首页 > 最新文献

Inflammation Research最新文献

英文 中文
Dissection of pro-tumoral macrophage subtypes and immunosuppressive cells participating in M2 polarization. 参与 M2 极化的促肿瘤巨噬细胞亚型和免疫抑制细胞剖析
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-06-27 DOI: 10.1007/s00011-024-01907-3
Onurcan Sezginer, Nese Unver

Alternatively activated macrophage (M2) polarization can result in one of four subtypes based on cytokines and signaling pathways associated with macrophage activation: M2a, M2b, M2c, and M2d macrophages. The majority of M2 subtypes are anti-inflammatory and pro-angiogenic, secreting growth factors (VEGF, PDGF) and matrix metalloproteinases (MMP2, MMP9) which boost tumor growth, metastasis, and invasion. M2-polarized macrophages are associated with immune suppressor cells harboring Myeloid derived suppressor cells, Regulatory T cells (Tregs), Regulatory B cells as well as alternatively activated (N2) neutrophils. Treg cells selectively support the metabolic stability, mitochondrial integrity, and survival rate of M2-like TAMs in an indirect environment. Also, the contribution of Breg cells influences macrophage polarization towards the M2 direction. TAM is activated when TAN levels in the tumor microenvironment are insufficient or vice versa, suggesting that macrophage and its polarization are fine-tuned. Understanding the functions of immune suppressive cells, mediators, and signaling pathways involved with M2 polarization will allow us to identify potential strategies for targeting the TAM repolarization phenotype for innovative immunotherapy approaches. In this review, we have highlighted the critical factors for M2 macrophage polarization, differential cytokine/chemokine profiles of M1 and M2 macrophage subtypes, and other immune cells' impact on the polarization within the immunosuppressive niche.

根据与巨噬细胞活化相关的细胞因子和信号通路,另一种活化的巨噬细胞(M2)极化可产生四种亚型之一:M2a、M2b、M2c 和 M2d 巨噬细胞。大多数 M2 亚型具有抗炎和促进血管生成的作用,可分泌生长因子(血管内皮生长因子、促生长因子)和基质金属蛋白酶(MMP2、MMP9),促进肿瘤生长、转移和侵袭。M2 极化巨噬细胞与免疫抑制细胞有关,其中包括髓系衍生抑制细胞、调节性 T 细胞(Tregs)、调节性 B 细胞以及替代活化(N2)中性粒细胞。在间接环境中,Treg 细胞可选择性地支持 M2 类 TAM 的代谢稳定性、线粒体完整性和存活率。此外,Breg 细胞的贡献也会影响巨噬细胞向 M2 方向极化。当肿瘤微环境中的TAN水平不足时,TAM被激活,反之亦然,这表明巨噬细胞及其极化是微调的。了解与 M2 极化有关的免疫抑制细胞、介质和信号通路的功能,将使我们能够确定针对 TAM 再极化表型的潜在策略,从而采用创新的免疫疗法。在这篇综述中,我们强调了 M2 巨噬细胞极化的关键因素、M1 和 M2 巨噬细胞亚型的不同细胞因子/趋化因子谱,以及其他免疫细胞对免疫抑制龛内极化的影响。
{"title":"Dissection of pro-tumoral macrophage subtypes and immunosuppressive cells participating in M2 polarization.","authors":"Onurcan Sezginer, Nese Unver","doi":"10.1007/s00011-024-01907-3","DOIUrl":"10.1007/s00011-024-01907-3","url":null,"abstract":"<p><p>Alternatively activated macrophage (M2) polarization can result in one of four subtypes based on cytokines and signaling pathways associated with macrophage activation: M2a, M2b, M2c, and M2d macrophages. The majority of M2 subtypes are anti-inflammatory and pro-angiogenic, secreting growth factors (VEGF, PDGF) and matrix metalloproteinases (MMP2, MMP9) which boost tumor growth, metastasis, and invasion. M2-polarized macrophages are associated with immune suppressor cells harboring Myeloid derived suppressor cells, Regulatory T cells (Tregs), Regulatory B cells as well as alternatively activated (N2) neutrophils. Treg cells selectively support the metabolic stability, mitochondrial integrity, and survival rate of M2-like TAMs in an indirect environment. Also, the contribution of Breg cells influences macrophage polarization towards the M2 direction. TAM is activated when TAN levels in the tumor microenvironment are insufficient or vice versa, suggesting that macrophage and its polarization are fine-tuned. Understanding the functions of immune suppressive cells, mediators, and signaling pathways involved with M2 polarization will allow us to identify potential strategies for targeting the TAM repolarization phenotype for innovative immunotherapy approaches. In this review, we have highlighted the critical factors for M2 macrophage polarization, differential cytokine/chemokine profiles of M1 and M2 macrophage subtypes, and other immune cells' impact on the polarization within the immunosuppressive niche.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1411-1423"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11349836/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141456531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Shifting phenotype and differentiation of CD11b+Gr.1+ immature heterogeneous myeloid derived adjuster cells support inflammation and induce regulators of IL17A in imiquimod induced psoriasis. CD11b+Gr.1+未成熟异质性髓系衍生调节细胞的表型和分化变化支持炎症,并诱导咪喹莫特诱导的银屑病中的IL17A调节因子。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-07-25 DOI: 10.1007/s00011-024-01918-0
Debanjan Sarkar, Anik Pramanik, Dona Das, Sankar Bhattacharyya

Objective and design: The exact immunological mechanism of widespread chronic inflammatory skin disorder psoriasis has not been fully established. CD11b+Gr.1+ myeloid-derived cells are immature heterogeneous cells with T-cell suppressive property in neoplasia; however, influence of these cells on adaptive immunity is highly contextual; therefore, we dubbed these cells as myeloid-derived adjuster cells (MDAC). We studied imiquimod induced psoriasis in mouse model and evaluated for the first time the RORγt-NFAT1 axis in MDACs and the function, differentiation and interaction of these cells with T cells.

Materials and methods: The status of T cells and MDACs; their functionality and differentiation properties, and the roles of RORγt and NFAT1 in MDACs were evaluated using flow cytometry, qRT-PCR and confocal imaging.

Results: We found gradual increase in T cells and MDACs and an increase in the number of IL17 -secreting MDACs and T cells in the skin of psoriatic animals. We also noted that MDAC differentiation is biased toward M1 macrophages and DCs which perpetuate inflammation. We found that psoriatic MDACs were unable to suppress T-cell proliferation or activation but seemingly helped these T cells produce more IL17. Inhibition of the RORγt/NFAT1 axis in MDACs increased the suppressive nature of MDACs, allowing these cells to suppress the activity of psoriatic T-cells.

Conclusion: Our results indicate that altered MDAC properties in psoriatic condition sustains pathological inflammation and RORγt and NFAT1 as promising intervention target for psoriasis management.

