Journal of Neuroinflammation最新文献

筛选
英文 中文
Transcriptomic analysis unveils bona fide molecular signatures of microglia under conditions of homeostasis and viral encephalitis. 转录组分析揭示了小胶质细胞在平衡状态和病毒性脑炎条件下的真正分子特征。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-17 DOI: 10.1186/s12974-024-03197-2
Felix Mulenge, Olivia Luise Gern, Lena Mareike Busker, Angela Aringo, Luca Ghita, Inken Waltl, Andreas Pavlou, Ulrich Kalinke
{"title":"Transcriptomic analysis unveils bona fide molecular signatures of microglia under conditions of homeostasis and viral encephalitis.","authors":"Felix Mulenge, Olivia Luise Gern, Lena Mareike Busker, Angela Aringo, Luca Ghita, Inken Waltl, Andreas Pavlou, Ulrich Kalinke","doi":"10.1186/s12974-024-03197-2","DOIUrl":"10.1186/s12974-024-03197-2","url":null,"abstract":"<p><p>Microglia serve as a front-line defense against neuroinvasive viral infection, however, determination of their actual transcriptional profiles under conditions of health and disease is challenging. Here, we used various experimental approaches to delineate the transcriptional landscape of microglia during viral infection. Intriguingly, multiple activation genes were found to be artificially induced in sorted microglia and we demonstrated that shear stress encountered during cell sorting was one of the key inducers. Post-hoc analysis revealed that publicly available large-scale single-cell RNA sequencing datasets were significantly tainted by aberrant signatures that are associated with cell sorting. By exploiting the ribosomal tagging approach, we developed a strategy to enrich microglia-specific transcripts by comparing immunoprecipitated RNA with total RNA. Such enriched transcripts were instrumental in defining bona fide signatures of microglia under conditions of health and virus infection. These unified microglial signatures may serve as a benchmark to retrospectively assess ex vivo artefacts from available atlases. Leveraging the microglial translatome, we found enrichment of genes implicated in T-cell activation and cytokine production during the course of VSV infection. These data linked microglia with T-cell re-stimulation and further underscored that microglia are involved in shaping antiviral T-cell responses in the brain. Collectively, our study defines the transcriptional landscape of microglia under steady state and during viral encephalitis and highlights cellular interactions between microglia and T cells that contribute to the control of virus dissemination.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11330067/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141995921","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
Secoisolariciresinol diglucoside attenuates neuroinflammation and cognitive impairment in female Alzheimer's disease mice via modulating gut microbiota metabolism and GPER/CREB/BDNF pathway. Secoisolariciresinol diglucoside通过调节肠道微生物群代谢和GPER/CREB/BDNF途径减轻雌性阿尔茨海默病小鼠的神经炎症和认知障碍
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-12 DOI: 10.1186/s12974-024-03195-4
Mengzhen Jia, Fangjie Ning, Junqing Wen, Xiaorui Wang, Jiao Chen, Jun Hu, Xuhui Chen, Zhigang Liu
{"title":"Secoisolariciresinol diglucoside attenuates neuroinflammation and cognitive impairment in female Alzheimer's disease mice via modulating gut microbiota metabolism and GPER/CREB/BDNF pathway.","authors":"Mengzhen Jia, Fangjie Ning, Junqing Wen, Xiaorui Wang, Jiao Chen, Jun Hu, Xuhui Chen, Zhigang Liu","doi":"10.1186/s12974-024-03195-4","DOIUrl":"10.1186/s12974-024-03195-4","url":null,"abstract":"<p><strong>Background: </strong>Gender is a significant risk factor for late-onset Alzheimer's disease (AD), often attributed to the decline of estrogen. The plant estrogen secoisolariciresinol diglucoside (SDG) has demonstrated anti-inflammatory and neuroprotective effects. However, the protective effects and mechanisms of SDG in female AD remain unclear.