Journal of Neuroinflammation最新文献

筛选
英文 中文
B cells are not drivers of stromal cell activation during acute CNS infection. 急性中枢神经系统感染时,B细胞不是基质细胞活化的驱动因子。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-27 DOI: 10.1186/s12974-025-03491-7
Brendan T Boylan, Mihyun Hwang, Elyse Brozost, Hyunsuk Oh, Alexei V Tumanov, Antoine Louveau, Cornelia C Bergmann
{"title":"B cells are not drivers of stromal cell activation during acute CNS infection.","authors":"Brendan T Boylan, Mihyun Hwang, Elyse Brozost, Hyunsuk Oh, Alexei V Tumanov, Antoine Louveau, Cornelia C Bergmann","doi":"10.1186/s12974-025-03491-7","DOIUrl":"10.1186/s12974-025-03491-7","url":null,"abstract":"<p><strong>Background: </strong>CNS stromal cells, especially fibroblasts and endothelial cells, support leukocyte accumulation through upregulation of adhesion molecules and lymphoid chemokines. While chronically activated fibroblast networks can drive pathogenic immune cell aggregates known as tertiary lymphoid structures (TLS), early stromal cell activation during CNS infection can support anti-viral T cells. However, the cell types and factors driving early stromal cell activation is poorly explored.</p><p><strong>Aims: </strong>A neurotropic murine coronavirus (mCoV) infection model was used to better characterize signals that promote fibroblast networks supporting accumulation of antiviral lymphocytes. Based on the early appearance of IgD<sup>+</sup> B cells with unknown functions during several CNS infections, we probed their potential to activate stromal cells through lymphotoxin β (LTβ), a molecule critical in maintaining fibroblast-networks in lymphoid tissues as well as promoting TLS in autoimmunity and cancers.</p><p><strong>Results: </strong>Kinetic analysis of stromal cell activation in olfactory bulbs and brains revealed that upregulation of adhesion molecules and lymphoid chemokines Ccl19, Ccl21 and Cxcl13 closely tracked viral replication. Immunohistochemistry revealed that upregulation of the fibroblast marker podoplanin (PDPN) at meningeal and perivascular sites mirrored kinetics of RNA expression. Moreover, both B cells and T cells colocalized to areas of PDPN reactivity, supporting a potential role in regulating stromal cell activation. However, specific depletion of LTβ from B cells using Mb1-creERT2 x Ltβ<sup>fl/fl</sup> mice had no effect on T or B cell recruitment or viral replication. B cell depletion by anti-CD20 antibody also had no adverse effects. Surprisingly, LTβR agonism reduced viral control and parenchymal T cell localization despite increasing stromal cell lymphoid chemokines and PDPN. Additional assessment of direct stromal cell activation by the viral RNA mimic poly I:C showed induction of Pdpn and Ccl19 preceding Ltb.</p><p><strong>Conclusions: </strong>Neither B cell-derived LTβ or B cells are primary drivers of stromal cell activation networks in the CNS following mCoV infection. Although supplementary agonist mediated LTβR engagement confirmed a role for LTβ in enhancing PDPN and lymphoid chemokine expression, it impeded T cell migration to the CNS parenchyma and viral control. Our data overall indicate that stromal cells can integrate LTβR signals to tune their activation, but that LTβ is not necessarily essential and can even dysregulate protective antiviral T cell functions.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"165"},"PeriodicalIF":9.3,"publicationDate":"2025-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12203728/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144512060","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
N-Lactoyl-Phenylalanine modulates lipid metabolism in microglia/macrophage via the AMPK-PGC1α-PPARγ pathway to promote recovery in mice with spinal cord injury. n -乳酸-苯丙氨酸通过AMPK-PGC1α-PPARγ通路调节小胶质细胞/巨噬细胞脂质代谢,促进脊髓损伤小鼠的恢复。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-27 DOI: 10.1186/s12974-025-03495-3
