首页 > 最新文献

Journal of Neuroinflammation最新文献

英文 中文
PARK7/DJ-1 deficiency impairs microglial activation in response to LPS-induced inflammation. PARK7/DJ-1的缺乏会影响小胶质细胞对LPS诱导的炎症反应的激活。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-16 DOI: 10.1186/s12974-024-03164-x
Frida Lind-Holm Mogensen, Carole Sousa, Corrado Ameli, Katja Badanjak, Sandro L Pereira, Arnaud Muller, Tony Kaoma, Djalil Coowar, Andrea Scafidi, Suresh K Poovathingal, Maria Tziortziou, Paul M A Antony, Nathalie Nicot, Aurélien Ginolhac, Daniela M Vogt Weisenhorn, Wolfgang Wurst, Aurélie Poli, Petr V Nazarov, Alexander Skupin, Anne Grünewald, Alessandro Michelucci

Background: Specific microglia responses are thought to contribute to the development and progression of neurodegenerative diseases, including Parkinson's disease (PD). However, the phenotypic acquisition of microglial cells and their role during the underlying neuroinflammatory processes remain largely elusive. Here, according to the multiple-hit hypothesis, which stipulates that PD etiology is determined by a combination of genetics and various environmental risk factors, we investigate microglial transcriptional programs and morphological adaptations under PARK7/DJ-1 deficiency, a genetic cause of PD, during lipopolysaccharide (LPS)-induced inflammation.

Methods: Using a combination of single-cell RNA-sequencing, bulk RNA-sequencing, multicolor flow cytometry and immunofluorescence analyses, we comprehensively compared microglial cell phenotypic characteristics in PARK7/DJ-1 knock-out (KO) with wildtype littermate mice following 6- or 24-h intraperitoneal injection with LPS. For translational perspectives, we conducted corresponding analyses in human PARK7/DJ-1 mutant induced pluripotent stem cell (iPSC)-derived microglia and murine bone marrow-derived macrophages (BMDMs).

Results: By excluding the contribution of other immune brain resident and peripheral cells, we show that microglia acutely isolated from PARK7/DJ-1 KO mice display a distinct phenotype, specially related to type II interferon and DNA damage response signaling, when compared with wildtype microglia, in response to LPS. We also detected discrete signatures in human PARK7/DJ-1 mutant iPSC-derived microglia and BMDMs from PARK7/DJ-1 KO mice. These specific transcriptional signatures were reflected at the morphological level, with microglia in LPS-treated PARK7/DJ-1 KO mice showing a less amoeboid cell shape compared to wildtype mice, both at 6 and 24 h after acute inflammation, as also observed in BMDMs.

Conclusions: Taken together, our results show that, under inflammatory conditions, PARK7/DJ-1 deficiency skews microglia towards a distinct phenotype characterized by downregulation of genes involved in type II interferon signaling and a less prominent amoeboid morphology compared to wildtype microglia. These findings suggest that the underlying oxidative stress associated with the lack of PARK7/DJ-1 affects microglia neuroinflammatory responses, which may play a causative role in PD onset and progression.

背景:特定的小胶质细胞反应被认为有助于包括帕金森病(PD)在内的神经退行性疾病的发生和发展。然而,小胶质细胞的表型获得及其在潜在神经炎症过程中的作用在很大程度上仍然难以捉摸。这里,根据帕金森病病因由遗传和各种环境风险因素共同决定的多重打击假说,我们研究了帕金森病遗传病因--PARK7/DJ-1缺乏症在脂多糖(LPS)诱导的炎症过程中的小胶质细胞转录程序和形态适应:利用单细胞RNA测序、大容量RNA测序、多色流式细胞术和免疫荧光分析,我们全面比较了PARK7/DJ-1基因敲除(KO)小鼠和野生型同窝小鼠腹腔注射6或24小时LPS后的小胶质细胞表型特征。从转化的角度来看,我们对人类 PARK7/DJ-1 突变诱导多能干细胞(iPSC)衍生的小胶质细胞和小鼠骨髓衍生巨噬细胞(BMDMs)进行了相应的分析:通过排除其他免疫脑常驻细胞和外周细胞的贡献,我们发现与野生型小胶质细胞相比,从PARK7/DJ-1 KO小鼠体内急性分离出的小胶质细胞对LPS的反应显示出独特的表型,特别是与II型干扰素和DNA损伤反应信号有关。我们还在人 PARK7/DJ-1 突变 iPSC 衍生的小胶质细胞和 PARK7/DJ-1 KO 小鼠的 BMDMs 中检测到了离散特征。这些特异性转录特征反映在形态学水平上,与野生型小鼠相比,经 LPS 处理的 PARK7/DJ-1 KO 小鼠的小胶质细胞在急性炎症后 6 小时和 24 小时都显示出较少的变形虫细胞形状,在 BMDMs 中也观察到了这种情况:综上所述,我们的研究结果表明,在炎症条件下,PARK7/DJ-1 缺乏会使小胶质细胞偏向于一种独特的表型,其特征是参与 II 型干扰素信号转导的基因下调,并且与野生型小胶质细胞相比,阿米波状细胞形态不那么突出。这些发现表明,与 PARK7/DJ-1 缺乏相关的潜在氧化应激影响了小胶质细胞的神经炎症反应,这可能在帕金森病的发病和进展中起着致病作用。
{"title":"PARK7/DJ-1 deficiency impairs microglial activation in response to LPS-induced inflammation.","authors":"Frida Lind-Holm Mogensen, Carole Sousa, Corrado Ameli, Katja Badanjak, Sandro L Pereira, Arnaud Muller, Tony Kaoma, Djalil Coowar, Andrea Scafidi, Suresh K Poovathingal, Maria Tziortziou, Paul M A Antony, Nathalie Nicot, Aurélien Ginolhac, Daniela M Vogt Weisenhorn, Wolfgang Wurst, Aurélie Poli, Petr V Nazarov, Alexander Skupin, Anne Grünewald, Alessandro Michelucci","doi":"10.1186/s12974-024-03164-x","DOIUrl":"10.1186/s12974-024-03164-x","url":null,"abstract":"<p><strong>Background: </strong>Specific microglia responses are thought to contribute to the development and progression of neurodegenerative diseases, including Parkinson's disease (PD). However, the phenotypic acquisition of microglial cells and their role during the underlying neuroinflammatory processes remain largely elusive. Here, according to the multiple-hit hypothesis, which stipulates that PD etiology is determined by a combination of genetics and various environmental risk factors, we investigate microglial transcriptional programs and morphological adaptations under PARK7/DJ-1 deficiency, a genetic cause of PD, during lipopolysaccharide (LPS)-induced inflammation.</p><p><strong>Methods: </strong>Using a combination of single-cell RNA-sequencing, bulk RNA-sequencing, multicolor flow cytometry and immunofluorescence analyses, we comprehensively compared microglial cell phenotypic characteristics in PARK7/DJ-1 knock-out (KO) with wildtype littermate mice following 6- or 24-h intraperitoneal injection with LPS. For translational perspectives, we conducted corresponding analyses in human PARK7/DJ-1 mutant induced pluripotent stem cell (iPSC)-derived microglia and murine bone marrow-derived macrophages (BMDMs).</p><p><strong>Results: </strong>By excluding the contribution of other immune brain resident and peripheral cells, we show that microglia acutely isolated from PARK7/DJ-1 KO mice display a distinct phenotype, specially related to type II interferon and DNA damage response signaling, when compared with wildtype microglia, in response to LPS. We also detected discrete signatures in human PARK7/DJ-1 mutant iPSC-derived microglia and BMDMs from PARK7/DJ-1 KO mice. These specific transcriptional signatures were reflected at the morphological level, with microglia in LPS-treated PARK7/DJ-1 KO mice showing a less amoeboid cell shape compared to wildtype mice, both at 6 and 24 h after acute inflammation, as also observed in BMDMs.</p><p><strong>Conclusions: </strong>Taken together, our results show that, under inflammatory conditions, PARK7/DJ-1 deficiency skews microglia towards a distinct phenotype characterized by downregulation of genes involved in type II interferon signaling and a less prominent amoeboid morphology compared to wildtype microglia. These findings suggest that the underlying oxidative stress associated with the lack of PARK7/DJ-1 affects microglia neuroinflammatory responses, which may play a causative role in PD onset and progression.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11253405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141626999","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
Early depletion of gut microbiota shape oligodendrocyte response after traumatic brain injury. 肠道微生物群的早期消耗会影响脑外伤后少突胶质细胞的反应。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-15 DOI: 10.1186/s12974-024-03158-9
Kirill Shumilov, Allen Ni, Maria Garcia-Bonilla, Marta Celorrio, Stuart H Friess

