumonia.
Collapse
Affiliation(s)
- Kang Yan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiyun He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jia Tang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wei Peng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Beibei Dou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China
| | - Weicheng Bei
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
5
|
Sun X, Liu H, Li W, Li L, Tian Q, Cao Q, Meng Y, Shen Y, Che F, Chiu JC, Yu J, Hammock BD. The soluble epoxide hydrolase inhibitor TPPU alleviates Aβ-mediated neuroinflammatory responses in Drosophila melanogaster and cellular models of alzheimer's disease. J Inflamm (Lond) 2025; 22:25. [PMID: 40551105 PMCID: PMC12183823 DOI: 10.1186/s12950-025-00449-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/29/2025] [Indexed: 06/28/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a common neurodegenerative disease, and its pathogenesis is closely associated with neuroinflammation. The control of neuroinflammation in AD is the focus of current research. soluble epoxide hydrolase (sEH) protein is increased in the brain tissues of patients with AD and has been targeted by multiple genome wide association studies as a prime target for treating AD. Since sEH induces nerve inflammation by degrading epoxyeicosatrienoic acids (EETs), application of sEH inhibitor and sEH gene knockout are effective ways to improve the bioavailability of EETs and inhibit or even resolve neuroinflammation in AD. 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) is a potent sEH inhibitor that has been shown to be effective in preclinical animal models of a variety of chronic inflammatory diseases. This study aims to further explore whether TPPU can alleviate AD neuroinflammation. METHODS We established an Aβ42-transgenic Drosophila melanogaster model using the galactose-regulated upstream promoter element 4 (GAL4) / upstream active sequence (UAS) expression system and investigated the protective and anti-neuroinflammatory effects of TPPU against Aβ toxicity. We detected behavioral indexes (survival time, crawling ability, and olfactory memory) and biochemical indexes malondialdehyde (MDA) content and superoxide dismutase (SOD) activity in brain tissues of Aβ42 transgenic flies. Finally, we explored the anti-neuroinflammatory effect of TPPU and its possible mechanism by stimulating cocultures of human SH-SY5Y cells and HMC3 cells with Aβ(25-35) to model neuronal cell inflammation, and evaluated the cells by fluorescence microscopy, ELISA, Western Blot, and Real-time PCR. RESULTS We found that TPPU improved the survival time, crawling ability, and olfactory memory of Aβ42-transgenic flies. We also observed reduction of MDA content and elevation of SOD activity in the brain tissues of these flies. In human cell models, we found that TPPU improved cell viability, reduced cell apoptosis, decreased lipid oxidation, inhibited oxidative damage, thus playing a neuroprotective role. The inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6) and interleukin-18 (IL-18) were downregulated, and the mRNA expression of the M2 microglia markers CD206 and SOCS3 were upregulated by TPPU; thus, TPPU inhibited neuroinflammatory responses. TPPU exerted neuroprotective and anti-inflammatory effects by decreasing the protein expression of the sEH-encoding gene EPHX2 and increasing the levels of 11,12-epoxyeicosatrienoic acid (11,12-EET) and 14,15-epoxyeicosatrienoic acid (14,15-EET). The inhibitory effect of TPPU on Aβ(25-35)-mediated neuroinflammation was associated with inhibition of the toll like receptor 4 (TLR4)/nuclear transcription factor-κB (NF-κB) pathway and p38 mitogen activated protein kinases (MAPK)/NF-κB pathway. CONCLUSIONS We report that the sEH inhibitor TPPU exerts neuroprotective and anti-neuroinflammatory effects in AD models, and it is expected that this drug could potentially be used for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Xiaowen Sun
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Hongxiang Liu
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Wei Li
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Lin Li
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Qian Tian
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Qingyang Cao
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Yun Meng
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Yan Shen
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China
| | - Joanna C Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA, 95616, USA
| | - Jixu Yu
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong, China.
- Shandong Provincial Clinical Research Center for Geriatric Diseases, Linyi, 276000, Shandong, China.
| | - Bruce D Hammock
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
6
|
Yamauchi K, Koike M, Hioki H. A three dimensional immunolabeling method with peroxidase-fused nanobodies and fluorochromized tyramide-glucose oxidase signal amplification. Commun Biol 2025; 8:903. [PMID: 40533487 PMCID: PMC12177075 DOI: 10.1038/s42003-025-08317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 05/29/2025] [Indexed: 06/22/2025] Open
Abstract
Three dimensional immunohistochemistry (3D-IHC), immunolabeling of 3D tissues, reveals the spatial organization of molecular and cellular assemblies in the context of the tissue architecture. Deep and rapid penetration of antibodies into 3D tissues and highly sensitive detection are critical for high-throughput imaging analysis of immunolabeled 3D tissues. Here, we report a nanobody (nAb)-based 3D-IHC, POD-nAb/FT-GO 3D-IHC, for high-speed and high-sensitive detection of targets within 3D tissues. Peroxidase-fused nAbs (POD-nAbs) enhanced immunolabeling depth and allowed for highly sensitive detection by combined with a fluorescent tyramide signal amplification system, Fluorochromized Tyramide-Glucose Oxidase (FT-GO). Multiplex labeling was implemented to the 3D-IHC by quenching POD with sodium azide. Using the 3D-IHC technique, we successfully visualized somata and processes of neuronal and glial cells in millimeter-thick mouse brain tissues within three days. Given its high-speed and high-sensitive detection, our 3D-IHC protocol, POD-nAb/FT-GO 3D-IHC, would provide a useful platform for histochemical analysis in 3D tissues.
Collapse
Affiliation(s)
- Kenta Yamauchi
- Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Hioki
- Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Multi-Scale Brain Structure Imaging, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
7
|
Ye F, Li M, Liu M, Wu X, Tian F, Gong Y, Cao Y, Zhang J, Zhang X, Qin C, Zhang L. Co-Aggregation of Syndecan-3 with β-Amyloid Aggravates Neuroinflammation and Cognitive Impairment in 5×FAD Mice. Int J Mol Sci 2025; 26:5502. [PMID: 40564963 DOI: 10.3390/ijms26125502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 05/17/2025] [Accepted: 06/06/2025] [Indexed: 06/28/2025] Open
Abstract
Abnormal deposition of β-amyloid (Aβ) is a core pathological feature of Alzheimer's disease (AD). Syndecan-3 (SDC3), a type I transmembrane heparan sulfate proteoglycan (HSPG), is abnormally overexpressed in the brains of AD patients and model animals, specifically accumulating in the peri-plaque region of amyloid plaques. However, its regulatory mechanism in the process of Aβ deposition remains unclear. This study aims to clearly define the role of SDC3 in Aβ aggregation and neuroinflammation, two critical processes in AD pathogenesis. Specifically, we investigate how SDC3 modulates Aβ aggregation and its interaction with neuroinflammatory pathways, which may contribute to the progression of AD. By elucidating the mechanisms underlying SDC3's involvement in these processes, we seek to provide new insights into potential therapeutic targets for AD. In this study, a 5×FAD mouse model with downregulated SDC3 expression was constructed. Behavioral assessments and synaptic function tests were performed to explore the effects of SDC3 on cognition in 5×FAD mice. Immunofluorescence co-localization technology was utilized to analyze the pathological co-deposition of SDC3 and Aβ in the hippocampus, cortex, and meningeal blood vessels. Quantitative assessments of pro-inflammatory cytokines such as Tnf-α and Cxcl10 in the brain were performed through histopathological analysis combined with qPCR. Western blotting was used to examine the phosphorylation status of STAT1/STAT3 and the expression changes of IBA1/GFAP to systematically analyze the molecular mechanisms through which SDC3 regulates AD pathology. This study revealed that SDC3 expression was significantly upregulated in the brain regions of the 5×FAD model mice and co-localized pathologically with Aβ. Cell lineage tracing analysis showed that the elevated SDC3 expression primarily originated from glial cells. Behavioral and pathological results demonstrated that downregulation of SDC3 significantly improved cognitive dysfunction in the model mice and effectively reduced the Aβ burden in the brain. Molecular mechanism studies showed that downregulation of SDC3 reduced the phosphorylation of STAT1 and STAT3, thereby inhibiting the activation of the JAK-STAT and cGAS-STING signaling pathways, reducing the activation of microglia/astrocytes and suppressing the expression of pro-inflammatory cytokines such as Tnf-α and Cxcl10. This study reveals that SDC3 co-localizes with Aβ pathology and synergistically exacerbates neuroinflammation. Knockdown of SDC3 can simultaneously reduce both Aβ deposition and the release of inflammatory factors from glial cells. Mechanistic research indicates that SDC3 drives a "glial activation-cytokine release" vicious cycle through the JAK-STAT and cGAS-STING signaling pathways. These findings suggest that SDC3 may serve as a key hub coordinating amyloid pathology and neuroinflammation in AD, providing new insights for the development of combination therapies targeting the HSPG network.
Collapse
Affiliation(s)
- Fan Ye
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Mingfeng Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Min Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Xinghan Wu
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Fan Tian
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Yanju Gong
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Yan Cao
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Jingtai Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Xueling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and lnnovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing 100730, China
| |
Collapse
|
8
|
Henningfield CM, Ngo M, Murray KM, Kwang NE, Tsourmas KI, Neumann J, Pashkutz ZA, Kawauchi S, Swarup V, Lane TE, MacGregor GR, Green KN. Generation of an Inducible Destabilized-Domain Cre Mouse Line to Target Disease Associated Microglia. Glia 2025; 73:1272-1287. [PMID: 39988890 DOI: 10.1002/glia.70004] [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: 09/24/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
The function of microglia during progression of Alzheimer's disease (AD) can be investigated using mouse models that enable genetic manipulation of microglial subpopulations in a temporal manner. We developed mouse lines that express either Cre recombinase (Cre) for constitutive targeting, or destabilized-domain Cre recombinase (DD-Cre) for inducible targeting from the Cst7 locus (Cst7 DD-Cre) to specifically manipulate disease associated microglia (DAM) and crossed with Ai14 tdTomato cre-reporter line mice. Cst7Cre was found to target all brain resident myeloid cells, due to transient developmental expression of Cst7, but no expression was found in the inducible Cst7 DD-Cre mice. Further crossing of this line with 5xFAD mice combined with dietary administration of trimethoprim to induce DD-Cre activity produces long-term labeling in DAM without evidence of leakiness, with tdTomato-expression restricted to cells surrounding plaques. Using this model, we found that DAMs are a subset of plaque-associated microglia (PAMs) and their transition to DAM increases with age and disease stage. Spatial transcriptomic analysis revealed that tdTomato+ cells show higher expression of disease and inflammatory genes compared to other microglial populations, including non-labeled PAMs. These models allow either complete cre-loxP targeting of all brain myeloid cells (Cst7Cre), or inducible targeting of DAMs, without leakiness (Cst7 DD-Cre).