Weiyang Ying, Weidong Weng, Peifang Wang, Chi Pan, Jiani Qiu, Qianqian Huang, Gonghao Zhan, Xiaoli Chen
{"title":"N-Lactoyl-Phenylalanine modulates lipid metabolism in microglia/macrophage via the AMPK-PGC1α-PPARγ pathway to promote recovery in mice with spinal cord injury.","authors":"Weiyang Ying, Weidong Weng, Peifang Wang, Chi Pan, Jiani Qiu, Qianqian Huang, Gonghao Zhan, Xiaoli Chen","doi":"10.1186/s12974-025-03495-3","DOIUrl":"10.1186/s12974-025-03495-3","url":null,"abstract":"<p><p>The accumulation of lipids in microglia/macrophage-induced inflammation exacerbation represents a pivotal factor contributing to secondary injury following spinal cord injury (SCI). N-Lactoyl-Phenylalanine (L-P), a metabolic byproduct of exercise, exhibits the capacity to regulate carbohydrate and lipid metabolism and may serve as a potential regulator of lipid metabolism in microglia/macrophage. This study investigates the role of L-P in modulating lipid homeostasis in microglia/macrophage and its therapeutic implications for SCI recovery. By establishing a mouse model of SCI, we confirmed that L-P administration markedly altered lipid metabolism in microglia/macrophage. This metabolic reprogramming was mediated through the activation of the AMPK-PGC1α-PPARγ signaling pathway, which plays a crucial role in regulating cellular energy metabolism and inflammatory responses. Our findings demonstrate that L-P treatment enhances the lipid metabolic capacity of microglia/macrophage, thereby attenuating neuroinflammation and promoting tissue repair after injury. Moreover, the polarization of microglia/macrophage shifts toward the anti-inflammatory M2 phenotype, providing substantial support for the regenerative process of the injured spinal cord. Functional analysis revealed that mice treated with L-P exhibited significantly improved motor function compared to the control group. Collectively, these results underscore the therapeutic potential of L-P in SCI and suggest its utility as a metabolic intervention strategy by modulating microglia/macrophage lipid metabolism to accelerate recovery.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"167"},"PeriodicalIF":9.3,"publicationDate":"2025-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144512062","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Depression exacerbates AD pathology through lactate-dependent activation of microglial Kv1.3 to promote Aβ-containing exosome spreading. 抑郁症通过乳酸依赖性激活小胶质细胞Kv1.3,促进含a β的外泌体扩散,从而加剧AD病理。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-27 DOI: 10.1186/s12974-025-03488-2
Xiaoli Liu, Huijin Wang, Xi Tian, Yingqi Luo, Minmin Ma, Zilong Zheng, Yaping Wang, Shi Feng, Qiushi Wang, Zhuo Xu, Wen Yao, Siqiang Ren
{"title":"Depression exacerbates AD pathology through lactate-dependent activation of microglial Kv1.3 to promote Aβ-containing exosome spreading.","authors":"Xiaoli Liu, Huijin Wang, Xi Tian, Yingqi Luo, Minmin Ma, Zilong Zheng, Yaping Wang, Shi Feng, Qiushi Wang, Zhuo Xu, Wen Yao, Siqiang Ren","doi":"10.1186/s12974-025-03488-2","DOIUrl":"10.1186/s12974-025-03488-2","url":null,"abstract":"<p><p>Depression has been widely recognized as an important accelerating factor contributing to the aggravation of cognitive decline in Alzheimer's disease (AD) patients. Previous studies show that microglia-mediated neuroinflammation is a common and critical event in the etiology of both depression and dementia, but whether and how microglia participate in the process of depression-exacerbating AD pathology is largely unknown. By establishing the learned helplessness depression model on 5×FAD mice, we confirmed that depression can indeed promote Aβ plaque deposition and deteriorate the cognitive performance of the AD mice. Importantly, we found that microglial lactate concentration is dramatically increased in the depressed AD brain, leading to activation of potassium channel Kv1.3 likely through non-direct-lactylation. The activated Kv1.3 further facilitates Aβ-containing exosome spreading from microglia in the vicinity of Aβ plaque into the surrounding brain tissue. Notably, conditional knock-out of Kv1.3 in microglia can reverse the depression-induced acceleration of AD pathology and cognitive decline. Together, our study highlights an important function of microglia Kv1.3 in the promotion of Aβ propagation in the context of depression-exacerbating AD pathology.