White matter injury (WMI) is thought to be a major contributor to long-term cognitive dysfunctions after traumatic brain injury (TBI). This damage occurs partly due to apoptotic death of oligodendrocyte lineage cells (OLCs) after the injury, triggered directly by the trauma or in response to degenerating axons. Recent research suggests that the gut microbiota modulates the inflammatory response through the regulation of peripheral immune cell infiltration after TBI. Additionally, T-cells directly impact OLCs differentiation and proliferation. Therefore, we hypothesized that the gut microbiota plays a critical role in regulating the OLC response to WMI influencing T-cells differentiation and activation. Gut microbial depletion early after TBI chronically reduced re-myelination, acutely decreased OLCs proliferation, and was associated with increased myelin debris accumulation. Surprisingly, the absence of T-cells in gut microbiota depleted mice restored OLC proliferation and remyelination after TBI. OLCs co-cultured with T-cells derived from gut microbiota depleted mice resulted in impaired proliferation and increased expression of MHC-II compared with T cells from control-injured mice. Furthermore, MHC-II expression in OLCs appears to be linked to impaired proliferation under gut microbiota depletion and TBI conditions. Collectively our data indicates that depletion of the gut microbiota after TBI impaired remyelination, reduced OLCs proliferation with concomitantly increased OLC MHCII expression, and required the presence of T cells. This data suggests that T cells are an important mechanistic link by which the gut microbiota modulate the oligodendrocyte response and white matter recovery after TBI.

白质损伤(WMI)被认为是造成创伤性脑损伤(TBI)后长期认知功能障碍的主要原因。造成这种损伤的部分原因是受伤后少突胶质细胞系(OLCs)的凋亡,这种凋亡是由创伤直接引发的,或者是对退化的轴突的反应。最新研究表明,肠道微生物群通过调节创伤后外周免疫细胞浸润来调节炎症反应。此外,T 细胞会直接影响 OLCs 的分化和增殖。因此,我们假设肠道微生物群在调节 OLC 对 WMI 的反应中起着关键作用,影响着 T 细胞的分化和活化。创伤性脑损伤后早期的肠道微生物耗竭会慢性减少髓鞘再形成,急性减少 OLCs 增殖,并与髓鞘碎片堆积增加有关。令人惊讶的是,肠道微生物群耗竭的小鼠体内没有T细胞,却能恢复创伤性脑损伤后OLC的增殖和再髓鞘化。与来自对照组损伤小鼠的T细胞相比,OLC与来自肠道微生物群耗竭小鼠的T细胞共培养会导致增殖受损和MHC-II表达增加。此外,在肠道微生物群耗竭和创伤性脑损伤条件下,OLCs 中 MHC-II 的表达似乎与增殖受损有关。总之,我们的数据表明,创伤性脑损伤后肠道微生物群的耗竭会损害髓鞘再形成,减少 OLC 的增殖,同时增加 OLC MHCII 的表达,并且需要 T 细胞的存在。这些数据表明,T 细胞是肠道微生物群调节少突胶质细胞反应和创伤后白质恢复的重要机制环节。
{"title":"Early depletion of gut microbiota shape oligodendrocyte response after traumatic brain injury.","authors":"Kirill Shumilov, Allen Ni, Maria Garcia-Bonilla, Marta Celorrio, Stuart H Friess","doi":"10.1186/s12974-024-03158-9","DOIUrl":"10.1186/s12974-024-03158-9","url":null,"abstract":"<p><p>White matter injury (WMI) is thought to be a major contributor to long-term cognitive dysfunctions after traumatic brain injury (TBI). This damage occurs partly due to apoptotic death of oligodendrocyte lineage cells (OLCs) after the injury, triggered directly by the trauma or in response to degenerating axons. Recent research suggests that the gut microbiota modulates the inflammatory response through the regulation of peripheral immune cell infiltration after TBI. Additionally, T-cells directly impact OLCs differentiation and proliferation. Therefore, we hypothesized that the gut microbiota plays a critical role in regulating the OLC response to WMI influencing T-cells differentiation and activation. Gut microbial depletion early after TBI chronically reduced re-myelination, acutely decreased OLCs proliferation, and was associated with increased myelin debris accumulation. Surprisingly, the absence of T-cells in gut microbiota depleted mice restored OLC proliferation and remyelination after TBI. OLCs co-cultured with T-cells derived from gut microbiota depleted mice resulted in impaired proliferation and increased expression of MHC-II compared with T cells from control-injured mice. Furthermore, MHC-II expression in OLCs appears to be linked to impaired proliferation under gut microbiota depletion and TBI conditions. Collectively our data indicates that depletion of the gut microbiota after TBI impaired remyelination, reduced OLCs proliferation with concomitantly increased OLC MHCII expression, and required the presence of T cells. This data suggests that T cells are an important mechanistic link by which the gut microbiota modulate the oligodendrocyte response and white matter recovery after TBI.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251111/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141620118","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
Deletion of myeloid HDAC3 promotes efferocytosis to ameliorate retinal ischemic injury. 删除骨髓HDAC3可促进流出细胞,从而改善视网膜缺血性损伤。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-12 DOI: 10.1186/s12974-024-03159-8
Rami A Shahror, Esraa Shosha, Carol Morris, Melissa Wild, Shengyu Mu, Gabor Csanyi, Marjan Boerma, Nancy J Rusch, Abdelrahman Y Fouda

