1.The role of microglia activated by the deletion of immune checkpoint receptor CD200R1 gene in a mouse model of Parkinson's disease.
Jia-Li GUO ; Tao-Ying HUANG ; Zhen ZHANG ; Kun NIU ; Xarbat GONGBIKAI ; Xiao-Li GONG ; Xiao-Min WANG ; Ting ZHANG
Acta Physiologica Sinica 2025;77(1):13-24
The study aimed to investigate the effect of the CD200R1 gene deletion on microglia activation and nigrostriatal dopamine neuron loss in the Parkinson's disease (PD) process. The CRISPR-Cas9 technology was applied to construct the CD200R1-/- mice. The primary microglia cells of wild-type and CD200R1-/- mice were cultured and treated with bacterial lipopolysaccharide (LPS). Microglia phagocytosis level was assessed by a fluorescent microsphere phagocytosis assay. PD mouse model was prepared by nigral stereotaxic injection of recombinant adeno-associated virus vector carrying human α-synuclein (α-syn). The changes in the motor behavior of the mice with both genotypes were evaluated by cylinder test, open field test, and rotarod test. Immunohistochemical staining was used to assess the loss of dopamine neurons in substantia nigra. Immunofluorescence staining was used to detect the expression level of CD68 (a key molecule involved in phagocytosis) in microglia. The results showed that CD200R1 deletion markedly enhanced LPS-induced phagocytosis in vitro by the microglial cells. In the mouse model of PD, CD200R1 deletion exacerbated motor behavior impairment and dopamine neuron loss in substantia nigra. Fluorescence intensity analysis results revealed a significant increase in CD68 expression in microglia located in the substantia nigra of CD200R1-/- mice. The above results suggest that CD200R1 deletion may further activates microglia by promoting microglial phagocytosis, leading to increased loss of the nigrostriatal dopamine neurons in the PD model mice. Therefore, targeting CD200R1 could potentially serve as a novel therapeutic target for the treatment of early-stage PD.
Animals
;
Microglia/physiology*
;
Mice
;
Phagocytosis
;
Parkinson Disease/genetics*
;
Disease Models, Animal
;
Receptors, Cell Surface/physiology*
;
Dopaminergic Neurons/pathology*
;
Antigens, CD/metabolism*
;
Gene Deletion
;
Substantia Nigra
;
Mice, Inbred C57BL
;
Mice, Knockout
;
Cells, Cultured
;
Male
;
alpha-Synuclein
;
CD68 Molecule
;
Orexin Receptors
2.Research progress on ferroptosis mediated by microglia in hypoxic-ischemic brain damage.
Tao GUO ; Hanjun ZUO ; Xianfeng KUANG ; Shukun ZHANG ; Bolin CHEN ; Lixing LUO ; Xiao YANG ; Zhao WANG ; Juanjuan LI
Chinese Journal of Cellular and Molecular Immunology 2025;41(6):552-558
In hypoxic-ischemic brain damage (HIBD), the programmed cell death known as ferroptosis is significantly activated. Microglial cells demonstrate a high level of sensitivity to iron accumulation. Understanding how to regulate the dual role of microglia and transforming the microglial ferroptosis to a moderate and controllable process has considerable implications for the targeted treatment in HIBD. This paper serves as an overview of microglia-mediated ferroptosis in HIBD as a disease model. We discuss various aspects centered around microglia, including pathophysiological mechanisms, polarization and functions of microglia, molecular mechanisms of ferroptosis, signaling pathways, and therapeutic strategies. The review aims to provide a reference for studies of ferroptosis in microglia.
Microglia/physiology*
;
Ferroptosis/physiology*
;
Humans
;
Animals
;
Hypoxia-Ischemia, Brain/pathology*
;
Signal Transduction
3.EGR2 maintains neuropathic pain by promoting microglial phagocytosis.
Caiyun XI ; Jianxi ZHANG ; Zhifeng HUANG ; Liqiong HE ; Kailu ZOU ; Xiaoping XU ; Qulian GUO ; Bei SUN ; Changsheng HUANG
Journal of Central South University(Medical Sciences) 2025;50(4):586-601
OBJECTIVES:
Neuropathic pain (NP) is one of the most common forms of chronic pain, yet current treatment options are limited in effectiveness. Peripheral nerve injury activates spinal microglia, altering their inflammatory response and phagocytic functions, which contributes to the progression of NP. Most current research on NP focuses on microglial inflammation, with relatively little attention to their phagocytic function. Early growth response factor 2 (EGR2) has been shown to regulate microglial phagocytosis, but its specific role in NP remains unclear. This study aims to investigate how EGR2 modulates microglial phagocytosis and its involvement in NP, with the goal of identifying potential therapeutic targets.
