首页 > 最新文献

Cellular &Molecular Immunology最新文献

英文 中文
Unsynchronized butyrophilin molecules dictate cancer cell evasion of Vγ9Vδ2 T-cell killing 不同步的丁酵素分子决定了癌细胞对 Vγ9Vδ2 T 细胞杀伤的逃避。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-02-20 DOI: 10.1038/s41423-024-01135-z
Zeguang Wu, Qiezhong Lamao, Meichao Gu, Xuanxuan Jin, Ying Liu, Feng Tian, Ying Yu, Pengfei Yuan, Shuaixin Gao, Thomas S. Fulford, Adam P. Uldrich, Catherine CL Wong, Wensheng Wei
Vγ9Vδ2 T cells are specialized effector cells that have gained prominence as immunotherapy agents due to their ability to target and kill cells with altered pyrophosphate metabolites. In our effort to understand how cancer cells evade the cell-killing activity of Vγ9Vδ2 T cells, we performed a comprehensive genome-scale CRISPR screening of cancer cells. We found that four molecules belonging to the butyrophilin (BTN) family, specifically BTN2A1, BTN3A1, BTN3A2, and BTN3A3, are critically important and play unique, nonoverlapping roles in facilitating the destruction of cancer cells by primary Vγ9Vδ2 T cells. The coordinated function of these BTN molecules was driven by synchronized gene expression, which was regulated by IFN-γ signaling and the RFX complex. Additionally, an enzyme called QPCTL was shown to play a key role in modifying the N-terminal glutamine of these BTN proteins and was found to be a crucial factor in Vγ9Vδ2 T cell killing of cancer cells. Through our research, we offer a detailed overview of the functional genomic mechanisms that underlie how cancer cells escape Vγ9Vδ2 T cells. Moreover, our findings shed light on the importance of the harmonized expression and function of gene family members in modulating T-cell activity.
Vγ9Vδ2 T 细胞是一种特化的效应细胞,由于它们能够靶向杀伤焦磷酸代谢物发生改变的细胞,因此作为免疫疗法药物而备受瞩目。为了了解癌细胞如何逃避 Vγ9Vδ2 T 细胞的细胞杀伤活性,我们对癌细胞进行了全面的基因组规模 CRISPR 筛选。我们发现,属于丁嗜蛋白(BTN)家族的四个分子,特别是 BTN2A1、BTN3A1、BTN3A2 和 BTN3A3,在促进原代 Vγ9Vδ2 T 细胞消灭癌细胞的过程中至关重要,并发挥着独特而不重叠的作用。这些 BTN 分子的协调功能由同步基因表达驱动,而同步基因表达则受 IFN-γ 信号传导和 RFX 复合物的调控。此外,一种名为 QPCTL 的酶被证明在修饰这些 BTN 蛋白的 N 端谷氨酰胺方面起着关键作用,并被发现是 Vγ9Vδ2 T 细胞杀死癌细胞的关键因素。通过我们的研究,我们详细概述了癌细胞如何逃避 Vγ9Vδ2 T 细胞的功能基因组机制。此外,我们的研究结果还揭示了基因家族成员的协调表达和功能在调节 T 细胞活性方面的重要性。
{"title":"Unsynchronized butyrophilin molecules dictate cancer cell evasion of Vγ9Vδ2 T-cell killing","authors":"Zeguang Wu, Qiezhong Lamao, Meichao Gu, Xuanxuan Jin, Ying Liu, Feng Tian, Ying Yu, Pengfei Yuan, Shuaixin Gao, Thomas S. Fulford, Adam P. Uldrich, Catherine CL Wong, Wensheng Wei","doi":"10.1038/s41423-024-01135-z","DOIUrl":"10.1038/s41423-024-01135-z","url":null,"abstract":"Vγ9Vδ2 T cells are specialized effector cells that have gained prominence as immunotherapy agents due to their ability to target and kill cells with altered pyrophosphate metabolites. In our effort to understand how cancer cells evade the cell-killing activity of Vγ9Vδ2 T cells, we performed a comprehensive genome-scale CRISPR screening of cancer cells. We found that four molecules belonging to the butyrophilin (BTN) family, specifically BTN2A1, BTN3A1, BTN3A2, and BTN3A3, are critically important and play unique, nonoverlapping roles in facilitating the destruction of cancer cells by primary Vγ9Vδ2 T cells. The coordinated function of these BTN molecules was driven by synchronized gene expression, which was regulated by IFN-γ signaling and the RFX complex. Additionally, an enzyme called QPCTL was shown to play a key role in modifying the N-terminal glutamine of these BTN proteins and was found to be a crucial factor in Vγ9Vδ2 T cell killing of cancer cells. Through our research, we offer a detailed overview of the functional genomic mechanisms that underlie how cancer cells escape Vγ9Vδ2 T cells. Moreover, our findings shed light on the importance of the harmonized expression and function of gene family members in modulating T-cell activity.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139905189","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
CD200Rhigh neutrophils with dysfunctional autophagy establish systemic immunosuppression by increasing regulatory T cells 自噬功能失调的 CD200R 高中性粒细胞会通过增加调节性 T 细胞来建立全身免疫抑制。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-02-05 DOI: 10.1038/s41423-024-01136-y
Ye Seon Kim, Yu Sun Jeong, Geon Ho Bae, Ji Hyeon Kang, Mingyu Lee, Brian A. Zabel, Yoe-Sik Bae
Distinct neutrophil populations arise during certain pathological conditions. The generation of dysfunctional neutrophils during sepsis and their contribution to septicemia-related systemic immune suppression remain unclear. In this study, using an experimental sepsis model that features immunosuppression, we identified a novel population of pathogenic CD200Rhigh neutrophils that are generated during the initial stages of sepsis and contribute to systemic immune suppression by enhancing regulatory T (Treg) cells. Compared to their CD200Rlow counterparts, sepsis-generated CD200Rhigh neutrophils exhibit impaired autophagy and dysfunction, with reduced chemotactic migration, superoxide anion production, and TNF-α production. Increased soluble CD200 blocks autophagy and neutrophil maturation in the bone marrow during experimental sepsis, and recombinant CD200 treatment in vitro can induce neutrophil dysfunction similar to that observed in CD200Rhigh neutrophils. The administration of an α-CD200R antibody effectively reversed neutrophil dysfunction by enhancing autophagy and protecting against a secondary infection challenge, leading to increased survival. Transcriptome analysis revealed that CD200Rhigh neutrophils expressed high levels of Igf1, which elicits the generation of Treg cells, while the administration of an α-CD200R antibody inhibited Treg cell generation in a secondary infection model. Taken together, our findings revealed a novel CD200Rhigh neutrophil population that mediates the pathogenesis of sepsis-induced systemic immunosuppression by generating Treg cells.