Ischemia-induced retinopathy is a hallmark finding of common visual disorders including diabetic retinopathy (DR) and central retinal artery and vein occlusions. Treatments for ischemic retinopathies fail to improve clinical outcomes and the design of new therapies will depend on understanding the underlying disease mechanisms. Histone deacetylases (HDACs) are an enzyme class that removes acetyl groups from histone and non-histone proteins, thereby regulating gene expression and protein function. HDACs have been implicated in retinal neurovascular injury in preclinical studies in which nonspecific HDAC inhibitors mitigated retinal injury. Histone deacetylase 3 (HDAC3) is a class I histone deacetylase isoform that plays a central role in the macrophage inflammatory response. We recently reported that myeloid cells upregulate HDAC3 in a mouse model of retinal ischemia-reperfusion (IR) injury. However, whether this cellular event is an essential contributor to retinal IR injury is unknown. In this study, we explored the role of myeloid HDAC3 in ischemia-induced retinal neurovascular injury by subjecting myeloid-specific HDAC3 knockout (M-HDAC3 KO) and floxed control mice to retinal IR. The M-HDAC3 KO mice were protected from retinal IR injury as shown by the preservation of inner retinal neurons, vascular integrity, and retinal thickness. Electroretinography confirmed that this neurovascular protection translated to improved retinal function. The retinas of M-HDAC3 KO mice also showed less proliferation and infiltration of myeloid cells after injury. Interestingly, myeloid cells lacking HDAC3 more avidly engulfed apoptotic cells in vitro and after retinal IR injury in vivo compared to wild-type myeloid cells, suggesting that HDAC3 hinders the reparative phagocytosis of dead cells, a process known as efferocytosis. Further mechanistic studies indicated that although HDAC3 KO macrophages upregulate the reparative enzyme arginase 1 (A1) that enhances efferocytosis, the inhibitory effect of HDAC3 on efferocytosis is not solely dependent on A1. Finally, treatment of wild-type mice with the HDAC3 inhibitor RGFP966 ameliorated the retinal neurodegeneration and thinning caused by IR injury. Collectively, our data show that HDAC3 deletion enhances macrophage-mediated efferocytosis and protects against retinal IR injury, suggesting that inhibiting myeloid HDAC3 holds promise as a novel therapeutic strategy for preserving retinal integrity after ischemic insult.

缺血引起的视网膜病变是糖尿病视网膜病变(DR)和视网膜中央动脉和静脉闭塞症等常见视觉疾病的标志性发现。缺血性视网膜病变的治疗方法无法改善临床疗效,新疗法的设计将取决于对潜在疾病机理的了解。组蛋白去乙酰化酶(HDACs)是一种酶,能从组蛋白和非组蛋白中去除乙酰基,从而调节基因表达和蛋白质功能。临床前研究发现,HDAC 与视网膜神经血管损伤有关,非特异性 HDAC 抑制剂可减轻视网膜损伤。组蛋白去乙酰化酶 3(HDAC3)是一种 I 类组蛋白去乙酰化酶同工酶,在巨噬细胞炎症反应中发挥着核心作用。我们最近报告说,在小鼠视网膜缺血再灌注(IR)损伤模型中,髓系细胞上调 HDAC3。然而,这一细胞事件是否是视网膜 IR 损伤的重要因素尚不清楚。在本研究中,我们通过让髓细胞特异性 HDAC3 敲除(M-HDAC3 KO)小鼠和荧光对照小鼠接受视网膜 IR 试验,探讨了髓细胞 HDAC3 在缺血诱导的视网膜神经血管损伤中的作用。M-HDAC3 KO小鼠的视网膜内神经元、血管完整性和视网膜厚度都得到了保护,从而避免了视网膜红外损伤。视网膜电图证实,这种神经血管保护可改善视网膜功能。损伤后,M-HDAC3 KO 小鼠视网膜中髓系细胞的增殖和浸润也较少。有趣的是,与野生型髓系细胞相比,缺乏 HDAC3 的髓系细胞在体外和视网膜红外损伤后更热衷于吞噬凋亡细胞,这表明 HDAC3 阻碍了对死亡细胞的修复性吞噬,这一过程被称为流出吞噬。进一步的机理研究表明,虽然 HDAC3 KO 巨噬细胞会上调可增强流出细胞吞噬功能的修复酶精氨酸酶 1(A1),但 HDAC3 对流出细胞吞噬功能的抑制作用并不完全依赖于 A1。最后,用 HDAC3 抑制剂 RGFP966 处理野生型小鼠可改善红外损伤引起的视网膜神经变性和变薄。总之,我们的数据表明,HDAC3 基因缺失能增强巨噬细胞介导的流出细胞功能,保护视网膜免受红外损伤,这表明抑制骨髓 HDAC3 有望成为缺血损伤后保护视网膜完整性的一种新型治疗策略。
{"title":"Deletion of myeloid HDAC3 promotes efferocytosis to ameliorate retinal ischemic injury.","authors":"Rami A Shahror, Esraa Shosha, Carol Morris, Melissa Wild, Shengyu Mu, Gabor Csanyi, Marjan Boerma, Nancy J Rusch, Abdelrahman Y Fouda","doi":"10.1186/s12974-024-03159-8","DOIUrl":"10.1186/s12974-024-03159-8","url":null,"abstract":"<p><p>Ischemia-induced retinopathy is a hallmark finding of common visual disorders including diabetic retinopathy (DR) and central retinal artery and vein occlusions. Treatments for ischemic retinopathies fail to improve clinical outcomes and the design of new therapies will depend on understanding the underlying disease mechanisms. Histone deacetylases (HDACs) are an enzyme class that removes acetyl groups from histone and non-histone proteins, thereby regulating gene expression and protein function. HDACs have been implicated in retinal neurovascular injury in preclinical studies in which nonspecific HDAC inhibitors mitigated retinal injury. Histone deacetylase 3 (HDAC3) is a class I histone deacetylase isoform that plays a central role in the macrophage inflammatory response. We recently reported that myeloid cells upregulate HDAC3 in a mouse model of retinal ischemia-reperfusion (IR) injury. However, whether this cellular event is an essential contributor to retinal IR injury is unknown. In this study, we explored the role of myeloid HDAC3 in ischemia-induced retinal neurovascular injury by subjecting myeloid-specific HDAC3 knockout (M-HDAC3 KO) and floxed control mice to retinal IR. The M-HDAC3 KO mice were protected from retinal IR injury as shown by the preservation of inner retinal neurons, vascular integrity, and retinal thickness. Electroretinography confirmed that this neurovascular protection translated to improved retinal function. The retinas of M-HDAC3 KO mice also showed less proliferation and infiltration of myeloid cells after injury. Interestingly, myeloid cells lacking HDAC3 more avidly engulfed apoptotic cells in vitro and after retinal IR injury in vivo compared to wild-type myeloid cells, suggesting that HDAC3 hinders the reparative phagocytosis of dead cells, a process known as efferocytosis. Further mechanistic studies indicated that although HDAC3 KO macrophages upregulate the reparative enzyme arginase 1 (A1) that enhances efferocytosis, the inhibitory effect of HDAC3 on efferocytosis is not solely dependent on A1. Finally, treatment of wild-type mice with the HDAC3 inhibitor RGFP966 ameliorated the retinal neurodegeneration and thinning caused by IR injury. Collectively, our data show that HDAC3 deletion enhances macrophage-mediated efferocytosis and protects against retinal IR injury, suggesting that inhibiting myeloid HDAC3 holds promise as a novel therapeutic strategy for preserving retinal integrity after ischemic insult.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11241909/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141600221","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
Integrating single-nucleus RNA sequencing and spatial transcriptomics to elucidate a specialized subpopulation of astrocytes, microglia and vascular cells in brains of mouse model of lipopolysaccharide-induced sepsis-associated encephalopathy. 整合单核 RNA 测序和空间转录组学,阐明脂多糖诱导败血症相关脑病小鼠模型大脑中星形胶质细胞、小胶质细胞和血管细胞的特殊亚群。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-03 DOI: 10.1186/s12974-024-03161-0
Yanyan Zhu, Yin Zhang, Sheng He, Sanjun Yi, Hao Feng, Xianzhu Xia, Xiaodong Fang, Xiaoqian Gong, Pingsen Zhao

Background: Understanding the mechanism behind sepsis-associated encephalopathy (SAE) remains a formidable task. This study endeavors to shed light on the complex cellular and molecular alterations that occur in the brains of a mouse model with SAE, ultimately unraveling the underlying mechanisms of this condition.

