首页 > 最新文献

Cellular &Molecular Immunology最新文献

英文 中文
Neutrophils disrupt B-1a cell homeostasis by targeting Siglec-G to exacerbate sepsis 中性粒细胞通过靶向 Siglec-G 破坏 B-1a 细胞稳态,从而加剧败血症。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-24 DOI: 10.1038/s41423-024-01165-7
Chuyi Tan, Bridgette Reilly, Gaifeng Ma, Atsushi Murao, Alok Jha, Monowar Aziz, Ping Wang
B-1a cells, an innate-like cell population, are crucial for pathogen defense and the regulation of inflammation through their release of natural IgM and IL-10. In sepsis, B-1a cell numbers are decreased in the peritoneal cavity as they robustly migrate to the spleen. Within the spleen, migrating B-1a cells differentiate into plasma cells, leading to alterations in their original phenotype and functionality. We discovered a key player, sialic acid-binding immunoglobulin-like lectin-G (Siglec-G), which is expressed predominantly on B-1a cells and negatively regulates B-1a cell migration to maintain homeostasis. Siglec-G interacts with CXCR4/CXCL12 to modulate B-1a cell migration. Neutrophils aid B-1a cell migration via neutrophil elastase (NE)-mediated Siglec-G cleavage. Human studies revealed increased NE expression in septic patients. We identified an NE cleavage sequence in silico, leading to the discovery of a decoy peptide that protects Siglec-G, preserves peritoneal B-1a cells, reduces inflammation, and enhances sepsis survival. The role of Siglec-G in inhibiting B-1a cell migration to maintain their inherent phenotype and function is compromised by NE in sepsis, offering valuable insights into B-1a cell homeostasis. Employing a small decoy peptide to prevent NE-mediated Siglec-G cleavage has emerged as a promising strategy to sustain peritoneal B-1a cell homeostasis, alleviate inflammation, and ultimately improve outcomes in sepsis patients.
B-1a 细胞是一种先天性类细胞群,通过释放天然 IgM 和 IL-10 对病原体防御和炎症调节至关重要。在败血症时,腹腔中的 B-1a 细胞数量会减少,因为它们会大量迁移到脾脏。在脾脏内,迁移的 B-1a 细胞分化成浆细胞,导致其原有的表型和功能发生改变。我们发现了一种关键的作用因子--唾液酸结合免疫球蛋白样凝集素-G(Siglec-G),它主要在B-1a细胞上表达,负向调节B-1a细胞的迁移以维持体内平衡。Siglec-G 与 CXCR4/CXCL12 相互作用,调节 B-1a 细胞的迁移。中性粒细胞通过中性粒细胞弹性蛋白酶(NE)介导的 Siglec-G 裂解来帮助 B-1a 细胞迁移。人体研究显示,脓毒症患者体内 NE 表达增加。我们在硅学中确定了 NE 的裂解序列,从而发现了一种诱饵肽,它能保护 Siglec-G、保护腹膜 B-1a 细胞、减轻炎症反应并提高脓毒症患者的存活率。Siglec-G在抑制B-1a细胞迁移以维持其固有表型和功能方面的作用在脓毒症中会受到NE的损害,这为了解B-1a细胞的稳态提供了宝贵的信息。采用一种小型诱饵肽来阻止 NE 介导的 Siglec-G 裂解,已成为一种有希望维持腹膜 B-1a 细胞稳态、缓解炎症并最终改善脓毒症患者预后的策略。
{"title":"Neutrophils disrupt B-1a cell homeostasis by targeting Siglec-G to exacerbate sepsis","authors":"Chuyi Tan, Bridgette Reilly, Gaifeng Ma, Atsushi Murao, Alok Jha, Monowar Aziz, Ping Wang","doi":"10.1038/s41423-024-01165-7","DOIUrl":"10.1038/s41423-024-01165-7","url":null,"abstract":"B-1a cells, an innate-like cell population, are crucial for pathogen defense and the regulation of inflammation through their release of natural IgM and IL-10. In sepsis, B-1a cell numbers are decreased in the peritoneal cavity as they robustly migrate to the spleen. Within the spleen, migrating B-1a cells differentiate into plasma cells, leading to alterations in their original phenotype and functionality. We discovered a key player, sialic acid-binding immunoglobulin-like lectin-G (Siglec-G), which is expressed predominantly on B-1a cells and negatively regulates B-1a cell migration to maintain homeostasis. Siglec-G interacts with CXCR4/CXCL12 to modulate B-1a cell migration. Neutrophils aid B-1a cell migration via neutrophil elastase (NE)-mediated Siglec-G cleavage. Human studies revealed increased NE expression in septic patients. We identified an NE cleavage sequence in silico, leading to the discovery of a decoy peptide that protects Siglec-G, preserves peritoneal B-1a cells, reduces inflammation, and enhances sepsis survival. The role of Siglec-G in inhibiting B-1a cell migration to maintain their inherent phenotype and function is compromised by NE in sepsis, offering valuable insights into B-1a cell homeostasis. Employing a small decoy peptide to prevent NE-mediated Siglec-G cleavage has emerged as a promising strategy to sustain peritoneal B-1a cell homeostasis, alleviate inflammation, and ultimately improve outcomes in sepsis patients.