Collapse
Affiliation(s)
- Caden M Henningfield
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Minh Ngo
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kaitlin M Murray
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Nellie E Kwang
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kate I Tsourmas
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Zachary A Pashkutz
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Shimako Kawauchi
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Thomas E Lane
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
- Center for Virus Research, University of California, Irvine, USA
| | - Grant R MacGregor
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
9
|
Hou J, Hess JL, Zhang C, van Rooij JGJ, Hearn GC, Fan CC, Faraone SV, Fennema-Notestine C, Lin SJ, Escott-Price V, Seshadri S, Holmans P, Tsuang MT, Kremen WS, Gaiteri C, Glatt SJ. Meta-Analysis of Transcriptomic Studies of Blood and Six Brain Regions Identifies a Consensus of 15 Cross-Tissue Mechanisms in Alzheimer's Disease and Suggests an Origin of Cross-Study Heterogeneity. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33019. [PMID: 39679839 PMCID: PMC12048288 DOI: 10.1002/ajmg.b.33019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
The comprehensive genome-wide nature of transcriptome studies in Alzheimer's disease (AD) should provide a reliable description of disease molecular states. However, the genes and molecular systems nominated by transcriptomic studies do not always overlap. Even when results do align, it is not clear if those observations represent true consensus across many studies. A couple of sources of variation have been proposed to explain this variability, including tissue-of-origin and cohort type, but its basis remains uncertain. To address this variability and extract reliable results, we utilized all publicly available blood or brain transcriptomic datasets of AD, comprised of 24 brain studies with 4007 samples from six different brain regions, and eight blood studies with 1566 samples. We identified a consensus of AD-associated genes across brain regions and AD-associated gene-sets across blood and brain, generalizable machine learning and linear scoring classifiers, and significant contributors to biological diversity in AD datasets. While AD-associated genes did not significantly overlap between blood and brain, our findings highlighted 15 dysregulated processes shared across blood and brain in AD. The top five most significantly dysregulated processes were DNA replication, metabolism of proteins, protein localization, cell cycle, and programmed cell death. Conversely, addressing the discord across studies, we found that large-scale gene co-regulation patterns can account for a significant fraction of variability in AD datasets. Overall, this study ranked and characterized a compilation of genes and molecular systems consistently identified across a large assembly of AD transcriptome studies in blood and brain, providing potential candidate biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jiahui Hou
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jonathan L Hess
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Chunling Zhang
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jeroen G J van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gentry C Hearn
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Chun Chieh Fan
- Department of Cognitive Science, University of California San Diego, La Jolla, California, USA
| | - Stephen V Faraone
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Shu-Ju Lin
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Valentina Escott-Price
- Dementia Research Institute, School of Medicine, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurology and Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Sudha Seshadri
- Department of Neurology, School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Peter Holmans
- Division of Psychological Medicine and Clinical Neurology and Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Ming T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Chris Gaiteri
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Stephen J Glatt
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Public Health and Preventive Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
10
|
He F, Shi WJ, Liu W, Fan JX, He ZG, Zhang YQ, Xiao J, Ruan WW, Gai YK, Zhang HL, Yang BB, Qin Y, Wang H, Li J, Wang JL, Liu S, Shi LP, Chen ZX, Jiang WJ, An N, Xue PJ, Wang ZH, Yang RJ, Tian PY, Chen Z, Xiao L, Yang ZS, Feng KB, Tan WY, Sun ZM, Xu W, Shu H, Wang JZ. A mass-producible macaque model displays a durable Alzheimer-like cognitive deficit and hallmark amyloid-β/tau/neurofilament light chain pathologies. J Alzheimers Dis 2025; 105:1432-1446. [PMID: 40267273 DOI: 10.1177/13872877251334316] [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: 04/25/2025]
Abstract
BackgroundAlzheimer's disease (AD) is the most prevalent neurodegenerative disorder characterized by cognitive deficit and pathological accumulation of amyloid-β (Aβ) and tau proteins. The rodent models have contributed greatly to unravel AD pathogenesis, but these AD models have been shown a modest clinical translational effectiveness.ObjectiveTherefore, developing mass-producible primate AD models is promising for more effective drug development.MethodsHere, we constructed the AD monkey models by simultaneously infusing AAV-Tau and Aβ into different brain regions.ResultsThe induced monkeys showed a durable cognitive impairment lasting for at least 10 months after the modeling. Simultaneously, the increased levels of total tau and hyperphosphorylated tau (pTau) at several AD-associated sites, and neurofilament light chains (NfL) with altered Aβ level were detected at different time points in cerebrospinal fluid and/or plasma by using MSD kits. The increased brain accumulation of Aβ and tau proteins was also detected by positron emission tomography/magnetic resonance imaging and immunohistochemical staining. The model monkeys also had significant glial activation; an indicator of inflammation commonly seen in the brains of AD patients.ConclusionsTogether, this study provides mass-producible monkey models showing durable AD-like hallmark pathologies (Aβ, tau, NfL, i.e., ATN) and cognitive deficits. As monkeys are genetically and metabolically the closest to humans, these models will offer more effective drug discovery and development for AD.
Collapse
Affiliation(s)
- Feng He
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Wen-Jiao Shi
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Wen Liu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| | - Jing-Xin Fan
- Department of Neurosurgery, Hubei Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, P. R. China
| | - Zhi-Gang He
- Department of Emergency Medicine/Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ya-Qi Zhang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Jing Xiao
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Wei-Wei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, P. R. China
| | - Yong-Kang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, P. R. China
| | - Hong-Li Zhang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Bin-Bin Yang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Yao Qin
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Hao Wang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Jia Li
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Jun-Li Wang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Sha Liu
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Li-Ping Shi
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Zhong-Xu Chen
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Wei-Jie Jiang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Ni An
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Peng-Jing Xue
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Zi-Hao Wang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Rui-Jie Yang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Peng-Yu Tian
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Zhu Chen
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Ling Xiao
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Zheng-Sheng Yang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Kang-Bo Feng
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Wei-Ye Tan
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Zhan-Meng Sun
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Wei Xu
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Research Institute, Hubei Topgene Biotechnology, Wuhan, P. R. China
- Department of Pathophysiology, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
11
|
Cao T, Liao P, Lu J, Liang G, Wei Q, Song W, Lan Y, Zeng J, Zou C, Pan M, Su L, Zou D. Single-nucleus RNA sequencing and network pharmacology reveal the mediation of fisetin on neuroinflammation in Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156724. [PMID: 40215814 DOI: 10.1016/j.phymed.2025.156724] [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: 01/06/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by a progressive decline in cognitive function and memory. This study explores cellular subgroups in AD using single-nucleus RNA sequencing (snRNA-seq). It integrates the pharmacological network of traditional Chinese medicine (TCM) to identify potential therapeutic targets, providing a theoretical basis for the development of clinical AD. METHODS We obtained data information from the Gene Expression Omnibus (GEO) for snRNA-seq analysis. Enrichment and pseudotime analysis were performed to explore the functions and differentiation pathways of cellular subgroups. Cellular communication networks were mapped to reveal subgroup interactions. Additionally, a pharmacological network for AD was constructed using the TCM pharmacology database. RESULTS We identified several cell subgroups associated with AD pathology, contributing to disease progression in various ways. Notably, the TNC+ CD44+ astrocyte subgroup activated the I-kappa B kinase/ NF-κB signaling pathway, leading to increased expression of inflammatory cytokines. In the pharmacological network, fisetin was identified as a promising compound with the potential to bind to the CD44 protein, mitigating the inflammatory response and preventing further neuronal damage. CONCLUSIONS By exploring the ecological landscape of various cellular subgroups in AD and investigating the roles and mechanisms, combined with molecular docking and pharmacological network screening, our findings provide new insights and therapeutic possibilities for AD treatment.
Collapse
Affiliation(s)
- Tingting Cao
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Peiling Liao
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China; Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jia Lu
- School of Basic Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Guining Liang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Qingyan Wei
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Wenyi Song
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Chun Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China
| | - Li Su
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, Guangxi 533000, China.
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nannning, Guangxi 530007, China.
| |
Collapse
|
12
|
Liu Y, Gao L, Chen W, Yan Y, Ye Z, Li Z. "Armed in-vitro retina"-generating microglial retinal organoids, where are we now? Front Cell Dev Biol 2025; 13:1574283. [PMID: 40519267 PMCID: PMC12162952 DOI: 10.3389/fcell.2025.1574283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 05/21/2025] [Indexed: 06/18/2025] Open
Abstract
The objective of organoid research is to develop in vitro models that accurately replicate the microenvironment of tissues and organs in vivo. Although techniques for culturing retinal organoids (ROs) have advanced significantly, they still fall short of incorporating all cell types necessary for maintaining retinal homeostasis, particularly immune cells like microglia. Standardizing the inclusion of immune cells in RO cultures would greatly enhance research into the mechanisms underlying retinal diseases and the discovery of therapeutic targets. This review examines recent advancements in co-culturing ROs with immune cells to mimic the physiological and pathological microenvironments of the retina, focusing on tissue structure and function. Furthermore, it emphasizes the importance of cutting-edge organoid technologies, such as microfluidics and organ-on-chip systems, in propelling research in this field. The goal is to equip researchers with a more profound understanding of microglial ROs and their potential applications in scientific investigations.
Collapse
Affiliation(s)
- Yaohong Liu
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wenqian Chen
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Chinese People's Liberation Army Medical School, Beijing, China
| | - Yuhan Yan
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Chinese People's Liberation Army Medical School, Beijing, China
| | - Zi Ye
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- Chinese People's Liberation Army Medical School, Beijing, China
| | - Zhaohui Li
- Department of Ophthalmology, Third Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- Chinese People's Liberation Army Medical School, Beijing, China
| |
Collapse
|
13
|
Takatori S, Kondo M, Tomita T. Unraveling the complex role of microglia in Alzheimer's disease: amyloid β metabolism and plaque formation. Inflamm Regen 2025; 45:16. [PMID: 40448228 PMCID: PMC12123843 DOI: 10.1186/s41232-025-00383-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by amyloid β (Aβ) accumulation in the brain. Recent genome-wide association studies have identified numerous AD risk genes highly expressed in microglia, highlighting their potential role in AD pathogenesis. Although microglia possess phagocytic capacity and have been implicated in Aβ clearance, accumulating evidence suggests their contribution to AD pathogenesis is more complex than initially anticipated. MAIN BODY This review synthesizes current knowledge on microglial Aβ metabolism in AD, reconciling conflicting data from various studies. We examine evidence supporting the role of microglia in Aβ clearance, including studies on AD risk genes like TREM2 and their impact on microglial phagocytosis. Conversely, we explore findings that challenge this view, such as microglial depletion experiments resulting in unchanged or decreased Aβ accumulation. We propose that the contribution of microglia to Aβ metabolism is context-dependent, varying with disease progression, genetic background, and experimental conditions. Notably, microglia may promote parenchymal amyloid accumulation in early disease stages, while this accumulation-promoting effect may diminish in later stages. We discuss potential mechanisms for this paradoxical effect, including intracellular Aβ aggregation and release of pro-aggregation factors. Additionally, we explore the interplay between microglia-mediated Aβ metabolism and other clearance pathways, such as the glymphatic system, highlighting a potential compensatory relationship between parenchymal amyloid deposition and cerebral amyloid angiopathy. CONCLUSION Our review underscores the complex and dynamic role of microglia in AD pathogenesis. Understanding the stage-specific functions of microglia in Aβ metabolism is crucial for developing targeted interventions. Future research should focus on elucidating the mechanisms of microglial functional changes throughout disease progression and determining the pathological significance of these changes. Exploring potential therapeutic strategies that selectively enhance beneficial microglial functions while mitigating their detrimental effects remains an important goal.
Collapse
Affiliation(s)
- Sho Takatori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mayuna Kondo
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
14
|
Mukhina KA, Kechko OI, Osypov AA, Petrushanko IY, Makarov AA, Mitkevich VA, Popova IY. Short-Term Inhibition of NOX2 Prevents the Development of Aβ-Induced Pathology in Mice. Antioxidants (Basel) 2025; 14:663. [PMID: 40563297 DOI: 10.3390/antiox14060663] [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: 04/18/2025] [Revised: 05/20/2025] [Accepted: 05/29/2025] [Indexed: 06/28/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized by the formation of neurotoxic beta-amyloid (Aβ) oligomers in the central nervous system. One of the earliest pathological effects of Aβ is the induction of oxidative stress in brain tissue, mediated by NADPH oxidase 2 (NOX2). This study aimed to determine whether short-term inhibition of NOX2 could disrupt the pathological cascade and prevent the development of Aβ-induced pathology. We demonstrated that suppressing NOX2 activity by GSK2795039 during the first three days after intracerebral Aβ administration prevented the development of the pathological process in mice. Two weeks after the induction of Aβ pathology, animals treated with GSK2795039 showed no neuropsychiatric-like behavioral changes, which correlated with the absence of chronic oxidative damage in brain tissue. Moreover, GSK2795039 prevented microglial activation and reduced microglia-associated neuroinflammation. These findings indicate that short-term NOX2 inhibition effectively suppresses the development of Aβ-induced pathology, suggesting that NOX2 is a potential target for treatment and prevention of AD pathology.