Methods: We established a murine model using intraperitoneal injection of lipopolysaccharide (LPS) in wild type and Anxa1-/- mice and collected brain tissues for analysis at 0-hour, 12-hour, 24-hour, and 72-hour post-injection. Utilizing advanced techniques such as single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq, we conducted a comprehensive characterization of the cellular responses and molecular patterns within the brain.

Results: Our study uncovered notable temporal differences in the response to LPS challenge between Anxa1-/- (annexin A1 knockout) and wild type mice, specifically at the 12-hour and 24-hour time points following injection. We observed a significant increase in the proportion of Astro-2 and Micro-2 cells in these mice. These cells exhibited a colocalization pattern with the vascular subtype Vas-1, forming a distinct region known as V1A2M2, where Astro-2 and Micro-2 cells surrounded Vas-1. Moreover, through further analysis, we discovered significant upregulation of ligands and receptors such as Timp1-Cd63, Timp1-Itgb1, Timp1-Lrp1, as well as Ccl2-Ackr1 and Cxcl2-Ackr1 within this region. In addition, we observed a notable increase in the expression of Cd14-Itgb1, Cd14-Tlr2, and Cd14-C3ar1 in regions enriched with Micro-2 cells. Additionally, Cxcl10-Sdc4 showed broad upregulation in brain regions containing both Micro-2 and Astro-2 cells. Notably, upon LPS challenge, there was an observed increase in Anxa1 expression in the mouse brain. Furthermore, our study revealed a noteworthy increase in mortality rates following Anxa1 knockdown. However, we did not observe substantial differences in the types, numbers, or distribution of other brain cells between Anxa1-/- and wildtype mice over time. Nevertheless, when comparing the 24-hour post LPS injection time point, we observed a significant decrease in the proportion and distribution of Micro-2 and Astro-2 cells in the vicinity of blood vessels in Anxa1-/- mice. Additionally, we noted reduced expression levels of several ligand-receptor pairs including Cd14-Tlr2, Cd14-C3ar1, Cd14-Itgb1, Cxcl10-Sdc4, Ccl2-Ackr1, and Cxcl2-Ackr1.

Conclusions: By combining snRNA-seq and Stereo-seq techniques, our study successfully identified a distinctive cellular colocalization, referred to as a special pathological niche, comprising Astro-2, Micro-2, and Vas-1 cells. Furthermore, we observed an upregulation of ligand-receptor pairs within this niche. These findings suggest a potential association between this cellular arrangement and the underlying m

背景:了解败血症相关脑病(SAE)背后的机制仍然是一项艰巨的任务。本研究试图揭示脓毒症相关脑病小鼠模型大脑中发生的复杂的细胞和分子变化,最终揭示该病症的内在机制:我们利用腹腔注射脂多糖(LPS)的方法建立了野生型和 Anxa1-/- 小鼠模型,并收集了注射后 0 小时、12 小时、24 小时和 72 小时的脑组织进行分析。利用单核 RNA 测序(snRNA-seq)和立体测序(Stereo-seq)等先进技术,我们对脑内的细胞反应和分子模式进行了全面鉴定:我们的研究发现,Anxa1-/-(annexin A1基因敲除)和野生型小鼠对LPS挑战的反应存在明显的时间差异,特别是在注射后的12小时和24小时时间点。我们观察到这些小鼠中 Astro-2 和 Micro-2 细胞的比例明显增加。这些细胞表现出与血管亚型 Vas-1 的共定位模式,形成一个称为 V1A2M2 的独特区域,其中 Astro-2 和 Micro-2 细胞包围着 Vas-1。此外,通过进一步分析,我们发现在这一区域内,Timp1-Cd63、Timp1-Itgb1、Timp1-Lrp1 以及 Ccl2-Ackr1 和 Cxcl2-Ackr1 等配体和受体显著上调。此外,我们还观察到,在 Micro-2 细胞富集的区域,Cd14-Itgb1、Cd14-Tlr2 和 Cd14-C3ar1 的表达明显增加。此外,在含有 Micro-2 和 Astro-2 细胞的脑区,Cxcl10-Sdc4 也出现了广泛的上调。值得注意的是,在小鼠脑部受到 LPS 挑战时,可以观察到 Anxa1 的表达增加。此外,我们的研究还发现,在敲除 Anxa1 后,死亡率显著上升。然而,随着时间的推移,我们并没有观察到 Anxa1-/- 和野生型小鼠在其他脑细胞的类型、数量或分布上存在实质性差异。然而,在注射 LPS 后 24 小时的时间点进行比较时,我们观察到 Anxa1-/- 小鼠血管附近的 Micro-2 和 Astro-2 细胞的比例和分布明显减少。此外,我们还注意到一些配体-受体对的表达水平降低,包括 Cd14-Tlr2、Cd14-C3ar1、Cd14-Itgb1、Cxcl10-Sdc4、Ccl2-Ackr1 和 Cxcl2-Ackr1:通过结合 snRNA-seq 和 Stereo-seq 技术,我们的研究成功地发现了一种独特的细胞共定位,即由 Astro-2、Micro-2 和 Vas-1 细胞组成的特殊病理生态位。此外,我们还观察到配体-受体对在这个龛位中上调。这些发现表明,这种细胞排列与导致 SAE 或 Anxa1 基因敲除小鼠死亡率升高的潜在机制之间存在潜在联系。
{"title":"Integrating single-nucleus RNA sequencing and spatial transcriptomics to elucidate a specialized subpopulation of astrocytes, microglia and vascular cells in brains of mouse model of lipopolysaccharide-induced sepsis-associated encephalopathy.","authors":"Yanyan Zhu, Yin Zhang, Sheng He, Sanjun Yi, Hao Feng, Xianzhu Xia, Xiaodong Fang, Xiaoqian Gong, Pingsen Zhao","doi":"10.1186/s12974-024-03161-0","DOIUrl":"10.1186/s12974-024-03161-0","url":null,"abstract":"<p><strong>Background: </strong>Understanding the mechanism behind sepsis-associated encephalopathy (SAE) remains a formidable task. This study endeavors to shed light on the complex cellular and molecular alterations that occur in the brains of a mouse model with SAE, ultimately unraveling the underlying mechanisms of this condition.</p><p><strong>Methods: </strong>We established a murine model using intraperitoneal injection of lipopolysaccharide (LPS) in wild type and Anxa1<sup>-/-</sup> mice and collected brain tissues for analysis at 0-hour, 12-hour, 24-hour, and 72-hour post-injection. Utilizing advanced techniques such as single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq, we conducted a comprehensive characterization of the cellular responses and molecular patterns within the brain.</p><p><strong>Results: </strong>Our study uncovered notable temporal differences in the response to LPS challenge between Anxa1<sup>-/-</sup> (annexin A1 knockout) and wild type mice, specifically at the 12-hour and 24-hour time points following injection. We observed a significant increase in the proportion of Astro-2 and Micro-2 cells in these mice. These cells exhibited a colocalization pattern with the vascular subtype Vas-1, forming a distinct region known as V1A2M2, where Astro-2 and Micro-2 cells surrounded Vas-1. Moreover, through further analysis, we discovered significant upregulation of ligands and receptors such as Timp1-Cd63, Timp1-Itgb1, Timp1-Lrp1, as well as Ccl2-Ackr1 and Cxcl2-Ackr1 within this region. In addition, we observed a notable increase in the expression of Cd14-Itgb1, Cd14-Tlr2, and Cd14-C3ar1 in regions enriched with Micro-2 cells. Additionally, Cxcl10-Sdc4 showed broad upregulation in brain regions containing both Micro-2 and Astro-2 cells. Notably, upon LPS challenge, there was an observed increase in Anxa1 expression in the mouse brain. Furthermore, our study revealed a noteworthy increase in mortality rates following Anxa1 knockdown. However, we did not observe substantial differences in the types, numbers, or distribution of other brain cells between Anxa1<sup>-/-</sup> and wildtype mice over time. Nevertheless, when comparing the 24-hour post LPS injection time point, we observed a significant decrease in the proportion and distribution of Micro-2 and Astro-2 cells in the vicinity of blood vessels in Anxa1<sup>-/-</sup> mice. Additionally, we noted reduced expression levels of several ligand-receptor pairs including Cd14-Tlr2, Cd14-C3ar1, Cd14-Itgb1, Cxcl10-Sdc4, Ccl2-Ackr1, and Cxcl2-Ackr1.</p><p><strong>Conclusions: </strong>By combining snRNA-seq and Stereo-seq techniques, our study successfully identified a distinctive cellular colocalization, referred to as a special pathological niche, comprising Astro-2, Micro-2, and Vas-1 cells. Furthermore, we observed an upregulation of ligand-receptor pairs within this niche. These findings suggest a potential association between this cellular arrangement and the underlying m","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11223438/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141498297","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
Redefining the ontogeny of hyalocytes as yolk sac-derived tissue-resident macrophages of the vitreous body. 重新定义透明细胞作为卵黄囊衍生的玻璃体组织驻留巨噬细胞的本体。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-03 DOI: 10.1186/s12974-024-03110-x
Dennis-Dominik Rosmus, Jana Koch, Annika Hausmann, Aude Chiot, Franz Arnhold, Takahiro Masuda, Katrin Kierdorf, Stefanie Marie Hansen, Heidrun Kuhrt, Janine Fröba, Julian Wolf, Stefaniya Boneva, Martin Gericke, Bahareh Ajami, Marco Prinz, Clemens Lange, Peter Wieghofer