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141092447","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bridging the gap with multispecific immune cell engagers in cancer and infectious diseases 利用多特异性免疫细胞参与剂弥合癌症和传染病领域的差距。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-24 DOI: 10.1038/s41423-024-01176-4
Camille Rolin, Jacques Zimmer, Carole Seguin-Devaux
By binding to multiple antigens simultaneously, multispecific antibodies are expected to substantially improve both the activity and long-term efficacy of antibody-based immunotherapy. Immune cell engagers, a subclass of antibody-based constructs, consist of engineered structures designed to bridge immune effector cells to their target, thereby redirecting the immune response toward the tumor cells or infected cells. The increasing number of recent clinical trials evaluating immune cell engagers reflects the important role of these molecules in new therapeutic approaches for cancer and infections. In this review, we discuss how different immune cell types (T and natural killer lymphocytes, as well as myeloid cells) can be bound by immune cell engagers in immunotherapy for cancer and infectious diseases. Furthermore, we explore the preclinical and clinical advancements of these constructs, and we discuss the challenges in translating the current knowledge from cancer to the virology field. Finally, we speculate on the promising future directions that immune cell engagers may take in cancer treatment and antiviral therapy.
通过同时与多种抗原结合,多特异性抗体有望大幅提高抗体免疫疗法的活性和长期疗效。免疫细胞吞噬剂是抗体构建物的一个亚类,由工程结构组成,旨在将免疫效应细胞与靶细胞连接起来,从而将免疫反应转向肿瘤细胞或受感染细胞。最近,越来越多的临床试验对免疫细胞吸引剂进行了评估,这反映出这些分子在癌症和感染的新疗法中发挥着重要作用。在这篇综述中,我们将讨论在癌症和感染性疾病的免疫疗法中,不同类型的免疫细胞(T 淋巴细胞、自然杀伤淋巴细胞以及髓系细胞)如何与免疫细胞吞噬剂结合。此外,我们还探讨了这些构建物的临床前和临床进展,并讨论了将现有知识从癌症转化到病毒学领域所面临的挑战。最后,我们推测了免疫细胞吞噬剂在癌症治疗和抗病毒治疗中的未来发展方向。
{"title":"Bridging the gap with multispecific immune cell engagers in cancer and infectious diseases","authors":"Camille Rolin, Jacques Zimmer, Carole Seguin-Devaux","doi":"10.1038/s41423-024-01176-4","DOIUrl":"10.1038/s41423-024-01176-4","url":null,"abstract":"By binding to multiple antigens simultaneously, multispecific antibodies are expected to substantially improve both the activity and long-term efficacy of antibody-based immunotherapy. Immune cell engagers, a subclass of antibody-based constructs, consist of engineered structures designed to bridge immune effector cells to their target, thereby redirecting the immune response toward the tumor cells or infected cells. The increasing number of recent clinical trials evaluating immune cell engagers reflects the important role of these molecules in new therapeutic approaches for cancer and infections. In this review, we discuss how different immune cell types (T and natural killer lymphocytes, as well as myeloid cells) can be bound by immune cell engagers in immunotherapy for cancer and infectious diseases. Furthermore, we explore the preclinical and clinical advancements of these constructs, and we discuss the challenges in translating the current knowledge from cancer to the virology field. Finally, we speculate on the promising future directions that immune cell engagers may take in cancer treatment and antiviral therapy.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11214628/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141092312","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of plasmacytoid dendritic cells (pDCs) in immunity during viral infections and beyond 质体树突状细胞(pDCs)在病毒感染期间及以后的免疫中的作用。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-22 DOI: 10.1038/s41423-024-01167-5
