首页 > 最新文献

Journal of Neuroinflammation最新文献

英文 中文
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 Q1 IMMUNOLOGY 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

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.

围产期感染会对大脑发育产生不利影响,使婴儿易患缺血性中风,并造成终生后果。我们以前曾证实,富含 n-3 多不饱和脂肪酸(n-3 PUFA)的饮食能改变后代的脑脂质组成,并保护新生儿大脑免受中风的影响,部分原因是它能抑制损伤性免疫反应。血管尤其是内皮细胞对大脑和全身循环之间的界面至关重要,它们支持大脑的平衡并为全身感染提供屏障。在此,我们研究了母体富含 PUFA 的膳食是否会在使用 LPS 模拟感染后对出生后第 9 天小鼠的内皮细胞信号进行重编程。研究人员对从妊娠第 1 天起使用 3 种不同母体膳食(标准膳食、富含 n-3 脂肪酸膳食或富含 n-6 脂肪酸膳食)的母鼠所产幼鼠大脑中分离出的微血管进行了转录组分析。根据饮食的不同,LPS 对内皮细胞中与免疫反应、细胞周期、细胞外基质和血管生成有关的通路产生了不同的调节作用。N-3 PUFA饮食可提高脑血管的免疫反应性,同时防止标准饮食中伴随炎症反应的细胞周期调节和细胞外基质级联失衡。细胞因子分析表明,食用富含 n-3 脂肪酸膳食的母鼠后代血液和大脑中的 LPS 反应减弱。对后代体内脑血管的分析表明,血管密度没有差异。然而,在标准膳食和 n-6 膳食中,72 h 后代的血管复杂性对 LPS 的反应有所降低。因此,LPS 在生命早期会调节后代脑血管的特定转录组变化,而不是主要的血管结构特征。富含 N-3 PUFA 的母体膳食在一定程度上防止了体内平衡过程的失衡,改变了炎症,并最终减轻了感染导致的表面血管网络复杂性的变化。重要的是,母体饮食可能预示着后代神经血管日后的结果。
{"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
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 Q1 IMMUNOLOGY 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

背景:我们最近报道了多巴胺(DA)类似物CA140可调节脂多糖注射野生型(WT)小鼠和3月龄5xFAD小鼠(一种阿尔茨海默病(AD)模型)的神经炎症反应。然而,CA140对Aβ/tau病理学和突触/认知功能的影响及其分子作用机制尚不清楚:为了研究CA140对认知和突触功能以及AD病理学的影响,3个月大的WT小鼠或8个月大(老龄)的5xFAD小鼠每天注射载体(10% DMSO)或CA140(30 mg/kg,i.p.),持续10天、14天或17天。对小鼠进行了行为测试、ELISA、电生理学、RNA测序、实时PCR、高尔基体染色、免疫荧光染色和Western印迹分析:结果:在老年5xFAD小鼠(AD病理模型)中,CA140通过抑制NLRP3活化,显著减少了Aβ/tau纤维化、Aβ斑块数量、tau高磷酸化和神经炎症。此外,CA140还能下调5xFAD小鼠中与AD相关的反应性星形胶质细胞(RAs)的标记物cxcl10和RAs与疾病相关小胶质细胞(DAMs)相互作用的标记物c1qa的表达。CA140 处理还抑制了 5xFAD 小鼠原代星形胶质细胞中与 AD 相关的 RA 的标志物 s100β 和 cxcl10 的 mRNA 水平。在来自 5xFAD 小鼠的原代小胶质细胞中,CA140 处理提高了稳态小胶质细胞标记物(cx3cr1 和 p2ry12)的 mRNA 水平,降低了增殖区相关小胶质细胞标记物(gpnmb)和脂滴累积小胶质细胞标记物(cln3)的 mRNA 水平。重要的是,CA140治疗能挽救东莨菪碱(SCO)介导的长期记忆缺陷、树突棘数量和LTP损伤。在年老的5xFAD小鼠中,CA140治疗对认知/突触功能和AD病理学的这些影响受多巴胺D1受体(DRD1)/Elk1信号传导的调节。在初级海马神经元和WT小鼠中,CA140治疗通过影响DRD1/CaMKIIα和/或ERK信号传导,促进了长期记忆和树突棘的形成:我们的研究结果表明,CA140可通过调节原代海马神经元、原代星形胶质细胞/小胶质细胞、WT小鼠和老龄5xFAD小鼠的DRD1信号传导,改善神经元/突触/认知功能,改善Aβ/tau病理学和神经炎症。
{"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
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 Q1 IMMUNOLOGY 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

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.