</p><p><strong>Methods: </strong>Ten-month-old female APPswe/PSEN1dE9 (APP/PS1) transgenic mice were treated with SDG to assess its potential ameliorative effects on cognitive impairments in a female AD model through a series of behavioral and biochemical experiments. Serum levels of gut microbial metabolites enterodiol (END) and enterolactone (ENL) were quantified using HPLC-MS. Correlation analysis and broad-spectrum antibiotic cocktail (ABx) treatment were employed to demonstrate the involvement of END and ENL in SDG's cognitive improvement effects in female APP/PS1 mice. Additionally, an acute neuroinflammation model was constructed in three-month-old C57BL/6J mice treated with lipopolysaccharide (LPS) and subjected to i.c.v. injection of G15, an inhibitor of G protein-coupled estrogen receptor (GPER), to investigate the mediating role of the estrogen receptor GPER in the cognitive benefits conferred by SDG.</p><p><strong>Results: </strong>SDG administration resulted in significant improvements in spatial, recognition, and working memory in female APP/PS1 mice. Neuroprotective effects were observed, including enhanced expression of CREB/BDNF and PSD-95, reduced β-amyloid (Aβ) deposition, and decreased levels of TNF-α, IL-6, and IL-10. SDG also altered gut microbiota composition, increasing serum levels of END and ENL. Correlation analysis indicated significant associations between END, ENL, cognitive performance, hippocampal Aβ-related protein mRNA expression, and cortical neuroinflammatory cytokine levels. The removal of gut microbiota inhibited END and ENL production and eliminated the neuroprotective effects of SDG. Furthermore, GPER was found to mediate the inhibitory effects of SDG on neuroinflammatory responses.</p><p><strong>Conclusion: </strong>These findings suggest that SDG promotes the production of gut microbial metabolites END and ENL, which inhibit cerebral β-amyloid deposition, activate GPER to enhance CREB/BDNF signaling pathways, and suppress neuroinflammatory responses. Consequently, SDG exerts neuroprotective effects and ameliorates cognitive impairments associated with AD in female mice.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11320852/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141971274","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
The dopamine analogue CA140 alleviates AD pathology, neuroinflammation, and rescues synaptic/cognitive functions by modulating DRD1 signaling or directly binding to Abeta. 多巴胺类似物 CA140 可通过调节 DRD1 信号或直接与 Abeta 结合,减轻注意力缺失症的病理和神经炎症,并挽救突触/认知功能。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-11 DOI: 10.1186/s12974-024-03180-x
Sehyun Chae, Hyun-Ju Lee, Ha-Eun Lee, Jieun Kim, Yoo Joo Jeong, Yuxi Lin, Hye Yun Kim, Geoffray Leriche, Rachel S Ehrlich, Sascha Castro Lingl, Min-Duk Seo, Young-Ho Lee, Jerry Yang, Jae-Ick Kim, Hyang-Sook Hoe
{"title":"The dopamine analogue CA140 alleviates AD pathology, neuroinflammation, and rescues synaptic/cognitive functions by modulating DRD1 signaling or directly binding to Abeta.","authors":"Sehyun Chae, Hyun-Ju Lee, Ha-Eun Lee, Jieun Kim, Yoo Joo Jeong, Yuxi Lin, Hye Yun Kim, Geoffray Leriche, Rachel S Ehrlich, Sascha Castro Lingl, Min-Duk Seo, Young-Ho Lee, Jerry Yang, Jae-Ick Kim, Hyang-Sook Hoe","doi":"10.1186/s12974-024-03180-x","DOIUrl":"10.1186/s12974-024-03180-x","url":null,"abstract":"<p><strong>Background: </strong>We recently reported that the dopamine (DA) analogue CA140 modulates neuroinflammatory responses in lipopolysaccharide-injected wild-type (WT) mice and in 3-month-old 5xFAD mice, a model of Alzheimer's disease (AD). However, the effects of CA140 on Aβ/tau pathology and synaptic/cognitive function and its molecular mechanisms of action are unknown.</p><p><strong>Methods: </strong>To investigate the effects of CA140 on cognitive and synaptic function and AD pathology, 3-month-old WT mice or 8-month-old (aged) 5xFAD mice were injected with vehicle (10% DMSO) or CA140 (30 mg/kg, i.p.) daily for 10, 14, or 17 days. Behavioral tests, ELISA, electrophysiology, RNA sequencing, real-time PCR, Golgi staining, immunofluorescence staining, and western blotting were conducted.