Clémence Ngo, Clémence Garrec, Elena Tomasello, Marc Dalod
Type I and III interferons (IFNs) are essential for antiviral immunity and act through two different but complimentary pathways. First, IFNs activate intracellular antimicrobial programs by triggering the upregulation of a broad repertoire of viral restriction factors. Second, IFNs activate innate and adaptive immunity. Dysregulation of IFN production can lead to severe immune system dysfunction. It is thus crucial to identify and characterize the cellular sources of IFNs, their effects, and their regulation to promote their beneficial effects and limit their detrimental effects, which can depend on the nature of the infected or diseased tissues, as we will discuss. Plasmacytoid dendritic cells (pDCs) can produce large amounts of all IFN subtypes during viral infection. pDCs are resistant to infection by many different viruses, thus inhibiting the immune evasion mechanisms of viruses that target IFN production or their downstream responses. Therefore, pDCs are considered essential for the control of viral infections and the establishment of protective immunity. A thorough bibliographical survey showed that, in most viral infections, despite being major IFN producers, pDCs are actually dispensable for host resistance, which is achieved by multiple IFN sources depending on the tissue. Moreover, primary innate and adaptive antiviral immune responses are only transiently affected in the absence of pDCs. More surprisingly, pDCs and their IFNs can be detrimental in some viral infections or autoimmune diseases. This makes the conservation of pDCs during vertebrate evolution an enigma and thus raises outstanding questions about their role not only in viral infections but also in other diseases and under physiological conditions.
I 型和 III 型干扰素(IFNs)对抗病毒免疫至关重要,它们通过两种不同但互补的途径发挥作用。首先,IFNs 通过触发病毒限制因子的上调,激活细胞内的抗微生物程序。其次,IFNs 可激活先天性和适应性免疫。IFN 生成失调可导致严重的免疫系统功能障碍。因此,确定 IFNs 的细胞来源、作用及其调控方式并描述其特征至关重要,以促进其有益作用并限制其有害作用,这可能取决于感染或患病组织的性质,我们将对此进行讨论。类质体树突状细胞(pDCs)能在病毒感染期间产生大量的所有 IFN 亚型。pDCs 能抵抗多种不同病毒的感染,从而抑制病毒针对 IFN 产生或其下游反应的免疫逃避机制。因此,pDCs 被认为是控制病毒感染和建立保护性免疫的关键。一项详尽的文献调查显示,在大多数病毒感染中,尽管 pDCs 是 IFN 的主要产生者,但它们对宿主的抵抗力实际上是可有可无的,宿主的抵抗力是由不同组织的多种 IFN 来源实现的。此外,在缺乏 pDCs 的情况下,主要的先天性和适应性抗病毒免疫反应只会受到短暂的影响。更令人惊讶的是,pDCs 及其 IFNs 在某些病毒感染或自身免疫性疾病中可能是有害的。这使得 pDCs 在脊椎动物进化过程中的保存成为一个谜,并因此提出了关于它们不仅在病毒感染中,而且在其他疾病和生理条件下的作用的悬而未决的问题。
{"title":"The role of plasmacytoid dendritic cells (pDCs) in immunity during viral infections and beyond","authors":"Clémence Ngo, Clémence Garrec, Elena Tomasello, Marc Dalod","doi":"10.1038/s41423-024-01167-5","DOIUrl":"10.1038/s41423-024-01167-5","url":null,"abstract":"Type I and III interferons (IFNs) are essential for antiviral immunity and act through two different but complimentary pathways. First, IFNs activate intracellular antimicrobial programs by triggering the upregulation of a broad repertoire of viral restriction factors. Second, IFNs activate innate and adaptive immunity. Dysregulation of IFN production can lead to severe immune system dysfunction. It is thus crucial to identify and characterize the cellular sources of IFNs, their effects, and their regulation to promote their beneficial effects and limit their detrimental effects, which can depend on the nature of the infected or diseased tissues, as we will discuss. Plasmacytoid dendritic cells (pDCs) can produce large amounts of all IFN subtypes during viral infection. pDCs are resistant to infection by many different viruses, thus inhibiting the immune evasion mechanisms of viruses that target IFN production or their downstream responses. Therefore, pDCs are considered essential for the control of viral infections and the establishment of protective immunity. A thorough bibliographical