首页 > 最新文献

Journal of Neuroinflammation最新文献

英文 中文
Senolytic treatment diminishes microglia and decreases severity of experimental autoimmune encephalomyelitis. 溶酶治疗可减少小胶质细胞,减轻实验性自身免疫性脑脊髓炎的严重程度。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-11-01 DOI: 10.1186/s12974-024-03278-2
Sienna S Drake, Aliyah Zaman, Christine Gianfelice, Elizabeth M-L Hua, Kali Heale, Elia Afanasiev, Wendy Klement, Jo Anne Stratton, Alexandre Prat, Stephanie Zandee, Alyson E Fournier

Background: The role of senescence in disease contexts is complex, however there is considerable evidence that depletion of senescent cells improves outcomes in a variety of contexts particularly related to aging, cognition, and neurodegeneration. Much research has shown previously that inflammation can promote cellular senescence. Microglia are a central nervous system innate immune cell that undergo senescence with aging and during neurodegeneration. The contribution of senescent microglia to multiple sclerosis, an inflammatory neurodegenerative disease, is not clear, but microglia are strongly implicated in chronic active lesion pathology, tissue injury, and disease progression. Drugs that could specifically eliminate dysregulated microglia in multiple sclerosis are therefore of great interest to the field.

Results: A single-cell analysis of brain tissue from mice subjected to experimental autoimmune encephalomyelitis (EAE), a mouse model of CNS inflammation that models aspects of multiple sclerosis (MS), identified microglia with a strong transcriptional signature of senescence including the presence of BCL2-family gene transcripts. Microglia expressing Bcl2l1 had higher expression of pro-inflammatory and senescence associated genes than their Bcl2l1 negative counterparts in EAE, suggesting they may exacerbate inflammation. Notably, in human single-nucleus sequencing from MS, BCL2L1 positive microglia were enriched in lesions with active inflammatory pathology, and likewise demonstrated increased expression of immune genes suggesting they may be proinflammatory and contribute to disease processes in chronic active lesions. Employing a small molecule BCL2-family inhibitor, Navitoclax (ABT-263), significantly reduced the presence of microglia and macrophages in the EAE spinal cord, suggesting that these cells can be targeted by senolytic treatment. ABT-263 treatment had a profound effect on EAE mice: decreasing motor symptom severity, improving visual acuity, promoting neuronal survival, and decreasing white matter inflammation.

Conclusion: These results support the hypothesis that microglia and macrophages exhibit transcriptional features of cellular senescence in EAE and MS, and that microglia expressing Bcl2l1 demonstrate a proinflammatory signature that may exacerbate inflammation resulting in negative outcomes in neuroinflammatory disease. Depleting microglia and macrophages using a senolytic results in robust improvement in EAE disease severity, including across measures of neurodegeneration, inflammation, and demyelination, and may therefore represent a novel strategy to address disease progression in multiple sclerosis.

背景:衰老在疾病中的作用是复杂的,但有大量证据表明,衰老细胞的耗竭可改善各种疾病的治疗效果,尤其是与衰老、认知和神经退行性变有关的疾病。大量研究表明,炎症可促进细胞衰老。小胶质细胞是一种中枢神经系统先天性免疫细胞,会随着衰老和神经变性而衰老。衰老的小胶质细胞对多发性硬化症这种炎症性神经退行性疾病的影响尚不明确,但小胶质细胞与慢性活动性病变病理、组织损伤和疾病进展密切相关。因此,能特异性消除多发性硬化症中失调的小胶质细胞的药物引起了该领域的极大兴趣:实验性自身免疫性脑脊髓炎(EAE)是中枢神经系统炎症的一种小鼠模型,可模拟多发性硬化症(MS)的某些方面,对实验性自身免疫性脑脊髓炎小鼠的脑组织进行的单细胞分析发现,小胶质细胞具有强烈的衰老转录特征,包括存在 BCL2 家族基因转录物。在EAE中,表达Bcl2l1的小胶质细胞比Bcl2l1阴性的小胶质细胞有更高的促炎症和衰老相关基因表达,这表明它们可能会加剧炎症。值得注意的是,在多发性硬化症的人类单核测序中,BCL2L1 阳性的小胶质细胞富集在炎症病理活跃的病灶中,而且同样显示出免疫基因表达的增加,这表明它们可能是促炎症的,有助于慢性活跃病灶的疾病进程。使用小分子BCL2家族抑制剂Navitoclax(ABT-263)能显著减少EAE脊髓中小胶质细胞和巨噬细胞的存在,这表明这些细胞可以成为衰老治疗的靶点。ABT-263治疗对EAE小鼠产生了深远的影响:降低运动症状的严重程度,改善视力,促进神经元存活,减少白质炎症:这些结果支持以下假设:在 EAE 和多发性硬化症中,小胶质细胞和巨噬细胞表现出细胞衰老的转录特征,表达 Bcl2l1 的小胶质细胞显示出一种促炎特征,可能会加剧炎症,导致神经炎症性疾病的不良后果。使用衰老剂消耗小胶质细胞和巨噬细胞可显著改善 EAE 疾病的严重程度,包括神经变性、炎症和脱髓鞘等方面,因此可能是解决多发性硬化症疾病进展的一种新策略。
{"title":"Senolytic treatment diminishes microglia and decreases severity of experimental autoimmune encephalomyelitis.","authors":"Sienna S Drake, Aliyah Zaman, Christine Gianfelice, Elizabeth M-L Hua, Kali Heale, Elia Afanasiev, Wendy Klement, Jo Anne Stratton, Alexandre Prat, Stephanie Zandee, Alyson E Fournier","doi":"10.1186/s12974-024-03278-2","DOIUrl":"10.1186/s12974-024-03278-2","url":null,"abstract":"<p><strong>Background: </strong>The role of senescence in disease contexts is complex, however there is considerable evidence that depletion of senescent cells improves outcomes in a variety of contexts particularly related to aging, cognition, and neurodegeneration. Much research has shown previously that inflammation can promote cellular senescence. Microglia are a central nervous system innate immune cell that undergo senescence with aging and during neurodegeneration. The contribution of senescent microglia to multiple sclerosis, an inflammatory neurodegenerative disease, is not clear, but microglia are strongly implicated in chronic active lesion pathology, tissue injury, and disease progression. Drugs that could specifically eliminate dysregulated microglia in multiple sclerosis are therefore of great interest to the field.</p><p><strong>Results: </strong>A single-cell analysis of brain tissue from mice subjected to experimental autoimmune encephalomyelitis (EAE), a mouse model of CNS inflammation that models aspects of multiple sclerosis (MS), identified microglia with a strong transcriptional signature of senescence including the presence of BCL2-family gene transcripts. Microglia expressing Bcl2l1 had higher expression of pro-inflammatory and senescence associated genes than their Bcl2l1 negative counterparts in EAE, suggesting they may exacerbate inflammation. Notably, in human single-nucleus sequencing from MS, BCL2L1 positive microglia were enriched in lesions with active inflammatory pathology, and likewise demonstrated increased expression of immune genes suggesting they may be proinflammatory and contribute to disease processes in chronic active lesions. Employing a small molecule BCL2-family inhibitor, Navitoclax (ABT-263), significantly reduced the presence of microglia and macrophages in the EAE spinal cord, suggesting that these cells can be targeted by senolytic treatment. ABT-263 treatment had a profound effect on EAE mice: decreasing motor symptom severity, improving visual acuity, promoting neuronal survival, and decreasing white matter inflammation.</p><p><strong>Conclusion: </strong>These results support the hypothesis that microglia and macrophages exhibit transcriptional features of cellular senescence in EAE and MS, and that microglia expressing Bcl2l1 demonstrate a proinflammatory signature that may exacerbate inflammation resulting in negative outcomes in neuroinflammatory disease. Depleting microglia and macrophages using a senolytic results in robust improvement in EAE disease severity, including across measures of neurodegeneration, inflammation, and demyelination, and may therefore represent a novel strategy to address disease progression in multiple sclerosis.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11529445/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142564436","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
Ethanol-activated microglial exosomes induce MCP1 signaling mediated death of stress-regulatory proopiomelanocortin neurons in the developing hypothalamus. 乙醇激活的小胶质细胞外泌体可诱导 MCP1 信号介导下丘脑发育期应激调节性前绒毛膜促皮质素神经元死亡。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-30 DOI: 10.1186/s12974-024-03274-6
Prashant Tarale, Shaista Chaudhary, Sayani Mukherjee, Dipak K Sarkar

Background: Microglia, a type of resident immune cells within the central nervous system, have been implicated in ethanol-activated neuronal death of the stress regulatory proopiomelanocortin (POMC) neuron-producing β-endorphin peptides in the hypothalamus in a postnatal rat model of fetal alcohol spectrum disorders. We determined if microglial extracellular vesicles (exosomes) are involved in the ethanol-induced neuronal death of the β-endorphin neuron via secreting elevated levels of the chemokine monocyte chemoattractant protein 1 (MCP1), a key regulator of neuroinflammation.