目的和设计:广泛存在的慢性炎症性皮肤病银屑病的确切免疫学机制尚未完全确定。CD11b+Gr.1+髓源性细胞是一种未成熟的异型细胞,在肿瘤中具有抑制T细胞的特性;然而,这些细胞对适应性免疫的影响与环境有很大关系;因此,我们将这些细胞称为髓源性调节细胞(MDAC)。我们研究了咪喹莫特诱导的银屑病小鼠模型,并首次评估了 MDACs 中的 RORγt-NFAT1 轴以及这些细胞的功能、分化和与 T 细胞的相互作用:采用流式细胞术、qRT-PCR和共聚焦成像技术评估了T细胞和MDACs的状态、功能和分化特性,以及RORγt和NFAT1在MDACs中的作用:结果:我们发现银屑病动物皮肤中的 T 细胞和 MDACs 逐渐增多,分泌 IL17 的 MDACs 和 T 细胞数量增加。我们还注意到,MDAC 的分化偏向于 M1 巨噬细胞和 DCs,它们会使炎症持续存在。我们发现,银屑病 MDAC 无法抑制 T 细胞的增殖或活化,但似乎有助于这些 T 细胞产生更多的 IL17。抑制MDACs中的RORγt/NFAT1轴增加了MDACs的抑制性,使这些细胞能够抑制银屑病T细胞的活性:我们的研究结果表明,银屑病患者的MDAC特性改变会维持病理炎症,而RORγt和NFAT1是治疗银屑病的有希望的干预靶点。
{"title":"Shifting phenotype and differentiation of CD11b<sup>+</sup>Gr.1<sup>+</sup> immature heterogeneous myeloid derived adjuster cells support inflammation and induce regulators of IL17A in imiquimod induced psoriasis.","authors":"Debanjan Sarkar, Anik Pramanik, Dona Das, Sankar Bhattacharyya","doi":"10.1007/s00011-024-01918-0","DOIUrl":"10.1007/s00011-024-01918-0","url":null,"abstract":"<p><strong>Objective and design: </strong>The exact immunological mechanism of widespread chronic inflammatory skin disorder psoriasis has not been fully established. CD11b<sup>+</sup>Gr.1<sup>+</sup> myeloid-derived cells are immature heterogeneous cells with T-cell suppressive property in neoplasia; however, influence of these cells on adaptive immunity is highly contextual; therefore, we dubbed these cells as myeloid-derived adjuster cells (MDAC). We studied imiquimod induced psoriasis in mouse model and evaluated for the first time the RORγt-NFAT1 axis in MDACs and the function, differentiation and interaction of these cells with T cells.</p><p><strong>Materials and methods: </strong>The status of T cells and MDACs; their functionality and differentiation properties, and the roles of RORγt and NFAT1 in MDACs were evaluated using flow cytometry, qRT-PCR and confocal imaging.</p><p><strong>Results: </strong>We found gradual increase in T cells and MDACs and an increase in the number of IL17 -secreting MDACs and T cells in the skin of psoriatic animals. We also noted that MDAC differentiation is biased toward M1 macrophages and DCs which perpetuate inflammation. We found that psoriatic MDACs were unable to suppress T-cell proliferation or activation but seemingly helped these T cells produce more IL17. Inhibition of the RORγt/NFAT1 axis in MDACs increased the suppressive nature of MDACs, allowing these cells to suppress the activity of psoriatic T-cells.</p><p><strong>Conclusion: </strong>Our results indicate that altered MDAC properties in psoriatic condition sustains pathological inflammation and RORγt and NFAT1 as promising intervention target for psoriasis management.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1581-1599"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141758429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SDS3 regulates microglial inflammation by modulating the expression of the upstream kinase ASK1 in the p38 MAPK signaling pathway. SDS3 通过调节 p38 MAPK 信号通路上游激酶 ASK1 的表达来调节小胶质细胞炎症。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-07-15 DOI: 10.1007/s00011-024-01913-5
Jian Shen, Wenjia Lai, Zeyang Li, Wenyuan Zhu, Xue Bai, Zihao Yang, Qingsong Wang, Jianguo Ji

Background: Microglia, the main innate immune cells in the central nervous system, are key drivers of neuroinflammation, which plays a crucial role in the pathogenesis of neurodegenerative diseases. The Sin3/histone deacetylase (HDAC) complex, a highly conserved multiprotein co-repressor complex, primarily performs transcriptional repression via deacetylase activity; however, the function of SDS3, which maintains the integrity of the complex, in microglia remains unclear.

Methods: To uncover the regulatory role of the transcriptional co-repressor SDS3 in microglial inflammation, we used chromatin immunoprecipitation to identify SDS3 target genes and combined with transcriptomics and proteomics analysis to explore expression changes in cells following SDS3 knocking down. Subsequently, we validated our findings through experimental assays.

Results: Our analysis revealed that SDS3 modulates the expression of the upstream kinase ASK1 of the p38 MAPK pathway, thus regulating the activation of signaling pathways and ultimately influencing inflammation.

Conclusions: Our findings provide important evidence of the contributions of SDS3 toward microglial inflammation and offer new insights into the regulatory mechanisms of microglial inflammatory responses.

背景:小胶质细胞是中枢神经系统的主要先天性免疫细胞,是神经炎症的关键驱动因素,在神经退行性疾病的发病机制中起着至关重要的作用。Sin3/组蛋白去乙酰化酶(HDAC)复合物是一种高度保守的多蛋白共抑制复合物,主要通过去乙酰化酶活性进行转录抑制;然而,维持该复合物完整性的SDS3在小胶质细胞中的功能仍不清楚:为了揭示转录共抑制因子 SDS3 在小胶质细胞炎症中的调控作用,我们利用染色质免疫沉淀技术鉴定了 SDS3 的靶基因,并结合转录组学和蛋白质组学分析探讨了 SDS3 被敲除后细胞中的表达变化。随后,我们通过实验验证了我们的发现:我们的分析发现,SDS3 可调节 p38 MAPK 通路上游激酶 ASK1 的表达,从而调节信号通路的激活,最终影响炎症:我们的研究结果为 SDS3 对小胶质细胞炎症的贡献提供了重要证据,并为了解小胶质细胞炎症反应的调控机制提供了新的视角。
{"title":"SDS3 regulates microglial inflammation by modulating the expression of the upstream kinase ASK1 in the p38 MAPK signaling pathway.","authors":"Jian Shen, Wenjia Lai, Zeyang Li, Wenyuan Zhu, Xue Bai, Zihao Yang, Qingsong Wang, Jianguo Ji","doi":"10.1007/s00011-024-01913-5","DOIUrl":"10.1007/s00011-024-01913-5","url":null,"abstract":"<p><strong>Background: </strong>Microglia, the main innate immune cells in the central nervous system, are key drivers of neuroinflammation, which plays a crucial role in the pathogenesis of neurodegenerative diseases. The Sin3/histone deacetylase (HDAC) complex, a highly conserved multiprotein co-repressor complex, primarily performs transcriptional repression via deacetylase activity; however, the function of SDS3, which maintains the integrity of the complex, in microglia remains unclear.</p><p><strong>Methods: </strong>To uncover the regulatory role of the transcriptional co-repressor SDS3 in microglial inflammation, we used chromatin immunoprecipitation to identify SDS3 target genes and combined with transcriptomics and proteomics analysis to explore expression changes in cells following SDS3 knocking down. Subsequently, we validated our findings through experimental assays.</p><p><strong>Results: </strong>Our analysis revealed that SDS3 modulates the expression of the upstream kinase ASK1 of the p38 MAPK pathway, thus regulating the activation of signaling pathways and ultimately influencing inflammation.</p><p><strong>Conclusions: </strong>Our findings provide important evidence of the contributions of SDS3 toward microglial inflammation and offer new insights into the regulatory mechanisms of microglial inflammatory responses.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1547-1564"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11349808/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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 emerging role of fibrin(ogen) in cardiovascular disease. 纤维蛋白(原)在心血管疾病中的新作用。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-07-18 DOI: 10.1007/s00011-024-01916-2
Hong Lan, Shengtao Zhao, Yuting Xiong, Xiang-Zhen Yan

Objective: A coagulation factor called fibrinogen is produced by the liver and is proteolyzed by thrombin to become fibrin. The latest studies have revealed that fibrin(ogen) palys an essential role in the regulation of cardiovascular disease. Understanding the relationship and mechanism between fibrin(ogen) and cardiovascular disease is of great significance for maintaining overall health. The objective of this review is to discuss the specific involvement and underlying mechanisms of fibrin(ogen) in cardiovascular disease.

Methods: A review was conducted using the PubMed database to identify and analyze the emerging role of fibrinogen in cardiovascular disease.

Results: The literature review revealed that fibrin(ogen) plays a pivotal role in maintaining cardiovascular disease and are involved in the pathogenesis of cardiovascular disease. Fibrin(ogen) mainly influence various pathophysiological processes, such as participating in thrombosis formation, stimulating the inflammatory response, and other molecular pathways.