survey showed that, in most viral infections, despite being major IFN producers, pDCs are actually dispensable for host resistance, which is achieved by multiple IFN sources depending on the tissue. Moreover, primary innate and adaptive antiviral immune responses are only transiently affected in the absence of pDCs. More surprisingly, pDCs and their IFNs can be detrimental in some viral infections or autoimmune diseases. This makes the conservation of pDCs during vertebrate evolution an enigma and thus raises outstanding questions about their role not only in viral infections but also in other diseases and under physiological conditions.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01167-5.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141079742","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
Liver kinase B1 (LKB1) staves off γδ T-cell-mediated autoimmune liver disease 肝激酶 B1 (LKB1) 阻止了 γδ T 细胞介导的自身免疫性肝病。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-05-16 DOI: 10.1038/s41423-024-01172-8
Yiwen Wang, Lan Wu, Luc Van Kaer
{"title":"Liver kinase B1 (LKB1) staves off γδ T-cell-mediated autoimmune liver disease","authors":"Yiwen Wang, Lan Wu, Luc Van Kaer","doi":"10.1038/s41423-024-01172-8","DOIUrl":"10.1038/s41423-024-01172-8","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140956417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chemerin is a key player in antimicrobial defense in skin 螯合素是皮肤抗菌防御的关键因素。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-05-16 DOI: 10.1038/s41423-024-01159-5
Silvano Sozzani, Francesca Sozio, Annalisa Del Prete
{"title":"Chemerin is a key player in antimicrobial defense in skin","authors":"Silvano Sozzani, Francesca Sozio, Annalisa Del Prete","doi":"10.1038/s41423-024-01159-5","DOIUrl":"10.1038/s41423-024-01159-5","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140956416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neutrophil–macrophage communication via extracellular vesicle transfer promotes itaconate accumulation and ameliorates cytokine storm syndrome 中性粒细胞-巨噬细胞通过细胞外囊泡转移进行交流,促进了伊塔康酸的积累并改善了细胞因子风暴综合征。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-14 DOI: 10.1038/s41423-024-01174-6
Haixia Kang, Ting Liu, Yuanyuan Wang, Wenjuan Bai, Yan Luo, Jing Wang
Cytokine storm syndrome (CSS) is a life-threatening systemic inflammatory syndrome involving innate immune hyperactivity triggered by various therapies, infections, and autoimmune conditions. However, the potential interplay between innate immune cells is not fully understood. Here, using poly I:C and lipopolysaccharide (LPS)-induced cytokine storm models, a protective role of neutrophils through the modulation of macrophage activation was identified in a CSS model. Intravital imaging revealed neutrophil-derived extracellular vesicles (NDEVs) in the liver and spleen, which were captured by macrophages. NDEVs suppressed proinflammatory cytokine production by macrophages when cocultured in vitro or infused into CSS models. Metabolic profiling of macrophages treated with NDEV revealed elevated levels of the anti-inflammatory metabolite, itaconate, which is produced from cis-aconitate in the Krebs cycle by cis-aconitate decarboxylase (Acod1, encoded by Irg1). Irg1 in macrophages, but not in neutrophils, was critical for the NDEV-mediated anti-inflammatory effects. Mechanistically, NDEVs delivered miR-27a-3p, which suppressed the expression of Suclg1, the gene encoding the enzyme that metabolizes itaconate, thereby resulting in the accumulation of itaconate in macrophages. These findings demonstrated that neutrophil-to-macrophage communication mediated by extracellular vesicles is critical for promoting the anti-inflammatory reprogramming of macrophages in CSS and may have potential implications for the treatment of this fatal condition.