Methods: We employed an in vitro model, consisting of primary culture of hypothalamic microglia prepared from postnatal day 2 (PND2) rat hypothalami and treated with or without 50 mM ethanol for 24 h, and an in vivo animal model in which microglia were obtained from hypothalami of PND6 rats fed daily with 2.5 mg/kg ethanol or control milk formula for five days prior to use. Exosomes were extracted and characterized with nanosight tracking analysis (NTA), transmission electron microscopy and western blot. Chemokine multiplex immunoassay and ELISA were used for quantitative estimation of MCP1 level. Neurotoxic ability of exosome was tested using primary cultures of β-endorphin neurons and employing nucleosome assay and immunocytochemistry. Elevated plus maze, open field and restraint tests were used to assess anxiety-related behaviors.

Results: Ethanol elevated MCP1 levels in microglial exosomes both in vitro and in vivo models. Ethanol-activated microglial exosomes when introduced into primary cultures of β-endorphin neurons, increased cellular levels of MCP1 and the chemokine receptor CCR2 related signaling molecules including inflammatory cytokines and apoptotic genes as well as apoptotic death of β-endorphin neurons. These effects of microglial exosomes on β-endorphin neurons were suppressed by a CCR2 antagonist RS504393. Furthermore, RS504393 when injected in postnatal rats prior to feeding with ethanol it reduced alcohol-induced β-endorphin neuronal death in the hypothalamus. RS504393 also suppressed corticosterone response to stress and anxiety-like behaviors in postnatally alcohol-fed rats during adult period.

Conclusion: These data suggest that alcohol exposures during the developmental period elevates MCP1 levels in microglial exosomes that promote MCP1/CCR2 signaling to increase the apoptosis of β-endorphin neurons and resulting in hormonal and behavioral stress responses.

背景:小胶质细胞是中枢神经系统内的一种常驻免疫细胞,在胎儿酒精中毒谱系障碍的产后大鼠模型中,小胶质细胞与乙醇激活下丘脑中产生β-内啡肽的应激调节原绒毛膜促皮质素(POMC)神经元的神经元死亡有关。我们确定了小胶质细胞胞外囊泡(外泌体)是否通过分泌高水平的趋化因子单核细胞趋化蛋白1(MCP1)参与了乙醇诱导的β-内啡肽神经元死亡:我们采用了一种体外模型和一种体内动物模型,前者是从出生后第 2 天(PND2)大鼠下丘脑制备的下丘脑小胶质细胞原代培养物中提取的小胶质细胞,用或不用 50 mM 乙醇处理 24 小时;后者是从出生后第 6 天(PND6)大鼠下丘脑中提取的小胶质细胞,在使用前每天用 2.5 mg/kg 乙醇或对照奶粉喂养 5 天。外泌体被提取出来,并通过纳米光跟踪分析(NTA)、透射电子显微镜和免疫印迹进行表征。化学趋化因子多重免疫分析法和酶联免疫吸附法用于定量评估 MCP1 的水平。利用β-内啡肽神经元的原代培养物、核糖体检测法和免疫细胞化学法检测了外泌体的神经毒性能力。高架迷宫、开阔地和束缚试验用于评估焦虑相关行为:结果:在体外和体内模型中,乙醇都会升高小胶质细胞外泌体中的 MCP1 水平。乙醇激活的小胶质细胞外泌体被引入到β-内啡肽神经元的原代培养物中时,会增加细胞中MCP1和趋化因子受体CCR2相关信号分子的水平,包括炎症细胞因子和凋亡基因,以及β-内啡肽神经元的凋亡。CCR2拮抗剂RS504393抑制了小胶质细胞外泌体对β-内啡肽神经元的这些影响。此外,在喂食乙醇之前给出生后的大鼠注射 RS504393,可减少乙醇诱导的下丘脑β-内啡肽神经元死亡。RS504393还能抑制出生后喂食酒精的大鼠成年后对应激和焦虑样行为的皮质酮反应:这些数据表明,发育期接触酒精会使小胶质细胞外泌体中的 MCP1 水平升高,从而促进 MCP1/CCR2 信号转导,增加 β 内啡肽神经元的凋亡,导致激素和行为应激反应。
{"title":"Ethanol-activated microglial exosomes induce MCP1 signaling mediated death of stress-regulatory proopiomelanocortin neurons in the developing hypothalamus.","authors":"Prashant Tarale, Shaista Chaudhary, Sayani Mukherjee, Dipak K Sarkar","doi":"10.1186/s12974-024-03274-6","DOIUrl":"10.1186/s12974-024-03274-6","url":null,"abstract":"<p><strong>Background: </strong>Microglia, a type of resident immune cells within the central nervous system, have been implicated in ethanol-activated neuronal death of the stress regulatory proopiomelanocortin (POMC) neuron-producing β-endorphin peptides in the hypothalamus in a postnatal rat model of fetal alcohol spectrum disorders. We determined if microglial extracellular vesicles (exosomes) are involved in the ethanol-induced neuronal death of the β-endorphin neuron via secreting elevated levels of the chemokine monocyte chemoattractant protein 1 (MCP1), a key regulator of neuroinflammation.</p><p><strong>Methods: </strong>We employed an in vitro model, consisting of primary culture of hypothalamic microglia prepared from postnatal day 2 (PND2) rat hypothalami and treated with or without 50 mM ethanol for 24 h, and an in vivo animal model in which microglia were obtained from hypothalami of PND6 rats fed daily with 2.5 mg/kg ethanol or control milk formula for five days prior to use. Exosomes were extracted and characterized with nanosight tracking analysis (NTA), transmission electron microscopy and western blot. Chemokine multiplex immunoassay and ELISA were used for quantitative estimation of MCP1 level. Neurotoxic ability of exosome was tested using primary cultures of β-endorphin neurons and employing nucleosome assay and immunocytochemistry. Elevated plus maze, open field and restraint tests were used to assess anxiety-related behaviors.</p><p><strong>Results: </strong>Ethanol elevated MCP1 levels in microglial exosomes both in vitro and in vivo models. Ethanol-activated microglial exosomes when introduced into primary cultures of β-endorphin neurons, increased cellular levels of MCP1 and the chemokine receptor CCR2 related signaling molecules including inflammatory cytokines and apoptotic genes as well as apoptotic death of β-endorphin neurons. These effects of microglial exosomes on β-endorphin neurons were suppressed by a CCR2 antagonist RS504393. Furthermore, RS504393 when injected in postnatal rats prior to feeding with ethanol it reduced alcohol-induced β-endorphin neuronal death in the hypothalamus. RS504393 also suppressed corticosterone response to stress and anxiety-like behaviors in postnatally alcohol-fed rats during adult period.</p><p><strong>Conclusion: </strong>These data suggest that alcohol exposures during the developmental period elevates MCP1 levels in microglial exosomes that promote MCP1/CCR2 signaling to increase the apoptosis of β-endorphin neurons and resulting in hormonal and behavioral stress responses.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526652/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142546031","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
Japanese encephalitis virus-induced DNA methylation contributes to blood-brain barrier permeability by modulating tight junction protein expression. 日本脑炎病毒诱导的 DNA 甲基化通过调节紧密连接蛋白的表达促进血脑屏障的通透性。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-28 DOI: 10.1186/s12974-024-03266-6
Xiao Xiang, Du Yu, Zhuangzhuang Li, Jelke J Fros, Jianchao Wei, Ke Liu, Zongjie Li, Donghua Shao, Beibei Li, Jeroen Kortekaas, Monique M van Oers, Zhiyong Ma, Gorben P Pijlman, Yafeng Qiu