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"166"},"PeriodicalIF":9.3,"publicationDate":"2025-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144512061","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IFP35, a novel DAMP, aggravates neuroinflammation following acute ischemic stroke via TLR4/NF-κB/NLRP3 signaling. IFP35是一种新型的DAMP,可通过TLR4/NF-κB/NLRP3信号通路加重急性缺血性卒中后的神经炎症。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-25 DOI: 10.1186/s12974-025-03492-6
Mengmeng Zhang, Bingnan Guo, Xiaowei Zhang, Dong Han, Lanxin Lv, Xiaoqing Yan, Chenglei Su, Dafei Chai, Ningjun Zhao, Xianliang Yan, Shuqun Hu
{"title":"IFP35, a novel DAMP, aggravates neuroinflammation following acute ischemic stroke via TLR4/NF-κB/NLRP3 signaling.","authors":"Mengmeng Zhang, Bingnan Guo, Xiaowei Zhang, Dong Han, Lanxin Lv, Xiaoqing Yan, Chenglei Su, Dafei Chai, Ningjun Zhao, Xianliang Yan, Shuqun Hu","doi":"10.1186/s12974-025-03492-6","DOIUrl":"10.1186/s12974-025-03492-6","url":null,"abstract":"<p><strong>Background: </strong>Acute ischemic stroke is a disastrous disease characterized by damaging blood flow in the brain, leading to acute brain injury. Acute brain ischemia elicits severe inflammation, thus in turn, aggravates neural injury. Interferon-Induced Protein 35 (IFP35), is a 35 kDa protein, a novel type of DAMP that trigger inflammatory responses, exacerbating acute and chronic inflammatory disease. This study aimed to investigate the potential neuroinflammation role of IFP35 in acute ischemic stroke in a mouse model of MCAO.</p><p><strong>Methods: </strong>C57BL/6 male mice were subjected to middle cerebral artery occlusion (MCAO) to establish an animal model of acute ischemic stroke. Leveraging serum from stroke patients, serum and brain tissue after MCAO mice, IFP35 was released. Immunofluorescence assay was used to investigated the cell sources of IFP35 expression after MCAO. The impact of IFP35 on neuroinflammation and neural injury was assessed by siRNA-mediated cerebral IFP35 knockdown. Behavioral tests, and brain tissues were harvested for histological analysis and biochemical assays. TUNEL assays were used to evaluate neuronal damage. TTC staining was performed to assess infarction volumes. Additionally, using western blotting and immunofluorescence assays, we further assessed the contribution of TLR4/NF-κB/NLRP3 signaling in MCAO mice and BV2 cells.</p><p><strong>Results: </strong>IFP35 was accumulated in peripheral blood of cerebral ischaemia patients, ischemia mice serum, as well as peri-infarct regions in focal cerebral ischemia mice. Although endothelial cells, microglia, and astrocytes are capable of expressing IFP35, cerebral neural cells seem to express and release more IFP35 compare to other cell types. Knockdown of IFP35 alleviated the production of neuroinflammatory cytokines, decreased neuronal death, and minimized infarct volumes, ultimately leading to improved neurological outcomes. Importantly, IFP35 triggered the activation of NF-κΒ and NLRP3 signaling, exacerbating neuroinflammation and brain injury by binding its receptor TLR4.</p><p><strong>Conclusions: </strong>This study revealed IFP35 as a novel DAMP released during cerebral ischemia that promotes neuroinflammation and injury, expanding the current understanding of inflammatory networks following stroke.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"164"},"PeriodicalIF":9.3,"publicationDate":"2025-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12188676/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144497345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Group 2 innate lymphoid cells drive inhibitory synapse formation with lasting effects on learning and memory. 2组先天淋巴样细胞驱动抑制性突触的形成,对学习和记忆有持久的影响。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-23 DOI: 10.1186/s12974-025-03485-5