Conclusion: This review focuses on the involvement of fibrin(ogen) in cardiovascular disease, with a particular emphasis on the main functions and underlying mechanisms by which fibrin(ogen) influence the pathogenesis and progression of these conditions. This review underscores the potential of fibrin(ogen) as therapeutic targets in managing cardiovascular disease.

目的:一种名为纤维蛋白原的凝血因子由肝脏产生,经凝血酶蛋白水解后成为纤维蛋白。最新研究表明,纤维蛋白原在心血管疾病的调控中发挥着重要作用。了解纤维蛋白(原)与心血管疾病之间的关系和机制对维护整体健康具有重要意义。本综述旨在讨论纤维蛋白(原)在心血管疾病中的具体参与情况和内在机制:方法:使用 PubMed 数据库进行综述,以确定和分析纤维蛋白原在心血管疾病中的新作用:结果:文献综述显示,纤维蛋白原在维持心血管疾病方面起着关键作用,并参与了心血管疾病的发病机制。纤维蛋白原主要影响各种病理生理过程,如参与血栓形成、刺激炎症反应等分子通路:本综述侧重于纤维蛋白(原)参与心血管疾病的研究,特别强调了纤维蛋白(原)影响这些疾病的发病和进展的主要功能和潜在机制。这篇综述强调了纤维蛋白原作为心血管疾病治疗靶点的潜力。
{"title":"The emerging role of fibrin(ogen) in cardiovascular disease.","authors":"Hong Lan, Shengtao Zhao, Yuting Xiong, Xiang-Zhen Yan","doi":"10.1007/s00011-024-01916-2","DOIUrl":"10.1007/s00011-024-01916-2","url":null,"abstract":"<p><strong>Objective: </strong>A coagulation factor called fibrinogen is produced by the liver and is proteolyzed by thrombin to become fibrin. The latest studies have revealed that fibrin(ogen) palys an essential role in the regulation of cardiovascular disease. Understanding the relationship and mechanism between fibrin(ogen) and cardiovascular disease is of great significance for maintaining overall health. The objective of this review is to discuss the specific involvement and underlying mechanisms of fibrin(ogen) in cardiovascular disease.</p><p><strong>Methods: </strong>A review was conducted using the PubMed database to identify and analyze the emerging role of fibrinogen in cardiovascular disease.</p><p><strong>Results: </strong>The literature review revealed that fibrin(ogen) plays a pivotal role in maintaining cardiovascular disease and are involved in the pathogenesis of cardiovascular disease. Fibrin(ogen) mainly influence various pathophysiological processes, such as participating in thrombosis formation, stimulating the inflammatory response, and other molecular pathways.</p><p><strong>Conclusion: </strong>This review focuses on the involvement of fibrin(ogen) in cardiovascular disease, with a particular emphasis on the main functions and underlying mechanisms by which fibrin(ogen) influence the pathogenesis and progression of these conditions. This review underscores the potential of fibrin(ogen) as therapeutic targets in managing cardiovascular disease.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1435-1444"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141633426","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular profiling of core immune-escape genes highlights TNFAIP3 as an immune-related prognostic biomarker in neuroblastoma. 核心免疫排斥基因的分子剖析显示 TNFAIP3 是神经母细胞瘤中与免疫相关的预后生物标志物。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-07-19 DOI: 10.1007/s00011-024-01914-4
Linyu Yang, Kai Huang, Lijian Cao, Yue Ma, Suwen Li, Jianwu Zhou, Zhenzhen Zhao, Shan Wang

Background: Neuroblastoma (NB) is the most prevalent and deadliest pediatric solid tumor. With of over 50% of high-risk neuroblastoma cases relapse, the imperative for novel drug targets and therapeutic strategies is accentuated. In neuroblastoma, the existence of tumor-associated macrophages (TAMs) correlates with an unfavorable patient prognosis. However, the clinical relevance and prognostic implications of regulatory genes linked to TAMs infiltration in neuroblastoma remain unclear, and further study is required.

Methods: We conducted a comprehensive analysis utilizing transcriptome expression profiles from three primary datasets associated with neuroblastoma (GSE45547, GSE49710, TARGET) to identify hub genes implicated in immune evasion within neuroblastoma. Subsequently, we utilized single-cell RNA sequencing analysis on 17 clinical neuroblastoma samples to investigate the expression and distribution of these hub genes, leading to the identification of TNFAIP3. The above three public databases were merged to allowed for the validation of TNFAIP3's molecular functions through GO and KEGG analysis. Furthermore, we assessed TNFAIP3's correlation with immune infiltration and its potential immunotherapeutic impact by multiple algorithms. Our single-cell transcriptome data revealed the role of TNFAIP3 in macrophage polarization. Finally, preliminary experimental verifications to confirm the biological functions of TNFAIP3-mediated TAMs in NB.

Results: A total of 6 genes related to immune evasion were screened and we found that TNFAIP3 exhibited notably higher expression in macrophages than other immune cell types, based on the scRNA-sequencing data. GO and KEGG analysis showed that low expression of TNFAIP3 significantly correlated with the activation of multiple oncogenic pathways as well as immune-related pathways. Then validation affirmed that individuals within the TNFAIP3 high-expression cohort could potentially derive greater advantages from immunotherapeutic interventions, alongside exhibiting heightened immune responsiveness. Deciphering the pseudotime trajectory of macrophages, we revealed the potential of TNFAIP3 in inducing the polarization of macrophages towards the M1 phenotype. Finally, we confirmed that patients in the TNFAIP3 high expression group might benefit more from immunotherapy or chemotherapy as substantiated by RT-qPCR and immunofluorescence examinations. Moreover, the role of TNFAIP3 in macrophage polarization was validated. Preliminary experiment showed that TNFAIP3-mediated TAMs inhibit the proliferation, migration and invasion capabilities of NB cells.

Conclusions: Our results suggest that TNFAIP3 was first identified as a promising biomarker for immunotherapy and potential molecular target in NB. Besides, the presence of TNFAIP3 within TAMs may offer a novel therapeutic strategy for NB.