Collapse
Affiliation(s)
- Kristina A Mukhina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Olga I Kechko
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander A Osypov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, 117485 Moscow, Russia
| | - Irina Yu Petrushanko
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina Yu Popova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, 119991 Moscow, Russia
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russia
| |
Collapse
|
15
|
Fołta J, Rzepka Z, Wrześniok D. The Role of Inflammation in Neurodegenerative Diseases: Parkinson's Disease, Alzheimer's Disease, and Multiple Sclerosis. Int J Mol Sci 2025; 26:5177. [PMID: 40507988 PMCID: PMC12154089 DOI: 10.3390/ijms26115177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Neurodegenerative diseases are a group of conditions that have in common the progressive damage and degeneration of neurons in the central nervous system. This group includes Parkinson's disease, Alzheimer's disease, and multiple sclerosis, among others. In recent years, increasing evidence has pointed to the important role of inflammation in the pathogenesis of these conditions. The occurrence of inflammation in the brain, which is often triggered by pro-inflammatory activation of microglia or astrocytes, can consequently lead to a chronic inflammatory response that contributes to the development of neurodegenerative processes. Inflammatory processes themselves, both within the nervous system and throughout the human body, appear to be central to the initiation and progression of neuronal damage. Understanding the role of inflammation in these diseases may open up new perspectives and opportunities in the future in the development of effective therapies to improve patients' quality of life as the vast majority of cases of patients affected by neurodegenerative diseases continue to be treated symptomatically since causal treatments are lacking. In this review, we provide information on the impact of inflammation on the pathogenesis, course, and potential therapeutic options for selected neurodegenerative diseases. In addition, this article provides a general description of neuroinflammation and the involvement and role of specific cells in the central nervous system.
Collapse
Affiliation(s)
| | | | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (J.F.); (Z.R.)
| |
Collapse
|
16
|
Chen W, Liu X, Muñoz VR, Kahn CR. Loss of insulin signaling in microglia impairs cellular uptake of Aβ and neuroinflammatory response exacerbating AD-like neuropathology. Proc Natl Acad Sci U S A 2025; 122:e2501527122. [PMID: 40388612 DOI: 10.1073/pnas.2501527122] [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: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
Insulin receptors are present on cells throughout the body, including the brain. Dysregulation of insulin signaling in neurons and astrocytes has been implicated in altered mood, cognition, and the pathogenesis of Alzheimer's disease (AD). To define the role of insulin signaling in microglia, the primary phagocytes in the brain critical for maintenance and damage repair, we created mice with an inducible microglia-specific insulin receptor knockout (MG-IRKO). RiboTag profiling of microglial mRNAs revealed that loss of insulin signaling results in alterations of gene expression in pathways related to innate immunity and cellular metabolism. In vitro, loss of insulin signaling in microglia results in metabolic reprogramming with an increase in glycolysis and impaired uptake of Aβ. In vivo, MG-IRKO mice exhibit alterations in mood and social behavior, and when crossed with the 5xFAD mouse model of AD, the resultant mice exhibit increased levels of Aβ plaque and elevated neuroinflammation. Thus, insulin signaling in microglia plays a key role in microglial cellular metabolism and the ability of the cells to take up Aβ, such that reduced insulin signaling in microglia alters mood and social behavior and accelerates AD pathogenesis. Together, these data indicate key roles of insulin action in microglia and the potential of targeting insulin signaling in microglia in treatment of AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Xiangyu Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Vitor Rosetto Muñoz
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Laboratory of Molecular Biology of Exercise, University of Campinas, Limeira, São Paulo 13484-350, Brazil
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
17
|
Fan L, Zhang X, Song X, Yang L, Liu H, Zhang Y, Li K, Jin X, Lei H, Zhang Z, Zhang F, Song J. HMGB1/RAGE signaling mediates the activation of microglia and participates in depressive-like behaviors and cognitive deficits in rats after ischemia-reperfusion. Behav Brain Res 2025; 492:115662. [PMID: 40419119 DOI: 10.1016/j.bbr.2025.115662] [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] [Revised: 05/07/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Post-stroke depression (PSD) is a common psychiatric complication that occurs after stroke, especially in ischemic stroke (I/S). It has been reported that high-mobility group box-1 (HMGB1) is highly expressed in clinical PSD patients, but the exact molecular mechanism of its involvement in PSD is not completely clear, the neuroinflammation may participate in its development. Thus, we established a PSD rat model, observed behavioral and cognitive deficits, and found that the HMGB1/ receptor for advanced glycation end products (RAGE) pathway were activated in microglia. Glycyrrhizin acid (GA), an inhibitor of HMGB1, inhibited microglial activation, reversed the expression of HMGB1/RAGE, and ameliorated depressive-like behaviors in PSD rats. GA also reduced the expression of MAPK and NF-κB, which further led to decreased expression of IL-1β and NLRP3 inflammasome. These results suggested that the HMGB1/RAGE pathway was involved in microglial activation in the PSD model, promoting neuroinflammation and depressive-like behaviors.
Collapse
Affiliation(s)
- Lifei Fan
- The First Affiliated Hospital of Xinxiang Medical College, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Xinyue Zhang
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Xiaojia Song
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Lina Yang
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China
| | - Huanhuan Liu
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Yunfei Zhang
- The First Affiliated Hospital of Xinxiang Medical College, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Kun Li
- The First Affiliated Hospital of Xinxiang Medical College, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Xuejiao Jin
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Hao Lei
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Zhaohui Zhang
- The First Affiliated Hospital of Xinxiang Medical College, Xinxiang 453000, China
| | - Fuping Zhang
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China.
| | - Jinggui Song
- The Second Affiliated Hospital of Xinxiang Medical College, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China.
| |
Collapse
|
18
|
Liu X, Baxley S, Hebron M, Turner RS, Moussa C. Resveratrol Attenuates CSF Markers of Neurodegeneration and Neuroinflammation in Individuals with Alzheimer's Disease. Int J Mol Sci 2025; 26:5044. [PMID: 40507855 PMCID: PMC12155158 DOI: 10.3390/ijms26115044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2025] [Revised: 05/16/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by amyloid-beta (Aβ) accumulation and neuroinflammation. A previous multicenter, phase 2, double-blind, placebo-controlled trial randomized 179 participants into placebo or resveratrol over 52 weeks. Sub-analysis of CSF biomarkers of neuronal damage, inflammation, and microglial activity was performed in a subset of patients treated with a placebo (n = 21) versus resveratrol (n = 30). Markers of neuronal damage, including neuron-specific enolase and hyperphosphorylated neurofilaments, were reduced. Microglial activation was measured via a triggering receptor expressed on myeloid cells (TREM)-2 at baseline and after resveratrol treatment. Resveratrol significantly reduced CSF TREM2 levels and decreased inflammation and tissue damage, including matrix metalloprotease (MMP)-9. Cathepsin D, a lysosomal marker of autophagy, was reduced in the resveratrol group compared with placebo, while angiogenin, a marker of vascular angiogenesis, was increased. These data suggest that resveratrol may exert anti-inflammatory and neuroprotective effects in AD by reducing CSF TREM2 and other markers of neuronal damage. Further research is needed to assess the significance of these biomarker changes on clinical outcomes in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
| | - Sean Baxley
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Michaeline Hebron
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
| | - Raymond Scott Turner
- Memory Disorders Program, Department of Neurology, Georgetown University, Washington, DC 20057, USA;
| | - Charbel Moussa
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
| |
Collapse
|
19
|
Gu L, Mao X, Tian C, Yang Y, Yang K, Canfield SG, Zhu D, Gu M, Guo F. Engineering blood-brain barrier microphysiological systems to model Alzheimer's disease monocyte penetration and infiltration. Biomater Sci 2025. [PMID: 40391576 DOI: 10.1039/d5bm00204d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Alzheimer's disease (AD) is a progressive and neurodegenerative disease, predominantly causing dementia. Despite increasing clinical evidence suggesting the involvement of peripheral immune cells such as monocytes in AD pathology, the dynamic penetration and infiltration of monocytes crossing blood-brain barrier (BBB) and inducing neuroinflammation is largely understudied in an AD brain. Herein, we engineer BBB-like microphysiological system (BBB-MPS) models for recapitulating the dynamic penetration and infiltration of monocytes in an AD patient's brain. Each BBB-MPS model can be engineered by integrating a functional BBB-like structure on a human cortical organoid using a 3D-printed device within a well of a plate. By coculturing these BBB-MPS models with monocytes from AD patients and age-matched healthy donors, we found that AD monocytes exhibit significantly greater BBB penetration and brain infiltration compared to age-matched control monocytes. Moreover, we also tested the interventions including Minocycline and Bindarit, and found they can effectively inhibit AD monocyte infiltration, subsequently reducing neuroinflammation and neuronal apoptosis. We believe these scalable and user-friendly BBB-MPS models may hold promising potential in modeling and advancing therapeutics for neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Xiangdi Mao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Yang Yang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Kaiyuan Yang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Scott G Canfield
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Terre Haute, IN, 47809, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, University of Stony Brook, 100 Nicolls Rd, Stony Brook, NY, 11794, USA
| | - Mingxia Gu
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, University of California, Los Angeles, CA 90095, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
20
|
Shen T, Tai W, Jiang D, Ma S, Zhong X, Zou Y, Zhang CL. GADD45G operates as a pathological sensor orchestrating reactive gliosis and neurodegeneration. Neuron 2025:S0896-6273(25)00345-9. [PMID: 40409253 DOI: 10.1016/j.neuron.2025.04.033] [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: 09/05/2024] [Revised: 03/28/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025]
Abstract
Reactive gliosis is a hallmark of neuropathology and offers a potential target for addressing numerous neurological diseases. Here, we show that growth arrest and DNA damage inducible gamma (GADD45G), a stress sensor in astrocytes, is a nodal orchestrator of reactive gliosis and neurodegeneration. GADD45G expression in astrocytes is sufficient to incite astrogliosis, microgliosis, synapse loss, compromised animal behavior, and the aggravation of Alzheimer's disease (AD). Conversely, silencing GADD45G specifically in astrocytes preserves synapses and rescues the histological and behavioral phenotypes of AD. Mechanistically, GADD45G controls the mitogen-activated protein kinase kinase kinase 4 (MAP3K4) and neuroimmune signaling pathways, including nuclear factor κB (NF-κB) and interferon regulatory factor 3 (IRF3), leading to profound molecular changes and the secretion of various factors that regulate both cell-autonomous and cell-nonautonomous reactive gliosis and glia-neuron interactions. These results uncover GADD45G signaling as a promising therapeutic target for AD and potentially for numerous other neurological disorders.