Background: The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive.

Results: In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes.

Conclusion: Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.

背景:眼睛是一个高度特化的感觉器官,它包括作为中枢神经系统一部分的视网膜,但也包括非神经区,如确保眼球稳定和清晰光轴的透明玻璃体。透明细胞是玻璃体内的组织驻留巨噬细胞,被认为在玻璃体视网膜界面的健康和疾病(如增殖性玻璃体视网膜病变或糖尿病视网膜病变)中发挥着关键作用。然而,与其他眼部巨噬细胞不同的是,它们的胚胎起源以及这些髓系细胞在多大程度上可能由循环单核细胞补充的问题仍未解决:在这项研究中,我们将转基因报告小鼠、胚胎和成体命运图谱方法以及同种异体实验与多色免疫荧光标记和共聚焦激光扫描显微镜相结合,全面描述了小鼠透明细胞群体在整个发育过程和成年期的特征。我们发现,小鼠透明细胞表达许多众所周知的髓细胞标记,但同时也显示出不同于视网膜小胶质细胞的独特免疫表型。胚胎脉冲标记显示,小鼠透明细胞起源于卵黄囊,其前体在出生前为发育中的眼睛播下种子。最后,产后标记和同种异体培养确定了透明细胞的寿命,透明细胞的维持依赖于集落刺激因子 1 受体(CSF1R)信号,与血源性单核细胞无关:我们的研究发现透明细胞是卵黄囊造血的长寿后代,并强调了它们作为眼球先天免疫系统重要成员的作用。由于透明质细胞寿命长,免疫衰老过程可能会导致透明质细胞功能障碍,从而引发玻璃体视网膜疾病。因此,髓系细胞靶向疗法通过改变透明质细胞的特性来达到治疗效果,可能是减轻玻璃体视网膜界面疾病造成的负担的一种有趣方法。
{"title":"Redefining the ontogeny of hyalocytes as yolk sac-derived tissue-resident macrophages of the vitreous body.","authors":"Dennis-Dominik Rosmus, Jana Koch, Annika Hausmann, Aude Chiot, Franz Arnhold, Takahiro Masuda, Katrin Kierdorf, Stefanie Marie Hansen, Heidrun Kuhrt, Janine Fröba, Julian Wolf, Stefaniya Boneva, Martin Gericke, Bahareh Ajami, Marco Prinz, Clemens Lange, Peter Wieghofer","doi":"10.1186/s12974-024-03110-x","DOIUrl":"10.1186/s12974-024-03110-x","url":null,"abstract":"<p><strong>Background: </strong>The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive.</p><p><strong>Results: </strong>In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes.</p><p><strong>Conclusion: </strong>Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11223341/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141498298","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
Dysregulated expression of miR-140 and miR-122 compromised microglial chemotaxis and led to reduced restriction of AD pathology. miR-140 和 miR-122 的表达失调会损害小胶质细胞的趋化性,并导致减少对 AD 病理学的限制。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-02 DOI: 10.1186/s12974-024-03162-z
Chao Song, Shufang Li, Yingren Mai, Linpeng Li, Guoku Dai, Yuan Zhou, Xiaosheng Liang, Olivia Meilan Zou, Ya Wang, Libing Zhou, Jun Liu, Yi Zou

Background: Deposition of amyloid β, which is produced by amyloidogenic cleavage of APP by β- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology.

Results: In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aβ were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aβ phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets.

Conclusions: Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.