背景:神经母细胞瘤(NB神经母细胞瘤(NB)是发病率最高、死亡率最高的儿科实体瘤。50%以上的高危神经母细胞瘤病例会复发,因此寻找新的药物靶点和治疗策略变得尤为重要。在神经母细胞瘤中,肿瘤相关巨噬细胞(TAMs)的存在与患者的不良预后相关。然而,与神经母细胞瘤中TAMs浸润相关的调控基因的临床意义和预后影响仍不清楚,需要进一步研究:我们利用与神经母细胞瘤相关的三个主要数据集(GSE45547、GSE49710、TARGET)中的转录组表达谱进行了综合分析,以确定神经母细胞瘤中与免疫逃避相关的枢纽基因。随后,我们利用 17 个临床神经母细胞瘤样本的单细胞 RNA 测序分析,研究了这些枢纽基因的表达和分布,最终确定了 TNFAIP3。合并上述三个公共数据库后,我们通过 GO 和 KEGG 分析验证了 TNFAIP3 的分子功能。此外,我们还通过多种算法评估了 TNFAIP3 与免疫浸润的相关性及其潜在的免疫治疗影响。我们的单细胞转录组数据揭示了 TNFAIP3 在巨噬细胞极化中的作用。最后,通过初步实验验证了TNFAIP3介导的TAMs在NB中的生物学功能:根据scRNA测序数据,我们发现TNFAIP3在巨噬细胞中的表达明显高于其他免疫细胞类型。GO和KEGG分析表明,TNFAIP3的低表达与多种致癌通路以及免疫相关通路的激活密切相关。随后的验证证实,TNFAIP3高表达组群中的个体有可能从免疫治疗干预中获得更大的优势,同时表现出更高的免疫反应性。通过解密巨噬细胞的假时轨迹,我们发现 TNFAIP3 有可能诱导巨噬细胞向 M1 表型极化。最后,我们通过 RT-qPCR 和免疫荧光检查证实,TNFAIP3 高表达组患者可能从免疫疗法或化疗中获益更多。此外,我们还验证了TNFAIP3在巨噬细胞极化中的作用。初步实验表明,TNFAIP3 介导的 TAMs 可抑制 NB 细胞的增殖、迁移和侵袭能力:我们的研究结果表明,TNFAIP3首次被确定为一种有希望的免疫治疗生物标志物和NB的潜在分子靶点。此外,TAMs中TNFAIP3的存在可能为NB提供了一种新的治疗策略。
{"title":"Molecular profiling of core immune-escape genes highlights TNFAIP3 as an immune-related prognostic biomarker in neuroblastoma.","authors":"Linyu Yang, Kai Huang, Lijian Cao, Yue Ma, Suwen Li, Jianwu Zhou, Zhenzhen Zhao, Shan Wang","doi":"10.1007/s00011-024-01914-4","DOIUrl":"10.1007/s00011-024-01914-4","url":null,"abstract":"<p><strong>Background: </strong>Neuroblastoma (NB) is the most prevalent and deadliest pediatric solid tumor. With of over 50% of high-risk neuroblastoma cases relapse, the imperative for novel drug targets and therapeutic strategies is accentuated. In neuroblastoma, the existence of tumor-associated macrophages (TAMs) correlates with an unfavorable patient prognosis. However, the clinical relevance and prognostic implications of regulatory genes linked to TAMs infiltration in neuroblastoma remain unclear, and further study is required.</p><p><strong>Methods: </strong>We conducted a comprehensive analysis utilizing transcriptome expression profiles from three primary datasets associated with neuroblastoma (GSE45547, GSE49710, TARGET) to identify hub genes implicated in immune evasion within neuroblastoma. Subsequently, we utilized single-cell RNA sequencing analysis on 17 clinical neuroblastoma samples to investigate the expression and distribution of these hub genes, leading to the identification of TNFAIP3. The above three public databases were merged to allowed for the validation of TNFAIP3's molecular functions through GO and KEGG analysis. Furthermore, we assessed TNFAIP3's correlation with immune infiltration and its potential immunotherapeutic impact by multiple algorithms. Our single-cell transcriptome data revealed the role of TNFAIP3 in macrophage polarization. Finally, preliminary experimental verifications to confirm the biological functions of TNFAIP3-mediated TAMs in NB.</p><p><strong>Results: </strong>A total of 6 genes related to immune evasion were screened and we found that TNFAIP3 exhibited notably higher expression in macrophages than other immune cell types, based on the scRNA-sequencing data. GO and KEGG analysis showed that low expression of TNFAIP3 significantly correlated with the activation of multiple oncogenic pathways as well as immune-related pathways. Then validation affirmed that individuals within the TNFAIP3 high-expression cohort could potentially derive greater advantages from immunotherapeutic interventions, alongside exhibiting heightened immune responsiveness. Deciphering the pseudotime trajectory of macrophages, we revealed the potential of TNFAIP3 in inducing the polarization of macrophages towards the M1 phenotype. Finally, we confirmed that patients in the TNFAIP3 high expression group might benefit more from immunotherapy or chemotherapy as substantiated by RT-qPCR and immunofluorescence examinations. Moreover, the role of TNFAIP3 in macrophage polarization was validated. Preliminary experiment showed that TNFAIP3-mediated TAMs inhibit the proliferation, migration and invasion capabilities of NB cells.</p><p><strong>Conclusions: </strong>Our results suggest that TNFAIP3 was first identified as a promising biomarker for immunotherapy and potential molecular target in NB. Besides, the presence of TNFAIP3 within TAMs may offer a novel therapeutic strategy for NB.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1529-1545"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141723605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Kinin B1 receptor and TLR4 interaction in inflammatory response. 激肽 B1 受体和 TLR4 在炎症反应中的相互作用
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-07-04 DOI: 10.1007/s00011-024-01909-1
Carolina Batista, João Victor Roza Cruz, Joice Stipursky, Fabio de Almeida Mendes, João Bosco Pesquero

Objective: We aimed to broaden our understanding of a potential interaction between B1R and TLR4, considering earlier studies suggesting that lipopolysaccharide (LPS) may trigger B1R stimulation.

Methods: We assessed the impact of DBK and LPS on the membrane potential of thoracic aortas from C57BL/6, B1R, or TLR4 knockout mice. Additionally, we examined the staining patterns of these receptors in the thoracic aortas of C57BL/6 and in endothelial cells (HBMEC).

Results: DBK does not affect the resting membrane potential of aortic rings in C57BL/6 mice, but it hyperpolarizes preparations in B1KO and TLR4KO mice. The hyperpolarization mechanism in B1KO mice involves B2R, and the TLR4KO response is independent of cytoplasmic calcium influx but relies on potassium channels. Conversely, LPS hyperpolarizes thoracic aorta rings in both C57BL/6 and B1KO mice, with the response unaffected by a B1R antagonist. Interestingly, the absence of B1R alters the LPS response to potassium channels. These activities are independent of nitric oxide synthase (NOS). While exposure to DBK and LPS does not alter B1R and TLR4 mRNA expression, treatment with these agonists increases B1R staining in endothelial cells of thoracic aortic rings and modifies the staining pattern of B1R and TLR4 in endothelial cells. Proximity ligation assay suggests a interaction between the receptors.

Conclusion: Our findings provide additional support for a putative connection between B1R and TLR4 signaling. Given the involvement of these receptors and their agonists in inflammation, it suggests that drugs and therapies targeting their effects could be promising therapeutic avenues worth exploring.