Collapse
Affiliation(s)
- Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dongfang Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
21
|
Ebrahimi R, Shahrokhi Nejad S, Falah Tafti M, Karimi Z, Sadr SR, Ramadhan Hussein D, Talebian N, Esmaeilpour K. Microglial activation as a hallmark of neuroinflammation in Alzheimer's disease. Metab Brain Dis 2025; 40:207. [PMID: 40381069 DOI: 10.1007/s11011-025-01631-9] [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: 03/24/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
Microglial activation has emerged as a hallmark of neuroinflammation in Alzheimer's disease (AD). Central to this process is the formation and accumulation of amyloid beta (Aβ) peptide and neurofibrillary tangles, both of which contribute to synaptic dysfunction and neuronal cell death. Aβ oligomers trigger microglial activation, leading to the release of pro-inflammatory cytokines, which further exacerbates neuroinflammation and neuronal damage. Importantly, the presence of activated microglia surrounding amyloid plaques is correlated with heightened production of cytokines such as interleukin (IL)-1β and tumor necrosis factor-alpha (TNF-α), creating a vicious cycle of inflammation. While microglia play a protective role by clearing Aβ plaques during the early stages of AD, their chronic activation can lead to detrimental outcomes, including enhanced tau pathology and neuronal apoptosis. Recent studies have highlighted the dualistic nature of microglial activation, showcasing both inflammatory (M1) and anti-inflammatory (M2) phenotypes that fluctuate based on the surrounding microenvironment. Disruption in microglial function and regulation can lead to neurovascular dysfunction, further contributing to the cognitive decline seen in AD. Moreover, emerging biomarkers and imaging techniques are unveiling the complexity of microglial responses in AD, providing avenues for targeted therapeutics aimed at modulating these cells. Understanding the intricate interplay between microglia, Aβ, and tau pathology is vital for developing potential interventions to mitigate neuroinflammation and its impact on cognitive decline in AD. This review synthesizes current findings regarding microglial activation and its implications for AD pathogenesis, offering insights into future therapeutic strategies.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mahdi Falah Tafti
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Karimi
- Ross and Carol Nese College of Nursing, Pennsylvania State University, University Park, PA, USA
| | - Seyyedeh Reyhaneh Sadr
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Niki Talebian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khadijeh Esmaeilpour
- Department of Psychology, University of Toronto Mississagua, Mississauga, ON, Canada.
| |
Collapse
|
22
|
Park Y, Moon S, Jung H, Park S, Kim JW, Song DG, In YH, Han SW, Sohn JH, Lee CH. Mirodenafil improves cognitive function by reducing microglial activation and blood-brain barrier permeability in ApoE4 KI mice. Front Aging Neurosci 2025; 17:1579411. [PMID: 40443793 PMCID: PMC12119498 DOI: 10.3389/fnagi.2025.1579411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Alzheimer's disease (AD) has significant public health concerns in the aging society. AD can compromise brain function and lead to severe neurological abnormalities associated with dementia. The human Apolipoprotein E (ApoE4) gene is a strong risk factor for AD. However, comprehensive analyses and improvements of mouse models expressing ApoE4 remain largely unexplored. Methods ApoE4 knock-in (KI) mice were used to investigate the role of humanized ApoE4 in hippocampal histological changes and cognitive impairment. Cerebrovascular perfusion, blood-brain barrier (BBB) integrity, microgliosis, and amyloid-beta 42 (Aβ42) accumulation were examined. Cognitive functions were assessed using the Morris water maze, Y-maze, and novel object recognition tests. Mirodenafil, a potent and selective phosphodiesterase 5 inhibitor (PDE5i), was orally administered to ApoE4 KI mice for 4 weeks. An in vitro BBB model and BV2 microglial cells were used to investigate endothelial permeability and inflammation. Results ApoE4 KI mice exhibited not only reduced cerebrovascular perfusion and CLN-5 expression but also increased microgliosis and Aβ42 accumulation in the hippocampus. These phenomena were accompanied by impaired cognitive functions. Mirodenafil administration reversed the histological and behavioral alterations induced by ApoE4 KI. In vitro, mirodenafil treatment mitigated Aβ42-induced endothelial permeability and lipopolysaccharide-induced microglial inflammation. Discussion These findings suggest that mirodenafil enhances cerebrovascular function, preserves BBB integrity, and mitigates neuroinflammation in ApoE4 KI mice, leading to cognitive improvement. PDE5 inhibition may serve as a promising therapeutic approach for addressing ApoE4-associated cerebrovascular and cognitive dysfunction.
Collapse
Affiliation(s)
- Yejin Park
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Subin Moon
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Harry Jung
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Songyi Park
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Ju Won Kim
- AriBio Co., Ltd., Seongnam-si, Gyeonggi-Do, Republic of Korea
| | - Dan-Gyeong Song
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Ho In
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sang Won Han
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Jong-Hee Sohn
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Chan Hee Lee
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
23
|
Gauron MC, Prokopenko D, Lee S, Wolfe SA, Hecker J, Willett J, Waqas M, Lordén G, Yang Y, Mayfield JE, Castanho I, Mullin K, Morgan S, Hahn G, Demeo DL, Hide W, Bertram L, Lange C, Newton AC, Tanzi RE. Protein kinase C eta enhances Golgi-localized signaling and is associated with Alzheimer's disease using a recessive mode of inheritance. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.13.25327562. [PMID: 40463529 PMCID: PMC12132147 DOI: 10.1101/2025.05.13.25327562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The identification of Alzheimer's disease (AD)-associated genomic variants has provided powerful insight into disease etiology. Genome-wide association studies (GWAS) for AD have successfully identified new targets but have almost exclusively utilized additive genetic models. Here, we performed a family-based GWAS under a recessive inheritance model using whole genome sequencing from families affected by AD. We found that the variant, rs7161410, located in an intron of the PRKCH gene, encoding protein kinase C eta (PKCη), was associated with AD risk (p-value=1.41 × 10-7). Further analysis revealed a rare PRKCH missense mutation K65R in linkage disequilibrium with rs7161410, which was present in homozygous carriers of the rs7161410 risk allele. We show that this mutation leads to enhanced localization and signaling of PKCη at the Golgi. The novel genetically-validated association of aberrant PKCη signaling with AD opens avenues for new therapeutic targets aimed at prevention and treatment.
Collapse
Affiliation(s)
- Maria Celeste Gauron
- Department of Pharmacology, University of California; San Diego, La Jolla, CA, USA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital; Charlestown, MA, USA
- Harvard Medical School; Boston, MA, USA
| | - Sanghun Lee
- Department of Medical Consilience, Graduate School, Dankook University; South Korea
- Department of Biostatistics, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Sarah A. Wolfe
- Department of Pharmacology, University of California; San Diego, La Jolla, CA, USA
| | - Julian Hecker
- Harvard Medical School; Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital; Boston, MA, USA
| | - Julian Willett
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital; Charlestown, MA, USA
- Harvard Medical School; Boston, MA, USA
| | - Mohammad Waqas
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital; Charlestown, MA, USA
| | - Gema Lordén
- Department of Pharmacology, University of California; San Diego, La Jolla, CA, USA
| | - Yimin Yang
- Department of Pharmacology, University of California; San Diego, La Jolla, CA, USA
| | - Joshua E. Mayfield
- Department of Pharmacology, University of California; San Diego, La Jolla, CA, USA
| | - Isabel Castanho
- Harvard Medical School; Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center; Boston, MA, USA
| | - Kristina Mullin
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital; Charlestown, MA, USA
| | - Sarah Morgan
- Department of Pathology, Beth Israel Deaconess Medical Center; Boston, MA, USA
- Blizard Institute, Department of Neuroscience, Surgery and Trauma, Queen Mary University of London; London, United Kingdom
| | - Georg Hahn
- Department of Biostatistics, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Dawn L. Demeo
- Harvard Medical School; Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital; Boston, MA, USA
| | - Winston Hide
- Harvard Medical School; Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center; Boston, MA, USA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck; Lübeck, Germany
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo; Oslo, Norway
| | - Christoph Lange
- Harvard Medical School; Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health; Boston, MA, USA
| | - Alexandra C. Newton
- Department of Pharmacology, University of California; San Diego, La Jolla, CA, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital; Charlestown, MA, USA
- Harvard Medical School; Boston, MA, USA
| |
Collapse
|
24
|
Hermine O, Gros L, Tran TA, Loussaief L, Flosseau K, Moussy A, Mansfield CD, Vermersch P. Tyrosine kinase inhibitor, masitinib, limits neuronal damage, as measured by serum neurofilament light chain concentration in a model of neuroimmune-driven neurodegenerative disease. PLoS One 2025; 20:e0322199. [PMID: 40367050 PMCID: PMC12077730 DOI: 10.1371/journal.pone.0322199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/17/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Masitinib is an orally administered tyrosine kinase inhibitor that targets activated cells of the innate neuroimmune system. We have studied the neuroprotective action of masitinib on the manifestations of experimental autoimmune encephalitis (EAE) induced axonal and neuronal damage. EAE is a model of neuroimmune-driven chronic neuroinflammation and therefore highly relevant to masitinib's mechanism of action in neurodegenerative diseases. Importantly, neuronal damage, or prevention thereof, can be rapidly assessed by measuring serum neurofilament light chain (NfL) concentration in EAE-induced mice. METHODS EAE induction was performed in healthy female C57BL/6 mice via active MOG 35-55 peptide immunization. Treatments were initiated 14 days post EAE induction. On day-0, 39 mice with established EAE symptoms were randomly assigned to 3 treatment groups (n = 13): EAE control, masitinib 50 mg/kg/day (M50), and masitinib 100 mg/kg/day (M100). The treatment started on day-1 and ended on day-15. Blood samples were collected on day-1 and day-8, via tail vein sampling, and on day-15, via intracardiac puncture. Assessments included quantification of serum NfL levels along the disease duration, cytokine quantification at day-15, and clinical assessments. RESULTS Masitinib treatment significantly (p < 0.0001) limited NfL production with respect to control; specifically, relative change in serum NfL concentration at day-8 was 43% and 60% lower for the M50 and M100 groups, respectively. Likewise, for the assessment of absolute serum NfL at day-8 and day-15, there was a significantly lower NfL concentration for masitinib treatment as compared with control. Furthermore, EAE mice treated with masitinib showed significantly lower concentrations of several well-established pro-inflammatory cytokines relative to control at day-15. A beneficial effect of masitinib on functional performance was also observed, with both M50 and M100 groups showing significantly less relative deterioration in grip strength at day-15 as compared with control (p < 0.001). CONCLUSION This study is the first demonstration that masitinib, a drug that targets the innate as opposed to the adaptive neuroimmune system, can lower serum NfL levels, and by extension therefore, neuronal damage, in a neuroimmune-driven neurodegenerative disease model. Overall, findings indicate that masitinib has a neuroprotective effect under conditions of chronic neuroinflammation and therefore plausible disease-modifying activity across a broad range of neurodegenerative diseases.
Collapse
MESH Headings
- Animals
- Female
- Mice
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Neurofilament Proteins/blood
- Benzamides
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Mice, Inbred C57BL
- Disease Models, Animal
- Neurons/drug effects
- Neurons/pathology
- Thiazoles/pharmacology
- Thiazoles/therapeutic use
- Neurodegenerative Diseases/drug therapy
- Neurodegenerative Diseases/blood
- Neurodegenerative Diseases/pathology
- Pyridines
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Tyrosine Kinase Inhibitors
- Piperidines
Collapse
Affiliation(s)
- Olivier Hermine
- Imagine Institute, INSERM UMR 1163, University of Paris, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implication, Hôpital Necker, Paris, France
- Department of Hematology, Necker Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | | | | | | | | | | | - Patrick Vermersch
- Univ. Lille, UMR Inserm U1172, CHU Lille, FHU Precise, Lille, France
| |
Collapse
|
25
|
Ding S, Choi SH, Miller YI. Amyloid β-Induced Inflammarafts in Alzheimer's Disease. Int J Mol Sci 2025; 26:4592. [PMID: 40429737 PMCID: PMC12111532 DOI: 10.3390/ijms26104592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The formation of amyloid beta (Aβ) plaques is a central process in the development of Alzheimer's disease (AD). Although its causative role or the effectiveness of therapeutic targeting is still debated, the key involvement of Aβ in the pathogenesis of neuroinflammation and neurodegeneration in AD is broadly accepted. In this review, we emphasize the role of lipid rafts, both in APP cleavage producing Aβ in neurons and in mediating Aβ inflammatory signaling in microglia. We introduce the term inflammarafts to characterize the Aβ-driven formation of enlarged, cholesterol-rich lipid rafts in activated microglia, which support protein-protein and lipid-protein interactions of inflammatory receptors. Examples reviewed include toll-like receptors (TLR2, TLR4), scavenger receptors (CD36, RAGE), and TREM2. The downstream pathways lead to the production of cytokines and reactive oxygen species, intensifying neuroinflammation and resulting in neuronal injury and cognitive decline. We further summarize emerging therapeutic strategies and emphasize the utility of apolipoprotein A-I binding protein (AIBP) in selective targeting of inflammarafts and attenuation of microglia-driven inflammation. Unlike the targeting of a single inflammatory receptor or a secretase, selective disruption of inflammarafts and preservation of physiological lipid rafts offer a novel approach to targeting multiple components and processes that contribute to neuroinflammation in AD.