细胞因子风暴综合征(CSS)是一种危及生命的全身性炎症综合征,涉及由各种疗法、感染和自身免疫疾病引发的先天性免疫亢进。然而,先天性免疫细胞之间潜在的相互作用尚未完全明了。本文利用多聚 I:C 和脂多糖(LPS)诱导的细胞因子风暴模型,确定了中性粒细胞在 CSS 模型中通过调节巨噬细胞活化起到的保护作用。肉眼成像显示肝脏和脾脏中有中性粒细胞衍生的细胞外囊泡 (NDEV),这些囊泡被巨噬细胞捕获。NDEVs在体外共培养或注入CSS模型时可抑制巨噬细胞产生促炎细胞因子。经 NDEV 处理的巨噬细胞的代谢图谱显示,抗炎代谢物伊他康酸的水平升高,伊他康酸是由顺式乌头酸脱羧酶(Acod1,由 Irg1 编码)在克雷布斯循环中从顺式乌头酸产生的。巨噬细胞(而非中性粒细胞)中的 Irg1 对 NDEV 介导的抗炎作用至关重要。从机制上讲,NDEV 传递 miR-27a-3p,抑制了 Suclg1 的表达,而 Suclg1 是伊他康酸代谢酶的编码基因,因此导致了伊他康酸在巨噬细胞中的积累。这些研究结果表明,由细胞外囊泡介导的中性粒细胞与巨噬细胞之间的通讯对于促进 CSS 中巨噬细胞的抗炎重编程至关重要,可能对治疗这种致命疾病具有潜在的意义。
{"title":"Neutrophil–macrophage communication via extracellular vesicle transfer promotes itaconate accumulation and ameliorates cytokine storm syndrome","authors":"Haixia Kang, Ting Liu, Yuanyuan Wang, Wenjuan Bai, Yan Luo, Jing Wang","doi":"10.1038/s41423-024-01174-6","DOIUrl":"10.1038/s41423-024-01174-6","url":null,"abstract":"Cytokine storm syndrome (CSS) is a life-threatening systemic inflammatory syndrome involving innate immune hyperactivity triggered by various therapies, infections, and autoimmune conditions. However, the potential interplay between innate immune cells is not fully understood. Here, using poly I:C and lipopolysaccharide (LPS)-induced cytokine storm models, a protective role of neutrophils through the modulation of macrophage activation was identified in a CSS model. Intravital imaging revealed neutrophil-derived extracellular vesicles (NDEVs) in the liver and spleen, which were captured by macrophages. NDEVs suppressed proinflammatory cytokine production by macrophages when cocultured in vitro or infused into CSS models. Metabolic profiling of macrophages treated with NDEV revealed elevated levels of the anti-inflammatory metabolite, itaconate, which is produced from cis-aconitate in the Krebs cycle by cis-aconitate decarboxylase (Acod1, encoded by Irg1). Irg1 in macrophages, but not in neutrophils, was critical for the NDEV-mediated anti-inflammatory effects. Mechanistically, NDEVs delivered miR-27a-3p, which suppressed the expression of Suclg1, the gene encoding the enzyme that metabolizes itaconate, thereby resulting in the accumulation of itaconate in macrophages. These findings demonstrated that neutrophil-to-macrophage communication mediated by extracellular vesicles is critical for promoting the anti-inflammatory reprogramming of macrophages in CSS and may have potential implications for the treatment of this fatal condition.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140921447","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel insights into the sexual dimorphism-associated immune response 对性双态相关免疫反应的新认识。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-13 DOI: 10.1038/s41423-024-01177-3
José M. Izquierdo
{"title":"Novel insights into the sexual dimorphism-associated immune response","authors":"José M. Izquierdo","doi":"10.1038/s41423-024-01177-3","DOIUrl":"10.1038/s41423-024-01177-3","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140916179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Eomesodermin spatiotemporally orchestrates the early and late stages of NK cell development by targeting KLF2 and T-bet, respectively Eomesodermin通过分别靶向KLF2和T-bet,在时空上协调NK细胞发育的早期和晚期阶段。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-13 DOI: 10.1038/s41423-024-01164-8
Junming He, Donglin Chen, Wei Xiong, Xinlei Hou, Yuhe Quan, Meixiang Yang, Zhongjun Dong