</p><p><strong>Results: </strong>In aged 5xFAD mice, a model of AD pathology, CA140 treatment significantly reduced Aβ/tau fibrillation, Aβ plaque number, tau hyperphosphorylation, and neuroinflammation by inhibiting NLRP3 activation. In addition, CA140 treatment downregulated the expression of cxcl10, a marker of AD-associated reactive astrocytes (RAs), and c1qa, a marker of the interaction of RAs with disease-associated microglia (DAMs) in 5xFAD mice. CA140 treatment also suppressed the mRNA levels of s100β and cxcl10, markers of AD-associated RAs, in primary astrocytes from 5xFAD mice. In primary microglial cells from 5xFAD mice, CA140 treatment increased the mRNA levels of markers of homeostatic microglia (cx3cr1 and p2ry12) and decreased the mRNA levels of a marker of proliferative region-associated microglia (gpnmb) and a marker of lipid-droplet-accumulating microglia (cln3). Importantly, CA140 treatment rescued scopolamine (SCO)-mediated deficits in long-term memory, dendritic spine number, and LTP impairment. In aged 5xFAD mice, these effects of CA140 treatment on cognitive/synaptic function and AD pathology were regulated by dopamine D1 receptor (DRD1)/Elk1 signaling. In primary hippocampal neurons and WT mice, CA140 treatment promoted long-term memory and dendritic spine formation via effects on DRD1/CaMKIIα and/or ERK signaling.</p><p><strong>Conclusions: </strong>Our results indicate that CA140 improves neuronal/synaptic/cognitive function and ameliorates Aβ/tau pathology and neuroinflammation by modulating DRD1 signaling in primary hippocampal neurons, primary astrocytes/microglia, WT mice, and aged 5xFAD mice.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11317008/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141916992","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
Maternal n-3 enriched diet reprograms the offspring neurovascular transcriptome and blunts inflammation induced by endotoxin in the neonate. 母体富含 n-3 的饮食可重编后代神经血管转录组,并减缓新生儿由内毒素诱发的炎症。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-11 DOI: 10.1186/s12974-024-03191-8
Tetyana Chumak, Amandine Jullienne, C Joakim Ek, Maryam Ardalan, Pernilla Svedin, Ryan Quan, Arjang Salehi, Sirus Salari, Andre Obenaus, Zinaida S Vexler, Carina Mallard
{"title":"Maternal n-3 enriched diet reprograms the offspring neurovascular transcriptome and blunts inflammation induced by endotoxin in the neonate.","authors":"Tetyana Chumak, Amandine Jullienne, C Joakim Ek, Maryam Ardalan, Pernilla Svedin, Ryan Quan, Arjang Salehi, Sirus Salari, Andre Obenaus, Zinaida S Vexler, Carina Mallard","doi":"10.1186/s12974-024-03191-8","DOIUrl":"10.1186/s12974-024-03191-8","url":null,"abstract":"<p><p>Infection during the perinatal period can adversely affect brain development, predispose infants to ischemic stroke and have lifelong consequences. We previously demonstrated that diet enriched in n-3 polyunsaturated fatty acids (n-3 PUFA) transforms brain lipid composition in the offspring and protects the neonatal brain from stroke, in part by blunting injurious immune responses. Critical to the interface between the brain and systemic circulation is the vasculature, endothelial cells in particular, that support brain homeostasis and provide a barrier to systemic infection. Here, we examined whether maternal PUFA-enriched diets exert reprograming of endothelial cell signalling in postnatal day 9 mice after modeling aspects of infection using LPS. Transcriptome analysis was performed on microvessels isolated from brains of pups from dams maintained on 3 different maternal diets from gestation day 1: standard, n-3 enriched or n-6 enriched diets. Depending on the diet, in endothelial cells LPS produced distinct regulation of pathways related to immune response, cell cycle, extracellular matrix, and angiogenesis. N-3 PUFA diet enabled higher immune reactivity in brain vasculature, while preventing imbalance of cell cycle regulation and extracellular matrix cascades that accompanied inflammatory response in standard diet. Cytokine analysis revealed a blunted LPS response in blood and brain of offspring from dams on n-3 enriched diet. Analysis of cerebral vasculature in offspring in vivo revealed no differences in vessel density. However, vessel complexity was decreased in response to LPS at 72 h in standard and n-6 diets. Thus, LPS modulates specific transcriptomic changes in brain vessels of offspring rather than major structural vessel characteristics during early life. N-3 PUFA-enriched maternal diet in part prevents an imbalance in homeostatic processes, alters inflammation and ultimately mitigates changes to the complexity of surface vessel networks that result from infection. Importantly, maternal diet may presage offspring neurovascular outcomes later in life.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11316986/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141916991","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