Japanese encephalitis virus (JEV) is a neurotropic and neuroinvasive flavivirus causing viral encephalitis, which seriously threatens the development of animal husbandry and human health. DNA methylation is a major epigenetic modification involved in viral pathogenesis, yet how DNA methylation affects JEV infection remains unknown. Here, we show genome-wide DNA methylation profiles in the brains of JEV-infected mice compared to mock-infected mice. JEV can significantly increase the overall DNA methylation levels in JEV-infected mouse brains. A total of 14,781 differentially methylated regions associated genes (DMGs) have been identified. Subsequently, KEGG pathway analysis suggested that DNA methylation modulates the tight junction signaling pathway, which can potentially impact the permeability of the blood-brain barrier (BBB). We demonstrate that hypermethylation of the tight junction gene Afdn promoter inhibited AFDN expression and increased monolayer permeability of mouse brain microvascular endothelial (bEnd.3) cells in an in vitro transwell assay. Collectively, this study reveals that DNA methylation is increased in a murine Japanese encephalitis model and that modulation of Afdn expression promotes BBB permeability.

日本脑炎病毒(JEV)是一种具有神经侵袭性和神经侵入性的黄病毒,可引起病毒性脑炎,严重威胁畜牧业的发展和人类健康。DNA 甲基化是病毒致病过程中的主要表观遗传修饰,但 DNA 甲基化如何影响 JEV 感染仍是未知数。在这里,我们展示了与模拟感染小鼠相比,JEV 感染小鼠大脑中的全基因组 DNA 甲基化图谱。JEV能明显增加JEV感染小鼠大脑中DNA甲基化的整体水平。共鉴定出 14781 个差异甲基化区域相关基因(DMGs)。随后,KEGG通路分析表明,DNA甲基化可调节紧密连接信号通路,从而可能影响血脑屏障(BBB)的通透性。我们证明,在体外透孔试验中,紧密连接基因 Afdn 启动子的高甲基化抑制了 AFDN 的表达,并增加了小鼠脑微血管内皮细胞(bEnd.3)的单层渗透性。总之,这项研究揭示了 DNA 甲基化在小鼠日本脑炎模型中会增加,而调节 Afdn 的表达会促进 BBB 的通透性。
{"title":"Japanese encephalitis virus-induced DNA methylation contributes to blood-brain barrier permeability by modulating tight junction protein expression.","authors":"Xiao Xiang, Du Yu, Zhuangzhuang Li, Jelke J Fros, Jianchao Wei, Ke Liu, Zongjie Li, Donghua Shao, Beibei Li, Jeroen Kortekaas, Monique M van Oers, Zhiyong Ma, Gorben P Pijlman, Yafeng Qiu","doi":"10.1186/s12974-024-03266-6","DOIUrl":"10.1186/s12974-024-03266-6","url":null,"abstract":"<p><p>Japanese encephalitis virus (JEV) is a neurotropic and neuroinvasive flavivirus causing viral encephalitis, which seriously threatens the development of animal husbandry and human health. DNA methylation is a major epigenetic modification involved in viral pathogenesis, yet how DNA methylation affects JEV infection remains unknown. Here, we show genome-wide DNA methylation profiles in the brains of JEV-infected mice compared to mock-infected mice. JEV can significantly increase the overall DNA methylation levels in JEV-infected mouse brains. A total of 14,781 differentially methylated regions associated genes (DMGs) have been identified. Subsequently, KEGG pathway analysis suggested that DNA methylation modulates the tight junction signaling pathway, which can potentially impact the permeability of the blood-brain barrier (BBB). We demonstrate that hypermethylation of the tight junction gene Afdn promoter inhibited AFDN expression and increased monolayer permeability of mouse brain microvascular endothelial (bEnd.3) cells in an in vitro transwell assay. Collectively, this study reveals that DNA methylation is increased in a murine Japanese encephalitis model and that modulation of Afdn expression promotes BBB permeability.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11520778/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142522169","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
Ganglion cell-derived LysoPS induces retinal neovascularisation by activating the microglial GPR34-PI3K-AKT-NINJ1 axis. 神经节细胞衍生的 LysoPS 通过激活小胶质细胞 GPR34-PI3K-AKT-NINJ1 轴诱导视网膜新生血管。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-28 DOI: 10.1186/s12974-024-03265-7
Lushu Chen, HuiYing Zhang, Ying Zhang, Xiumiao Li, MeiHuan Wang, Yaming Shen, Yuan Cao, Yong Xu, Jin Yao

Retinal neovascularisation is a major cause of blindness in patients with proliferative diabetic retinopathy (PDR). It is mediated by the complex interaction between dysfunctional ganglion cells, microglia, and vascular endothelial cells. Notably, retinal microglia, the intrinsic immune cells of the retina, play a crucial role in the pathogenesis of retinopathy. In this study, we found that lysophosphatidylserines (LysoPS) released from injured ganglion cells induced microglial extracellular trap formation and retinal neovascularisation. Mechanistically, LysoPS activated the GPR34-PI3K-AKT-NINJ1 signalling axis by interacting with the GPR34 receptor on the microglia. This activation upregulated the expression of inflammatory cytokines, such as IL-6, IL-8, VEGFA, and FGF2, and facilitated retinal vascular endothelial cell angiogenesis. As a result, inhibition of the GPR34-PI3K-AKT-NINJ1 axis significantly decreased microglial extracellular trap formation and neovascularisation by suppressing LysoPS-induced microglial inflammatory responses, both in vitro and in vivo. This study reveals the crucial role of apoptotic ganglion cells in activating microglial inflammation in PDR, thereby enhancing our understanding of the pathogenesis of retinal neovascularisation.