Eomesodermin (Eomes) is a critical factor in the development of natural killer (NK) cells, but its precise role in temporal and spatial coordination during this process remains unclear. Our study revealed that Eomes plays distinct roles during the early and late stages of NK cell development. Specifically, the early deletion of Eomes via the CD122-Cre transgene resulted in significant blockade at the progenitor stage due to the downregulation of KLF2, another important transcription factor. ChIP-seq revealed direct binding of Eomes to the conserved noncoding sequence (CNS) of Klf2. Utilizing the CHimeric IMmune Editing (CHIME) technique, we found that deletion of the CNS region of Klf2 via CRISPRi led to a reduction in the NK cell population and developmental arrest. Moreover, constitutive activation of this specific CNS region through CRISPRa significantly reversed the severe defects in NK cell development caused by Eomes deficiency. Conversely, Ncr1-Cre-mediated terminal deletion of Eomes expedited the transition of NK cell subsets from the CD27+CD11b+ phenotype to the CD27−CD11b+ phenotype. Late-stage deficiency of Eomes led to a significant increase in T-bet expression, which subsequently increased the expression of the transcription factor Zeb2. Genetic deletion of one allele of Tbx21, encoding T-bet, effectively reversed the aberrant differentiation of Eomes-deficient NK cells. In summary, we utilized two innovative genetic models to elucidate the intricate mechanisms underlying Eomes-mediated NK cell commitment and differentiation.
Eomesodermin(Eomes)是天然杀伤细胞(NK)发育过程中的一个关键因素,但它在这一过程中的时间和空间协调方面的确切作用仍不清楚。我们的研究发现,Eomes在NK细胞发育的早期和晚期发挥着不同的作用。具体来说,通过CD122-Cre转基因早期删除Eomes会导致另一个重要转录因子KLF2下调,从而在祖细胞阶段造成显著阻滞。ChIP-seq 发现,Eomes 与 Klf2 的保守非编码序列 (CNS) 直接结合。利用嵌合 IMmune 编辑(CHIME)技术,我们发现通过 CRISPRi 删除 Klf2 的 CNS 区域会导致 NK 细胞数量减少和发育停滞。此外,通过CRISPRa对这一特异性CNS区域进行组成性激活,可显著逆转Eomes缺乏导致的NK细胞发育严重缺陷。相反,Ncr1-Cre介导的Eomes末端缺失加速了NK细胞亚群从CD27+CD11b+表型向CD27-CD11b+表型的转变。编码 T-bet 的 Tbx21 的一个等位基因的基因缺失有效逆转了 Eomes 缺失的 NK 细胞的异常分化。总之,我们利用两种创新的遗传模型阐明了Eomes介导的NK细胞承诺和分化的复杂机制。
{"title":"Eomesodermin spatiotemporally orchestrates the early and late stages of NK cell development by targeting KLF2 and T-bet, respectively","authors":"Junming He, Donglin Chen, Wei Xiong, Xinlei Hou, Yuhe Quan, Meixiang Yang, Zhongjun Dong","doi":"10.1038/s41423-024-01164-8","DOIUrl":"10.1038/s41423-024-01164-8","url":null,"abstract":"Eomesodermin (Eomes) is a critical factor in the development of natural killer (NK) cells, but its precise role in temporal and spatial coordination during this process remains unclear. Our study revealed that Eomes plays distinct roles during the early and late stages of NK cell development. Specifically, the early deletion of Eomes via the CD122-Cre transgene resulted in significant blockade at the progenitor stage due to the downregulation of KLF2, another important transcription factor. ChIP-seq revealed direct binding of Eomes to the conserved noncoding sequence (CNS) of Klf2. Utilizing the CHimeric IMmune Editing (CHIME) technique, we found that deletion of the CNS region of Klf2 via CRISPRi led to a reduction in the NK cell population and developmental arrest. Moreover, constitutive activation of this specific CNS region through CRISPRa significantly reversed the severe