背景:淀粉样β是由β-和γ-分泌酶裂解APP产生的淀粉样蛋白,它的沉积是AD病理学的主要特征之一。APP也可先后被α-和γ-分泌酶处理,生成sAPPα,而sAPPα已被证明具有神经保护作用,能促进神经元突起生长和神经元存活等。方法:利用 miRNA-seq 技术分析了 11 名 AD 患者和 14 名年龄与性别匹配的认知正常志愿者血浆样本中 miRNA 的全局表达谱。然后,在体内和体外过表达 miR-140 和 miR-122,并在体外敲除 miR-140 和 miR-122 的内源性表达。采用分子生物学、免疫组化等多种技术检测miRNA对AD病理的影响:结果:在这项研究中,我们发现在AD患者的血浆中,两种miRNA(miR-140-3p和miR-122-5p)都出现了上调,这两种miRNA都靶向中枢神经系统中主要的α-分泌酶ADAM10。在小鼠大脑中过表达这两种miRNA会诱导野生型C57BL/6J小鼠认知能力下降,并加剧APP/PS1小鼠的认知障碍。虽然没有检测到APP和总Aβ的明显变化,但在过表达miR-140/miR-122的小鼠脑中观察到ADAM10及其非淀粉样蛋白生成产物sAPPα的明显下调。免疫组织学分析表明,这些小鼠海马中神经元萎缩的增加与小胶质细胞趋化性的降低有关,而与参与调节小胶质细胞免疫活性的另外两种 ADAM10 底物(神经元 CX3CL1 和小胶质细胞 TREM2)无关。进一步的体外分析表明,过表达miR-140/miR-122的小鼠胚胎神经元细胞的神经外生能力降低,以及与过表达miR-140/miR-122的HT22细胞共培养的小胶质细胞的Aβ吞噬能力降低,都可以通过过表达miR-140/miR-122 TuD的特异性抑制序列以及添加sAPPα来挽救,从而使这些miRNAs成为潜在的治疗靶点:我们的研究结果表明,具有神经保护作用的sAPPα是miR-140和miR-122表达失调诱导神经病理进展的关键因素。以这些 miRNA 为靶点可能是治疗 AD 的一种有前景的治疗策略。
{"title":"Dysregulated expression of miR-140 and miR-122 compromised microglial chemotaxis and led to reduced restriction of AD pathology.","authors":"Chao Song, Shufang Li, Yingren Mai, Linpeng Li, Guoku Dai, Yuan Zhou, Xiaosheng Liang, Olivia Meilan Zou, Ya Wang, Libing Zhou, Jun Liu, Yi Zou","doi":"10.1186/s12974-024-03162-z","DOIUrl":"10.1186/s12974-024-03162-z","url":null,"abstract":"<p><strong>Background: </strong>Deposition of amyloid β, which is produced by amyloidogenic cleavage of APP by β- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology.</p><p><strong>Results: </strong>In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aβ were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aβ phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets.</p><p><strong>Conclusions: </strong>Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11218311/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492340","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
Intermittent hypoxia exacerbates anxiety in high-fat diet-induced diabetic mice by inhibiting TREM2-regulated IFNAR1 signaling. 间歇性缺氧通过抑制 TREM2 调节的 IFNAR1 信号传导,加剧高脂饮食诱导的糖尿病小鼠的焦虑。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-02 DOI: 10.1186/s12974-024-03160-1
Wenyu Ni, Yun Niu, Sitong Cao, Chunsun Fan, Jian Fan, Li Zhu, Xueting Wang

Background: Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM.

Methods: A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR.

Results: Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation.

Conclusions: HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.

背景:2 型糖尿病(T2DM)和阻塞性睡眠呼吸暂停(OSA)是相互影响的危险因素,这两种疾病都会诱发认知障碍和焦虑。然而,OSA 是否会加重 T2DM 患者的认知障碍和焦虑仍不清楚。此外,TREM2 的上调被认为在减轻 T2DM 小鼠的小胶质细胞激活和改善突触功能方面起到保护作用。本研究旨在探索 TREM2 的调控机制以及 OSA 合并 T2DM 小鼠的认知和焦虑样行为变化:方法:通过60%千卡高脂饮食(HFD)和间歇性缺氧(IH)治疗小鼠,建立了T2DM合并OSA模型。对小鼠的空间学习记忆能力和焦虑进行了研究。大脑神经元损伤是通过突触密度的数量、大脑小胶质细胞的数量和形态以及促炎因子来确定的。为了探索其机制,研究人员用高糖(HG)和IH共同处理小胶质细胞,建立了T2DM合并OSA的体外模型。通过 RNA 测序和 qRT-PCR 确定了 TREM2 对 IFNAR1-STAT1 通路的调控作用:结果表明:高脂饮食小鼠表现出明显的认知功能障碍和焦虑样行为,并伴有明显的突触丢失。此外,还观察到大脑小胶质细胞明显活化,小胶质细胞吞噬突触的能力增强。此外,研究还发现 IH 能显著加重高氟酸小鼠的焦虑。HG 治疗的机制可能是促进 TREM2 上调,进而通过抑制 IFNAR1-STAT1 通路减轻促炎性小胶质细胞。相反,IH 联合处理的 HFD 小鼠和 HG 处理的小胶质细胞中 TREM2 的显著减少导致 IFNAR1-STAT1 通路的进一步激活,从而增加了促炎性小胶质细胞的激活:结论:HFD 会上调 IFNAR1-STAT1 通路并诱导促炎性小胶质细胞,从而导致突触损伤,造成焦虑和认知障碍。T2DM小鼠脑内上调的TREM2对IFNAR1-STAT1通路具有负向调节作用。T2DM合并OSA的小鼠通过下调TREM2加剧了焦虑,导致IFNAR1-STAT1通路激活,从而增加了促炎性小胶质细胞。
{"title":"Intermittent hypoxia exacerbates anxiety in high-fat diet-induced diabetic mice by inhibiting TREM2-regulated IFNAR1 signaling.","authors":"Wenyu Ni, Yun Niu, Sitong Cao, Chunsun Fan, Jian Fan, Li Zhu, Xueting Wang","doi":"10.1186/s12974-024-03160-1","DOIUrl":"10.1186/s12974-024-03160-1","url":null,"abstract":"<p><strong>Background: </strong>Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM.</p><p><strong>Methods: </strong>A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR.</p><p><strong>Results: </strong>Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation.</p><p><strong>Conclusions: </strong>HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11218348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492366","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
Traumatic brain injury alters the effects of class II invariant peptide (CLIP) antagonism on chronic meningeal CLIP + B cells, neuropathology, and neurobehavioral impairment in 5xFAD mice. 创伤性脑损伤改变了 II 类不变肽(CLIP)拮抗剂对 5xFAD 小鼠慢性脑膜 CLIP + B 细胞、神经病理学和神经行为障碍的影响。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-27 DOI: 10.1186/s12974-024-03146-z
Jaclyn Iannucci, Reagan Dominy, Shreya Bandopadhyay, E Madison Arthur, Brenda Noarbe, Amandine Jullienne, Margret Krkasharyan, Richard P Tobin, Aleksandr Pereverzev, Samantha Beevers, Lavanya Venkatasamy, Karienn A Souza, Daniel C Jupiter, Alan Dabney, Andre Obenaus, M Karen Newell-Rogers, Lee A Shapiro

Background: Traumatic brain injury (TBI) is a significant risk factor for Alzheimer's disease (AD), and accumulating evidence supports a role for adaptive immune B and T cells in both TBI and AD pathogenesis. We previously identified B cell and major histocompatibility complex class II (MHCII)-associated invariant chain peptide (CLIP)-positive B cell expansion after TBI. We also showed that antagonizing CLIP binding to the antigen presenting groove of MHCII after TBI acutely reduced CLIP + splenic B cells and was neuroprotective. The current study investigated the chronic effects of antagonizing CLIP in the 5xFAD Alzheimer's mouse model, with and without TBI.