Johannes Steffen, Divija Deshpande, Henning Peter Düsedau, Janna Schmitz, Caio Andreeta Figueiredo, Laura Velleman, Claudia Pitzer, Christoph S N Klose, Ildiko R Dunay
{"title":"Group 2 innate lymphoid cells drive inhibitory synapse formation with lasting effects on learning and memory.","authors":"Johannes Steffen, Divija Deshpande, Henning Peter Düsedau, Janna Schmitz, Caio Andreeta Figueiredo, Laura Velleman, Claudia Pitzer, Christoph S N Klose, Ildiko R Dunay","doi":"10.1186/s12974-025-03485-5","DOIUrl":"10.1186/s12974-025-03485-5","url":null,"abstract":"<p><p>The immune system provides multiple layers of protection that extend beyond conventional pathogen defense, including context-dependent modulation of behavior. However, the mechanisms driving these immune-mediated behavioral modifications remain incompletely understood. Here, we demonstrate that group 2 innate lymphoid cells (ILC2s) shape hippocampal synaptic development during early postnatal stages, with lasting effects on adult behavior, learning, and memory.Using flow synaptometry, we identified a selective reduction in hippocampal VGAT<sup>+</sup> GABAergic/glycinergic inhibitory synapse frequency at postnatal day 15 in ILC2-deficient mice, while the proportions of inhibitory GABAergic (NL2<sup>+</sup>) or excitatory glutamatergic (GluR1<sup>+</sup>) synapses remained unaltered. These synaptic changes occurred without detectable phenotypical changes in cortical and hippocampal microglia. In adulthood, ILC2-deficient mice displayed significant impairments in hippocampus-dependent tasks, such as active place avoidance and operant conditioning, reflecting deficits in learning and memory.Our findings reveal a critical role for ILC2s in the formation of inhibitory synapses in the hippocampus, highlighting the impact of immune signaling on neuronal network maturation during a crucial period of brain development. This early immune-mediated modulation may have lasting effects on neuronal circuitry and cognitive functions that persist into adulthood, emphasizing the long-term implications of neuro-immune interactions for normal cognitive development and function.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"163"},"PeriodicalIF":9.3,"publicationDate":"2025-06-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12183876/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144475691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting the RAGE-RIPK1 binding site attenuates diabetes-associated cognitive deficits. 靶向RAGE-RIPK1结合位点可减轻糖尿病相关认知缺陷。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-21 DOI: 10.1186/s12974-025-03489-1
Lin Gao, Shidi Wu, Bin Hu, Qiuyu Zhang, Yifei Wu, Hui Li, Ye Qian, Chengyu Huang, Xiangru Wen, Hui Li, Aifang Cheng, Yuanjian Song, Changjiang Ying, Xiaoyan Zhou
{"title":"Targeting the RAGE-RIPK1 binding site attenuates diabetes-associated cognitive deficits.","authors":"Lin Gao, Shidi Wu, Bin Hu, Qiuyu Zhang, Yifei Wu, Hui Li, Ye Qian, Chengyu Huang, Xiangru Wen, Hui Li, Aifang Cheng, Yuanjian Song, Changjiang Ying, Xiaoyan Zhou","doi":"10.1186/s12974-025-03489-1","DOIUrl":"10.1186/s12974-025-03489-1","url":null,"abstract":"<p><p>Microglial activation can cause neuroinflammation and the consequent neurological impairments play prominent roles in diabetes-associated cognitive deficits. Receptor-interacting protein kinase 1 (RIPK1) phosphorylation is involved in this deleterious microglial activation, but the exact molecular mechanisms are not clear. Here, RIPK1 expression was increased in diabetic patients with cognitive