目的考虑到先前的研究表明脂多糖(LPS)可能会触发 B1R 刺激,我们旨在扩大我们对 B1R 和 TLR4 之间潜在相互作用的理解:我们评估了 DBK 和 LPS 对 C57BL/6、B1R 或 TLR4 基因敲除小鼠胸主动脉膜电位的影响。此外,我们还检查了这些受体在 C57BL/6 小鼠主动脉和内皮细胞(HBMEC)中的染色模式:结果:DBK 不影响 C57BL/6 小鼠主动脉环的静息膜电位,但它能使 B1KO 和 TLR4KO 小鼠的制备过度极化。B1KO 小鼠的超极化机制涉及 B2R,而 TLR4KO 的反应与胞质钙离子流入无关,而是依赖于钾通道。相反,LPS 可使 C57BL/6 和 B1KO 小鼠的胸主动脉环超极化,而 B1R 拮抗剂不会影响这种反应。有趣的是,B1R 的缺失会改变 LPS 对钾通道的反应。这些活动与一氧化氮合酶(NOS)无关。虽然暴露于 DBK 和 LPS 不会改变 B1R 和 TLR4 mRNA 的表达,但用这些激动剂处理会增加胸主动脉环内皮细胞中的 B1R 染色,并改变内皮细胞中 B1R 和 TLR4 的染色模式。近接实验表明受体之间存在相互作用:我们的研究结果为 B1R 和 TLR4 信号传导之间的假定联系提供了更多支持。鉴于这些受体及其激动剂在炎症中的参与,这表明针对其效应的药物和疗法可能是值得探索的有前途的治疗途径。
{"title":"Kinin B<sub>1</sub> receptor and TLR4 interaction in inflammatory response.","authors":"Carolina Batista, João Victor Roza Cruz, Joice Stipursky, Fabio de Almeida Mendes, João Bosco Pesquero","doi":"10.1007/s00011-024-01909-1","DOIUrl":"10.1007/s00011-024-01909-1","url":null,"abstract":"<p><strong>Objective: </strong>We aimed to broaden our understanding of a potential interaction between B1R and TLR4, considering earlier studies suggesting that lipopolysaccharide (LPS) may trigger B1R stimulation.</p><p><strong>Methods: </strong>We assessed the impact of DBK and LPS on the membrane potential of thoracic aortas from C57BL/6, B1R, or TLR4 knockout mice. Additionally, we examined the staining patterns of these receptors in the thoracic aortas of C57BL/6 and in endothelial cells (HBMEC).</p><p><strong>Results: </strong>DBK does not affect the resting membrane potential of aortic rings in C57BL/6 mice, but it hyperpolarizes preparations in B<sub>1</sub>KO and TLR4KO mice. The hyperpolarization mechanism in B<sub>1</sub>KO mice involves B2R, and the TLR4KO response is independent of cytoplasmic calcium influx but relies on potassium channels. Conversely, LPS hyperpolarizes thoracic aorta rings in both C57BL/6 and B<sub>1</sub>KO mice, with the response unaffected by a B1R antagonist. Interestingly, the absence of B1R alters the LPS response to potassium channels. These activities are independent of nitric oxide synthase (NOS). While exposure to DBK and LPS does not alter B1R and TLR4 mRNA expression, treatment with these agonists increases B1R staining in endothelial cells of thoracic aortic rings and modifies the staining pattern of B1R and TLR4 in endothelial cells. Proximity ligation assay suggests a interaction between the receptors.</p><p><strong>Conclusion: </strong>Our findings provide additional support for a putative connection between B1R and TLR4 signaling. Given the involvement of these receptors and their agonists in inflammation, it suggests that drugs and therapies targeting their effects could be promising therapeutic avenues worth exploring.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1459-1476"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141534370","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TRIM25-mediated XRCC1 ubiquitination accelerates atherosclerosis by inducing macrophage M1 polarization and programmed death. TRIM25 介导的 XRCC1 泛素化通过诱导巨噬细胞 M1 极化和程序性死亡加速动脉粥样硬化。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-06-19 DOI: 10.1007/s00011-024-01906-4
Hongxian Wu, Wei Gao, Yuanji Ma, Xin Zhong, Juying Qian, Dong Huang, Junbo Ge
<p><strong>Background: </strong>Macrophage-mediated cleaning up of dead cells is a crucial determinant in reducing coronary artery inflammation and maintaining vascular homeostasis. However, this process also leads to programmed death of macrophages. So far, the role of macrophage death in the progression of atherosclerosis remains controversial. Also, the underlying mechanism by which transcriptional regulation and reprogramming triggered by macrophage death pathways lead to changes in vascular inflammation and remodeling are still largely unknown. TRIM25-mediated RIG-I signaling plays a key role in regulation of macrophages fate, however the role of TRIM25 in macrophage death-mediated atherosclerotic progression remains unclear. This study aims to investigate the relationship between TRIM25 and macrophage death in atherosclerosis.</p><p><strong>Methods: </strong>A total of 34 blood samples of patients with coronary stent implantation, including chronic total occlusion (CTO) leisions (n = 14) or with more than 50% stenosis of a coronary artery but without CTO leisions (n = 20), were collected, and the serum level of TRIM25 was detected by ELISA. Apoe<sup>-/-</sup> mice with or without TRIM25 gene deletion were fed with the high-fat diet (HFD) for 12 weeks and the plaque areas, necrotic core size, aortic fibrosis and inflammation were investigated. TRIM25 wild-type and deficient macrophages were isolated, cultured and stimulated with ox-LDL, RNA-seq, real-time PCR, western blot and FACS experiments were used to screen and validate signaling pathways caused by TRIM25 deletion.</p><p><strong>Results: </strong>Downregulation of TRIM25 was observed in circulating blood of CTO patients and also in HFD-induced mouse aortas. After HFD for 12 weeks, TRIM25<sup>-/-</sup>ApoeE<sup>-/-</sup> mice developed smaller atherosclerotic plaques, less inflammation, lower collagen content and aortic fibrosis compared with TRIM25<sup>+/+</sup>ApoeE<sup>-/-</sup> mice. By RNA-seq and KEGG enrichment analysis, we revealed that deletion of TRIM25 mainly affected pyroptosis and necroptosis pathways in ox-LDL-induced macrophages, and the expressions of PARP1 and RIPK3, were significantly decreased in TRIM25 deficient macrophages. Overexpression of TRIM25 promoted M1 polarization and necroptosis of macrophages, while inhibition of PARP1 reversed this process. Further, we observed that XRCC1, a repairer of DNA damage, was significantly upregulated in TRIM25 deficient macrophages, inhibiting PARP1 activity and PARP1-mediated pro-inflammatory change, M1 polarization and necroptosis of macrophages. By contrast, TRIM25 overexpression mediated ubiquitination of XRCC1, and the inhibition of XRCC1 released PARP1, and activated macrophage M1 polarization and necroptosis, which accelerated aortic inflammation and atherosclerotic plaque progression.</p><p><strong>Conclusions: </strong>Our study has uncovered a crucial role of the TRIM25-XRCC1<sup>Ub</sup>-PARP1-RIPK3 axis in regulati
背景:巨噬细胞介导的死亡细胞清理是减轻冠状动脉炎症和维持血管平衡的关键因素。然而,这一过程也会导致巨噬细胞的程序性死亡。迄今为止,巨噬细胞死亡在动脉粥样硬化进展过程中的作用仍存在争议。此外,由巨噬细胞死亡途径引发的转录调控和重编程导致血管炎症和重塑变化的潜在机制在很大程度上仍不为人所知。TRIM25 介导的 RIG-I 信号在调控巨噬细胞命运中发挥着关键作用,但 TRIM25 在巨噬细胞死亡介导的动脉粥样硬化进展中的作用仍不清楚。本研究旨在探讨TRIM25与动脉粥样硬化中巨噬细胞死亡之间的关系:方法:收集34例冠状动脉支架植入患者的血样,包括慢性全闭塞(CTO)左冠状动脉(14例)或冠状动脉狭窄超过50%但无CTO左冠状动脉(20例)的患者,用ELISA法检测血清中TRIM25的水平。用高脂饮食(HFD)喂养缺失或未缺失TRIM25基因的载脂蛋白/-小鼠12周,并调查斑块面积、坏死核心大小、主动脉纤维化和炎症情况。分离、培养TRIM25野生型和缺失型巨噬细胞并用ox-LDL刺激,通过RNA-seq、实时PCR、Western印迹和FACS实验筛选和验证TRIM25基因缺失导致的信号通路:结果:TRIM25在CTO患者的循环血液和HFD诱导的小鼠主动脉中均出现下调。与 TRIM25+/+ApoeE-/- 小鼠相比,高频分解膳食 12 周后,TRIM25-/-ApoeE-/- 小鼠的动脉粥样硬化斑块更小、炎症更少、胶原蛋白含量更低且主动脉纤维化程度更轻。通过RNA-seq和KEGG富集分析,我们发现缺失TRIM25主要影响ox-LDL诱导的巨噬细胞中的热凋亡和坏死通路,缺失TRIM25的巨噬细胞中PARP1和RIPK3的表达显著下降。过表达 TRIM25 可促进巨噬细胞的 M1 极化和坏死,而抑制 PARP1 则可逆转这一过程。此外,我们还观察到,DNA损伤修复者XRCC1在TRIM25缺陷的巨噬细胞中显著上调,抑制了PARP1的活性和PARP1介导的巨噬细胞促炎变化、M1极化和坏死。相比之下,TRIM25过表达介导XRCC1泛素化,抑制XRCC1释放PARP1,激活巨噬细胞M1极化和坏死,从而加速主动脉炎症和动脉粥样硬化斑块的进展:我们的研究揭示了TRIM25-XRCC1Ub-PARP1-RIPK3轴在动脉粥样硬化过程中调控巨噬细胞死亡的关键作用,并强调了巨噬细胞重编程调控在预防动脉粥样硬化发展中的潜在治疗意义。
{"title":"TRIM25-mediated XRCC1 ubiquitination accelerates atherosclerosis by inducing macrophage M1 polarization and programmed death.","authors":"Hongxian Wu, Wei Gao, Yuanji Ma, Xin Zhong, Juying Qian, Dong Huang, Junbo Ge","doi":"10.1007/s00011-024-01906-4","DOIUrl":"10.1007/s00011-024-01906-4","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Macrophage-mediated cleaning up of dead cells is a crucial determinant in reducing coronary artery inflammation and maintaining vascular homeostasis. However, this process also leads to programmed death of macrophages. So far, the role of macrophage death in the progression of atherosclerosis remains controversial. Also, the underlying mechanism by which transcriptional regulation and reprogramming triggered by macrophage death pathways lead to changes in vascular inflammation and remodeling are still largely unknown. TRIM25-mediated RIG-I signaling plays a key role in regulation of macrophages fate, however the role of TRIM25 in macrophage death-mediated atherosclerotic progression remains unclear. This study aims to investigate the relationship between TRIM25 and macrophage death in atherosclerosis.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;A total of 34 blood samples of patients with coronary stent implantation, including chronic total occlusion (CTO) leisions (n = 14) or with more than 50% stenosis of a coronary artery but without CTO leisions (n = 20), were collected, and the serum level of TRIM25 was detected by ELISA. Apoe&lt;sup&gt;-/-&lt;/sup&gt; mice with or without TRIM25 gene deletion were fed with the high-fat diet (HFD) for 12 weeks and the plaque areas, necrotic core size, aortic fibrosis and inflammation were investigated. TRIM25 wild-type and deficient macrophages were isolated, cultured and stimulated with ox-LDL, RNA-seq, real-time PCR, western blot and FACS experiments were used to screen and validate signaling pathways caused by TRIM25 deletion.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Downregulation of TRIM25 was observed in circulating blood of CTO patients and also in HFD-induced mouse aortas. After HFD for 12 weeks, TRIM25&lt;sup&gt;-/-&lt;/sup&gt;ApoeE&lt;sup&gt;-/-&lt;/sup&gt; mice developed smaller atherosclerotic plaques, less inflammation, lower collagen content and aortic fibrosis compared with TRIM25&lt;sup&gt;+/+&lt;/sup&gt;ApoeE&lt;sup&gt;-/-&lt;/sup&gt; mice. By RNA-seq and KEGG enrichment analysis, we revealed that deletion of TRIM25 mainly affected pyroptosis and necroptosis pathways in ox-LDL-induced macrophages, and the expressions of PARP1 and RIPK3, were significantly decreased in TRIM25 deficient macrophages. Overexpression of TRIM25 promoted M1 polarization and necroptosis of macrophages, while inhibition of PARP1 reversed this process. Further, we observed that XRCC1, a repairer of DNA damage, was significantly upregulated in TRIM25 deficient macrophages, inhibiting PARP1 activity and PARP1-mediated pro-inflammatory change, M1 polarization and necroptosis of macrophages. By contrast, TRIM25 overexpression mediated ubiquitination of XRCC1, and the inhibition of XRCC1 released PARP1, and activated macrophage M1 polarization and necroptosis, which accelerated aortic inflammation and atherosclerotic plaque progression.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;Our study has uncovered a crucial role of the TRIM25-XRCC1&lt;sup&gt;Ub&lt;/sup&gt;-PARP1-RIPK3 axis in regulati","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1445-1458"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141418788","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Alpha-galactosylceramide pre-treatment attenuates clinical symptoms of LPS-induced acute neuroinflammation by converting pathogenic iNKT cells to anti-inflammatory iNKT10 cells in the brain. α-半乳糖甘油酰胺预处理通过将脑内致病性 iNKT 细胞转化为抗炎性 iNKT10 细胞,减轻 LPS 诱导的急性神经炎症的临床症状。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-09-01 Epub Date: 2024-07-19 DOI: 10.1007/s00011-024-01915-3
Tae-Cheol Kim, Hyun Jung Park, Sung Won Lee, Yun Hoo Park, Luc Van Kaer, Seokmann Hong