METHODS:
Adult male Sprague-Dawley (SD) rats were used to establish a chronic constriction injury (CCI) model of the sciatic nerve. Pain behaviors were assessed on days 1, 3, 7, 10, and 14 post-surgery to confirm successful model induction. The temporal and spatial expression of EGR2 in the spinal cord was examined using real-time quantitative PCR (RT-qPCR), Western blotting, and immunofluorescence staining. Adeno-associated virus (AAV) was used to overexpress EGR2 in the spinal cord, and behavioral assessments were performed to evaluate the effects of EGR2 modulation of NP. CCI and lipopolysaccharide (LPS) models were established in animals and microglial cell lines, respectively, and changes in phagocytic activity were measured using RT-qPCR and fluorescent latex bead uptake assays. After confirming the involvement of microglial phagocytosis in NP, AAV was used to overexpress EGR2 in both in vivo and in vitro models, and phagocytic activity was further evaluated. Finally, eukaryotic transcriptome sequencing was conducted to screen differentially expressed mRNAs, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses to identify potential downstream effectors of EGR2.
RESULTS:
The CCI model successfully induced NP. Following CCI, EGR2 expression in the spinal cord was upregulated in parallel with NP development. Overexpression of EGR2 via spinal AAV injection enhanced microglial phagocytic activity and increased pain hypersensitivity in rats. Both animal and cellular models showed that CCI or LPS stimulation enhanced microglial phagocytosis, which was further amplified by EGR2 overexpression. Transcriptomic analysis of spinal cord tissues from CCI rats overexpressing EGR2 revealed upregulation of numerous genes associated with microglial phagocytosis and pain regulation. Among them, Lag3 emerged as a potential downstream target of EGR2.
CONCLUSIONS
EGR2 contributes to the maintenance of NP by enhancing microglial phagocytosis in the spinal dorsal horn.
Animals
;
Microglia/metabolism*
;
Phagocytosis/physiology*
;
Rats, Sprague-Dawley
;
Neuralgia/physiopathology*
;
Early Growth Response Protein 2/metabolism*
;
Male
;
Rats
;
Spinal Cord/metabolism*
;
Sciatic Nerve/injuries*
4.Mechanisms of spinal microglia and astrocytes in exercise-induced analgesia.
Shuang HU ; Haojun YOU ; Jing LEI
Journal of Central South University(Medical Sciences) 2025;50(8):1455-1464
Exercise-induced analgesia (EIA) refers to the elevation of pain thresholds and reduction in sensitivity to noxious stimuli achieved through exercise training. As a non-pharmacological treatment strategy, exercise therapy has demonstrated positive effects on both acute and chronic pain. Increasing evidence indicates that modulation of glial cell activity is an important mechanism underlying analgesia. Spinal glial cells contribute to the development and maintenance of pathological pain by promoting pain signal transmission through inflammatory responses and synaptic remodeling. Exercise can differentially regulate microglia and astrocyte activity, inhibiting multiple inflammatory signaling pathways, such as P2X4/P2X7 purinergic receptors, brain-derived neurotrophic factor (BDNF)/phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR), interleukin (IL)-6/Janus kinase (JAK) 2/signal transducer and activator of transcription 3 (STAT3), p38-mitogen-activated protein kinases (MAPK), and Toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB), thereby reducing the release of pro-inflammatory cytokines, decreasing inflammatory and nociceptive hypersensitivity, and alleviating pathological pain. This review also summarized the effects of different exercise intensities, durations, and frequencies on glial cell responses in order to provide a theoretical foundation for optimizing exercise-based interventions for pathological pain conditions.
Humans
;
Microglia/metabolism*
;
Astrocytes/metabolism*
;
Exercise/physiology*
;
Signal Transduction
;
Analgesia/methods*
;
Spinal Cord/cytology*
;
Exercise Therapy
;
Pain Management/methods*
;
Animals
;
Brain-Derived Neurotrophic Factor/metabolism*
5.OGT-Mediated O-GlcNAcylation of ATF2 Protects Against Sepsis-Associated Encephalopathy by Inhibiting Microglial Pyroptosis.
Huan YAO ; Caixia LIANG ; Xueting WANG ; Chengwei DUAN ; Xiao SONG ; Yanxing SHANG ; Mingyang ZHANG ; Yiyun PENG ; Dongmei ZHANG
Neuroscience Bulletin 2025;41(10):1761-1778
Microglial pyroptosis and neuroinflammation have been implicated in the pathogenesis of sepsis-associated encephalopathy (SAE). OGT-mediated O-GlcNAcylation is involved in neurodevelopment and injury. However, its regulatory function in microglial pyroptosis and involvement in SAE remains unclear. In this study, we demonstrated that OGT deficiency augmented microglial pyroptosis and exacerbated secondary neuronal injury. Furthermore, OGT inhibition impaired cognitive function in healthy mice and accelerated the progression in SAE mice. Mechanistically, OGT-mediated O-GlcNAcylation of ATF2 at Ser44 inhibited its phosphorylation and nuclear translocation, thereby amplifying NLRP3 inflammasome activation and promoting inflammatory cytokine production in microglia in response to LPS/Nigericin stimulation. In conclusion, this study uncovers the critical role of OGT-mediated O-GlcNAcylation in modulating microglial activity through the regulation of ATF2 and thus protects against SAE progression.