星形胶质细胞会对神经炎症做出反应,形成炎症反应状态。尽管最近的研究已经描述了与这些反应状态相关的基因表达特征,但对炎症反应性星形胶质细胞表型所依据的细胞生物学特性的研究仍然不足。在这里,我们使用基于 CRISPR 的方法筛选人 iPSC 衍生的星形胶质细胞,发现 mTOR 激活是细胞因子诱导的内溶酶体系统重塑的驱动因素,表现为内溶酶体隔室碱化、自噬通量减少以及某些内溶酶体载体的外排增加。通过内溶酶体蛋白质组学,我们发现并重点研究了其中一种载体--IL-32,这是一种与疾病相关的促炎细胞因子,在啮齿类动物中并不存在,其分泌机制尚不十分清楚。我们发现,IL-32部分分泌于细胞外囊泡中,可能是外泌体。此外,我们还发现 IL-32 参与了炎症反应性星形胶质细胞状态的极化,并在多发性硬化病变的星形胶质细胞中上调。我们相信,我们的研究结果有助于我们了解炎症反应性星形胶质细胞表型的细胞生物学通路,并确定潜在的治疗靶点。
{"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 Q1 IMMUNOLOGY 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

背景:重症肌无力(MG)是一种自身免疫性疾病,其特征是针对神经肌肉接头结构的致病性抗体。有证据表明,由转录因子(TFs)介导的长非编码 RNAs(lncRNAs)的调控在 MG 的病理生理学中起着关键作用。然而,lncRNAs在MG中的详细分子机制在很大程度上仍未确定:方法:我们使用微阵列分析方法分析了MG中的lncRNA水平。通过生物信息学分析,我们发现LINC01566可能在MG中发挥重要作用。首先,进行了 qRT-PCR 验证 LINC1566 在 MG 患者中的表达。然后,荧光原位杂交确定了LINC01566在CD4 + T细胞中的定位。最后,还利用流式细胞术和 CCK-8 检测法分析了 LINC01566 敲除或过表达对 CD4 + T 细胞功能的影响。双荧光素酶报告实验用于验证 TF FOSL1 与 LINC01566 启动子的结合:基于 lncRNA 微阵列和差异表达分析,我们在 MG 中发现了 563 个差异表达(DE)的 lncRNA、450 个 DE mRNA 和 19 个 DE TF。然后,我们构建了一个lncRNA-TF-mRNA网络。通过网络分析,我们发现LINC01566可能通过调控T细胞相关通路在MG中发挥关键作用。进一步的实验表明,LINC01566在MG患者中的表达水平较低。在功能上,LINC01566 主要分布在细胞核中,可促进 CD4 + T 细胞凋亡并抑制细胞增殖。从机理上讲,我们推测 LINC01566 可能会负向调节参与 T 细胞活化途径的 DUSP3、CCR2、FADD、SIRPB1、LGALS3 和 SIRPB1 的表达,从而进一步影响 MG 的细胞增殖和凋亡。此外,我们发现LINC01566对MG中CD4 + T细胞的影响是由TF FOSL1介导的,体外实验表明FOSL1能与LINC01566的启动子区域结合:总之,我们的研究在临床样本和细胞实验中揭示了 LINC01566 的保护作用,说明了 FOSL1/LINC01566 负向调节 MG 中 CD4 + T 细胞活化的潜在作用和机制。
{"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 Q1 IMMUNOLOGY 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

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.