Collapse
Affiliation(s)
| | | | - Yury I. Miller
- Department of Medicine, University of California, San Diego, CA 92093, USA; (S.D.); (S.-H.C.)
| |
Collapse
|
26
|
Han X, Cao X, Ju Q, Ge C, Lin Y, Shi J, Zhang X, Sun C, Li H. Microglial TAK1 promotes neurotoxic astrocytes and cognitive impairment in LPS-induced hippocampal neuroinflammation. J Biol Chem 2025:110225. [PMID: 40349778 DOI: 10.1016/j.jbc.2025.110225] [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: 11/25/2024] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
The peripheral immune system has a strong effect on the central nervous system (CNS). Systemic lipopolysaccharides (LPS) administration triggers robust microglial activation and induces significant inflammatory responses in the hippocampus. This study investigates the role of Transforming Growth Factor-β-Activated Kinase 1 (TAK1) in mediating LPS-induced hippocampal neuroinflammation and cognitive impairment. Our findings reveal that LPS induces activation of microglial TAK1, which in turn actives downstream effector NF-κB/p65 to release pro-inflammatory cytokines. The activated microglia also promote astrocytes to polarize into a neurotoxic phenotype (A1-like phenotype), and cause the loss of newborn neurons in the hippocampal dentate gyrus (DG). However, TAK1 reduction inhibits microglial responses, limits neurotoxic astrocytes, rescues newborn neurons, and subsequently improves LPS-induced cognitive deficits, suggesting that targeting TAK1 may be an effective strategy for alleviating neuroinflammation. The interaction between TAK1 activation, microglial responses, and the transition of neurotoxic astrocytes enhances our understanding of the cellular dynamics driving LPS-induced neuroinflammation, suggesting that TAK1 may be a therapeutic target for treating cognitive impairment.
Collapse
Affiliation(s)
- Xiao Han
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Xin Cao
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Qianqian Ju
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Chengxin Ge
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yongqi Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Jinhong Shi
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinhua Zhang
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| | - Haoming Li
- Department of Human Anatomy, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
27
|
Wang X, Zhang H, Wan Z, Li X, Ibáñez CF, Xie M. A single-cell transcriptomic atlas of all cell types in the brain of 5xFAD Alzheimer mice in response to dietary inulin supplementation. BMC Biol 2025; 23:124. [PMID: 40346662 PMCID: PMC12065180 DOI: 10.1186/s12915-025-02230-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease that is a major threat to the aging population. Due to lack of effective therapy, preventive treatments are important strategies to limit AD onset and progression, of which dietary regimes have been implicated as a key factor. Diet with high fiber content is known to have beneficial effects on cognitive decline in AD. However, a global survey on microbiome and brain cell dynamics in response to high fiber intake at single-cell resolution in AD mouse models is still missing. RESULTS Here, we show that dietary inulin supplementation synergized with AD progression to specifically increase the abundance of Akkermansia muciniphila in gut microbiome of 5 × Familial AD (FAD) mice. By performing single-nucleus RNA sequencing on different regions of the whole brain with three independent biological replicates, we reveal region-specific changes in the proportion of neuron, astrocyte, and granule cell subpopulations upon inulin supplementation in 5xFAD mice. In addition, we find that astrocytes have more pronounced region-specific diversity than microglia. Intriguingly, such dietary change reduces amyloid-β plaque burden and alleviates microgliosis in the forebrain region, without affecting the spatial learning and memory. CONCLUSIONS These results provide a comprehensive overview on the transcriptomic changes in individual cells of the entire mouse brain in response to high fiber intake and a resourceful foundation for future mechanistic studies on the influence of diet and gut microbiome on the brain during neurodegeneration.
Collapse
Affiliation(s)
- Xiaoyan Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Chinese Institute for Brain Research, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Houyu Zhang
- Chinese Institute for Brain Research, Zhongguancun Life Science Park, Beijing, 102206, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Zhou Wan
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xuetong Li
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Carlos F Ibáñez
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Chinese Institute for Brain Research, Zhongguancun Life Science Park, Beijing, 102206, China.
- School of Life Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, China.
- Department of Neuroscience, Karolinska Institute, 17165, Stockholm, Sweden.
| | - Meng Xie
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, China.
- Beijing Key Laboratory of Behavior and Mental Health, School of Psychological and Cognitive Sciences, Peking University, Beijing, 100871, China.
- Department of Medicine Huddinge, Karolinska Institute, 14183, Stockholm, Sweden.
| |
Collapse
|
28
|
Ji Y, Chen X, Wang Z, Meek CJ, McLean JL, Yang Y, Yuan C, Rochet JC, Liu F, Xu R. Alzheimer's disease patient brain extracts induce multiple pathologies in novel vascularized neuroimmune organoids for disease modeling and drug discovery. Mol Psychiatry 2025:10.1038/s41380-025-03041-w. [PMID: 40316675 DOI: 10.1038/s41380-025-03041-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 04/10/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
Alzheimer's Disease (AD) is the most common cause of dementia, afflicting 55 million individuals worldwide, with limited treatment available. Current AD models mainly focus on familial AD (fAD), which is due to genetic mutations. However, models for studying sporadic AD (sAD), which represents over 95% of AD cases without specific genetic mutations, are severely limited. Moreover, the fundamental species differences between humans and animals might significantly contribute to clinical failures for AD therapeutics that have shown success in animal models, highlighting the urgency to develop more translational human models for studying AD, particularly sAD. In this study, we developed a complex human pluripotent stem cell (hPSC)-based vascularized neuroimmune organoid model, which contains multiple cell types affected in human AD brains, including human neurons, microglia, astrocytes, and blood vessels. Importantly, we demonstrated that brain extracts from individuals with sAD can effectively induce multiple AD pathologies in organoids four weeks post-exposure, including amyloid beta (Aβ) plaque-like aggregates, tau tangle-like aggregates, neuroinflammation, elevated microglial synaptic pruning, synapse/neuronal loss, and impaired neural network activity. Proteomics analysis also revealed disrupted AD-related pathways in our vascularized AD neuroimmune organoids. Furthermore, after treatment with Lecanemab, an FDA-approved antibody drug targeting Aβ, AD brain extracts exposed organoids showed a significant reduction of amyloid burden, along with an elevated vascular inflammation response. Thus, the vascularized neuroimmune organoid model provides a unique opportunity to study AD, particularly sAD, under a pathophysiological relevant three-dimensional (3D) human cell environment. It also holds great promise to facilitate AD drug development, particularly for immunotherapies.
Collapse
Affiliation(s)
- Yanru Ji
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, IN, 47907, USA
| | - Xiaoling Chen
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, IN, 47907, USA
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Zhen Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Connor Joseph Meek
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Jenna Lillie McLean
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Yang Yang
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, IN, 47907, USA
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Chongli Yuan
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, IN, 47907, USA
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jean-Christophe Rochet
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, IN, 47907, USA
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY, 10314, USA
| | - Ranjie Xu
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
29
|
Augusto-Oliveira M, Arrifano GDP, Leal-Nazaré CG, Chaves-Filho A, Santos-Sacramento L, Lopes-Araujo A, Tremblay MÈ, Crespo-Lopez ME. Morphological diversity of microglia: Implications for learning, environmental adaptation, ageing, sex differences and neuropathology. Neurosci Biobehav Rev 2025; 172:106091. [PMID: 40049541 DOI: 10.1016/j.neubiorev.2025.106091] [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: 11/06/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Microglia are the brain resident macrophages that respond rapidly to any insult. These non-neuroectodermal cells are decorated with plenty of receptors allowing them to recognise and respond precisely to a multitude of stimuli. To do so, microglia undergo structural and functional changes aiming to actively keep the brain's homeostasis. However, some microglial responses, when sustained or exacerbated, can contribute to neuropathology and neurodegeneration. Many microglial molecular and cellular changes were identified that display a strong correlation with neuronal damage and neuroinflammation/disease status, as well as present key sex-related differences that modulate microglial outcomes. Nevertheless, the relationship between microglial structural and functional features is just beginning to be unravelled. Several reports show that microglia undergo soma and branch remodelling in response to environmental stimuli, ageing, neurodegenerative diseases, trauma, and systemic inflammation, suggesting a complex form and function link. Also, it is reasonable overall to suppose that microglia diminishing their process length and ramification also reduce their monitoring activity of synapses, which is critical for detecting any synaptic disturbance and performing synaptic remodelling. Elucidating the complex interactions between microglial morphological plasticity and its functional implications appears essential for the understanding of complex cognitive and behavioural processes in health and neuropathological conditions.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| | - Gabriela de Paula Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Amanda Lopes-Araujo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec, Qubec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada; College Member of the Royal Society of Canada, Canada.
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| |
Collapse
|
30
|
Chauhan P, Begum MY, Narapureddy BR, Gupta S, Wadhwa K, Singh G, Kumawat R, Sharma N, Ballal S, Jha SK, Abomughaid MM, B D, Ojha S, Jha NK. Unveiling the Involvement of Herpes Simplex Virus-1 in Alzheimer's Disease: Possible Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:5850-5874. [PMID: 39648189 DOI: 10.1007/s12035-024-04535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Viruses pose a significant challenge and threat to human health, as demonstrated by the current COVID-19 pandemic. Neurodegeneration, particularly in the case of Alzheimer's disease (AD), is significantly influenced by viral infections. AD is a neurodegenerative disease that affects people of all ages and poses a significant threat to millions of individuals worldwide. The precise mechanism behind its development is not yet fully understood; however, the emergence and advancement of AD can be hastened by various environmental factors, such as bacterial and viral infections. There has been a longstanding suspicion that the herpes simplex virus-1 (HSV-1) may have a role to play in the development or advancement of AD. Reactivation of HSV-1 could potentially lead to damage to neurons, either by direct means or indirectly by triggering inflammation. This article provides an overview of the connection between HSV-1 infections and immune cells (astrocytes, microglia, and oligodendrocytes) in the progression of AD. It summarizes recent scientific research on how HSV-1 affects neurons, which could potentially shed light on the clinical features and treatment options for AD. In addition, the paper has explored the impact of HSV-1 on neurons and its role in various aspects of AD, such as Aβ secretion, tau hyperphosphorylation, metabolic dysregulation, oxidative damage, apoptosis, and autophagy. It is believed that the immune response triggered by HSV-1 reactivation plays a role in the development of neurodegeneration in AD. Despite the lack of a cure for AD, researchers have made significant efforts to study the clinical and pathological aspects of the disease, identify biomarkers, and gain insight into its underlying causes. The goal is to achieve early diagnosis and develop treatments that can modify the progression of the disease. The current article discusses the most promising therapy for combating the viral impacts, which provides additional evidence for the frequent reactivations of latent HSV-1 in the AD brain. However, further research is still required to establish the molecular and cellular mechanisms underlying the development of AD through the reactivation of HSV-1. This could potentially lead to new insights in drug development aimed at preventing HSV-1 reactivation and the subsequent development and progression of AD.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajsthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Suhas Ballal
- Departmant of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Dheepak B
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology, Galgotias University, Greater Noida, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
31
|
King KE, Haeri M, Swerdlow RH, Wozniak AL. RILP cleavage links an inflammatory state to enhanced tau propagation in a cell culture model of Alzheimer's disease. Mol Biol Cell 2025; 36:br15. [PMID: 40137558 PMCID: PMC12086573 DOI: 10.1091/mbc.e24-04-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by the progressive spread of tau pathology throughout the brain. Inflammation has been demonstrated to be present in the disease state as well as changes in endocytic trafficking. Here we identify the Rab7 effector RILP, a protein at the intersection of inflammatory states and endocytic trafficking, as a novel player in tau propagation. We show that RILP is cleaved in AD brain and this cleavage correlates to increases in hyperphosphorylated tau. Cleavage can be induced in both BE(2) neuron-like cells as well as a microglia cell line when they are treated with the inflammatory mediators lipopolysaccharide (LPS) and ATP. This inflammatory state also enhances tau propagation between BE(2) cells, an effect that is mitigated by overexpressing a noncleavable RILP. Furthermore, microglial cells contribute to intercellular tau propagation through both the release of inflammation-associated factors and the direct uptake and secretion of tau, potentially via extracellular vesicles (EVs). In HMC3 microglial cells, RILP cleavage led to impaired tau degradation, increasing intracellular tau accumulation. Additionally, the RILP cleavage status influences EV secretion in microglia. These findings suggest that RILP cleavage alters the endocytic trafficking of tau causing increased cell-cell propagation in a cell-culture model of AD.