Animals
;
Microglia/metabolism*
;
Pyroptosis/physiology*
;
Mice
;
Sepsis-Associated Encephalopathy/prevention & control*
;
Activating Transcription Factor 2/metabolism*
;
N-Acetylglucosaminyltransferases/genetics*
;
Mice, Inbred C57BL
;
Male
;
Mice, Knockout
6.Modulation of Ryanodine Receptors on Microglial Ramification, Migration, and Phagocytosis in an Alzheimer's Disease Mouse Model.
Yulin OUYANG ; Zihao CHEN ; Qiang HUANG ; Hai ZHANG ; Haolin SONG ; Xinnian WANG ; Wenxiu DONG ; Yong TANG ; Najeebullah SHAH ; Shimin SHUAI ; Yang ZHAN
Neuroscience Bulletin 2025;41(11):2063-2077
Microglial functions are linked to Ca2+ signaling, with endoplasmic reticulum (ER) calcium stores playing a crucial role. Microglial abnormality is a hallmark of Alzheimer's disease (AD), but how ER Ca2+ receptors regulate microglial functions under physiological and AD conditions remains unclear. We found reduced ryanodine receptor 2 (Ryr2) expression in microglia from an AD mouse model. Modulation of RyR2 using S107, a RyR-Calstabin stabilizer, blunted spontaneous Ca2+ transients in controls and normalized Ca2+ transients in AD mice. S107 enhanced ATP-induced migration and phagocytosis while reducing ramification in control microglia; however, these effects were absent in AD microglia. Our findings indicate that RyR2 stabilization promotes an activation state shift in control microglia, a mechanism impaired in AD. These results highlight the role of ER Ca2+ receptors in both homeostatic and AD microglia, providing insights into microglial Ca2+ malfunctions in AD.
Animals
;
Microglia/pathology*
;
Alzheimer Disease/pathology*
;
Phagocytosis/drug effects*
;
Ryanodine Receptor Calcium Release Channel/metabolism*
;
Disease Models, Animal
;
Mice
;
Cell Movement/drug effects*
;
Mice, Transgenic
;
Calcium Signaling/physiology*
;
Calcium/metabolism*
;
Mice, Inbred C57BL
;
Male
;
Endoplasmic Reticulum/metabolism*
7.Fibroblast Growth Factor 8 Suppresses Neurotoxic Astrocytes and Alleviates Neuropathic Pain via Spinal FGFR3 Signaling.
Huizhu LIU ; Lanxing YI ; Guiling LI ; Kangli WANG ; Hongsheng WANG ; Yuqiu ZHANG ; Benlong LIU
Neuroscience Bulletin 2025;41(12):2218-2232
Astrocytes in the spinal dorsal horn (SDH) exhibit diverse reactive phenotypes under neuropathic conditions, yet the mechanisms driving this diversity and its implications in chronic pain remain unclear. Here, we report that spared nerve injury (SNI) induces marked upregulation of both complement component 3 (C3⁺, A1-like) and S100 calcium-binding protein A10 (S100A10⁺, A2-like) astrocyte subpopulations in the SDH, with elevated microglial cytokines including interleukin-1α, tumor necrosis factor-α, and complement component 1q. Transcriptomic, immunohistochemical, and Western blot analyses reveal co-activation of multiple reactive astrocyte states over a unidirectional shift toward an A1-like phenotype. Fibroblast growth factor 8 (FGF8), a neuroprotective factor via FGFR3, mitigated microglia-induced C3⁺ astrocyte reactivity in vitro and suppressed spinal C3 expression and mechanical allodynia following intrathecal administration in SNI mice. These findings reveal a microglia-astrocyte signaling axis that promotes A1 reactivity and position FGF8 as a promising therapeutic candidate for neuropathic pain by modulating astrocyte heterogeneity.
Animals
;
Astrocytes/drug effects*
;
Neuralgia/pathology*
;
Receptor, Fibroblast Growth Factor, Type 3/metabolism*
;
Signal Transduction/physiology*
;
Male
;
Mice
;
Microglia/drug effects*
;
Fibroblast Growth Factor 8/pharmacology*
;
Mice, Inbred C57BL
;
Hyperalgesia/drug therapy*
;
Spinal Cord/drug effects*
;
Complement C3/metabolism*
;
Spinal Cord Dorsal Horn/metabolism*
8.Single-Cell Mapping of Brain Myeloid Cell Subsets Reveals Key Transcriptomic Changes Favoring Neuroplasticity after Ischemic Stroke.