视网膜新生血管是增生性糖尿病视网膜病变(PDR)患者失明的主要原因。它是由功能失调的神经节细胞、小胶质细胞和血管内皮细胞之间复杂的相互作用介导的。值得注意的是,视网膜小胶质细胞是视网膜固有的免疫细胞,在视网膜病变的发病机制中起着至关重要的作用。在这项研究中,我们发现受伤的神经节细胞释放的溶血磷脂酰丝氨酸(LysoPS)会诱导小胶质细胞胞外陷阱的形成和视网膜新生血管的形成。从机制上讲,LysoPS 通过与小胶质细胞上的 GPR34 受体相互作用,激活了 GPR34-PI3K-AKT-NINJ1 信号轴。这种激活会上调炎性细胞因子(如 IL-6、IL-8、VEGFA 和 FGF2)的表达,并促进视网膜血管内皮细胞的血管生成。因此,抑制 GPR34-PI3K-AKT-NINJ1 轴可通过抑制 LysoPS 诱导的小胶质细胞炎症反应,在体外和体内显著减少小胶质细胞胞外陷阱的形成和新生血管的形成。这项研究揭示了凋亡的神经节细胞在激活 PDR 中的小胶质细胞炎症中的关键作用,从而加深了我们对视网膜新生血管发病机制的理解。
{"title":"Ganglion cell-derived LysoPS induces retinal neovascularisation by activating the microglial GPR34-PI3K-AKT-NINJ1 axis.","authors":"Lushu Chen, HuiYing Zhang, Ying Zhang, Xiumiao Li, MeiHuan Wang, Yaming Shen, Yuan Cao, Yong Xu, Jin Yao","doi":"10.1186/s12974-024-03265-7","DOIUrl":"10.1186/s12974-024-03265-7","url":null,"abstract":"<p><p>Retinal neovascularisation is a major cause of blindness in patients with proliferative diabetic retinopathy (PDR). It is mediated by the complex interaction between dysfunctional ganglion cells, microglia, and vascular endothelial cells. Notably, retinal microglia, the intrinsic immune cells of the retina, play a crucial role in the pathogenesis of retinopathy. In this study, we found that lysophosphatidylserines (LysoPS) released from injured ganglion cells induced microglial extracellular trap formation and retinal neovascularisation. Mechanistically, LysoPS activated the GPR34-PI3K-AKT-NINJ1 signalling axis by interacting with the GPR34 receptor on the microglia. This activation upregulated the expression of inflammatory cytokines, such as IL-6, IL-8, VEGFA, and FGF2, and facilitated retinal vascular endothelial cell angiogenesis. As a result, inhibition of the GPR34-PI3K-AKT-NINJ1 axis significantly decreased microglial extracellular trap formation and neovascularisation by suppressing LysoPS-induced microglial inflammatory responses, both in vitro and in vivo. This study reveals the crucial role of apoptotic ganglion cells in activating microglial inflammation in PDR, thereby enhancing our understanding of the pathogenesis of retinal neovascularisation.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11520652/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142522168","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
Fenebrutinib, a Bruton's tyrosine kinase inhibitor, blocks distinct human microglial signaling pathways. 布鲁顿酪氨酸激酶抑制剂 Fenebrutinib 可阻断不同的人类小胶质细胞信号通路。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-27 DOI: 10.1186/s12974-024-03267-5
Julie Langlois, Simona Lange, Martin Ebeling, Will Macnair, Roland Schmucki, Cenxiao Li, Jonathan DeGeer, Tania J J Sudharshan, V Wee Yong, Yun-An Shen, Christopher Harp, Ludovic Collin, James Keaney

Background: Bruton's tyrosine kinase (BTK) is an intracellular signaling enzyme that regulates B-lymphocyte and myeloid cell functions. Due to its involvement in both innate and adaptive immune compartments, BTK inhibitors have emerged as a therapeutic option in autoimmune disorders such as multiple sclerosis (MS). Brain-penetrant, small-molecule BTK inhibitors may also address compartmentalized neuroinflammation, which is proposed to underlie MS disease progression. BTK is expressed by microglia, which are the resident innate immune cells of the brain; however, the precise roles of microglial BTK and impact of BTK inhibitors on microglial functions are still being elucidated. Research on the effects of BTK inhibitors has been limited to rodent disease models. This is the first study reporting effects in human microglia.

Methods: Here we characterize the pharmacological and functional properties of fenebrutinib, a potent, highly selective, noncovalent, reversible, brain-penetrant BTK inhibitor, in human microglia and complex human brain cell systems, including brain organoids.

Results: We find that fenebrutinib blocks the deleterious effects of microglial Fc gamma receptor (FcγR) activation, including cytokine and chemokine release, microglial clustering and neurite damage in diverse human brain cell systems. Gene expression analyses identified pathways linked to inflammation, matrix metalloproteinase production and cholesterol metabolism that were modulated by fenebrutinib treatment. In contrast, fenebrutinib had no significant impact on human microglial pathways linked to Toll-like receptor 4 (TLR4) and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) signaling or myelin phagocytosis.

Conclusions: Our study enhances the understanding of BTK functions in human microglial signaling that are relevant to MS pathogenesis and suggests that fenebrutinib could attenuate detrimental microglial activity associated with FcγR activation in people with MS.

背景:布鲁顿酪氨酸激酶(BTK)是一种细胞内信号酶,可调节B淋巴细胞和髓细胞的功能。由于 BTK 参与先天性免疫和适应性免疫,BTK 抑制剂已成为多发性硬化症(MS)等自身免疫性疾病的一种治疗选择。具有脑穿透力的小分子 BTK 抑制剂还可以解决分区性神经炎症问题,而神经炎症被认为是多发性硬化症疾病进展的基础。BTK 由小胶质细胞表达,而小胶质细胞是大脑的常驻先天性免疫细胞;然而,小胶质细胞 BTK 的确切作用以及 BTK 抑制剂对小胶质细胞功能的影响仍有待阐明。有关 BTK 抑制剂影响的研究仅限于啮齿类动物疾病模型。方法:在此,我们描述了一种强效、高选择性、非共价、可逆、脑穿透性 BTK 抑制剂--芬布替尼在人类小胶质细胞和复杂的人类脑细胞系统(包括脑器官组织)中的药理学和功能特性:结果:我们发现非尼布替尼能阻断小胶质细胞Fcγ受体(FcγR)活化的有害影响,包括细胞因子和趋化因子的释放、小胶质细胞集聚以及不同人脑细胞系统中的神经元损伤。基因表达分析确定了与炎症、基质金属蛋白酶生成和胆固醇代谢有关的通路,这些通路受到非尼布替尼治疗的调节。相比之下,非尼布替尼对与Toll样受体4(TLR4)和含NACHT、LRR和PYD结构域的蛋白3(NLRP3)信号转导或髓鞘吞噬有关的人类小胶质细胞通路没有明显影响:我们的研究加深了人们对与多发性硬化症发病机制相关的BTK在人类微胶质细胞信号传导中的功能的了解,并表明非尼布替尼可以减轻多发性硬化症患者与FcγR激活相关的有害微胶质细胞活动。
{"title":"Fenebrutinib, a Bruton's tyrosine kinase inhibitor, blocks distinct human microglial signaling pathways.","authors":"Julie Langlois, Simona Lange, Martin Ebeling, Will Macnair, Roland Schmucki, Cenxiao Li, Jonathan DeGeer, Tania J J Sudharshan, V Wee Yong, Yun-An Shen, Christopher Harp, Ludovic Collin, James Keaney","doi":"10.1186/s12974-024-03267-5","DOIUrl":"10.1186/s12974-024-03267-5","url":null,"abstract":"<p><strong>Background: </strong>Bruton's tyrosine kinase (BTK) is an intracellular signaling enzyme that regulates B-lymphocyte and myeloid cell functions. Due to its involvement in both innate and adaptive immune compartments, BTK inhibitors have emerged as a therapeutic option in autoimmune disorders such as multiple sclerosis (MS). Brain-penetrant, small-molecule BTK inhibitors may also address compartmentalized neuroinflammation, which is proposed to underlie MS disease progression. BTK is expressed by microglia, which are the resident innate immune cells of the brain; however, the precise roles of microglial BTK and impact of BTK inhibitors on microglial functions are still being elucidated. Research on the effects of BTK inhibitors has been limited to rodent disease models. This is the first study reporting effects in human microglia.</p><p><strong>Methods: </strong>Here we characterize the pharmacological and functional properties of fenebrutinib, a potent, highly selective, noncovalent, reversible, brain-penetrant BTK inhibitor, in human microglia and complex human brain cell systems, including brain organoids.</p><p><strong>Results: </strong>We find that fenebrutinib blocks the deleterious effects of microglial Fc gamma receptor (FcγR) activation, including cytokine and chemokine release, microglial clustering and neurite damage in diverse human brain cell systems. Gene expression analyses identified pathways linked to inflammation, matrix metalloproteinase production and cholesterol metabolism that were modulated by fenebrutinib treatment. In contrast, fenebrutinib had no significant impact on human microglial pathways linked to Toll-like receptor 4 (TLR4) and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) signaling or myelin phagocytosis.</p><p><strong>Conclusions: </strong>Our study enhances the understanding of BTK functions in human microglial signaling that are relevant to MS pathogenesis and suggests that fenebrutinib could attenuate detrimental microglial activity associated with FcγR activation in people with MS.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11514909/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142502342","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
α-Synuclein disrupts microglial autophagy through STAT1-dependent suppression of Ulk1 transcription. α-突触核蛋白通过 STAT1 依赖性抑制 Ulk1 转录来破坏小胶质细胞自噬。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-26 DOI: 10.1186/s12974-024-03268-4
Chong-Shuang Pei, Xiao-Ou Hou, Zhen-Yuan Ma, Hai-Yue Tu, Hai-Chun Qian, Yang Li, Kai Li, Chun-Feng Liu, Liang Ouyang, Jun-Yi Liu, Li-Fang Hu

Background: Autophagy dysfunction in glial cells is implicated in the pathogenesis of Parkinson's disease (PD). The previous study reported that α-synuclein (α-Syn) disrupted autophagy in cultured microglia. However, the mechanism of microglial autophagy dysregulation is poorly understood.