Collapse
Affiliation(s)
- Kayla E. King
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Liver Center, University of Kansas Medical Center, Kansas City, KS 66160
| | - Mohammad Haeri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, KS 66160
| | - Russell H. Swerdlow
- Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Ann L. Wozniak
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Liver Center, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
32
|
Shwab EK, Man Z, Gingerich DC, Gamache J, Garrett ME, Serrano GE, Beach TG, Crawford GE, Ashley‐Koch AE, Chiba‐Falek O. Comparative mapping of single-cell transcriptomic landscapes in neurodegenerative diseases. Alzheimers Dement 2025; 21:e70012. [PMID: 40344336 PMCID: PMC12061851 DOI: 10.1002/alz.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease (PD) represent a spectrum of neurodegenerative diseases (NDDs). Here, we performed the first direct comparison of their transcriptomic landscapes. METHODS We profiled whole transcriptomes of NDD cortical tissue by single-nucleus RNA sequencing, using computational analyses to identify common and distinct differentially expressed genes (DEGs), pathways, vulnerable and disease-driver cell subtypes, and altered cell-to-cell interactions. RESULTS The same inhibitory neuron subtype was depleted in both AD and DLB. Potentially disease-driving neuronal cell subtypes were identified in both PD and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. DEGs were most commonly shared across NDDs within inhibitory neuron subtypes. Overall, AD and PD showed greatest transcriptomic divergence, while DLB exhibited an intermediate signature. DISCUSSION These results may help explain the clinicopathological spectrum of these NDDs and provide unique insights into shared and distinct molecular mechanisms underlying pathogenesis. HIGHLIGHTS The same vulnerable inhibitory neuron subtype population was depleted in both Alzheimer's disease (AD) and dementia with Lewy bodies (DLB). Potentially disease-driving neuronal cell subtypes were discovered in both Parkinson's disease (PD) and DLB. Cell-cell communication was predicted to be increased in AD but decreased in DLB and PD. Differentially expressed genes were most commonly shared across neurodegenerative diseases in inhibitory neuron types. AD and PD had the greatest transcriptomic divergence, with DLB showing an intermediate signature.
Collapse
Affiliation(s)
- E. Keats Shwab
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Zhaohui Man
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Daniel C. Gingerich
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Julia Gamache
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Melanie E. Garrett
- Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNorth CarolinaUSA
| | | | | | - Gregory E. Crawford
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of PediatricsDivision of Medical GeneticsDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Advanced Genomic TechnologiesDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Allison E. Ashley‐Koch
- Duke Molecular Physiology InstituteDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of MedicineDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ornit Chiba‐Falek
- Division of Translational Brain SciencesDepartment of NeurologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Center for Genomic and Computational BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| |
Collapse
|
33
|
Canet G, Zussy C, Vitalis M, Morin F, Chevallier N, Hunt H, Claeysen S, Blaquière M, Marchi N, Planel E, Meijer OC, Desrumaux C, Givalois L. Advancing Alzheimer's disease pharmacotherapy: efficacy of glucocorticoid modulation with dazucorilant (CORT113176) in preclinical mouse models. Br J Pharmacol 2025; 182:1930-1956. [PMID: 39891319 DOI: 10.1111/bph.17457] [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: 08/27/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND AND PURPOSE Exposure to chronic stress and high levels of glucocorticoid hormones in adulthood has been associated with cognitive deficits and increased risk of Alzheimer's disease (AD). Dazucorilant has recently emerged as a selective glucocorticoid receptor (NR3C1) modulator, exhibiting efficacy in counteracting amyloid-β toxicity in an acute model of AD. We aim to assess the therapeutic potential of dazucorilant in reversing amyloid and tau pathologies through the inhibition of glucocorticoid receptor pathological activity, and providing additional evidence for its consideration in AD treatment. EXPERIMENTAL APPROACH The efficacy of dazucorilant was evaluated in two transgenic mouse models of amyloid pathology. The slowly progressing J20 and the aggressively pathological 5xFAD mice. Behavioural analysis was conducted to evaluate welfare, cognitive performances and anxiety levels. The activity of the glucocorticoid receptor system, neuroinflammation, amyloid burden and tau phosphorylation were examined in hippocampi. KEY RESULTS In both AD models, chronic treatment with dazucorilant improved working and long-term spatial memories along with the inhibition of glucocorticoid receptor-dependent pathogenic processes and the normalization of plasma glucocorticoid levels. Dazucorilant treatment also resulted in a reduction in tau hyperphosphorylation and amyloid production and aggregation. Additionally, dazucorilant seemed to mediate a specific re-localization of activated glial cells onto amyloid plaques in J20 mice, suggesting a restoration of physiological neuroinflammatory processes. CONCLUSION AND IMPLICATIONS Dazucorilant exhibited sustained disease-modifying effects in two AD models. Given that this compound has demonstrated safety and tolerability in human subjects, our results provide pre-clinical support for conducting clinical trials to evaluate its potential in AD.
Collapse
Affiliation(s)
- Geoffrey Canet
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
| | - Charleine Zussy
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
| | - Mathieu Vitalis
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
| | - Françoise Morin
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
| | | | - Hazel Hunt
- Corcept Therapeutics, Menlo Park, California, USA
| | | | | | - Nicola Marchi
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Emmanuel Planel
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
| | - Onno C Meijer
- Einthoven Laboratory, Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catherine Desrumaux
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
- LIPSTIC LabEx, Dijon, France
| | - Laurent Givalois
- MMDN, Univ Montpellier, EPHE-PSL, INSERM, Montpellier, France
- Faculty of Medicine, Department of Psychiatry and Neurosciences, CR-CHUQ, Laval University, Québec City, Quebec, Canada
- CNRS, Paris, France
| |
Collapse
|
34
|
Yu Y, Lv J, Ma D, Han Y, Zhang Y, Wang S, Wang Z. Microglial ApoD-induced NLRC4 inflammasome activation promotes Alzheimer's disease progression. Animal Model Exp Med 2025; 8:773-783. [PMID: 38520135 DOI: 10.1002/ame2.12361] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/20/2023] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease with no effective therapies. It is well known that chronic neuroinflammation plays a critical role in the onset and progression of AD. Well-balanced neuronal-microglial interactions are essential for brain functions. However, determining the role of microglia-the primary immune cells in the brain-in neuroinflammation in AD and the associated molecular basis has been challenging. METHODS Inflammatory factors in the sera of AD patients were detected and their association with microglia activation was analyzed. The mechanism for microglial inflammation was investigated. IL6 and TNF-α were found to be significantly increased in the AD stage. RESULTS Our analysis revealed that microglia were extensively activated in AD cerebra, releasing sufficient amounts of cytokines to impair the neural stem cells (NSCs) function. Moreover, the ApoD-induced NLRC4 inflammasome was activated in microglia, which gave rise to the proinflammatory phenotype. Targeting the microglial ApoD promoted NSC self-renewal and inhibited neuron apoptosis. These findings demonstrate the critical role of ApoD in microglial inflammasome activation, and for the first time reveal that microglia-induced inflammation suppresses neuronal proliferation. CONCLUSION Our studies establish the cellular basis for microglia activation in AD progression and shed light on cellular interactions important for AD treatment.
Collapse
Affiliation(s)
- Yaliang Yu
- Department of Neurology, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| | - Jianzhou Lv
- Department of Neurology, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| | - Dan Ma
- Department of Neurology, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| | - Ya Han
- Department of Neurology, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| | - Yaheng Zhang
- Department of Neurology, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| | - Shanlong Wang
- Clinical Lab, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| | - Zhitao Wang
- Department of Neurology, The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, P. R. China
| |
Collapse
|
35
|
Li B, Wang L, Xiao Y, Wang Y, Wang Y, Peng Y, Zhang A, Tang Z, Qi X. Gastrodin Ameliorates Tau Pathology and BBB Dysfunction in 3xTg-AD Transgenic Mice by Regulating the ADRA1/NF-κB/NLRP3 Pathway to Reduce Neuroinflammation. Phytother Res 2025; 39:1996-2016. [PMID: 39963073 DOI: 10.1002/ptr.8461] [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: 11/16/2024] [Revised: 01/13/2025] [Accepted: 02/08/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND AND AIM Gastrodin, an active compound derived from the traditional Chinese herbal medicine Gastrodia, demonstrates a variety of pharmacological effects, particularly in the enhancement of neural functions. Thus, the aim of this study is to explore the therapeutic effects of gastrodin on Alzheimer's disease (AD) and its underlying molecular mechanisms. EXPERIMENTAL PROCEDURE Cognitive function was assessed via Morris water maze and Y-maze tests. Tau pathology, neuroinflammation, and BBB dysfunction were analyzed using various techniques, including Western blot, immunohistochemistry, and ELISA. ADRA1 overexpression was induced by lentiviral infection, and gastrodin's impact on NF-κB p65, NLRP3, IL-1β, and IL-18 levels was evaluated. KEY RESULTS In the in vivo experiment, gastrodin enhanced learning and spatial memory in 3xTg-AD mice, as well as reducing p-Tau protein expression in the hippocampus and cortex. Gastrodin inhibited the ADRA1/NF-κB/NLRP3 pathway, which decreased glial cell activation and inflammatory cytokines IL-1β and IL-18, improving neuron and BBB function. In the in vitro experiment, gastrodin inhibited the activation of the NF-κB/NLRP3 pathway due to ADRA1 overexpression and prevented the Aβ42-induced increase in ADRA1/NF-κB/NLRP3 protein expression in SH-SY5Y cells. It also reduced IL-1β and IL-18 cytokine release, restoring tight junction protein expression in bEnd.3 cells. CONCLUSIONS AND IMPLICATIONS gastrodin ameliorates learning and memory abilities by alleviating neuroinflammation and tau pathology, restoring the structure and function of neurons and BBB, suggesting that gastrodin may serve as an effective drug for the treatment of AD.