defects in NK cell development caused by Eomes deficiency. Conversely, Ncr1-Cre-mediated terminal deletion of Eomes expedited the transition of NK cell subsets from the CD27+CD11b+ phenotype to the CD27−CD11b+ phenotype. Late-stage deficiency of Eomes led to a significant increase in T-bet expression, which subsequently increased the expression of the transcription factor Zeb2. Genetic deletion of one allele of Tbx21, encoding T-bet, effectively reversed the aberrant differentiation of Eomes-deficient NK cells. In summary, we utilized two innovative genetic models to elucidate the intricate mechanisms underlying Eomes-mediated NK cell commitment and differentiation.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140916176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A novel target to turn cold tumors into hot tumors: lysosomal 25-hydroxycholesterol activates AMPKα and immunosuppressive tumor-associated macrophages 将冷肿瘤变为热肿瘤的新靶点:溶酶体 25- 羟基胆固醇激活 AMPKα 和免疫抑制性肿瘤相关巨噬细胞。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-13 DOI: 10.1038/s41423-024-01171-9
Shuangshuang Liu, Jiaqi Wu, Xiao Tong, Li-Hao Huang
{"title":"A novel target to turn cold tumors into hot tumors: lysosomal 25-hydroxycholesterol activates AMPKα and immunosuppressive tumor-associated macrophages","authors":"Shuangshuang Liu, Jiaqi Wu, Xiao Tong, Li-Hao Huang","doi":"10.1038/s41423-024-01171-9","DOIUrl":"10.1038/s41423-024-01171-9","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140916174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GITR exacerbates lysophosphatidylcholine-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3 GITR通过对NLRP3的翻译后调控,加剧脓毒症中溶血磷脂酰胆碱诱导的巨噬细胞脓毒症。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-05-13 DOI: 10.1038/s41423-024-01170-w
Siping Liang, Jinyu Zhou, Can Cao, Yiting Liu, Siqi Ming, Xi Liu, Yuqi Shang, Juanfeng Lao, Qin Peng, Jiahui Yang, Minhao Wu
The NLRP3 inflammasome functions as an inflammatory driver, but its relationship with lipid metabolic changes in early sepsis remains unclear. Here, we found that GITR expression in monocytes/macrophages was induced by lysophosphatidylcholine (LPC) and was positively correlated with the severity of sepsis. GITR is a costimulatory molecule that is mainly expressed on T cells, but its function in macrophages is largely unknown. Our in vitro data showed that GITR enhanced LPC uptake by macrophages and specifically enhanced NLRP3 inflammasome-mediated macrophage pyroptosis. Furthermore, in vivo studies using either cecal ligation and puncture (CLP) or LPS-induced sepsis models demonstrated that LPC exacerbated sepsis severity/lethality, while conditional knockout of GITR in myeloid cells or NLRP3/caspase-1/IL-1β deficiency attenuated sepsis severity/lethality. Mechanistically, GITR specifically enhanced inflammasome activation by regulating the posttranslational modification (PTM) of NLRP3. GITR competes with NLRP3 for binding to the E3 ligase MARCH7 and recruits MARCH7 to induce deacetylase SIRT2 degradation, leading to decreasing ubiquitination but increasing acetylation of NLRP3. Overall, these findings revealed a novel role of macrophage-derived GITR in regulating the PTM of NLRP3 and systemic inflammatory injury, suggesting that GITR may be a potential therapeutic target for sepsis and other inflammatory diseases.