impairment. Furthermore, in diabetic mice, RIPK1 death domain directly binds to C-terminal of the receptor for advanced glycation end products (ctRAGE) could regulate RIPK1 phosphorylation in microglia. This RAGE-RIPK1 complex activates inflammatory signaling, resulting in cascades that ultimately promote cognitive impairment in diabetic mice. An engineered brain-targeting RIPK1 peptide blocked binding of RIPK1 to RAGE, which inhibited RIPK1 phosphorylation, decreased neuroinflammation, improved neuronal morphology and function, and prevented diabetes-associated cognitive deficits in mice. This study uncovers a previously unknown mechanism of neuroinflammation and suggests a novel therapeutic avenue for treating cognitive deficits induced by hyperglycemia.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"162"},"PeriodicalIF":9.3,"publicationDate":"2025-06-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12182694/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144340216","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pro-repair macrophages driven by CGRP rescue white matter integrity following intracerebral hemorrhage. CGRP驱动巨噬细胞修复脑出血后脑白质完整性。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-21 DOI: 10.1186/s12974-025-03483-7
Huaping Huang, Yirui Kuang, Yang Chen, Yi Zhang, Jiayin Zhou, Xian Yu, Yonghe Zheng, Lingxin Cai, Wanglu Hu, Liansheng Gao, Haijian Wu, Hui Ling, Xiao Dong, Hang Zhou, Xiaobo Yu, Yucong Peng, Gao Chen, Xiaoyu Wang, Wei Yan
{"title":"Pro-repair macrophages driven by CGRP rescue white matter integrity following intracerebral hemorrhage.","authors":"Huaping Huang, Yirui Kuang, Yang Chen, Yi Zhang, Jiayin Zhou, Xian Yu, Yonghe Zheng, Lingxin Cai, Wanglu Hu, Liansheng Gao, Haijian Wu, Hui Ling, Xiao Dong, Hang Zhou, Xiaobo Yu, Yucong Peng, Gao Chen, Xiaoyu Wang, Wei Yan","doi":"10.1186/s12974-025-03483-7","DOIUrl":"10.1186/s12974-025-03483-7","url":null,"abstract":"","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"161"},"PeriodicalIF":9.3,"publicationDate":"2025-06-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12182684/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144340215","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
KChIP3 fosters neuroinflammation and synaptic dysfunction in the 5XFAD mouse model of Alzheimer's disease. KChIP3在阿尔茨海默病5XFAD小鼠模型中促进神经炎症和突触功能障碍。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-19 DOI: 10.1186/s12974-025-03426-2
Bolivar Arcos-Encarnación, Eladio Cortes-Flores, Isabel Barón-Mendoza, Jorge Luis Almazán, David Valle-García, Sol Díaz de León-Guerrero, Ladislav Hovan, Karla F Meza-Sosa, Nohemi Camacho-Concha, Jeovanis Gil, Marieke Lydia Kuijjer, Aliesha González-Arenas, Sergio Encarnación-Guevara, Gustavo Pedraza-Alva, Leonor Pérez-Martínez
{"title":"KChIP3 fosters neuroinflammation and synaptic dysfunction in the 5XFAD mouse model of Alzheimer's disease.","authors":"Bolivar Arcos-Encarnación, Eladio Cortes-Flores, Isabel Barón-Mendoza, Jorge Luis Almazán, David Valle-García, Sol Díaz de León-Guerrero, Ladislav Hovan, Karla F Meza-Sosa, Nohemi Camacho-Concha, Jeovanis Gil, Marieke Lydia Kuijjer, Aliesha González-Arenas, Sergio Encarnación-Guevara, Gustavo Pedraza-Alva, Leonor Pérez-Martínez","doi":"10.1186/s12974-025-03426-2","DOIUrl":"10.1186/s12974-025-03426-2","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a progressive neurodegenerative disorder marked by β-amyloid (βA) accumulation, neuroinflammation, excessive synaptic pruning, and cognitive decline. Despite extensive research, effective treatments remain elusive. Here, we identify potassium channel-interacting protein 3 (KChIP3) as a key driver of AD pathology using the 5XFAD mouse model. KChIP3 levels were significantly elevated in the hippocampus of 5XFAD mice, correlating with βA burden and neuroinflammation. This upregulation was triggered by inflammatory signaling via the NLRP3 inflammasome and Caspase-1 