Methods: Two α-Syn-based PD models were generated via AAV-mediated α-Syn delivery into the mouse substantia nigra and striatal α-Syn preformed fibril (PFF) injection. The levels of microglial UNC-51-like kinase 1 (Ulk1) and other autophagy-related genes in vitro and in PD mice, as well as in the peripheral blood mononuclear cells of PD patients and healthy controls, were determined via quantitative PCR, western blotting and immunostaining. The regulatory effect of signal transducer and activator of transcription 1 (STAT1) on Ulk1 transcription was determined via a luciferase reporter assay and other biochemical studies and was verified through Stat1 knockdown or overexpression. The effect of α-Syn on glial STAT1 activation was assessed by immunohistochemistry and western blotting. Changes in microglial status, proinflammatory molecule expression and dopaminergic neuron loss in the nigrostriatum of PD and control mice following microglial Stat1 conditional knockout (cKO) or treatment with the ULK1 activator BL-918 were evaluated by immunostaining and western blotting. Motor behaviors were determined via open field tests, rotarod tests and balance beam crossing.

Results: The transcription of microglial ULK1, a kinase that controls autophagy initiation, decreased in both in vitro and in vivo PD mouse models. STAT1 plays a critical role in suppressing Ulk1 transcription. Specifically, Stat1 overexpression downregulated Ulk1 transcription, while Stat1 knockdown increased ULK1 expression, along with an increase in LC3II and a decrease in the SQSTM1/p62 protein. α-Syn PFF caused toll-like receptor 4-dependent activation of STAT1 in microglia. Ablation of Stat1 alleviated the decrease in microglial ULK1 expression and disruption of autophagy caused by α-Syn PFF. Importantly, the ULK1 activator BL-918 and microglial Stat1 cKO attenuated neuroinflammation, dopaminergic neuronal damage and motor defects in PD models.

Conclusions: These findings reveal a novel mechanism by which α-Syn impairs microglial autophagy and indicate that targeting STAT1 or ULK1 may be a therapeutic strategy for PD.

背景:神经胶质细胞的自噬功能障碍与帕金森病(PD)的发病机制有关。之前的研究报告称,α-突触核蛋白(α-Syn)会破坏培养小胶质细胞的自噬功能。然而,人们对小胶质细胞自噬失调的机制知之甚少:方法:通过 AAV 介导将 α-Syn 运送到小鼠黑质和纹状体注射 α-Syn 预成纤维(PFF),产生了两种基于 α-Syn 的帕金森病模型。通过定量 PCR、Western 印迹和免疫染色法测定了小胶质细胞 UNC-51 样激酶 1(Ulk1)和其他自噬相关基因在体外、PD 小鼠体内以及 PD 患者和健康对照组外周血单核细胞中的水平。通过荧光素酶报告实验和其他生化研究确定了信号转导和激活转录1(STAT1)对Ulk1转录的调控作用,并通过敲除或过表达Stat1进行了验证。α-Syn对神经胶质STAT1活化的影响通过免疫组化和免疫印迹进行了评估。通过免疫染色和免疫印迹法评估了小胶质细胞Stat1条件性敲除(cKO)或ULK1激活剂BL-918治疗后,PD小鼠和对照组小鼠黑质中的小胶质细胞状态、促炎症分子表达和多巴胺能神经元丢失的变化。运动行为通过开阔地测试、旋转木马测试和平衡木穿越进行测定:结果:在体外和体内的帕金森病小鼠模型中,控制自噬启动的激酶--小胶质细胞ULK1的转录均有所下降。STAT1在抑制Ulk1转录方面起着关键作用。具体来说,Stat1过表达会下调Ulk1的转录,而敲除Stat1会增加ULK1的表达,同时增加LC3II和减少SQSTM1/p62蛋白。Stat1的消减减轻了α-Syn PFF引起的小胶质细胞ULK1表达的减少和自噬的破坏。重要的是,ULK1激活剂BL-918和小胶质细胞Stat1 cKO减轻了PD模型的神经炎症、多巴胺能神经元损伤和运动缺陷:这些发现揭示了α-Syn损害小胶质细胞自噬的新机制,并表明靶向STAT1或ULK1可能是一种治疗帕金森病的策略。
{"title":"α-Synuclein disrupts microglial autophagy through STAT1-dependent suppression of Ulk1 transcription.","authors":"Chong-Shuang Pei, Xiao-Ou Hou, Zhen-Yuan Ma, Hai-Yue Tu, Hai-Chun Qian, Yang Li, Kai Li, Chun-Feng Liu, Liang Ouyang, Jun-Yi Liu, Li-Fang Hu","doi":"10.1186/s12974-024-03268-4","DOIUrl":"10.1186/s12974-024-03268-4","url":null,"abstract":"<p><strong>Background: </strong>Autophagy dysfunction in glial cells is implicated in the pathogenesis of Parkinson's disease (PD). The previous study reported that α-synuclein (α-Syn) disrupted autophagy in cultured microglia. However, the mechanism of microglial autophagy dysregulation is poorly understood.</p><p><strong>Methods: </strong>Two α-Syn-based PD models were generated via AAV-mediated α-Syn delivery into the mouse substantia nigra and striatal α-Syn preformed fibril (PFF) injection. The levels of microglial UNC-51-like kinase 1 (Ulk1) and other autophagy-related genes in vitro and in PD mice, as well as in the peripheral blood mononuclear cells of PD patients and healthy controls, were determined via quantitative PCR, western blotting and immunostaining. The regulatory effect of signal transducer and activator of transcription 1 (STAT1) on Ulk1 transcription was determined via a luciferase reporter assay and other biochemical studies and was verified through Stat1 knockdown or overexpression. The effect of α-Syn on glial STAT1 activation was assessed by immunohistochemistry and western blotting. Changes in microglial status, proinflammatory molecule expression and dopaminergic neuron loss in the nigrostriatum of PD and control mice following microglial Stat1 conditional knockout (cKO) or treatment with the ULK1 activator BL-918 were evaluated by immunostaining and western blotting. Motor behaviors were determined via open field tests, rotarod tests and balance beam crossing.</p><p><strong>Results: </strong>The transcription of microglial ULK1, a kinase that controls autophagy initiation, decreased in both in vitro and in vivo PD mouse models. STAT1 plays a critical role in suppressing Ulk1 transcription. Specifically, Stat1 overexpression downregulated Ulk1 transcription, while Stat1 knockdown increased ULK1 expression, along with an increase in LC3II and a decrease in the SQSTM1/p62 protein. α-Syn PFF caused toll-like receptor 4-dependent activation of STAT1 in microglia. Ablation of Stat1 alleviated the decrease in microglial ULK1 expression and disruption of autophagy caused by α-Syn PFF. Importantly, the ULK1 activator BL-918 and microglial Stat1 cKO attenuated neuroinflammation, dopaminergic neuronal damage and motor defects in PD models.</p><p><strong>Conclusions: </strong>These findings reveal a novel mechanism by which α-Syn impairs microglial autophagy and indicate that targeting STAT1 or ULK1 may be a therapeutic strategy for PD.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515151/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142502347","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
IL-33/ST2 signaling in monocyte-derived macrophages maintains blood-brain barrier integrity and restricts infarctions early after ischemic stroke. 单核细胞衍生巨噬细胞中的 IL-33/ST2 信号可维持血脑屏障的完整性并限制缺血性脑卒中后早期的脑梗塞。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-24 DOI: 10.1186/s12974-024-03264-8
Miao Wang, Connor Dufort, Zhihong Du, Ruyu Shi, Fei Xu, Zhentai Huang, Ana Rios Sigler, Rehana K Leak, Xiaoming Hu

Background: Brain microglia and infiltrating monocyte-derived macrophages are vital in preserving blood vessel integrity after stroke. Understanding mechanisms that induce immune cells to adopt vascular-protective phenotypes may hasten the development of stroke treatments. IL-33 is a potent chemokine released from damaged cells, such as CNS glia after stroke. The activation of IL-33/ST2 signaling has been shown to promote neuronal viability and white matter integrity after ischemic stroke. The impact of IL-33/ST2 on blood-brain barrier (BBB) integrity, however, remains unknown. The current study fills this gap and reveals a critical role of IL-33/ST2 signaling in macrophage-mediated BBB protection after stroke.