Collapse
Affiliation(s)
- Bo Li
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Li Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
- School of Nursing, Guizhou Medical University, Guiyang, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Yang Wang
- The Department of Imaging, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yuanshang Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Yaqian Peng
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Anni Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
- The Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhi Tang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Constructed by the Province and Ministry, Guiyang, China
| |
Collapse
|
36
|
Doroszkiewicz J, Winkel I, Mroczko B. Comparative analysis of neuroinflammatory pathways in Alzheimer's disease, Parkinson's disease, and multiple sclerosis: insights into similarities and distinctions. Front Neurosci 2025; 19:1579511. [PMID: 40364858 PMCID: PMC12069400 DOI: 10.3389/fnins.2025.1579511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Neurodegenerative diseases, contributing to the significant socioeconomic burden due to aging society, are gaining increasing interest. Despite each disease having different etiologies, neuroinflammation is believed to play a crucial role in Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). In addition to the pathogenic function of inflammation in the brain there is growing evidence that immune responses are essential for neuroregeneration. This review compares and contrasts the neuroinflammatory pathways that selected neurodegenerative diseases share and have in common. In AD, tau tangles and beta-amyloid plaques cause microglia and astrocytes to become activated in an inflammatory response. Alpha-synuclein aggregation stimulate neuroinflammation in Parkinson's disease, especially in the substantia nigra. In Multiple Sclerosis an autoimmune attack on myelin is connected to inflammation via invading immune cells. Commonalities include the release of pro-inflammatory mediators like cytokines and activation of signaling pathways such as NF-κB and MAPK. Comprehending these common routes is essential for discovering early diagnostic possibilities for the diseases and possible tailored treatments. Our work underscores the potential for insights into disease mechanisms. Identifying common targets offers promise for advancing our understanding and potential future treatment approaches across these debilitating disorders.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, Scinawa, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
37
|
Colín-Martínez E, Arias C. Involvement of the VGF/BDNF axis in the neuropathology of Alzheimer's disease and its potential role in diagnosis and treatment. Rev Neurosci 2025; 36:267-278. [PMID: 39566031 DOI: 10.1515/revneuro-2024-0110] [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: 08/13/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024]
Abstract
The brain is a highly plastic organ that continually receives and integrates signals to generate functional and structural changes and homeostatic adaptations throughout life. Alterations in some signaling pathways that mediate these responses can impact brain plasticity, accelerate brain aging and potentially lead to neurodegeneration. There is substantial evidence that two important signaling pathways activated by neurotrophins, nonacronymic (VGF) and brain-derived neurotrophic factor (BDNF), are involved in substantial functions stimulating neuronal growth, differentiation, and circuit establishment during development and neuronal maintenance and plasticity in the mature brain. In this review, we present evidence that these two pathways and their interactions are central players in cognitive performance and alterations in pathological aging, particularly in conditions such as Alzheimer's disease (AD). Finally, we suggest specific avenues for future research on the basis of recent findings suggesting these molecules are diagnostic biomarkers and putative therapeutic tools to prevent, delay or improve AD neuropathology.
Collapse
Affiliation(s)
- Elizabeth Colín-Martínez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| |
Collapse
|
38
|
Zhan X, Wang S, Bèchet N, Gouras G, Wen G. Perivascular macrophages in the central nervous system: insights into their roles in health and disease. Cell Death Dis 2025; 16:350. [PMID: 40295513 PMCID: PMC12037809 DOI: 10.1038/s41419-025-07592-2] [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: 11/05/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
Perivascular macrophages (PVMs) are a specialized subset of macrophages situated near blood vessels in the brain. Their strategic positioning around these vessels enables them to perform key functions in immune surveillance and response to inflammation and injury. These cells are crucial for modulating the immune response within the brain, contributing to normal central nervous system (CNS) processes. In pathological conditions, the role of PVMs becomes more complex. Depending on the specific disease or injury, they may contribute to inflammation, blood-brain barrier (BBB) dysfunction, and the clearance of abnormal materials. PVMs are implicated in degenerative diseases, cerebrovascular impairment, and microhemorrhages associated with amyloid-β immunotherapy. Despite their important roles in the CNS, research on PVMs remains limited, and the mechanisms underlying their involvement in both physiological and pathological processes within the brain are not yet fully elucidated. Therefore, this review will focus on the current advancements in PVM research, including their origin, classification, roles in neuroinflammation and neuroprotection, and their potential roles as therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoni Zhan
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Shuying Wang
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Nicholas Bèchet
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Science, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Gunnar Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China.
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
39
|
Wang T, Liu Y, Lu Y, Chi L. NTN-1 attenuates amyloid-β-mediated microglial neuroinflammation and memory impairment via the NF-κB pathway and NLRP3 inflammasome in a rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1516399. [PMID: 40357234 PMCID: PMC12066592 DOI: 10.3389/fnagi.2025.1516399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Neuroinflammation driven by microglial activation represents a pivotal pathological mechanism underlying brain injury in Alzheimer's disease (AD), with NLRP3 inflammasome activation being a hallmark feature of this process. Netrin-1 (NTN-1) was recently shown to have potent anti-inflammatory and anti-apoptotic properties in a range of inflammatory diseases; however, its potential effect on neuroinflammation in AD treatment has not been well examined. Accordingly, this study aimed to investigate the effects of NTN-1 on cognitive impairment and to explore the anti-inflammatory properties related to the NLRP3 inflammasome and NF-κB signaling in Aβ1-42-induced rat models. Methods We assessed the effects of NTN-1 on neurobehavioral function, microglial activation and neuroinflammation mechanisms in Aβ1-42-treated rats using the Morris water maze test and Western blotting. Results Our results indicated that microinjections of NTN-1 attenuated Aβ1-42-induced memory and cognitive dysfunction and significantly inhibited microglial proliferation and NLRP3 inflammasome activation in the hippocampus and cortex of AD rats. Additionally, NTN-1 effectively prevented proinflammatory factor (IL1β and IL18) release and NF-κB signaling upstream activation. Discussion Overall, the results of the present study indicated that exogenous NTN-1 treatment prevented neuroinflammation and cognitive deficits by inhibiting microglial activation, which is possibly mediated by the NF-κB signaling pathway and NLRP3 inflammasome activation in Aβ1-42-simulated rat models. NTN-1 emerges as a promising therapeutic candidate for mitigating microglia-mediated neuropathology in AD through its anti-inflammatory properties.
Collapse
Affiliation(s)
| | | | | | - Lijun Chi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Saggu S, Pless A, Dew E, Ware D, Jiao K, Wang Q. Monoamine signaling and neuroinflammation: mechanistic connections and implications for neuropsychiatric disorders. Front Immunol 2025; 16:1543730. [PMID: 40356905 PMCID: PMC12066344 DOI: 10.3389/fimmu.2025.1543730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Monoamines, including norepinephrine, serotonin, and dopamine, orchestrate a broad spectrum of neurophysiological and homeostatic events. Recent research shows a pivotal role for monoaminergic signaling in modulating neuroinflammation by regulating proinflammatory cytokines and chemokines within the central nervous system. Importantly, this modulation is not unidirectional; released proinflammatory cytokines markedly "feedback" to influence the metabolism of monoamine neurotransmitters, impacting their synthesis, release, and reuptake. This bidirectional interplay significantly links monoaminergic pathways and neuroinflammatory responses. In this review, we summarize current knowledge of the dynamic interactions between monoamine signaling and neuroinflammation, as well as their critical implications for the pathophysiology of neuropsychiatric disorders, including Parkinson's Disease, Major Depressive Disorder, and Alzheimer's Disease. By integrating recent findings, we shed light on potential therapeutic targets within these interconnected pathways, providing insights into novel treatment strategies for these devastating disorders.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Emily Dew
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
41
|
Li HD, Zheng JY, Tan KW, Su JX, Chen W, Pang RK, Wu GL, Qiu YH, Li XX, Cai YF, Zhang SJ. Salvianolic acid B (SalB) improves high-fat diet (HFD)-caused cognitive impairment in mice by modulating the Trem2/Dap12 pathway in vivo and in vitro. Int Immunopharmacol 2025; 153:114461. [PMID: 40101423 DOI: 10.1016/j.intimp.2025.114461] [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: 12/13/2024] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Salvianolic acid B (SalB), which extracted from Salvia miltiorrhiza Bunge (Labiatae), is a traditional Chinese medicine. SalB is widely used in nervous system diseases. This study evaluated the protective effect of SalB on high-fat diet (HFD)-induced cognitive impairment and its mechanisms in vivo and in vitro. The behavior tests demonstrated that SalB alleviated motor skills and learning capacity in HFD mice. Animal experiments have confirmed that SalB reduced the mRNA expression of inflammatory markers and the Trem2/Dap12 pathway in HIP. Furthermore, SalB inhibited the microglia Trem2/Dap12 pathway in HIP. In vivo, palmitic acid (PA) was used to intervene in BV2 cells to construct an inflammatory. SalB reduced the mRNA expression of inflammatory markers and inhibited the Trem2/Dap12 pathway in BV2 cells. In conclusion, SalB treatment may serve as a possible therapy for cognitive impairment induced by HFD.
Collapse
Affiliation(s)
- Hong-Dan Li
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Kai-Wen Tan
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Jin-Xun Su
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Wei Chen
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Rui-Kang Pang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Guang-Liang Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Yu-Hui Qiu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Xiao-Xiao Li
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong 999077, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| |
Collapse
|
42
|
Wendt S, Lin AJ, Ebert SN, Brennan DJ, Cai W, Bai Y, Kong DY, Sorrentino S, Groten CJ, Lee C, Frew J, Choi HB, Karamboulas K, Delhaye M, Mackenzie IR, Kaplan DR, Miller FD, MacVicar BA, Nygaard HB. A 3D human iPSC-derived multi-cell type neurosphere system to model cellular responses to chronic amyloidosis. J Neuroinflammation 2025; 22:119. [PMID: 40275379 PMCID: PMC12023538 DOI: 10.1186/s12974-025-03433-3] [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: 01/18/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive amyloid beta (Aβ) deposition in the brain, with eventual widespread neurodegeneration. While the cell-specific molecular signature of end-stage AD is reasonably well characterized through autopsy material, less is known about the molecular pathways in the human brain involved in the earliest exposure to Aβ. Human model systems that not only replicate the pathological features of AD but also the transcriptional landscape in neurons, astrocytes and microglia are crucial for understanding disease mechanisms and for identifying novel therapeutic targets. METHODS In this study, we used a human 3D iPSC-derived neurosphere model to explore how resident neurons, microglia and astrocytes and their interplay are modified by chronic amyloidosis induced over 3-5 weeks by supplementing media with synthetic Aβ1 - 42 oligomers. Neurospheres under chronic Aβ exposure were grown with or without microglia to investigate the functional roles of microglia. Neuronal activity and oxidative stress were monitored using genetically encoded indicators, including GCaMP6f and roGFP1, respectively. Single nuclei RNA sequencing (snRNA-seq) was performed to profile Aβ and microglia driven transcriptional changes in neurons and astrocytes, providing a comprehensive analysis of cellular responses. RESULTS Microglia efficiently phagocytosed Aβ inside neurospheres and significantly reduced neurotoxicity, mitigating amyloidosis-induced oxidative stress and neurodegeneration following different exposure times to Aβ. The neuroprotective effects conferred by the presence of microglia was associated with unique gene expression profiles in astrocytes and neurons, including several known AD-associated genes such as APOE. These findings reveal how microglia can directly alter the molecular landscape of AD. CONCLUSIONS Our human 3D neurosphere culture system with chronic Aβ exposure reveals how microglia may be essential for the cellular and transcriptional responses in AD pathogenesis. Microglia are not only neuroprotective in neurospheres but also act as key drivers of Aβ-dependent APOE expression suggesting critical roles for microglia in regulating APOE in the AD brain. This novel, well characterized, functional in vitro platform offers unique opportunities to study the roles and responses of microglia to Aβ modelling key aspects of human AD. This tool will help identify new therapeutic targets, accelerating the transition from discovery to clinical applications.