activation. Notably, genetic deletion of KChIP3 (5XFAD/KChIP3<sup>-/-</sup>) markedly reduced βA plaque deposition, pro-inflammatory cytokines, reactive gliosis, and expression of inflammation-related proteins (APO, CLU, MDK). Transcriptomic and proteomic analyses revealed restored synaptic markers (CD47, CD200, CACNB4, GDA) and a shift of the disease-associated microglial (DAM-1) phenotype. Mechanistically, we propose that KChIP3 amplifies AD pathology through two key mechanisms: (1) sustaining neuroinflammation by upregulating pro-inflammatory genes and (2) impairing synaptic integrity by repressing genes critical for neuronal function. Consistently, KChIP3 deletion enhanced dendritic complexity, synaptic plasticity, and cognitive performance in 5XFAD mice. These findings position KChIP3 as a potential therapeutic target for mitigating neuroinflammation and synaptic dysfunction in AD and highlight its potential as a biomarker for disease progression.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"160"},"PeriodicalIF":9.3,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12178027/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144333326","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting glial fibrillary acidic protein in glaucoma: a monoclonal antibody approach to modulate glial reactivity and neuroinflammation for neuroprotection. 青光眼中靶向胶质原纤维酸性蛋白:单克隆抗体方法调节胶质反应性和神经炎症以实现神经保护。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-17 DOI: 10.1186/s12974-025-03482-8
Chaoqiang Guan, Linglin Zhang, Kristian Nzogang Fomo, Jie Yang, Norbert Pfeiffer, Franz H Grus
{"title":"Targeting glial fibrillary acidic protein in glaucoma: a monoclonal antibody approach to modulate glial reactivity and neuroinflammation for neuroprotection.","authors":"Chaoqiang Guan, Linglin Zhang, Kristian Nzogang Fomo, Jie Yang, Norbert Pfeiffer, Franz H Grus","doi":"10.1186/s12974-025-03482-8","DOIUrl":"10.1186/s12974-025-03482-8","url":null,"abstract":"<p><strong>Background: </strong>Glaucoma is a progressive neurodegenerative disorder that leads to irreversible vision loss, with neuroinflammation recognized as a key factor. Overexpression of glial fibrillary acidic protein (GFAP) is linked to glaucoma pathogenesis and plays a pivotal role in astrocyte-driven neuroinflammation. This study aimed to assess the neuroprotective effects of a monoclonal antibody (mAb) targeting GFAP in glaucoma and to elucidate the underlying mechanisms.</p><p><strong>Methods: </strong>An ocular hypertension (OHT) glaucoma model was established in female Sprague Dawley rats using episcleral vein occlusion. Three doses of GFAP mAb (2.5, 25, 50 µg) or vehicle were administered via intravitreal injection. Retinal nerve fiber layer (RNFL) thickness and photopic electroretinogram were monitored longitudinally. Retinal ganglion cell (RGC) survival and glial responses were evaluated with immunostaining. Western blot and microarray analyses were performed to investigate molecular and pathway alterations. Additionally, a cobalt chloride (CoCl<sub>2</sub>)-induced degenerative R28 cell model was used to validate the protective effects of GFAP mAb in vitro. A bioinformatics re-analysis of a public glaucomatous retina protein dataset was conducted using GSEA, GO, and Cytoscape with GENEMANIA.</p><p><strong>Results: </strong>OHT resulted in a significant loss of RNFL thickness, PhNR amplitude, and RGC survival, all of which were preserved by GFAP mAb treatment. Retinal astrocyte reactivity was inhibited by GFAPmAb in a dose-dependent manner by suppressing GFAP protein overexpression. Notably, 25 µg GFAP mAb effectively regulated both astrocyte and microglial reactivity, leading to a substantial attenuation of neuroinflammation. Mechanistically, GFAP mAb inhibited the p38 MAPK and NF-κB pathways and the NLRP3/Caspase-1/GSDMD axis. In vitro, GFAP mAb improved R28 cell viability under CoCl<sub>2</sub> exposure while reducing cell death via inhibition of pyroptosis. Bioinformatic re-analysis highlighted gliosis as a prominent pathway in the glaucomatous retina and indicated GFAP and Caspase1 as central nodes in the putative mechanism network modulated by GFAP mAb.