Methods: 12-week-old male wild type (WT) and 5xFAD mice were administered either CLIP antagonist peptide (CAP) or vehicle, once at 30 min after either sham or a lateral fluid percussion injury (FPI). Analyses included flow cytometric analysis of immune cells in dural meninges and spleen, histopathological analysis of the brain, magnetic resonance diffusion tensor imaging, cerebrovascular analysis, and assessment of motor and neurobehavioral function over the ensuing 6 months.

Results: 9-month-old 5xFAD mice had significantly more CLIP + B cells in the meninges compared to age-matched WT mice. A one-time treatment with CAP significantly reduced this population in 5xFAD mice. Importantly, CAP also improved some of the immune, histopathological, and neurobehavioral impairments in 5xFAD mice over the ensuing six months. Although FPI did not further elevate meningeal CLIP + B cells, it did negate the ability of CAP to reduce meningeal CLIP + B cells in the 5xFAD mice. FPI at 3 months of age exacerbated some aspects of AD pathology in 5xFAD mice, including further reducing hippocampal neurogenesis, increasing plaque deposition in CA3, altering microgliosis, and disrupting the cerebrovascular structure. CAP treatment after injury ameliorated some but not all of these FPI effects.

背景:创伤性脑损伤(TBI)是阿尔茨海默病(AD)的一个重要危险因素,越来越多的证据支持适应性免疫 B 细胞和 T 细胞在创伤性脑损伤和阿尔茨海默病发病机制中的作用。我们之前发现了创伤后B细胞和主要组织相容性复合体II类(MHCII)相关不变链肽(CLIP)阳性B细胞扩增。我们还发现,在创伤性脑损伤后,拮抗 CLIP 与 MHCII 的抗原递呈槽的结合可急性减少 CLIP + 脾脏 B 细胞,并具有神经保护作用。本研究调查了拮抗 CLIP 对 5xFAD 阿尔茨海默小鼠模型的慢性影响,包括有无 TBI。方法:在假性或侧液叩击伤(FPI)后 30 分钟,给 12 周大雄性野生型(WT)和 5xFAD 小鼠注射一次 CLIP 拮抗剂肽(CAP)或药物。分析包括硬脑膜和脾脏中免疫细胞的流式细胞分析、脑组织病理学分析、磁共振弥散张量成像、脑血管分析以及随后 6 个月的运动和神经行为功能评估:结果:与年龄匹配的WT小鼠相比,9个月大的5xFAD小鼠脑膜中的CLIP + B细胞明显增多。一次性使用 CAP 治疗后,5xFAD 小鼠脑膜中的 CLIP + B 细胞数量明显减少。重要的是,在随后的六个月中,CAP 还能改善 5xFAD 小鼠的一些免疫、组织病理学和神经行为损伤。虽然 FPI 没有进一步提高 5xFAD 小鼠脑膜 CLIP + B 细胞的数量,但它确实否定了 CAP 减少 5xFAD 小鼠脑膜 CLIP + B 细胞的能力。3月龄时的FPI加剧了5xFAD小鼠AD病理学的某些方面,包括进一步减少海马神经发生、增加CA3的斑块沉积、改变小胶质细胞和破坏脑血管结构。损伤后的 CAP 治疗可改善 FPI 的部分影响,但不是全部影响。
{"title":"Traumatic brain injury alters the effects of class II invariant peptide (CLIP) antagonism on chronic meningeal CLIP + B cells, neuropathology, and neurobehavioral impairment in 5xFAD mice.","authors":"Jaclyn Iannucci, Reagan Dominy, Shreya Bandopadhyay, E Madison Arthur, Brenda Noarbe, Amandine Jullienne, Margret Krkasharyan, Richard P Tobin, Aleksandr Pereverzev, Samantha Beevers, Lavanya Venkatasamy, Karienn A Souza, Daniel C Jupiter, Alan Dabney, Andre Obenaus, M Karen Newell-Rogers, Lee A Shapiro","doi":"10.1186/s12974-024-03146-z","DOIUrl":"10.1186/s12974-024-03146-z","url":null,"abstract":"<p><strong>Background: </strong>Traumatic brain injury (TBI) is a significant risk factor for Alzheimer's disease (AD), and accumulating evidence supports a role for adaptive immune B and T cells in both TBI and AD pathogenesis. We previously identified B cell and major histocompatibility complex class II (MHCII)-associated invariant chain peptide (CLIP)-positive B cell expansion after TBI. We also showed that antagonizing CLIP binding to the antigen presenting groove of MHCII after TBI acutely reduced CLIP + splenic B cells and was neuroprotective. The current study investigated the chronic effects of antagonizing CLIP in the 5xFAD Alzheimer's mouse model, with and without TBI.</p><p><strong>Methods: </strong>12-week-old male wild type (WT) and 5xFAD mice were administered either CLIP antagonist peptide (CAP) or vehicle, once at 30 min after either sham or a lateral fluid percussion injury (FPI). Analyses included flow cytometric analysis of immune cells in dural meninges and spleen, histopathological analysis of the brain, magnetic resonance diffusion tensor imaging, cerebrovascular analysis, and assessment of motor and neurobehavioral function over the ensuing 6 months.</p><p><strong>Results: </strong>9-month-old 5xFAD mice had significantly more CLIP + B cells in the meninges compared to age-matched WT mice. A one-time treatment with CAP significantly reduced this population in 5xFAD mice. Importantly, CAP also improved some of the immune, histopathological, and neurobehavioral impairments in 5xFAD mice over the ensuing six months. Although FPI did not further elevate meningeal CLIP + B cells, it did negate the ability of CAP to reduce meningeal CLIP + B cells in the 5xFAD mice. FPI at 3 months of age exacerbated some aspects of AD pathology in 5xFAD mice, including further reducing hippocampal neurogenesis, increasing plaque deposition in CA3, altering microgliosis, and disrupting the cerebrovascular structure. CAP treatment after injury ameliorated some but not all of these FPI effects.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11212436/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141468733","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Maternal SARS-CoV-2 impacts fetal placental macrophage programs and placenta-derived microglial models of neurodevelopment. 母体 SARS-CoV-2 影响胎儿胎盘巨噬细胞程序和胎盘源性微神经胶质细胞神经发育模型。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-25 DOI: 10.1186/s12974-024-03157-w
Lydia L Shook, Rebecca E Batorsky, Rose M De Guzman, Liam T McCrea, Sara M Brigida, Joy E Horng, Steven D Sheridan, Olha Kholod, Aidan M Cook, Jonathan Z Li, Donna K Slonim, Brittany A Goods, Roy H Perlis, Andrea G Edlow

Background: The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models.

Methods and results: We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls.

Conclusion: These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.