Methods: Transient middle cerebral artery occlusion (tMCAO) was performed to induce ischemic stroke in wildtype (WT) versus ST2 knockout (KO) male mice. IL-33 was applied intranasally to tMCAO mice with or without dietary PLX5622 to deplete microglia/macrophages. ST2 KO versus WT bone marrow or macrophage cell transplantations were used to test the involvement of ST2+ macrophages in BBB integrity. Macrophages were cocultured in transwells with brain endothelial cells (ECs) after oxygen-glucose deprivation (OGD) to test potential direct effects of IL33-treated macrophages on the BBB in vitro.

Results: The ST2 receptor was expressed in brain ECs, microglia, and infiltrating macrophages. Global KO of ST2 led to more IgG extravasation and loss of ZO-1 in cerebral microvessels 3 days post-tMCAO. Intranasal IL-33 administration reduced BBB leakage and infarct severity in microglia/macrophage competent mice, but not in microglia/macrophage depleted mice. Worse BBB injury was observed after tMCAO in chimeric WT mice reconstituted with ST2 KO bone marrow, and in WT mice whose monocytes were replaced by ST2 KO monocytes. Macrophages treated with IL-33 reduced in vitro barrier leakage and maintained tight junction integrity after OGD. In contrast, IL-33 exerted minimal direct effects on the endothelial barrier in the absence of macrophages. IL-33-treated macrophages demonstrated transcriptional upregulation of an array of protective factors, suggesting a shift towards favorable phenotypes.

Conclusion: Our results demonstrate that early-stage IL-33/ST2 signaling in infiltrating macrophages reduces the extent of acute BBB disruption after stroke. Intranasal IL-33 administration may represent a new strategy to reduce BBB leakage and infarct severity.

背景:脑小胶质细胞和浸润的单核细胞衍生巨噬细胞对中风后保护血管完整性至关重要。了解诱导免疫细胞采用血管保护表型的机制可加速中风治疗方法的开发。IL-33 是中风后中枢神经系统胶质细胞等受损细胞释放的一种强效趋化因子。事实证明,IL-33/ST2 信号的激活可促进缺血性中风后神经元的活力和白质的完整性。然而,IL-33/ST2 对血脑屏障(BBB)完整性的影响仍然未知。本研究填补了这一空白,揭示了IL-33/ST2信号在中风后巨噬细胞介导的血脑屏障保护中的关键作用:方法:对野生型(WT)和 ST2 基因敲除(KO)雄性小鼠分别进行瞬时大脑中动脉闭塞(tMCAO)以诱导缺血性中风。给 tMCAO 小鼠鼻内注射 IL-33 或不注射 PLX5622 以消耗小胶质细胞/巨噬细胞。ST2 KO 与 WT 骨髓或巨噬细胞移植被用来测试 ST2+ 巨噬细胞参与 BBB 的完整性。在氧气-葡萄糖剥夺(OGD)后,将巨噬细胞与脑内皮细胞(ECs)共培养在转孔中,以测试经IL33处理的巨噬细胞在体外对BBB的潜在直接影响:结果:ST2受体在脑EC、小胶质细胞和浸润巨噬细胞中均有表达。ST2 的全局性 KO 会导致更多的 IgG 外渗,并在 TMCAO 后 3 天导致脑微血管中 ZO-1 的缺失。鼻内注射IL-33可减少小胶质细胞/巨噬细胞能力小鼠的BBB渗漏和梗死严重程度,但不能减少小胶质细胞/巨噬细胞耗竭小鼠的BBB渗漏和梗死严重程度。用 ST2 KO 骨髓重组的嵌合 WT 小鼠和单核细胞被 ST2 KO 单核细胞取代的 WT 小鼠在 tMCAO 后观察到更严重的 BBB 损伤。用 IL-33 处理的巨噬细胞减少了体外屏障渗漏,并维持了 OGD 后紧密连接的完整性。相比之下,在没有巨噬细胞的情况下,IL-33 对内皮屏障的直接影响微乎其微。经 IL-33 处理的巨噬细胞表现出一系列保护因子的转录上调,这表明巨噬细胞向有利的表型转变:我们的研究结果表明,浸润巨噬细胞的早期 IL-33/ST2 信号传导可降低中风后急性 BBB 的破坏程度。鼻内注射 IL-33 可能是减少 BBB 渗漏和梗死严重程度的一种新策略。
{"title":"IL-33/ST2 signaling in monocyte-derived macrophages maintains blood-brain barrier integrity and restricts infarctions early after ischemic stroke.","authors":"Miao Wang, Connor Dufort, Zhihong Du, Ruyu Shi, Fei Xu, Zhentai Huang, Ana Rios Sigler, Rehana K Leak, Xiaoming Hu","doi":"10.1186/s12974-024-03264-8","DOIUrl":"10.1186/s12974-024-03264-8","url":null,"abstract":"<p><strong>Background: </strong>Brain microglia and infiltrating monocyte-derived macrophages are vital in preserving blood vessel integrity after stroke. Understanding mechanisms that induce immune cells to adopt vascular-protective phenotypes may hasten the development of stroke treatments. IL-33 is a potent chemokine released from damaged cells, such as CNS glia after stroke. The activation of IL-33/ST2 signaling has been shown to promote neuronal viability and white matter integrity after ischemic stroke. The impact of IL-33/ST2 on blood-brain barrier (BBB) integrity, however, remains unknown. The current study fills this gap and reveals a critical role of IL-33/ST2 signaling in macrophage-mediated BBB protection after stroke.</p><p><strong>Methods: </strong>Transient middle cerebral artery occlusion (tMCAO) was performed to induce ischemic stroke in wildtype (WT) versus ST2 knockout (KO) male mice. IL-33 was applied intranasally to tMCAO mice with or without dietary PLX5622 to deplete microglia/macrophages. ST2 KO versus WT bone marrow or macrophage cell transplantations were used to test the involvement of ST2<sup>+</sup> macrophages in BBB integrity. Macrophages were cocultured in transwells with brain endothelial cells (ECs) after oxygen-glucose deprivation (OGD) to test potential direct effects of IL33-treated macrophages on the BBB in vitro.</p><p><strong>Results: </strong>The ST2 receptor was expressed in brain ECs, microglia, and infiltrating macrophages. Global KO of ST2 led to more IgG extravasation and loss of ZO-1 in cerebral microvessels 3 days post-tMCAO. Intranasal IL-33 administration reduced BBB leakage and infarct severity in microglia/macrophage competent mice, but not in microglia/macrophage depleted mice. Worse BBB injury was observed after tMCAO in chimeric WT mice reconstituted with ST2 KO bone marrow, and in WT mice whose monocytes were replaced by ST2 KO monocytes. Macrophages treated with IL-33 reduced in vitro barrier leakage and maintained tight junction integrity after OGD. In contrast, IL-33 exerted minimal direct effects on the endothelial barrier in the absence of macrophages. IL-33-treated macrophages demonstrated transcriptional upregulation of an array of protective factors, suggesting a shift towards favorable phenotypes.</p><p><strong>Conclusion: </strong>Our results demonstrate that early-stage IL-33/ST2 signaling in infiltrating macrophages reduces the extent of acute BBB disruption after stroke. Intranasal IL-33 administration may represent a new strategy to reduce BBB leakage and infarct severity.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142502343","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
Impairment of spinal CSF flow precedes immune cell infiltration in an active EAE model. 在活动性 EAE 模型中,脊髓 CSF 流受损先于免疫细胞浸润。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-23 DOI: 10.1186/s12974-024-03247-9
Li Xin, Adrian Madarasz, Daniela C Ivan, Florian Weber, Simone Aleandri, Paola Luciani, Giuseppe Locatelli, Steven T Proulx