mTOR activation induces endolysosomal remodeling and nonclassical secretion of IL-32 via exosomes in inflammatory reactive astrocytes. 激活 mTOR 可诱导炎症反应性星形胶质细胞的内溶酶体重塑并通过外泌体非典型分泌 IL-32
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-08 DOI: 10.1186/s12974-024-03165-w
Kun Leng, Brendan Rooney, Frank McCarthy, Wenlong Xia, Indigo V L Rose, Sophie Bax, Marcus Chin, Saeed Fathi, Kari A Herrington, Manuel Leonetti, Aimee Kao, Stephen P J Fancy, Joshua E Elias, Martin Kampmann
{"title":"mTOR activation induces endolysosomal remodeling and nonclassical secretion of IL-32 via exosomes in inflammatory reactive astrocytes.","authors":"Kun Leng, Brendan Rooney, Frank McCarthy, Wenlong Xia, Indigo V L Rose, Sophie Bax, Marcus Chin, Saeed Fathi, Kari A Herrington, Manuel Leonetti, Aimee Kao, Stephen P J Fancy, Joshua E Elias, Martin Kampmann","doi":"10.1186/s12974-024-03165-w","DOIUrl":"10.1186/s12974-024-03165-w","url":null,"abstract":"<p><p>Astrocytes respond and contribute to neuroinflammation by adopting inflammatory reactive states. Although recent efforts have characterized the gene expression signatures associated with these reactive states, the cell biology underlying inflammatory reactive astrocyte phenotypes remains under-explored. Here, we used CRISPR-based screening in human iPSC-derived astrocytes to identify mTOR activation a driver of cytokine-induced endolysosomal system remodeling, manifesting as alkalinization of endolysosomal compartments, decreased autophagic flux, and increased exocytosis of certain endolysosomal cargos. Through endolysosomal proteomics, we identified and focused on one such cargo-IL-32, a disease-associated pro-inflammatory cytokine not present in rodents, whose secretion mechanism is not well understood. We found that IL-32 was partially secreted in extracellular vesicles likely to be exosomes. Furthermore, we found that IL-32 was involved in the polarization of inflammatory reactive astrocyte states and was upregulated in astrocytes in multiple sclerosis lesions. We believe that our results advance our understanding of cell biological pathways underlying inflammatory reactive astrocyte phenotypes and identify potential therapeutic targets.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11312292/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906867","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
FOSL1-mediated LINC01566 negatively regulates CD4+ T-cell activation in myasthenia gravis. FOSL1 介导的 LINC01566 负向调节重症肌无力患者 CD4+ T 细胞的活化。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-07 DOI: 10.1186/s12974-024-03194-5
Lifang Li, Danyang Li, Jingnan Jin, Fanfan Xu, Ni He, Yingjie Ren, Xiaokun Wang, Liting Tian, Biying Chen, Xiaoju Li, Zihong Chen, Lanxin Zhang, Lukuan Qiao, Lihua Wang, Jianjian Wang
{"title":"FOSL1-mediated LINC01566 negatively regulates CD4<sup>+</sup> T-cell activation in myasthenia gravis.","authors":"Lifang Li, Danyang Li, Jingnan Jin, Fanfan Xu, Ni He, Yingjie Ren, Xiaokun Wang, Liting Tian, Biying Chen, Xiaoju Li, Zihong Chen, Lanxin Zhang, Lukuan Qiao, Lihua Wang, Jianjian Wang","doi":"10.1186/s12974-024-03194-5","DOIUrl":"10.1186/s12974-024-03194-5","url":null,"abstract":"<p><strong>Background: </strong>Myasthenia gravis (MG) is an autoimmune disease characterized by pathogenic antibodies that target structures of the neuromuscular junction. The evidence suggests that the regulation of long noncoding RNAs (lncRNAs) that is mediated by transcription factors (TFs) plays a key role in the pathophysiology of MG. Nevertheless, the detailed molecular mechanisms of lncRNAs in MG remain largely undetermined.