Collapse
Affiliation(s)
- Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| | - Ada J Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Sarah N Ebert
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Declan J Brennan
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Wenji Cai
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Yanyang Bai
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Da Young Kong
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Stefano Sorrentino
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher J Groten
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher Lee
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Jonathan Frew
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Opalia Co, Montreal, QC, H2X 3Y7, Canada
| | - Hyun B Choi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Konstantina Karamboulas
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
| | - Mathias Delhaye
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Ian R Mackenzie
- Department of Pathology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - David R Kaplan
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1 A8, Canada
| | - Freda D Miller
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Brian A MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Haakon B Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| |
Collapse
|
43
|
Zhou P, Chao Q, Li C, Wang N, Guo S, Wang P, Ge P, Li C, Guo R, Yang N, Lu Z, Tang Z, Zhang Q, Fu T, Xiao Q, Zhu H. Microglia-targeting nanosystems that cooperatively deliver Chinese herbal ingredients alleviate behavioral and cognitive deficits in Alzheimer's disease model mice. J Nanobiotechnology 2025; 23:313. [PMID: 40275381 PMCID: PMC12020378 DOI: 10.1186/s12951-025-03385-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The effective treatment of Alzheimer's disease (AD) is challenging because of its complex and controversial pathological mechanisms. Moreover, multiple barriers, such as the blood-brain barrier (BBB), reduce drug delivery efficiency. Microglia-related neuroinflammation has recently attracted increasing attention as a possible cause of AD and has become a novel therapeutic target. Therefore, overcoming the BBB and targeted delivery of anti-inflammatory agents to microglia seem to be effective practical strategies for treating AD. A large proportion of natural active extracts possess exceptional immunomodulating capabilities. In this study, the cooperative delivery of berberine (Ber) and palmatine (Pal) by transferrin-decorated extracellular vesicles (Tf-hEVs-Ber/Pal), which can cross the BBB and precisely target microglia, was performed. This nanosystem effectively cleared amyloid β-protein (Aβ) aggregates, significantly regulated the neuroinflammatory environment both in vitro and in vivo and markedly altered the behavior and improved the cognitive and learning abilities of AD model mice. The efficacy of a microglia-targeting combined therapeutic approach for AD was demonstrated, which broadens the potential application of Chinese herbal ingredients.
Collapse
Affiliation(s)
- Peng Zhou
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Quan Chao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ningjing Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Siqi Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingping Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingyuan Ge
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Caihong Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nianyun Yang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhangdi Lu
- Polymer Science and Materials Chemistry, Exponent Ltd, Hong Kong, 999077, China
| | - Zhishu Tang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources, Shannxi University of Chinese Medicine, Xianyang, 712038, China
- China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qichun Zhang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tingming Fu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qingqing Xiao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Huaxu Zhu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
44
|
Papetti AV, Jin M, Ma Z, Stillitano AC, Jiang P. Chimeric brain models: Unlocking insights into human neural development, aging, diseases, and cell therapies. Neuron 2025:S0896-6273(25)00256-9. [PMID: 40300597 DOI: 10.1016/j.neuron.2025.03.036] [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: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Human-rodent chimeric brain models serve as a unique platform for investigating the pathophysiology of human cells within a living brain environment. These models are established by transplanting human tissue- or human pluripotent stem cell (hPSC)-derived macroglial, microglial, or neuronal lineage cells, as well as cerebral organoids, into the brains of host animals. This approach has opened new avenues for exploring human brain development, disease mechanisms, and regenerative processes. Here, we highlight recent advancements in using chimeric models to study human neural development, aging, and disease. Additionally, we explore the potential applications of these models for studying human glial cell-replacement therapies, studying in vivo human glial-to-neuron reprogramming, and harnessing single-cell omics and advanced functional assays to uncover detailed insights into human neurobiology. Finally, we discuss strategies to enhance the precision and translational relevance of these models, expanding their impact in stem cell and neuroscience research.
Collapse
Affiliation(s)
- Ava V Papetti
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alessandro C Stillitano
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA.
| |
Collapse
|
45
|
Ma HY, Wang J, Wang J, Guo Z, Qin XY, Lan R, Hu Y. Luteolin attenuates cadmium neurotoxicity by suppressing glial inflammation and supporting neuronal survival. Int Immunopharmacol 2025; 152:114406. [PMID: 40068520 DOI: 10.1016/j.intimp.2025.114406] [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: 12/09/2024] [Revised: 02/19/2025] [Accepted: 03/01/2025] [Indexed: 03/24/2025]
Abstract
Cadmium (Cd), a neurotoxic metal, is associated with the development of neurological disorders. This study investigated the neuroprotective effects of Luteolin against Cd-induced toxicity in cultured cells and mouse models. Our findings demonstrate that Luteolin protects hippocampal neurons from Cd toxicity and mitigates Cd-triggered inflammatory responses in microglial BV2 cells. In Cd-exposed mice, symptoms such as weight loss, motor retardation, multi-organ damage, and cognitive deficits were observed. Remarkably, Luteolin treatment reversed these effects, repaired organ damage, and restored learning and memory abilities. Mechanistically, Cd toxicity induced significant upregulation of pro-inflammatory factors and neuroinflammation in the hippocampus and prefrontal cortex, including elevated glial cell markers (IBA1, GFAP, and CD68) and reduced neuronal marker MAP2. Luteolin counteracted these adverse effects by inhibiting the Notch1/Hes1 inflammatory signaling axis and restoring the BDNF-TrkB/AKT1 signaling axis, thereby promoting neuronal survival. These results highlight the potential of Luteolin as a natural neuroprotective agent against Cd-induced neurotoxicity, offering a promising therapeutic strategy for mitigating Cd-related neurological damage.
Collapse
Affiliation(s)
- Hui-Yong Ma
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Jing Wang
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Jun Wang
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Zhe Guo
- Department of Cell Biology & Medical Genetics, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518060, China
| | - Xiao-Yan Qin
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Rongfeng Lan
- Department of Cell Biology & Medical Genetics, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518060, China.
| | - Yang Hu
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China..
| |
Collapse
|
46
|
Sirimaharaj N, Thiankhaw K, Chattipakorn N, Chattipakorn SC. Unveiling the Protective Roles of Melatonin on Glial Cells in the Battle Against Alzheimer's Disease-Insights from In Vivo and In Vitro Studies. Mol Neurobiol 2025:10.1007/s12035-025-04904-7. [PMID: 40208552 DOI: 10.1007/s12035-025-04904-7] [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: 11/09/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder that predominantly affects the elderly. Characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles, AD leads to memory loss, cognitive decline, and severe behavioral changes. As the most common form of dementia, AD imposes a significant global health burden, highlighting the need for interventions that address underlying disease mechanisms rather than only symptomatic treatment. Glial cells, including microglia and astrocytes, play a crucial role in AD progression by mediating neuroinflammatory responses and modulating Aβ clearance and neuronal health. Dysfunction in these cells can exacerbate neuroinflammation and neuronal damage, making glial cells an important target for therapeutic intervention. This review synthesizes findings from in vivo and in vitro studies on melatonin's effects on glial cell dysfunction in AD, emphasizing the multi-mechanistic nature of its neuroprotective properties. Recent studies highlight melatonin's potential as a therapeutic agent that addresses AD-related mechanisms through its interactions with glial cells. Melatonin has demonstrated protective effects, including reducing oxidative stress, apoptosis, and inflammation, inhibiting Aβ fibrillogenesis, and modulating amyloid precursor proteins. Additionally, its influence on glial cell activity, through melatonin receptor pathways, suggests it can alleviate neuroinflammation, a key component of AD progression. The collective evidence points to melatonin's promise as a therapeutic tool with potential roles in both preventive and adjunctive treatments for AD. However, further research is necessary to establish its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Nopdanai Sirimaharaj
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
47
|
Yoon CS. Natural Products in the Treatment of Neuroinflammation at Microglia: Recent Trend and Features. Cells 2025; 14:571. [PMID: 40277896 PMCID: PMC12026008 DOI: 10.3390/cells14080571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Natural products (NPs) are considered to be the oldest medicine in human history and numerous NPs have been investigated to search for therapeutic agents in various diseases. Neurodegenerative diseases such as dementia, Parkinson's, Alzheimer's, and Huntington's disease have been increasing following the extension of human lifespans. Neuroinflammation is a key factor in the genesis of several neurodegenerative diseases; therefore, many studies have been focused on finding therapeutics for the reduction in neuroinflammation. Microglia cells are found in the central nervous system (CNS) and these play a crucial role in the regulation of neuroinflammation; thus, the importance of microglia research has been recognized. This review focuses on recent research trends in finding neuroinflammatory regulators in microglia by using NPs.
Collapse
Affiliation(s)
- Chi-Su Yoon
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
48
|
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
|
49
|
Li Z, Gong C. NLRP3 inflammasome in Alzheimer's disease: molecular mechanisms and emerging therapies. Front Immunol 2025; 16:1583886. [PMID: 40260242 PMCID: PMC12009708 DOI: 10.3389/fimmu.2025.1583886] [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: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory impairment, and neuroinflammation, with no definitive cure currently available. The NLRP3 inflammasome, a key mediator of neuroinflammation, has emerged as a critical player in AD pathogenesis, contributing to the accumulation of β-amyloid (Aβ) plaques, tau hyperphosphorylation, and neuronal damage. This review explores the mechanisms by which the NLRP3 inflammasome is activated in AD, including its interactions with Aβ, tau, reactive oxygen species (ROS), and pyroptosis. Additionally, it highlights the role of the ubiquitin system, ion channels, autophagy, and gut microbiota in regulating NLRP3 activation. Therapeutic strategies targeting the NLRP3 inflammasome, such as IL-1β inhibitors, natural compounds, and novel small molecules, are discussed as promising approaches to mitigate neuroinflammation and slow AD progression. This review underscores the potential of NLRP3 inflammasome inhibition as a therapeutic avenue for AD.
Collapse
Affiliation(s)
- Zhitao Li
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunrong Gong
- Department of Rehabilitation Medicine, Linyi People’s Hospital, Linyi, China
| |
Collapse
|
50
|
Khan MS, Qureshi N, Khan R, Son YO, Maqbool T. CRISPR/Cas9-Based therapeutics as a promising strategy for management of Alzheimer's disease: progress and prospects. Front Cell Neurosci 2025; 19:1578138. [PMID: 40260080 PMCID: PMC12009953 DOI: 10.3389/fncel.2025.1578138] [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: 02/17/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
CRISPR/Cas9 technology has revolutionized genetic and biomedical research in recent years. It enables editing and modulation of gene function with an unparalleled precision and effectiveness. Among the various applications and prospects of this technology, the opportunities it offers in unraveling the molecular underpinnings of a myriad of central nervous system diseases, including neurodegenerative disorders, psychiatric conditions, and developmental abnormalities, are unprecedented. In this review, we highlight the applications of CRISPR/Cas9-based therapeutics as a promising strategy for management of Alzheimer's disease and transformative impact of this technology on AD research. Further, we emphasize the role of CRISPR/Cas9 in generating accurate AD models for identification of novel therapeutic targets, besides the role of CRISPR-based therapies aimed at correcting AD-associated mutations and modulating the neurodegenerative processes. Furthermore, various delivery systems are reviewed and potential of the non-viral nanotechnology-based carriers for overcoming the critical limitations of effective delivery systems for CRISPR/Cas9 is discussed. Overall, this review highlights the promise and prospects of CRISPR/Cas9 technology for unraveling the intricate molecular processes underlying the development of AD, discusses its limitations, ethical concerns and several challenges including efficient delivery across the BBB, ensuring specificity, avoiding off-target effects. This article can be helpful in better understanding the applications of CRISPR/Cas9 based therapeutic approaches and the way forward utilizing enormous potential of this technology in targeted, gene-specific treatments that could change the trajectory of this debilitating and incurable illness.
Collapse
Affiliation(s)
- Mohamad Sultan Khan
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Nousheen Qureshi
- Department of Higher Education, Government of Jammu and Kashmir, Srinagar, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences and Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|