NLRP3 炎性体是炎症的驱动因子,但它与早期脓毒症脂质代谢变化的关系仍不清楚。在这里,我们发现溶血磷脂酰胆碱(LPC)会诱导单核细胞/巨噬细胞中 GITR 的表达,并且与脓毒症的严重程度呈正相关。GITR 是一种成本刺激分子,主要在 T 细胞中表达,但其在巨噬细胞中的功能尚不清楚。我们的体外研究数据显示,GITR 可增强巨噬细胞对 LPC 的吸收,并特异性地增强 NLRP3 炎症体介导的巨噬细胞脓毒症。此外,使用盲肠结扎和穿刺(CLP)或 LPS 诱导的脓毒症模型进行的体内研究表明,LPC 会加重脓毒症的严重程度/致死率,而在骨髓细胞中条件性敲除 GITR 或 NLRP3/caspase-1/IL-1β 缺乏会减轻脓毒症的严重程度/致死率。从机制上讲,GITR通过调节NLRP3的翻译后修饰(PTM)特异性地增强了炎性体的激活。GITR与NLRP3竞争结合到E3连接酶MARCH7上,并招募MARCH7诱导去乙酰化酶SIRT2降解,导致NLRP3的泛素化减少而乙酰化增加。总之,这些发现揭示了巨噬细胞源性 GITR 在调节 NLRP3 的 PTM 和全身炎症损伤中的新作用,表明 GITR 可能是败血症和其他炎症性疾病的潜在治疗靶点。GITR 通过对 NLRP3 的翻译后调控,加剧了脓毒症中 LPC 诱导的巨噬细胞脓毒症。根据该模型,脓毒症早期 LPC 水平升高,诱导巨噬细胞表达 GITR。GITR 不仅会与 NLRP3 竞争与 E3 连接酶 MARCH7 的结合,还会招募 MARCH7 诱导去乙酰化酶 SIRT2 的降解,导致 NLRP3 的泛素化减少而乙酰化增加,从而加剧 LPC 诱导的 NLRP3 炎症小体激活、巨噬细胞脓毒症和全身炎症损伤。
{"title":"GITR exacerbates lysophosphatidylcholine-induced macrophage pyroptosis in sepsis via posttranslational regulation of NLRP3","authors":"Siping Liang, Jinyu Zhou, Can Cao, Yiting Liu, Siqi Ming, Xi Liu, Yuqi Shang, Juanfeng Lao, Qin Peng, Jiahui Yang, Minhao Wu","doi":"10.1038/s41423-024-01170-w","DOIUrl":"10.1038/s41423-024-01170-w","url":null,"abstract":"The NLRP3 inflammasome functions as an inflammatory driver, but its relationship with lipid metabolic changes in early sepsis remains unclear. Here, we found that GITR expression in monocytes/macrophages was induced by lysophosphatidylcholine (LPC) and was positively correlated with the severity of sepsis. GITR is a costimulatory molecule that is mainly expressed on T cells, but its function in macrophages is largely unknown. Our in vitro data showed that GITR enhanced LPC uptake by macrophages and specifically enhanced NLRP3 inflammasome-mediated macrophage pyroptosis. Furthermore, in vivo studies using either cecal ligation and puncture (CLP) or LPS-induced sepsis models demonstrated that LPC exacerbated sepsis severity/lethality, while conditional knockout of GITR in myeloid cells or NLRP3/caspase-1/IL-1β deficiency attenuated sepsis severity/lethality. Mechanistically, GITR specifically enhanced inflammasome activation by regulating the posttranslational modification (PTM) of NLRP3. GITR competes with NLRP3 for binding to the E3 ligase MARCH7 and recruits MARCH7 to induce deacetylase SIRT2 degradation, leading to decreasing ubiquitination but increasing acetylation of NLRP3. Overall, these findings revealed a novel role of macrophage-derived GITR in regulating the PTM of NLRP3 and systemic inflammatory injury, suggesting that GITR may be a potential therapeutic target for sepsis and other inflammatory diseases.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140916178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cellular &Molecular Immunology
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