Accumulation of immune cells and proteins in the subarachnoid space (SAS) is found during multiple sclerosis and in the animal model experimental autoimmune encephalomyelitis (EAE). Whether the flow of cerebrospinal fluid (CSF) along the SAS of the spinal cord is impacted is yet unknown. Combining intravital near-infrared (NIR) imaging with histopathological analyses, we observed a significantly impaired bulk flow of CSF tracers within the SAS of the spinal cord prior to EAE onset, which persisted until peak stage and was only partially recovered during chronic disease. The impairment of spinal CSF flow coincided with the appearance of fibrin aggregates in the SAS, however, it preceded immune cell infiltration and breakdown of the glia limitans superficialis. Conversely, cranial CSF efflux to cervical lymph nodes was not altered during the disease course. Our study highlights an early and persistent impairment of spinal CSF flow and suggests it as a sensitive imaging biomarker for pathological changes within the leptomeninges.

在多发性硬化症和动物模型实验性自身免疫性脑脊髓炎(EAE)中,免疫细胞和蛋白质在蛛网膜下腔(SAS)聚集。脊髓蛛网膜下腔(SAS)的脑脊液(CSF)流动是否受到影响尚不清楚。通过将视内近红外(NIR)成像与组织病理学分析相结合,我们观察到在EAE发病前,脊髓SAS内的CSF示踪剂大量流动明显受损,这种情况一直持续到高峰期,在慢性病期间才部分恢复。脊髓CSF流动的障碍与SAS中纤维蛋白聚集的出现同时发生,但它先于免疫细胞浸润和浅表神经胶质的破坏。相反,颅脑 CSF 流向颈淋巴结的情况在病程中没有改变。我们的研究强调了脊髓CSF流的早期和持续性损害,并建议将其作为一种敏感的成像生物标志物,以反映脑膜内的病理变化。
{"title":"Impairment of spinal CSF flow precedes immune cell infiltration in an active EAE model.","authors":"Li Xin, Adrian Madarasz, Daniela C Ivan, Florian Weber, Simone Aleandri, Paola Luciani, Giuseppe Locatelli, Steven T Proulx","doi":"10.1186/s12974-024-03247-9","DOIUrl":"10.1186/s12974-024-03247-9","url":null,"abstract":"<p><p>Accumulation of immune cells and proteins in the subarachnoid space (SAS) is found during multiple sclerosis and in the animal model experimental autoimmune encephalomyelitis (EAE). Whether the flow of cerebrospinal fluid (CSF) along the SAS of the spinal cord is impacted is yet unknown. Combining intravital near-infrared (NIR) imaging with histopathological analyses, we observed a significantly impaired bulk flow of CSF tracers within the SAS of the spinal cord prior to EAE onset, which persisted until peak stage and was only partially recovered during chronic disease. The impairment of spinal CSF flow coincided with the appearance of fibrin aggregates in the SAS, however, it preceded immune cell infiltration and breakdown of the glia limitans superficialis. Conversely, cranial CSF efflux to cervical lymph nodes was not altered during the disease course. Our study highlights an early and persistent impairment of spinal CSF flow and suggests it as a sensitive imaging biomarker for pathological changes within the leptomeninges.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11520187/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142502345","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 deficient CLEC5A ameliorates the behavioral and pathological deficits via the microglial Aβ clearance in Alzheimer's disease mouse model. 在阿尔茨海默病小鼠模型中,CLEC5A的缺陷可通过清除小胶质细胞Aβ改善行为和病理缺陷。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-23 DOI: 10.1186/s12974-024-03253-x
Yu-Yi Lin, Wen-Han Chang, Shie-Liang Hsieh, Irene Han-Juo Cheng

Background: Alzheimer's disease (AD) is a neurodegenerative disease that causes cognitive dysfunction in older adults. One of the AD pathological factors, β-Amyloid (Aβ), triggers inflammatory responses and phagocytosis of microglia. C-type lectin domain family 5 member A (CLEC5A) induces over-reactive inflammatory responses in several virus infections. Yet, the role of CLEC5A in AD progression remains unknown. This study aimed to elucidate the contribution of CLEC5A to Aβ-induced microglial activation and behavioral deficits.

Methods: The AD mouse model was crossed with Clec5a knockout mice for subsequent behavioral and pathological tests. The memory deficit was revealed by the Morris water maze, while the nociception abnormalities were examined by the von Frey filament and hotplate test. The Aβ deposition and microglia recruitment were identified by ELISA and immunohistochemistry. The inflammatory signals were identified by ELISA and western blotting. In the Clec5a knockdown microglial cell model and Clec5a knockout primary microglia, the microglial phagocytosis was revealed using the fluorescent-labeled Aβ.

Results: The AD mice with Clec5a knockout improved Aβ-induced memory deficit and abnormal nociception. These mice have reduced Aβ deposition and increased microglia coverage surrounding the amyloid plaque, suggesting the involvement of CLEC5A in AD progression and Aβ clearance. Moreover, the phagocytosis was also increased in the Aβ-stressed Clec5a knockdown microglial cell lines and Clec5a knockout primary microglia.

Conclusion: The Clec5a knockout ameliorates AD-like deficits by modulating microglial Aβ clearance. This study implies that targeting microglial Clec5a could offer a promising approach to mitigate AD progression.