背景:SARS-CoV-2 病毒会激活母体和胎盘的免疫反应。众所周知,在怀孕期间发生其他感染时,这种激活会影响胎儿的大脑发育。母体免疫激活对神经发育的影响至少部分是由胎儿大脑小胶质细胞介导的。然而,小胶质细胞无法直接进行分析,也没有经过验证的非侵入性替代模型来评估子宫内小胶质细胞的启动和功能。我们之前在小鼠模型中证实了小胶质细胞和霍夫鲍尔细胞(HBCs,或胎儿胎盘巨噬细胞)之间共享转录程序:我们评估了母体 SARS-CoV-2 对从 24 个足月胎盘(N = 10 个 SARS-CoV-2 阳性病例,14 个阴性对照)中分离出的 HBCs 的影响。通过单细胞 RNA 测序,我们证明 HBC 亚群表现出不同的细胞程序,特定亚群受到 SARS-CoV-2 的不同影响。对不同表达基因的评估表明,某些亚群的吞噬功能受损,而吞噬功能是 HBC 和小胶质细胞的一项关键功能。利用先前验证的小胶质细胞突触修剪模型,我们发现从SARS-CoV-2阳性孕妇胎盘中分离出的HBCs可转分化为小胶质细胞样细胞(HBC-iMGs),与阴性对照的HBC模型相比,其突触修剪行为受损:这些研究结果表明,出生时分离的 HBC 可用于创建后代小胶质细胞编程的个性化细胞模型。
{"title":"Maternal SARS-CoV-2 impacts fetal placental macrophage programs and placenta-derived microglial models of neurodevelopment.","authors":"Lydia L Shook, Rebecca E Batorsky, Rose M De Guzman, Liam T McCrea, Sara M Brigida, Joy E Horng, Steven D Sheridan, Olha Kholod, Aidan M Cook, Jonathan Z Li, Donna K Slonim, Brittany A Goods, Roy H Perlis, Andrea G Edlow","doi":"10.1186/s12974-024-03157-w","DOIUrl":"10.1186/s12974-024-03157-w","url":null,"abstract":"<p><strong>Background: </strong>The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models.</p><p><strong>Methods and results: </strong>We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls.</p><p><strong>Conclusion: </strong>These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11197235/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450732","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
Therapeutic intervention in neuroinflammation for neovascular ocular diseases through targeting the cGAS-STING-necroptosis pathway. 通过靶向 cGAS-STING-necroptosis 通路对神经炎症进行治疗干预,以治疗新生血管性眼病。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-25 DOI: 10.1186/s12974-024-03155-y
Biyan Ni, Ziqi Yang, Tian Zhou, Hong Zhou, Yang Zhou, Shiya Lin, Huiyi Xu, Xiaojing Lin, Wei Yi, Chang He, Xialin Liu

The microglia-mediated neuroinflammation have been shown to play a crucial role in the ocular pathological angiogenesis process, but specific immunotherapies for neovascular ocular diseases are still lacking. This study proposed that targeting GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) might be a novel immunotherapy for these angiogenesis diseases. We found a significant upregulation of CGAS and STING genes in the RNA-seq data derived from retinal tissues of the patients with proliferative diabetic retinopathy. In experimental models of ocular angiogenesis including laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR), the cGAS-STING pathway was activated as angiogenesis progressed. Either genetic deletion or pharmacological inhibition of STING resulted in a remarkable suppression of neovascularization in both models. Furthermore, cGAS-STING signaling was specifically activated in myeloid cells, triggering the subsequent RIP1-RIP3-MLKL pathway activation and leading to necroptosis-mediated inflammation. Notably, targeted inhibition of the cGAS-STING pathway with C-176 or SN-011 could significantly suppress pathological angiogenesis in CNV and OIR. Additionally, the combination of C-176 or SN-011 with anti-VEGF therapy led to least angiogenesis, markedly enhancing the anti-angiogenic effectiveness. Together, our findings provide compelling evidence for the importance of the cGAS-STING-necroptosis axis in pathological angiogenesis, highlighting its potential as a promising immunotherapeutic target for treating neovascular ocular diseases.

小胶质细胞介导的神经炎症已被证明在眼部病理性血管生成过程中起着至关重要的作用,但目前仍缺乏针对新生血管性眼病的特异性免疫疗法。本研究提出,靶向 GMP-AMP 合成酶(cGAS)-干扰素基因刺激器(STING)可能是治疗这些血管生成疾病的一种新型免疫疗法。我们发现,在来自增殖性糖尿病视网膜病变患者视网膜组织的RNA-seq数据中,CGAS和STING基因明显上调。在包括激光诱导脉络膜新生血管(CNV)和氧诱导视网膜病变(OIR)在内的眼部血管生成实验模型中,随着血管生成的进展,cGAS-STING通路被激活。基因缺失或药物抑制 STING 可显著抑制这两种模型中的新生血管形成。此外,cGAS-STING 信号在骨髓细胞中被特异性激活,引发随后的 RIP1-RIP3-MLKL 通路激活,导致坏死介导的炎症。值得注意的是,用C-176或SN-011靶向抑制cGAS-STING通路可显著抑制CNV和OIR的病理性血管生成。此外,C-176或SN-011与抗血管内皮生长因子疗法联合使用可减少血管生成,明显提高抗血管生成的效果。总之,我们的研究结果为cGAS-STING-necroptosis轴在病理性血管生成中的重要性提供了令人信服的证据,凸显了其作为治疗新生血管性眼病的免疫治疗靶点的潜力。
{"title":"Therapeutic intervention in neuroinflammation for neovascular ocular diseases through targeting the cGAS-STING-necroptosis pathway.","authors":"Biyan Ni, Ziqi Yang, Tian Zhou, Hong Zhou, Yang Zhou, Shiya Lin, Huiyi Xu, Xiaojing Lin, Wei Yi, Chang He, Xialin Liu","doi":"10.1186/s12974-024-03155-y","DOIUrl":"10.1186/s12974-024-03155-y","url":null,"abstract":"<p><p>The microglia-mediated neuroinflammation have been shown to play a crucial role in the ocular pathological angiogenesis process, but specific immunotherapies for neovascular ocular diseases are still lacking. This study proposed that targeting GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) might be a novel immunotherapy for these angiogenesis diseases. We found a significant upregulation of CGAS and STING genes in the RNA-seq data derived from retinal tissues of the patients with proliferative diabetic retinopathy. In experimental models of ocular angiogenesis including laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR), the cGAS-STING pathway was activated as angiogenesis progressed. Either genetic deletion or pharmacological inhibition of STING resulted in a remarkable suppression of neovascularization in both models. Furthermore, cGAS-STING signaling was specifically activated in myeloid cells, triggering the subsequent RIP1-RIP3-MLKL pathway activation and leading to necroptosis-mediated inflammation. Notably, targeted inhibition of the cGAS-STING pathway with C-176 or SN-011 could significantly suppress pathological angiogenesis in CNV and OIR. Additionally, the combination of C-176 or SN-011 with anti-VEGF therapy led to least angiogenesis, markedly enhancing the anti-angiogenic effectiveness. Together, our findings provide compelling evidence for the importance of the cGAS-STING-necroptosis axis in pathological angiogenesis, highlighting its potential as a promising immunotherapeutic target for treating neovascular ocular diseases.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11197344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450733","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