多核小胶质细胞在感染、炎症和衰老等情况下均可观察到。虽然多核小胶质细胞通常与病理情况有关,但其较大的细胞体积可能会增强其吞噬功能,从而有可能帮助清除脑碎片,并建议重新评估其病理意义。为了评估多核小胶质细胞的吞噬能力及其对清除脑碎片的影响,我们使用药理抑制剂PF-431396抑制Pyk2的活性,诱导多核小胶质细胞的形成。多核小胶质细胞吞噬β淀粉样蛋白(Aβ)寡聚体的能力增强,证明其吞噬功能增强。与此同时,Pyk2抑制剂(Pyk2-Inh,PF-431396)可减少Aβ肽诱导的Pyk2磷酸化。此外,Pyk2-inh 处理后溶酶体标志物 Lamp1 的表达增加,表明蛋白水解活性增强。在体内,我们通过将 Aβ 注入 Iba-1 EGFP 转基因(Tg)小鼠的大脑,建立了急性阿尔茨海默病(AD)模型。注射Pyk2-Inh后,小胶质细胞向Iba-1 EGFP转基因小鼠大脑中的淀粉样蛋白沉积迁移增加,并伴有形态活化,这表明小胶质细胞对Aβ的亲和力增强。在人类小胶质细胞中,脂多糖(LPS)诱导的炎症反应显示,抑制 Pyk2 信号传导可显著减少促炎症标志物的转录和蛋白表达。这些结果表明,抑制 Pyk2 可以调节小胶质细胞的功能,从而有可能减轻神经炎症并帮助清除神经退行性疾病标志物。这凸显了 Pyk2 是治疗干预神经退行性疾病的一个有前景的靶点。
{"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 Q1 IMMUNOLOGY 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

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.

乳酸衍生的组蛋白乳酰化通过转录调控参与多种病理过程。乳酸衍生的组蛋白乳酸化在脊髓损伤(SCI)修复中的作用仍不清楚。在这里,我们报告了脊髓损伤后脊髓中整体乳酸水平和乳酸化上调的情况。值得注意的是,在损伤脊髓的小胶质细胞中,H4K12la明显升高,而外源乳酸盐处理进一步升高了SCI后小胶质细胞中的H4K12la。在功能上,乳酸盐处理促进了损伤脊髓后小胶质细胞的增殖、瘢痕形成、轴突再生和运动功能的恢复。从机制上讲,乳酸盐介导的 H4K12la 升高促进了小胶质细胞中 PD-1 的转录,从而促进了 SCI 的修复。此外,一系列抢救实验证实,PD-1抑制剂或小胶质细胞特异性AAV-sh-PD-1能显著逆转乳酸盐在SCI后的治疗效果。这项研究说明了乳酸/H4K12la/PD-1 信号在小胶质细胞介导的组织修复中的功能和机制,并为 SCI 治疗提供了一个新靶点。
{"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 Q1 IMMUNOLOGY 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
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.