背景:阿尔茨海默病(AD)是一种导致老年人认知功能障碍的神经退行性疾病。阿尔茨海默病的病理因素之一是β-淀粉样蛋白(Aβ),它会引发炎症反应和小胶质细胞的吞噬作用。在几种病毒感染中,C 型凝集素结构域家族 5 成员 A(CLEC5A)会诱发过度反应性炎症反应。然而,CLEC5A在AD进展中的作用仍然未知。本研究旨在阐明CLEC5A对Aβ诱导的小胶质细胞活化和行为障碍的贡献:方法:将AD小鼠模型与Clec5a基因敲除小鼠杂交,进行行为和病理测试。方法:将AD小鼠与Clec5a基因敲除小鼠杂交,进行行为和病理测试。Aβ沉积和小胶质细胞募集是通过酶联免疫吸附试验和免疫组化鉴定的。炎症信号通过 ELISA 和 Western 印迹法进行鉴定。在 Clec5a 基因敲除的小胶质细胞模型和 Clec5a 基因敲除的原代小胶质细胞中,使用荧光标记的 Aβ 揭示了小胶质细胞的吞噬功能:结果:Clec5a基因敲除的AD小鼠改善了Aβ诱导的记忆缺陷和异常痛觉。这些小鼠的Aβ沉积减少,淀粉样斑块周围的小胶质细胞覆盖率增加,表明CLEC5A参与了AD的进展和Aβ的清除。此外,在Aβ受压的Clec5a基因敲除小胶质细胞系和Clec5a基因敲除原代小胶质细胞中,吞噬作用也有所增加:结论:Clec5a基因敲除通过调节小胶质细胞对Aβ的清除而改善AD样缺陷。这项研究表明,以小胶质细胞Clec5a为靶点可为缓解AD进展提供一种前景广阔的方法。
{"title":"The deficient CLEC5A ameliorates the behavioral and pathological deficits via the microglial Aβ clearance in Alzheimer's disease mouse model.","authors":"Yu-Yi Lin, Wen-Han Chang, Shie-Liang Hsieh, Irene Han-Juo Cheng","doi":"10.1186/s12974-024-03253-x","DOIUrl":"10.1186/s12974-024-03253-x","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) is a neurodegenerative disease that causes cognitive dysfunction in older adults. One of the AD pathological factors, β-Amyloid (Aβ), triggers inflammatory responses and phagocytosis of microglia. C-type lectin domain family 5 member A (CLEC5A) induces over-reactive inflammatory responses in several virus infections. Yet, the role of CLEC5A in AD progression remains unknown. This study aimed to elucidate the contribution of CLEC5A to Aβ-induced microglial activation and behavioral deficits.</p><p><strong>Methods: </strong>The AD mouse model was crossed with Clec5a knockout mice for subsequent behavioral and pathological tests. The memory deficit was revealed by the Morris water maze, while the nociception abnormalities were examined by the von Frey filament and hotplate test. The Aβ deposition and microglia recruitment were identified by ELISA and immunohistochemistry. The inflammatory signals were identified by ELISA and western blotting. In the Clec5a knockdown microglial cell model and Clec5a knockout primary microglia, the microglial phagocytosis was revealed using the fluorescent-labeled Aβ.</p><p><strong>Results: </strong>The AD mice with Clec5a knockout improved Aβ-induced memory deficit and abnormal nociception. These mice have reduced Aβ deposition and increased microglia coverage surrounding the amyloid plaque, suggesting the involvement of CLEC5A in AD progression and Aβ clearance. Moreover, the phagocytosis was also increased in the Aβ-stressed Clec5a knockdown microglial cell lines and Clec5a knockout primary microglia.</p><p><strong>Conclusion: </strong>The Clec5a knockout ameliorates AD-like deficits by modulating microglial Aβ clearance. This study implies that targeting microglial Clec5a could offer a promising approach to mitigate AD progression.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515658/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142502346","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
Impairment of neuronal tyrosine phosphatase STEP worsens post-ischemic inflammation and brain injury under hypertensive condition. 神经元酪氨酸磷酸酶 STEP 的损伤会加重高血压条件下缺血后的炎症和脑损伤。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-22 DOI: 10.1186/s12974-024-03227-z
Prabu Paramasivam, Seong Won Choi, Ranjana Poddar, Surojit Paul

Hypertension is associated with poor outcome and higher mortality in patients with ischemic stroke. The impairment of adaptive vascular mechanisms under hypertensive condition compromises collateral blood flow after arterial occlusion in patients with acute ischemic stroke resulting in hypoperfusion. The increased oxidative stress caused by hypoperfusion is thought to be a trigger for the rapid evolution of ischemic infarct volume under hypertensive condition. However, the cellular factors and pathways that contribute to the exacerbation of ischemic brain injury under hypertensive condition is not yet understood. The current study reveals that predisposition to hypertension leads to basal loss of function of the neuron-specific tyrosine phosphatase STEP, which plays a crucial role in neuroprotection against excitotoxic insult. The findings further show that a mild ischemic insult in hypertensive rats triggers an early onset and sustained activation of the neuronal extracellular signal regulated kinase (ERK MAPK), a member of the mitogen activated protein kinase family and a substrate of STEP. This leads to rapid increase in the activation of neuronal NF-κB, expression of neuronal cyclooxygenase-2 and subsequent biosynthesis of the pro-inflammatory mediator prostaglandin E2, resulting in rapid morphological transformation of microglia to the pro-inflammatory state and subsequent exacerbation of ischemic brain injury. Restoration of STEP signaling with intravenous administration of a STEP-derived peptide mimetic reduces the pro-inflammatory response in neurons, activation of microglia, and ischemic brain injury. The findings suggest that the basal loss of STEP function under hypertensive condition contributes to the exacerbation of ischemic brain injury by enhancing post-ischemic inflammatory response. The study not only presents a novel role of STEP in regulating neuroimmune communication but also highlights the therapeutic potential of a STEP-mimetic in mitigating ischemic brain damage under hypertensive condition.

高血压与缺血性中风患者的不良预后和较高死亡率有关。在高血压状态下,血管适应机制受损,导致急性缺血性脑卒中患者动脉闭塞后侧支血流受损,造成低灌注。低灌注引起的氧化应激增加被认为是高血压条件下缺血性梗死体积快速演变的诱因。然而,导致高血压条件下缺血性脑损伤加重的细胞因素和途径尚不清楚。目前的研究揭示,易患高血压会导致神经元特异性酪氨酸磷酸酶 STEP 的基础功能丧失,而 STEP 在抗兴奋性毒性损伤的神经保护中发挥着关键作用。研究结果进一步表明,高血压大鼠的轻度缺血损伤会引发神经元细胞外信号调节激酶(ERK MAPK)的早期和持续激活,ERK MAPK 是丝裂原活化蛋白激酶家族的成员,也是 STEP 的底物。这导致神经元 NF-κB 的激活、神经元环氧化酶-2 的表达以及促炎介质前列腺素 E2 的生物合成迅速增加,从而使小胶质细胞的形态迅速转变为促炎状态,缺血性脑损伤随之加重。通过静脉注射一种 STEP 衍生肽模拟物来恢复 STEP 信号传导,可减轻神经元的促炎反应、小胶质细胞的活化和缺血性脑损伤。研究结果表明,高血压条件下 STEP 基础功能的丧失会通过增强缺血后的炎症反应加剧缺血性脑损伤。该研究不仅揭示了 STEP 在调节神经免疫通讯中的新作用,还强调了 STEP 模拟物在减轻高血压条件下缺血性脑损伤方面的治疗潜力。
{"title":"Impairment of neuronal tyrosine phosphatase STEP worsens post-ischemic inflammation and brain injury under hypertensive condition.","authors":"Prabu Paramasivam, Seong Won Choi, Ranjana Poddar, Surojit Paul","doi":"10.1186/s12974-024-03227-z","DOIUrl":"10.1186/s12974-024-03227-z","url":null,"abstract":"<p><p>Hypertension is associated with poor outcome and higher mortality in patients with ischemic stroke. The impairment of adaptive vascular mechanisms under hypertensive condition compromises collateral blood flow after arterial occlusion in patients with acute ischemic stroke resulting in hypoperfusion. The increased oxidative stress caused by hypoperfusion is thought to be a trigger for the rapid evolution of ischemic infarct volume under hypertensive condition. However, the cellular factors and pathways that contribute to the exacerbation of ischemic brain injury under hypertensive condition is not yet understood. The current study reveals that predisposition to hypertension leads to basal loss of function of the neuron-specific tyrosine phosphatase STEP, which plays a crucial role in neuroprotection against excitotoxic insult. The findings further show that a mild ischemic insult in hypertensive rats triggers an early onset and sustained activation of the neuronal extracellular signal regulated kinase (ERK MAPK), a member of the mitogen activated protein kinase family and a substrate of STEP. This leads to rapid increase in the activation of neuronal NF-κB, expression of neuronal cyclooxygenase-2 and subsequent biosynthesis of the pro-inflammatory mediator prostaglandin E2, resulting in rapid morphological transformation of microglia to the pro-inflammatory state and subsequent exacerbation of ischemic brain injury. Restoration of STEP signaling with intravenous administration of a STEP-derived peptide mimetic reduces the pro-inflammatory response in neurons, activation of microglia, and ischemic brain injury. The findings suggest that the basal loss of STEP function under hypertensive condition contributes to the exacerbation of ischemic brain injury by enhancing post-ischemic inflammatory response. The study not only presents a novel role of STEP in regulating neuroimmune communication but also highlights the therapeutic potential of a STEP-mimetic in mitigating ischemic brain damage under hypertensive condition.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11515672/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142502344","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