Background: Invariant natural killer T (iNKT) cells play protective or pathogenic roles in a variety of immune and inflammatory diseases. However, whether iNKT cells contribute to the progression of acute neuroinflammation remains unclear. Thus, we addressed this question with a mouse model of lipopolysaccharide (LPS)-induced acute neuroinflammation.

Methods: For induction of acute neuroinflammation, wild-type (WT) C57BL/6 (B6) mice were injected intraperitoneally (i.p.) with LPS for either three or five consecutive days, and then these mice were analyzed for brain-infiltrating leukocytes or mouse behaviors, respectively. To examine the role of iNKT cell activation in LPS-induced neuroinflammation, mice were injected i.p. with the iNKT cell agonist α-galactosylceramide (α-GalCer) seven days prior to LPS treatment. Immune cells infiltrated into the brain during LPS-induced neuroinflammation were determined by flow cytometry. In addition, LPS-induced clinical behavior symptoms such as depressive-like behavior and memory impairment in mice were evaluated by the open field and Y-maze tests, respectively.

Results: We found that iNKT cell-deficient Jα18 mutant mice display delayed disease progression and decreased leukocyte infiltration into the brain compared with WT mice, indicating that iNKT cells contribute to the pathogenesis of LPS-induced neuroinflammation. Since it has been reported that pre-treatment with α-GalCer, an iNKT cell agonist, can convert iNKT cells towards anti-inflammatory phenotypes, we next explored whether pre-activation of iNKT cells with α-GalCer can regulate LPS-induced neuroinflammation. Strikingly, we found that α-GalCer pre-treatment significantly delays the onset of clinical symptoms, including depression-like behavior and memory impairment, while decreasing brain infiltration of pro-inflammatory natural killer cells and neutrophils, in this model of LPS-induced neuroinflammation. Such anti-inflammatory effects of α-GalCer pre-treatment closely correlated with iNKT cell polarization towards IL4- and IL10-producing phenotypes. Furthermore, α-GalCer pre-treatment restored the expression of suppressive markers on brain regulatory T cells during LPS-induced neuroinflammation.

Conclusion: Our findings provide strong evidence that α-GalCer-induced pre-activation of iNKT cells expands iNKT10 cells, mitigating depressive-like behaviors and brain infiltration of inflammatory immune cells induced by LPS-induced acute neuroinflammation. Thus, we suggest the prophylactic potential of iNKT cells and α-GalCer against acute neuroinflammation.