慢性神经炎症和小胶质细胞活化是继发性轻度创伤性脑损伤(r-mTBI)后继发性损伤级联和认知障碍的关键介质。过氧化物酶体增殖激活受体-γ(PPARγ)在小胶质细胞和脑驻留髓系细胞类型上表达,其信号在调节小胶质细胞反应方面发挥着重要的抗炎作用。在损伤后的慢性时间点,组成型 PPARγ 信号被认为是失调的,从而解除了对慢性激活的小胶质细胞的抑制作用。越来越多的证据表明,噻唑烷二酮类化合物(TZDs)是一类被批准用于治疗糖尿病的化合物,它能通过激活过氧化物酶体增殖激活受体-γ(PPARγ)有效减轻神经炎症和慢性小胶质细胞活化。本研究采用闭合头颅 r-mTBI 模型,研究 TZD 吡格列酮对 r-mTBI 暴露后认知功能和神经炎症的影响。我们发现,吡格列酮治疗可减轻伤后6个月的空间学习和记忆损伤,并减少皮质、海马和胼胝体中反应性小胶质细胞和星形胶质细胞标记物的表达。我们随后研究了吡格列酮治疗是否会改变离体小胶质细胞的炎症信号转导机制,并证实了促炎症转录因子和细胞因子水平的下调。为了进一步研究PPARγ介导的神经保护的小胶质细胞特异性机制,我们产生了一种新型他莫昔芬诱导的小胶质细胞特异性PPARγ过表达小鼠品系,并研究了它对损伤后小胶质细胞表型的影响。通过 RNA 测序,我们发现 PPARγ 的过表达能改善小胶质细胞的活化,促进与伤口愈合和组织修复相关的通路(如 IL10、IL4 和 NGF 通路)的活化,并抑制疾病相关的小胶质细胞样(DAM-like)表型的形成。这项研究深入揭示了 PPARγ 作为小鼠 r-mTBI 后神经炎症级联的关键调节因子的作用,并证明使用 PPARγ 激动剂(如吡格列酮和新一代 TZDs)对预防 r-mTBI 的慢性神经退行性后遗症具有强大的治疗潜力。
{"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 Q1 IMMUNOLOGY 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
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.
慢性脑灌注不足(CCH)是一种困扰全球无数人的疾病,是造成认知障碍的主要原因,其发病机理至今仍不甚明了。布鲁顿酪氨酸激酶抑制(BTKi)被认为是调节脑内炎症反应的一种有前途的策略,而炎症反应被认为是推动缺血性脱髓鞘进展的一个关键过程。然而,迄今为止,BTKi 在脊髓灰质炎中的潜在作用尚未得到研究。在本研究中,我们阐明了 BTK 在体外缺氧和体内缺血性脱髓鞘模型中的潜在治疗作用。我们发现,脑灌注不足会诱导白质损伤、认知障碍、小胶质细胞 BTK 激活以及一系列与炎症、氧化应激、线粒体功能障碍和铁氧化相关的小胶质细胞反应。托乐布替尼治疗可抑制小胶质细胞的活化和小胶质细胞BTK的表达。同时,与小胶质细胞相关的炎症和铁蛋白沉积过程明显减轻,从而降低了疾病的严重程度。综上所述,BTKi 可通过使小胶质细胞极化向抗炎和平衡表型倾斜,以及减少小胶质细胞氧化应激损伤和铁蛋白沉积,改善慢性脑灌注不足诱导的白质损伤和认知障碍,对慢性脑灌注不足诱导的脱髓鞘具有良好的治疗潜力。
{"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
CD44 signaling in Müller cells impacts photoreceptor function and survival in healthy and diseased retinas. Müller 细胞中的 CD44 信号影响健康视网膜和患病视网膜中感光器的功能和存活。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s12974-024-03175-8
Monika Ayten, Tobias Straub, Lew Kaplan, Stefanie M Hauck, Antje Grosche, Susanne F Koch