</p><p><strong>Methods: </strong>Using microarray analysis, we analyzed the lncRNA levels in MG. By bioinformatics analysis, LINC01566 was found to potentially play an important role in MG. First, qRT‒PCR was performed to verify the LINC1566 expressions in MG patients. Then, fluorescence in situ hybridization was conducted to determine the localization of LINC01566 in CD4 + T cells. Finally, the impact of LINC01566 knockdown or overexpression on CD4 + T-cell function was also analyzed using flow cytometry and CCK-8 assay. A dual-luciferase reporter assay was used to validate the binding of the TF FOSL1 to the LINC01566 promoter.</p><p><strong>Results: </strong>Based on the lncRNA microarray and differential expression analyses, we identified 563 differentially expressed (DE) lncRNAs, 450 DE mRNAs and 19 DE TFs in MG. We then constructed a lncRNA-TF-mRNA network. Through network analysis, we found that LINC01566 may play a crucial role in MG by regulating T-cell-related pathways. Further experiments indicated that LINC01566 is expressed at low levels in MG patients. Functionally, LINC01566 is primarily distributed in the nucleus and can facilitate CD4 + T-cell apoptosis and inhibit cell proliferation. Mechanistically, we hypothesized that LINC01566 may negatively regulate the expressions of DUSP3, CCR2, FADD, SIRPB1, LGALS3 and SIRPB1, which are involved in the T-cell activation pathway, to further influence the cellular proliferation and apoptosis in MG. Moreover, we found that the effect of LINC01566 on CD4 + T cells in MG was mediated by the TF FOSL1, and in vitro experiments indicated that FOSL1 can bind to the promoter region of LINC01566.</p><p><strong>Conclusions: </strong>In summary, our research revealed the protective roles of LINC01566 in clinical samples and cellular experiments, illustrating the potential roles and mechanism by which FOSL1/LINC01566 negatively regulates CD4 + T-cell activation in MG.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11308467/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141902070","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
Enhanced phagocytosis associated with multinucleated microglia via Pyk2 inhibition in an acute β-amyloid infusion model. 在急性β淀粉样蛋白输注模型中通过抑制Pyk2增强与多核小胶质细胞相关的吞噬能力
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-06 DOI: 10.1186/s12974-024-03192-7
Ji-Won Lee, Kaito Mizuno, Haruhisa Watanabe, In-Hee Lee, Takuya Tsumita, Kyoko Hida, Yasutaka Yawaka, Yoshimasa Kitagawa, Akira Hasebe, Tadahiro Iimura, Sek Won Kong
{"title":"Enhanced phagocytosis associated with multinucleated microglia via Pyk2 inhibition in an acute β-amyloid infusion model.","authors":"Ji-Won Lee, Kaito Mizuno, Haruhisa Watanabe, In-Hee Lee, Takuya Tsumita, Kyoko Hida, Yasutaka Yawaka, Yoshimasa Kitagawa, Akira Hasebe, Tadahiro Iimura, Sek Won Kong","doi":"10.1186/s12974-024-03192-7","DOIUrl":"10.1186/s12974-024-03192-7","url":null,"abstract":"<p><p>Multinucleated microglia have been observed in contexts associated with infection, inflammation, and aging. Though commonly linked to pathological conditions, the larger cell size of multinucleated microglia might enhance their phagocytic functions, potentially aiding in the clearance of brain debris and suggesting a reassessment of their pathological significance. To assess the phagocytic capacity of multinucleated microglia and its implications for brain debris clearance, we induced their formation by inhibiting Pyk2 activity using the pharmacological inhibitor PF-431396, which triggers cytokinesis regression. Multinucleated microglia demonstrate enhanced phagocytic function, as evidenced by their increased capacity to engulf β-amyloid (Aβ) oligomers. Concurrently, the phosphorylation of Pyk2, induced by Aβ peptide, was diminished upon treatment with a Pyk2 inhibitor (Pyk2-Inh, PF-431396). Furthermore, the increased expression of Lamp1, a lysosomal marker, with Pyk2-inh treatment, suggests an enhancement in proteolytic activity. In vivo, we generated an acute Alzheimer's disease (AD) model by infusing Aβ into the brains of Iba-1 EGFP transgenic (Tg) mice. The administration of the Pyk2-Inh led to an increased migration of microglia toward amyloid deposits in the brains of Iba-1 EGFP Tg mice, accompanied by morphological activation, suggesting a heightened affinity for Aβ. In human microglia, lipopolysaccharide (LPS)-induced inflammatory responses showed that inhibition of Pyk2 signaling significantly reduced the transcription and protein expression of pro-inflammatory markers. These results suggest that Pyk2 inhibition can modulate microglial functions, potentially reducing neuroinflammation and aiding in the clearance of neurodegenerative disease markers. This highlights Pyk2 as a promising target for therapeutic intervention in neurodegenerative diseases.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11301859/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141897585","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