背景:不变自然杀伤 T 细胞(iNKT)在多种免疫和炎症疾病中发挥保护或致病作用。然而,iNKT 细胞是否有助于急性神经炎症的进展仍不清楚。因此,我们通过脂多糖(LPS)诱导急性神经炎症的小鼠模型来解决这个问题:为了诱导急性神经炎症,野生型(WT)C57BL/6(B6)小鼠腹腔注射(i.p.)LPS,连续注射三天或五天,然后分别分析这些小鼠的脑浸润白细胞或小鼠行为。为了研究 iNKT 细胞活化在 LPS 诱导的神经炎症中的作用,在 LPS 治疗前七天,给小鼠静脉注射 iNKT 细胞激动剂 α-半乳糖甘油酰胺(α-GalCer)。流式细胞术测定了在LPS诱导的神经炎症过程中渗入大脑的免疫细胞。此外,LPS诱导的小鼠临床表现症状,如抑郁样行为和记忆损伤,分别通过开阔地试验和Y迷宫试验进行了评估:结果:我们发现,与 WT 小鼠相比,iNKT 细胞缺失的 Jα18 突变小鼠的疾病进展延迟,脑内白细胞浸润减少,这表明 iNKT 细胞有助于 LPS 诱导的神经炎症的发病机制。据报道,用 iNKT 细胞激动剂 α-GalCer 预处理可使 iNKT 细胞向抗炎表型转化,因此我们接下来探讨了用α-GalCer 预激活 iNKT 细胞是否能调节 LPS 诱导的神经炎症。令人震惊的是,我们发现在这种 LPS 诱导的神经炎症模型中,α-GalCer 预处理能显著延缓抑郁样行为和记忆损伤等临床症状的出现,同时减少促炎性自然杀伤细胞和中性粒细胞在大脑中的浸润。α-GalCer预处理的这种抗炎作用与iNKT细胞向产生IL4和IL10表型的极化密切相关。此外,在 LPS 诱导的神经炎症中,α-GalCer 预处理可恢复脑调节性 T 细胞抑制性标记物的表达:我们的研究结果提供了强有力的证据,证明α-GalCer诱导的iNKT细胞预激活能扩增iNKT10细胞,减轻LPS诱导的急性神经炎症引起的抑郁样行为和脑内炎性免疫细胞的浸润。因此,我们认为 iNKT 细胞和 α-GalCer 对急性神经炎症具有预防潜力。
{"title":"Alpha-galactosylceramide pre-treatment attenuates clinical symptoms of LPS-induced acute neuroinflammation by converting pathogenic iNKT cells to anti-inflammatory iNKT10 cells in the brain.","authors":"Tae-Cheol Kim, Hyun Jung Park, Sung Won Lee, Yun Hoo Park, Luc Van Kaer, Seokmann Hong","doi":"10.1007/s00011-024-01915-3","DOIUrl":"10.1007/s00011-024-01915-3","url":null,"abstract":"<p><strong>Background: </strong>Invariant natural killer T (iNKT) cells play protective or pathogenic roles in a variety of immune and inflammatory diseases. However, whether iNKT cells contribute to the progression of acute neuroinflammation remains unclear. Thus, we addressed this question with a mouse model of lipopolysaccharide (LPS)-induced acute neuroinflammation.</p><p><strong>Methods: </strong>For induction of acute neuroinflammation, wild-type (WT) C57BL/6 (B6) mice were injected intraperitoneally (i.p.) with LPS for either three or five consecutive days, and then these mice were analyzed for brain-infiltrating leukocytes or mouse behaviors, respectively. To examine the role of iNKT cell activation in LPS-induced neuroinflammation, mice were injected i.p. with the iNKT cell agonist α-galactosylceramide (α-GalCer) seven days prior to LPS treatment. Immune cells infiltrated into the brain during LPS-induced neuroinflammation were determined by flow cytometry. In addition, LPS-induced clinical behavior symptoms such as depressive-like behavior and memory impairment in mice were evaluated by the open field and Y-maze tests, respectively.</p><p><strong>Results: </strong>We found that iNKT cell-deficient Jα18 mutant mice display delayed disease progression and decreased leukocyte infiltration into the brain compared with WT mice, indicating that iNKT cells contribute to the pathogenesis of LPS-induced neuroinflammation. Since it has been reported that pre-treatment with α-GalCer, an iNKT cell agonist, can convert iNKT cells towards anti-inflammatory phenotypes, we next explored whether pre-activation of iNKT cells with α-GalCer can regulate LPS-induced neuroinflammation. Strikingly, we found that α-GalCer pre-treatment significantly delays the onset of clinical symptoms, including depression-like behavior and memory impairment, while decreasing brain infiltration of pro-inflammatory natural killer cells and neutrophils, in this model of LPS-induced neuroinflammation. Such anti-inflammatory effects of α-GalCer pre-treatment closely correlated with iNKT cell polarization towards IL4- and IL10-producing phenotypes. Furthermore, α-GalCer pre-treatment restored the expression of suppressive markers on brain regulatory T cells during LPS-induced neuroinflammation.</p><p><strong>Conclusion: </strong>Our findings provide strong evidence that α-GalCer-induced pre-activation of iNKT cells expands iNKT10 cells, mitigating depressive-like behaviors and brain infiltration of inflammatory immune cells induced by LPS-induced acute neuroinflammation. Thus, we suggest the prophylactic potential of iNKT cells and α-GalCer against acute neuroinflammation.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1511-1527"},"PeriodicalIF":4.8,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141723604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A synthetic bioactive peptide of the C-terminal fragment of adhesion protein Fibulin7 attenuates the inflammatory functions of innate immune cells in LPS-induced systemic inflammation. 粘附蛋白 Fibulin7 C 端片段的合成生物活性肽可减轻 LPS 诱导的全身炎症中先天性免疫细胞的炎症功能。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-05 DOI: 10.1007/s00011-024-01903-7
Saloni Gupta, Nibedita Dalpati, Shubham Kumar Rai, Amit Sehrawat, Venkatesh Pai, Pranita P Sarangi

Objective: Systemic inflammation is associated with improper localization of hyperactive neutrophils and monocytes in visceral organs. Previously, a C-terminal fragment of adhesion protein Fibulin7 (Fbln7-C) was shown to regulate innate immune functionality during inflammation. Recently, a shorter bioactive peptide of Fbln7-C, FC-10, via integrin binding was shown to reduce ocular angiogenesis. However, the role of FC-10 in regulating the neutrophils and monocyte functionality during systemic inflammatory conditions is unknown. The study sought to explore the role of FC-10 peptide on the functionality of innate immune cells during inflammation and endotoxemic mice.

Methods: Neutrophils and monocytes were isolated from healthy donors and septic patient clinical samples and Cell adhesion assay was performed using a UV spectrophotometer. Gene expression studies were performed using qPCR. Protein level expression was measured using ELISA and flow cytometry. ROS assay, and activation markers analysis in vitro, and in vivo were done using flow cytometry.

Treatment: Cells were stimulated with LPS (100 ng/mL) and studied in the presence of peptides (10 μg, and 20 μg/mL) in vitro. In an in vivo study, mice were administered with LPS (36.8 mg/kg bw) and peptide (20 μg).

Results: This study demonstrates that human neutrophils and monocytes adhere to FC-10 via integrin β1, inhibit spreading, ROS, surface activation markers (CD44, CD69), phosphorylated Src kinase, pro-inflammatory genes, and protein expression, compared to scrambled peptide in cells isolated from healthy donors and clinical sample. In line with the in vitro data, FC-10 (20 μg) administration significantly decreases innate cell infiltration at inflammatory sites, improves survival in endotoxemia animals & reduces the inflammatory properties of neutrophils and monocytes isolated from septic patients.

Conclusion: FC-10 peptide can regulate neutrophils and monocyte functions and has potential to be used as an immunomodulatory therapeutic in inflammatory diseases.