</p><p><strong>Conclusions: </strong>This study demonstrates that GFAP mAb inhibits astrogliosis and glial-glial activation, exerting neuroprotection through the inhibition of inflammation and pyroptosis. The findings suggest that targeting GFAP represents a promising immunotherapeutic strategy for glaucoma treatment.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"159"},"PeriodicalIF":9.3,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12175471/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317139","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hypothalamic kisspeptin alleviates myasthenia gravis by regulating Th1/Th17/Treg balance through Inhibition of NF-κB signaling pathway. 下丘脑kisspeptin通过抑制NF-κB信号通路调节Th1/Th17/Treg平衡,减轻重症肌无力。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2025-06-16 DOI: 10.1186/s12974-025-03486-4
Dan Lu, Linqi Liu, Wenjun Que, Rui Fan, Pingyang Ke, Jing Dong, Yaoqi Gan, Fei Xiao
{"title":"Hypothalamic kisspeptin alleviates myasthenia gravis by regulating Th1/Th17/Treg balance through Inhibition of NF-κB signaling pathway.","authors":"Dan Lu, Linqi Liu, Wenjun Que, Rui Fan, Pingyang Ke, Jing Dong, Yaoqi Gan, Fei Xiao","doi":"10.1186/s12974-025-03486-4","DOIUrl":"10.1186/s12974-025-03486-4","url":null,"abstract":"<p><strong>Background: </strong>Myasthenia gravis (MG) is an autoimmune disorder affecting neuromuscular junctions. While neuroendocrine-immune system dysfunction plays a crucial role in the development of autoimmune diseases, its involvement in MG remains largely unexplored. Kisspeptin, a neuropeptide hormone and endogenous ligand for GPR54 receptor, has been demonstrated to regulate antitumor immunity, antiviral immunity, and several autoimmune diseases. However, the role and mechanism of kisspeptin in MG remain to be elucidated.</p><p><strong>Methods: </strong>Serum kisspeptin levels were measured by ELISA in MG patients and experimental autoimmune myasthenia gravis (EAMG) rats. EAMG rats were treated with KP10 (kisspeptin analog) to evaluate its effects on body weight, clinical scores, grip strength, antibody levels, and complement deposition. Hypothalamic Kiss1 expression was assessed using Western blot and immunofluorescence. Stereotactic injection of adeno-associated virus overexpressing Kiss1 was performed to study its regulatory effects on disease progression. CD4<sup>+</sup> T cell transfer via tail vein, Western blot, and flow cytometry were employed to investigate KP10's modulatory effects on CD4<sup>+</sup> T cell subsets and the NF-κB signaling pathway.</p><p><strong>Results: </strong>Kisspeptin expression was significantly decreased in both MG patient sera and EAMG rat sera, with reduced hypothalamic Kiss1 expression in EAMG rats. Either hypothalamic Kiss1 overexpression or intraperitoneal KP10 administration significantly improved clinical signs in EAMG rats. Further in vivo and in vitro studies revealed that KP10 ameliorated EAMG clinical signs by modulating Th1/Th17/Treg cell balance through inhibition of NF-κB signaling pathway activation in CD4<sup>+</sup> T cells.</p><p><strong>Conclusion: </strong>This study elucidates that Kisspeptin secreted by hypothalamic participates in MG pathogenesis through the Kisspeptin-GPR54-NF-κB signaling axis by regulating CD4<sup>+</sup> T cell subset balance, suggesting that the kisspeptin/GPR54 pathway may serve as a potential therapeutic target for MG treatment.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"22 1","pages":"158"},"PeriodicalIF":9.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12172211/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144310059","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信