Lactate promotes microglial scar formation and facilitates locomotor function recovery by enhancing histone H4 lysine 12 lactylation after spinal cord injury. 乳酸可促进小胶质细胞瘢痕的形成,并通过增强脊髓损伤后组蛋白 H4 赖氨酸 12 的乳化作用促进运动功能的恢复。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-03 DOI: 10.1186/s12974-024-03186-5
Xuyang Hu, Jinxin Huang, Ziyu Li, Jianjian Li, Fangru Ouyang, Zeqiang Chen, Yiteng Li, Yuanzhe Zhao, Jingwen Wang, Shuisheng Yu, Juehua Jing, Li Cheng
{"title":"Lactate promotes microglial scar formation and facilitates locomotor function recovery by enhancing histone H4 lysine 12 lactylation after spinal cord injury.","authors":"Xuyang Hu, Jinxin Huang, Ziyu Li, Jianjian Li, Fangru Ouyang, Zeqiang Chen, Yiteng Li, Yuanzhe Zhao, Jingwen Wang, Shuisheng Yu, Juehua Jing, Li Cheng","doi":"10.1186/s12974-024-03186-5","DOIUrl":"10.1186/s12974-024-03186-5","url":null,"abstract":"<p><p>Lactate-derived histone lactylation is involved in multiple pathological processes through transcriptional regulation. The role of lactate-derived histone lactylation in the repair of spinal cord injury (SCI) remains unclear. Here we report that overall lactate levels and lactylation are upregulated in the spinal cord after SCI. Notably, H4K12la was significantly elevated in the microglia of the injured spinal cord, whereas exogenous lactate treatment further elevated H4K12la in microglia after SCI. Functionally, lactate treatment promoted microglial proliferation, scar formation, axon regeneration, and locomotor function recovery after SCI. Mechanically, lactate-mediated H4K12la elevation promoted PD-1 transcription in microglia, thereby facilitating SCI repair. Furthermore, a series of rescue experiments confirmed that a PD-1 inhibitor or microglia-specific AAV-sh-PD-1 significantly reversed the therapeutic effects of lactate following SCI. This study illustrates the function and mechanism of lactate/H4K12la/PD-1 signaling in microglia-mediated tissue repair and provides a novel target for SCI therapy.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297795/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878902","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
PPARγ activation ameliorates cognitive impairment and chronic microglial activation in the aftermath of r-mTBI PPARγ 激活可改善 r-mTBI 后的认知障碍和慢性小胶质细胞活化
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-03 DOI: 10.1186/s12974-024-03173-w
Andrew Pearson, Milica Koprivica, Max Eisenbaum, Camila Ortiz, Mackenzie Browning, Tessa Vincennie, Cooper Tinsley, Michael Mullan, Fiona Crawford, Joseph Ojo
{"title":"PPARγ activation ameliorates cognitive impairment and chronic microglial activation in the aftermath of r-mTBI","authors":"Andrew Pearson, Milica Koprivica, Max Eisenbaum, Camila Ortiz, Mackenzie Browning, Tessa Vincennie, Cooper Tinsley, Michael Mullan, Fiona Crawford, Joseph Ojo","doi":"10.1186/s12974-024-03173-w","DOIUrl":"https://doi.org/10.1186/s12974-024-03173-w","url":null,"abstract":"Chronic neuroinflammation and microglial activation are key mediators of the secondary injury cascades and cognitive impairment that follow exposure to repetitive mild traumatic brain injury (r-mTBI). Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed on microglia and brain resident myeloid cell types and their signaling plays a major anti-inflammatory role in modulating microglial responses. At chronic timepoints following injury, constitutive PPARγ signaling is thought to be dysregulated, thus releasing the inhibitory brakes on chronically activated microglia. Increasing evidence suggests that thiazolidinediones (TZDs), a class of compounds approved from the treatment of diabetes mellitus, effectively reduce neuroinflammation and chronic microglial activation by activating the peroxisome proliferator-activated receptor-γ (PPARγ). The present study used a closed-head r-mTBI model to investigate the influence of the TZD Pioglitazone on cognitive function and neuroinflammation in the aftermath of r-mTBI exposure. We revealed that Pioglitazone treatment attenuated spatial learning and memory impairments at 6 months post-injury and reduced the expression of reactive microglia and astrocyte markers in the cortex, hippocampus, and corpus callosum. We then examined whether Pioglitazone treatment altered inflammatory signaling mechanisms in isolated microglia and confirmed downregulation of proinflammatory transcription factors and cytokine levels. To further investigate microglial-specific mechanisms underlying PPARγ-mediated neuroprotection, we generated a novel tamoxifen-inducible microglial-specific PPARγ overexpression mouse line and examined its influence on microglial phenotype following injury. Using RNA sequencing, we revealed that PPARγ overexpression ameliorates microglial activation, promotes the activation of pathways associated with wound healing and tissue repair (such as: IL10, IL4 and NGF pathways), and inhibits the adoption of a disease-associated microglia-like (DAM-like) phenotype. This study provides insight into the role of PPARγ as a critical regulator of the neuroinflammatory cascade that follows r-mTBI in mice and demonstrates that the use of PPARγ agonists such as Pioglitazone and newer generation TZDs hold strong therapeutic potential to prevent the chronic neurodegenerative sequelae of r-mTBI.","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141887059","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
Bruton’s tyrosine kinase inhibition ameliorated neuroinflammation during chronic white matter ischemia 布鲁顿酪氨酸激酶抑制剂可改善慢性白质缺血期间的神经炎症
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-03 DOI: 10.1186/s12974-024-03187-4
Lu-Lu Xu, Sheng Yang, Luo-Qi Zhou, Yun-Hui Chu, Xiao-Wei Pang, Yun-Fan You, Hang Zhang, Lu-Yang Zhang, Li-Fang Zhu, Lian Chen, Ke Shang, Jun Xiao, Wei Wang, Dai-Shi Tian, Chuan Qin
{"title":"Bruton’s tyrosine kinase inhibition ameliorated neuroinflammation during chronic white matter ischemia","authors":"Lu-Lu Xu, Sheng Yang, Luo-Qi Zhou, Yun-Hui Chu, Xiao-Wei Pang, Yun-Fan You, Hang Zhang, Lu-Yang Zhang, Li-Fang Zhu, Lian Chen, Ke Shang, Jun Xiao, Wei Wang, Dai-Shi Tian, Chuan Qin","doi":"10.1186/s12974-024-03187-4","DOIUrl":"https://doi.org/10.1186/s12974-024-03187-4","url":null,"abstract":"Chronic cerebral hypoperfusion (CCH), a disease afflicting numerous individuals worldwide, is a primary cause of cognitive deficits, the pathogenesis of which remains poorly understood. Bruton’s tyrosine kinase inhibition (BTKi) is considered a promising strategy to regulate inflammatory responses within the brain, a crucial process that is assumed to drive ischemic demyelination progression. However, the potential role of BTKi in CCH has not been investigated so far. In the present study, we elucidated potential therapeutic roles of BTK in both in vitro hypoxia and in vivo ischemic demyelination model. We found that cerebral hypoperfusion induced white matter injury, cognitive impairments, microglial BTK activation, along with a series of microglia responses associated with inflammation, oxidative stress, mitochondrial dysfunction, and ferroptosis. Tolebrutinib treatment suppressed both the activation of microglia and microglial BTK expression. Meanwhile, microglia-related inflammation and ferroptosis processes were attenuated evidently, contributing to lower levels of disease severity. Taken together, BTKi ameliorated white matter injury and cognitive impairments induced by CCH, possibly via skewing microglia polarization towards anti-inflammatory and homeostatic phenotypes, as well as decreasing microglial oxidative stress damage and ferroptosis, which exhibits promising therapeutic potential in chronic cerebral hypoperfusion-induced demyelination.","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141882268","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
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:481959085
Book学术官方微信