目的:全身性炎症与嗜中性粒细胞和单核细胞在内脏器官中的不当定位有关。此前,粘附蛋白 Fibulin7(Fbln7-C)的 C 端片段被证明能在炎症期间调节先天性免疫功能。最近,Fbln7-C 的一种较短的生物活性肽 FC-10 通过整合素结合被证明能减少眼部血管生成。然而,FC-10 在全身炎症条件下调节中性粒细胞和单核细胞功能的作用尚不清楚。本研究试图探索 FC-10 肽在炎症和内毒素血症小鼠体内对先天性免疫细胞功能的作用:方法:从健康供体和败血症患者的临床样本中分离出中性粒细胞和单核细胞,使用紫外分光光度计进行细胞粘附测定。使用 qPCR 进行基因表达研究。使用 ELISA 和流式细胞仪测量蛋白质水平表达。使用流式细胞仪进行 ROS 检测和体内外活化标志物分析:用 LPS(100 毫微克/毫升)刺激细胞,并在多肽(10 微克和 20 微克/毫升)存在的情况下进行体外研究。在体内研究中,给小鼠注射了 LPS(36.8 毫克/千克体重)和多肽(20 微克):本研究表明,在从健康供体和临床样本中分离出的细胞中,人中性粒细胞和单核细胞通过整合素β1粘附在FC-10上,与干扰肽相比,FC-10能抑制扩散、ROS、表面活化标志物(CD44、CD69)、磷酸化Src激酶、促炎基因和蛋白质表达。与体外数据一致,服用 FC-10(20 μg)能显著减少炎症部位的先天性细胞浸润,提高内毒素血症动物的存活率,并降低从脓毒症患者体内分离出的中性粒细胞和单核细胞的炎症特性:结论:FC-10 肽能调节中性粒细胞和单核细胞的功能,有望用作炎症性疾病的免疫调节疗法。
{"title":"A synthetic bioactive peptide of the C-terminal fragment of adhesion protein Fibulin7 attenuates the inflammatory functions of innate immune cells in LPS-induced systemic inflammation.","authors":"Saloni Gupta, Nibedita Dalpati, Shubham Kumar Rai, Amit Sehrawat, Venkatesh Pai, Pranita P Sarangi","doi":"10.1007/s00011-024-01903-7","DOIUrl":"10.1007/s00011-024-01903-7","url":null,"abstract":"<p><strong>Objective: </strong>Systemic inflammation is associated with improper localization of hyperactive neutrophils and monocytes in visceral organs. Previously, a C-terminal fragment of adhesion protein Fibulin7 (Fbln7-C) was shown to regulate innate immune functionality during inflammation. Recently, a shorter bioactive peptide of Fbln7-C, FC-10, via integrin binding was shown to reduce ocular angiogenesis. However, the role of FC-10 in regulating the neutrophils and monocyte functionality during systemic inflammatory conditions is unknown. The study sought to explore the role of FC-10 peptide on the functionality of innate immune cells during inflammation and endotoxemic mice.</p><p><strong>Methods: </strong>Neutrophils and monocytes were isolated from healthy donors and septic patient clinical samples and Cell adhesion assay was performed using a UV spectrophotometer. Gene expression studies were performed using qPCR. Protein level expression was measured using ELISA and flow cytometry. ROS assay, and activation markers analysis in vitro, and in vivo were done using flow cytometry.</p><p><strong>Treatment: </strong>Cells were stimulated with LPS (100 ng/mL) and studied in the presence of peptides (10 μg, and 20 μg/mL) in vitro. In an in vivo study, mice were administered with LPS (36.8 mg/kg bw) and peptide (20 μg).</p><p><strong>Results: </strong>This study demonstrates that human neutrophils and monocytes adhere to FC-10 via integrin β1, inhibit spreading, ROS, surface activation markers (CD44, CD69), phosphorylated Src kinase, pro-inflammatory genes, and protein expression, compared to scrambled peptide in cells isolated from healthy donors and clinical sample. In line with the in vitro data, FC-10 (20 μg) administration significantly decreases innate cell infiltration at inflammatory sites, improves survival in endotoxemia animals & reduces the inflammatory properties of neutrophils and monocytes isolated from septic patients.</p><p><strong>Conclusion: </strong>FC-10 peptide can regulate neutrophils and monocyte functions and has potential to be used as an immunomodulatory therapeutic in inflammatory diseases.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1333-1348"},"PeriodicalIF":4.8,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141247812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prostacyclin synthase deficiency exacerbates systemic inflammatory responses in lipopolysaccharide-induced septic shock in mice. 前列环素合成酶缺乏症会加剧脂多糖诱发小鼠脓毒性休克的全身炎症反应。
IF 4.8 3区 医学 Q2 CELL BIOLOGY Pub Date : 2024-08-01 Epub Date: 2024-06-04 DOI: 10.1007/s00011-024-01902-8
Tsubasa Ochiai, Toshiya Honsawa, Keishi Yamaguchi, Yuka Sasaki, Chieko Yokoyama, Hiroshi Kuwata, Shuntaro Hara

Objectives: Sepsis is a systemic inflammatory disorder characterized by life-threateningorgan dysfunction resulting from a dysregulated host response to infection. Prostacyclin (PGI2) is a bioactive lipid produced by PGI synthase (PGIS) and is known to play important roles in inflammatory reactions as well as cardiovascular regulation. However, little is known about the roles of PGIS and PGI2 in systemic inflammatory responses such as septic shock.

Methodology: Systemic inflammation was induced by intraperitoneal injection of 5 mg/kg lipopolysaccharide (LPS) in wild type (WT) or PGIS knockout (KO) mice. Selexipag, a selective PGI2 receptor (IP) agonist, was administered 2 h before LPS injection and again given every 12 h for 3 days.

Results: Intraperitoneal injection of LPS induced diarrhea, shivering and hypothermia. These symptoms were more severe in PGIS KO mice than in WT micqe. The expression of Tnf and Il6 genes was notably increased in PGIS KO mice. In contrast, over 95% of WT mice survived 72 h after the administration of LPS, whereas all of the PGIS KO mice had succumbed by that time. The mortality rate of LPS-administrated PGIS KO mice was improved by selexipag administration.

Conclusion: Our study suggests that PGIS-derived PGI2 negatively regulates LPS-induced symptoms via the IP receptor. PGIS-derived PGI2-IP signaling axis may be a new drug target for systemic inflammation in septic shock.

目的:败血症是一种全身性炎症性疾病,其特点是宿主对感染的反应失调导致器官功能障碍,危及生命。前列环素(PGI2)是由前列腺素合成酶(PGIS)产生的一种生物活性脂质,在炎症反应和心血管调节中发挥着重要作用。然而,人们对 PGIS 和 PGI2 在脓毒性休克等全身炎症反应中的作用知之甚少:方法:通过向野生型(WT)或 PGIS 基因敲除(KO)小鼠腹腔注射 5 mg/kg 脂多糖(LPS)诱导全身炎症反应。在注射 LPS 前 2 小时给小鼠注射选择性 PGI2 受体(IP)激动剂 Selexipag,然后每隔 12 小时再注射一次,连续注射 3 天:结果:腹腔注射 LPS 会导致腹泻、颤抖和体温过低。与 WT 小鼠相比,PGIS KO 小鼠的这些症状更为严重。PGIS KO小鼠的Tnf和Il6基因表达明显增加。相反,95%以上的 WT 小鼠在注射 LPS 72 小时后存活下来,而 PGIS KO 小鼠则全部死亡。服用 selexipag 后,服用 LPS 的 PGIS KO 小鼠的死亡率有所改善:我们的研究表明,PGIS 衍生的 PGI2 可通过 IP 受体负向调节 LPS 诱导的症状。结论:我们的研究表明,PGIS衍生的PGI2通过IP受体负向调节LPS诱导的症状,PGIS衍生的PGI2-IP信号轴可能是治疗脓毒性休克全身炎症的新药物靶点。
{"title":"Prostacyclin synthase deficiency exacerbates systemic inflammatory responses in lipopolysaccharide-induced septic shock in mice.","authors":"Tsubasa Ochiai, Toshiya Honsawa, Keishi Yamaguchi, Yuka Sasaki, Chieko Yokoyama, Hiroshi Kuwata, Shuntaro Hara","doi":"10.1007/s00011-024-01902-8","DOIUrl":"10.1007/s00011-024-01902-8","url":null,"abstract":"<p><strong>Objectives: </strong>Sepsis is a systemic inflammatory disorder characterized by life-threateningorgan dysfunction resulting from a dysregulated host response to infection. Prostacyclin (PGI<sub>2</sub>) is a bioactive lipid produced by PGI synthase (PGIS) and is known to play important roles in inflammatory reactions as well as cardiovascular regulation. However, little is known about the roles of PGIS and PGI<sub>2</sub> in systemic inflammatory responses such as septic shock.</p><p><strong>Methodology: </strong>Systemic inflammation was induced by intraperitoneal injection of 5 mg/kg lipopolysaccharide (LPS) in wild type (WT) or PGIS knockout (KO) mice. Selexipag, a selective PGI<sub>2</sub> receptor (IP) agonist, was administered 2 h before LPS injection and again given every 12 h for 3 days.</p><p><strong>Results: </strong>Intraperitoneal injection of LPS induced diarrhea, shivering and hypothermia. These symptoms were more severe in PGIS KO mice than in WT micqe. The expression of Tnf and Il6 genes was notably increased in PGIS KO mice. In contrast, over 95% of WT mice survived 72 h after the administration of LPS, whereas all of the PGIS KO mice had succumbed by that time. The mortality rate of LPS-administrated PGIS KO mice was improved by selexipag administration.</p><p><strong>Conclusion: </strong>Our study suggests that PGIS-derived PGI<sub>2</sub> negatively regulates LPS-induced symptoms via the IP receptor. PGIS-derived PGI<sub>2</sub>-IP signaling axis may be a new drug target for systemic inflammation in septic shock.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":" ","pages":"1349-1358"},"PeriodicalIF":4.8,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141237236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Inflammation Research
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