Retinitis pigmentosa (RP), an inherited retinal disease, affects 1,5 million people worldwide. The initial mutation-driven photoreceptor degeneration leads to chronic inflammation, characterized by Müller cell activation and upregulation of CD44. CD44 is a cell surface transmembrane glycoprotein and the primary receptor for hyaluronic acid. It is involved in many pathological processes, but little is known about CD44's retinal functions. CD44 expression is also increased in Müller cells from our Pde6bSTOP/STOP RP mouse model. To gain a more detailed understanding of CD44's role in healthy and diseased retinas, we analyzed Cd44-/- and Cd44-/-Pde6bSTOP/STOP mice, respectively. The loss of CD44 led to enhanced photoreceptor degeneration, reduced retinal function, and increased inflammatory response. To understand the underlying mechanism, we performed proteomic analysis on isolated Müller cells from Cd44-/- and Cd44-/-Pde6bSTOP/STOP retinas and identified a significant downregulation of glutamate transporter 1 (SLC1A2). This downregulation was accompanied by higher glutamate levels, suggesting impaired glutamate homeostasis. These novel findings indicate that CD44 stimulates glutamate uptake via SLC1A2 in Müller cells, which in turn, supports photoreceptor survival and function.

视网膜色素变性(RP)是一种遗传性视网膜疾病,影响着全球 150 万人。最初由突变驱动的感光细胞变性会导致慢性炎症,其特点是 Müller 细胞活化和 CD44 上调。CD44 是一种细胞表面跨膜糖蛋白,也是透明质酸的主要受体。它参与了许多病理过程,但人们对 CD44 的视网膜功能知之甚少。在我们的 Pde6bSTOP/STOP RP 小鼠模型的 Müller 细胞中,CD44 的表达也有所增加。为了更详细地了解 CD44 在健康和患病视网膜中的作用,我们分别对 Cd44-/- 和 Cd44-/-Pde6bSTOP/STOP 小鼠进行了分析。CD44 的缺失导致感光细胞变性增强、视网膜功能降低和炎症反应加剧。为了解其潜在机制,我们对来自 Cd44-/- 和 Cd44-/-Pde6bSTOP/STOP 视网膜的分离 Müller 细胞进行了蛋白质组学分析,发现谷氨酸转运体 1(SLC1A2)显著下调。谷氨酸转运体 1(SLC1A2)的下调伴随着谷氨酸水平的升高,这表明谷氨酸的稳态功能受损。这些新发现表明,CD44 通过 Müller 细胞中的 SLC1A2 刺激谷氨酸的摄取,进而支持感光细胞的存活和功能。
{"title":"CD44 signaling in Müller cells impacts photoreceptor function and survival in healthy and diseased retinas.","authors":"Monika Ayten, Tobias Straub, Lew Kaplan, Stefanie M Hauck, Antje Grosche, Susanne F Koch","doi":"10.1186/s12974-024-03175-8","DOIUrl":"10.1186/s12974-024-03175-8","url":null,"abstract":"<p><p>Retinitis pigmentosa (RP), an inherited retinal disease, affects 1,5 million people worldwide. The initial mutation-driven photoreceptor degeneration leads to chronic inflammation, characterized by Müller cell activation and upregulation of CD44. CD44 is a cell surface transmembrane glycoprotein and the primary receptor for hyaluronic acid. It is involved in many pathological processes, but little is known about CD44's retinal functions. CD44 expression is also increased in Müller cells from our Pde6b<sup>STOP/STOP</sup> RP mouse model. To gain a more detailed understanding of CD44's role in healthy and diseased retinas, we analyzed Cd44<sup>-/-</sup> and Cd44<sup>-/-</sup>Pde6b<sup>STOP/STOP</sup> mice, respectively. The loss of CD44 led to enhanced photoreceptor degeneration, reduced retinal function, and increased inflammatory response. To understand the underlying mechanism, we performed proteomic analysis on isolated Müller cells from Cd44<sup>-/-</sup> and Cd44<sup>-/-</sup>Pde6b<sup>STOP/STOP</sup> retinas and identified a significant downregulation of glutamate transporter 1 (SLC1A2). This downregulation was accompanied by higher glutamate levels, suggesting impaired glutamate homeostasis. These novel findings indicate that CD44 stimulates glutamate uptake via SLC1A2 in Müller cells, which in turn, supports photoreceptor survival and function.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297696/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878880","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
Toxoplasma infection induces an aged neutrophil population in the CNS that is associated with neuronal protection. 弓形虫感染会诱导中枢神经系统中的中性粒细胞老化,这与神经元保护有关。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s12974-024-03176-7
Kristina V Bergersen, Bill Kavvathas, Byron D Ford, Emma H Wilson

Background: Infection with the protozoan parasite Toxoplasma gondii leads to the formation of lifelong cysts in neurons that can have devastating consequences in the immunocompromised. In the immunocompetent individual, anti-parasitic effector mechanisms and a balanced immune response characterized by pro- and anti-inflammatory cytokine production establishes an asymptomatic infection that rarely leads to neurological symptoms. Several mechanisms are known to play a role in this successful immune response in the brain including T cell production of IFNγ and IL-10 and the involvement of CNS resident cells. This limitation of clinical neuropathology during chronic infection suggests a balance between immune response and neuroprotective mechanisms that collectively prevent clinical manifestations of disease. However, how these two vital mechanisms of protection interact during chronic Toxoplasma infection remains poorly understood.

Main text: This study demonstrates a previously undescribed connection between innate neutrophils found chronically in the brain, termed "chronic brain neutrophils" (CBNeuts), and neuroprotective mechanisms during Toxoplasma infection. Lack of CBNeuts during chronic infection, accomplished via systemic neutrophil depletion, led to enhanced infection and deleterious effects on neuronal regeneration and repair mechanisms in the brain. Phenotypic and transcriptomic analysis of CBNeuts identified them as distinct from peripheral neutrophils and revealed two main subsets of CBNeuts that display heterogeneity towards both classical effector and neuroprotective functions in an age-dependent manner. Further phenotypic profiling defined expression of the neuroprotective molecules NRG-1 andErbB4 by these cells, and the importance of this signaling pathway during chronic infection was demonstrated via NRG-1 treatment studies.