Fangxi LIU ; Xi CHENG ; Chuansheng ZHAO ; Xiaoqian ZHANG ; Chang LIU ; Shanshan ZHONG ; Zhouyang LIU ; Xinyu LIN ; Wei QIU ; Xiuchun ZHANG
Neuroscience Bulletin 2024;40(1):65-78
Interactions between brain-resident and peripheral infiltrated immune cells are thought to contribute to neuroplasticity after cerebral ischemia. However, conventional bulk sequencing makes it challenging to depict this complex immune network. Using single-cell RNA sequencing, we mapped compositional and transcriptional features of peri-infarct immune cells. Microglia were the predominant cell type in the peri-infarct region, displaying a more diverse activation pattern than the typical pro- and anti-inflammatory state, with axon tract-associated microglia (ATMs) being associated with neuronal regeneration. Trajectory inference suggested that infiltrated monocyte-derived macrophages (MDMs) exhibited a gradual fate trajectory transition to activated MDMs. Inter-cellular crosstalk between MDMs and microglia orchestrated anti-inflammatory and repair-promoting microglia phenotypes and promoted post-stroke neurogenesis, with SOX2 and related Akt/CREB signaling as the underlying mechanisms. This description of the brain's immune landscape and its relationship with neurogenesis provides new insight into promoting neural repair by regulating neuroinflammatory responses.
Humans
;
Ischemic Stroke
;
Brain/metabolism*
;
Macrophages
;
Brain Ischemia/metabolism*
;
Microglia/metabolism*
;
Gene Expression Profiling
;
Anti-Inflammatory Agents
;
Neuronal Plasticity/physiology*
;
Infarction/metabolism*
9.Research progress of signal pathways of microglia activation in sleep disorders.
Zhi-Jun SHU ; Quan-Yi ZHANG ; Yi-Peng XU ; Zheng-Yu ZHAO
Acta Physiologica Sinica 2023;75(4):569-574
Sleep is an extremely important physiological state to maintain human life. Sleep disorders can not only cause anxiety and depression, but also induce multi-system diseases that seriously affect brain function and physical health. The neuroinflammation is a key pathological process after sleep disorders, which can induce a series of nervous system diseases. In recent years, the role of microglia activation in neuroinflammation has been paid more and more attention and become a research hotspot in this field. The imbalance of the central microenvironment after sleep disorders leads to changes in the activation and polarization of microglia, which triggers neuroinflammatory response. The activation and polarization of microglia in the sleep disorders are regulated by multiple signaling pathways and complex molecular mechanisms. This paper summarizes five signaling pathways of microglia activation in central inflammation induced by sleep disorders, including P2X7 receptor (P2X7R), p38MAPK, Toll-like receptor 4 (TLR4)/NF-κB, JAK/STAT, and α7 nicotinic acetylcholine receptor (α7-nAChR) pathways, in order to provide reference for further research and clinical treatment targets selection of sleep disorders.
Humans
;
Neuroinflammatory Diseases
;
Microglia/metabolism*
;
Signal Transduction/physiology*
;
NF-kappa B/metabolism*
;
Inflammation/metabolism*
;
Sleep Wake Disorders/metabolism*
10.Chemogenetic and Optogenetic Manipulations of Microglia in Chronic Pain.
Sebastian PARUSEL ; Min-Hee YI ; Christine L HUNT ; Long-Jun WU
Neuroscience Bulletin 2023;39(3):368-378
Chronic pain relief remains an unmet medical need. Current research points to a substantial contribution of glia-neuron interaction in its pathogenesis. Particularly, microglia play a crucial role in the development of chronic pain. To better understand the microglial contribution to chronic pain, specific regional and temporal manipulations of microglia are necessary. Recently, two new approaches have emerged that meet these demands. Chemogenetic tools allow the expression of designer receptors exclusively activated by designer drugs (DREADDs) specifically in microglia. Similarly, optogenetic tools allow for microglial manipulation via the activation of artificially expressed, light-sensitive proteins. Chemo- and optogenetic manipulations of microglia in vivo are powerful in interrogating microglial function in chronic pain. This review summarizes these emerging tools in studying the role of microglia in chronic pain and highlights their potential applications in microglia-related neurological disorders.
Humans
;
Optogenetics
;
Brain/physiology*
;
Microglia
;
Chronic Pain/therapy*
;
Neurons/physiology*

Result Analysis
Print
Save
E-mail