Conclusions: In conclusion, this work identifies CBNeuts as a heterogenous population geared towards both classical immune responses and neuroprotection during chronic Toxoplasma infection and provides the foundation for future mechanistic studies of these cells.

背景:弓形虫原生寄生虫感染后会在神经元中形成终生囊肿,对免疫力低下的患者会产生破坏性后果。在免疫功能正常的个体中,抗寄生虫效应机制和以促炎症细胞因子和抗炎症细胞因子的产生为特征的平衡免疫反应可形成无症状感染,很少导致神经系统症状。已知有几种机制在这种成功的脑部免疫反应中发挥作用,包括 T 细胞产生 IFNγ 和 IL-10,以及中枢神经系统驻留细胞的参与。慢性感染期间临床神经病理学的局限性表明,免疫反应和神经保护机制之间的平衡共同防止了疾病的临床表现。然而,人们对这两种重要的保护机制在弓形虫慢性感染期间如何相互作用仍知之甚少:这项研究表明,在弓形虫感染期间,大脑中长期存在的先天性中性粒细胞(被称为 "慢性脑中性粒细胞"(CBNeuts))与神经保护机制之间存在着一种以前未曾描述过的联系。在慢性感染期间,通过全身中性粒细胞耗竭实现的CBNeuts缺失会导致感染增强,并对大脑神经元再生和修复机制产生有害影响。CBNeuts的表型和转录组分析确定了它们与外周中性粒细胞的区别,并揭示了CBNeuts的两个主要亚群,它们以年龄依赖的方式在经典效应和神经保护功能方面显示出异质性。进一步的表型分析确定了神经保护分子 NRG-1 和 ErbB4 在这些细胞中的表达,并通过 NRG-1 治疗研究证明了这一信号通路在慢性感染期间的重要性:总之,这项工作确定了 CBNeuts 是一个异源群体,在慢性弓形虫感染期间既能做出典型的免疫反应,又能起到神经保护作用,并为今后对这些细胞进行机理研究奠定了基础。
{"title":"Toxoplasma infection induces an aged neutrophil population in the CNS that is associated with neuronal protection.","authors":"Kristina V Bergersen, Bill Kavvathas, Byron D Ford, Emma H Wilson","doi":"10.1186/s12974-024-03176-7","DOIUrl":"10.1186/s12974-024-03176-7","url":null,"abstract":"<p><strong>Background: </strong>Infection with the protozoan parasite Toxoplasma gondii leads to the formation of lifelong cysts in neurons that can have devastating consequences in the immunocompromised. In the immunocompetent individual, anti-parasitic effector mechanisms and a balanced immune response characterized by pro- and anti-inflammatory cytokine production establishes an asymptomatic infection that rarely leads to neurological symptoms. Several mechanisms are known to play a role in this successful immune response in the brain including T cell production of IFNγ and IL-10 and the involvement of CNS resident cells. This limitation of clinical neuropathology during chronic infection suggests a balance between immune response and neuroprotective mechanisms that collectively prevent clinical manifestations of disease. However, how these two vital mechanisms of protection interact during chronic Toxoplasma infection remains poorly understood.</p><p><strong>Main text: </strong>This study demonstrates a previously undescribed connection between innate neutrophils found chronically in the brain, termed \"chronic brain neutrophils\" (CBNeuts), and neuroprotective mechanisms during Toxoplasma infection. Lack of CBNeuts during chronic infection, accomplished via systemic neutrophil depletion, led to enhanced infection and deleterious effects on neuronal regeneration and repair mechanisms in the brain. Phenotypic and transcriptomic analysis of CBNeuts identified them as distinct from peripheral neutrophils and revealed two main subsets of CBNeuts that display heterogeneity towards both classical effector and neuroprotective functions in an age-dependent manner. Further phenotypic profiling defined expression of the neuroprotective molecules NRG-1 andErbB4 by these cells, and the importance of this signaling pathway during chronic infection was demonstrated via NRG-1 treatment studies.</p><p><strong>Conclusions: </strong>In conclusion, this work identifies CBNeuts as a heterogenous population geared towards both classical immune responses and neuroprotection during chronic Toxoplasma infection and provides the foundation for future mechanistic studies of these cells.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297776/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878904","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
期刊
Journal of Neuroinflammation
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1