在某些病理条件下会出现不同的中性粒细胞群。脓毒症期间功能失调中性粒细胞的产生及其对脓毒症相关全身免疫抑制的贡献仍不清楚。在这项研究中,我们利用一种以免疫抑制为特征的实验性脓毒症模型,发现了一种新的致病性 CD200Rhigh 中性粒细胞群,它们在脓毒症初期产生,并通过增强调节性 T(Treg)细胞来促进全身免疫抑制。与 CD200R 低的中性粒细胞相比,脓毒症产生的 CD200R 高的中性粒细胞表现出自噬受损和功能障碍,趋化性迁移、超氧阴离子生成和 TNF-α 生成减少。在实验性脓毒症期间,可溶性 CD200 的增加会阻碍骨髓中的自噬和中性粒细胞成熟,体外重组 CD200 可诱导中性粒细胞功能障碍,与 CD200Rhigh 中性粒细胞中观察到的功能障碍相似。服用α-CD200R抗体可通过增强自噬和抵御二次感染挑战来有效逆转中性粒细胞功能障碍,从而提高存活率。转录组分析表明,CD200R高的中性粒细胞表达高水平的Igf1,而Igf1能诱导Treg细胞的生成,而服用α-CD200R抗体能抑制二次感染模型中Treg细胞的生成。总之,我们的研究结果揭示了一种新型的高CD200R中性粒细胞群,它通过生成Treg细胞介导脓毒症诱导的全身免疫抑制的发病机制。
{"title":"CD200Rhigh neutrophils with dysfunctional autophagy establish systemic immunosuppression by increasing regulatory T cells","authors":"Ye Seon Kim, Yu Sun Jeong, Geon Ho Bae, Ji Hyeon Kang, Mingyu Lee, Brian A. Zabel, Yoe-Sik Bae","doi":"10.1038/s41423-024-01136-y","DOIUrl":"10.1038/s41423-024-01136-y","url":null,"abstract":"Distinct neutrophil populations arise during certain pathological conditions. The generation of dysfunctional neutrophils during sepsis and their contribution to septicemia-related systemic immune suppression remain unclear. In this study, using an experimental sepsis model that features immunosuppression, we identified a novel population of pathogenic CD200Rhigh neutrophils that are generated during the initial stages of sepsis and contribute to systemic immune suppression by enhancing regulatory T (Treg) cells. Compared to their CD200Rlow counterparts, sepsis-generated CD200Rhigh neutrophils exhibit impaired autophagy and dysfunction, with reduced chemotactic migration, superoxide anion production, and TNF-α production. Increased soluble CD200 blocks autophagy and neutrophil maturation in the bone marrow during experimental sepsis, and recombinant CD200 treatment in vitro can induce neutrophil dysfunction similar to that observed in CD200Rhigh neutrophils. The administration of an α-CD200R antibody effectively reversed neutrophil dysfunction by enhancing autophagy and protecting against a secondary infection challenge, leading to increased survival. Transcriptome analysis revealed that CD200Rhigh neutrophils expressed high levels of Igf1, which elicits the generation of Treg cells, while the administration of an α-CD200R antibody inhibited Treg cell generation in a secondary infection model. Taken together, our findings revealed a novel CD200Rhigh neutrophil population that mediates the pathogenesis of sepsis-induced systemic immunosuppression by generating Treg cells.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139680666","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
Activation of leukotriene B4 receptor 1 is a prerequisite for complement receptor 3-mediated antifungal responses of neutrophils 激活白三烯 B4 受体 1 是补体受体 3 介导的中性粒细胞抗真菌反应的先决条件
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-31 DOI: 10.1038/s41423-024-01130-4
Yan Xin, Sihan Xiong, Linghong Zhou, Xin Lin
Invasive fungal infections are life-threatening, and neutrophils are vital cells of the innate immune system that defend against them. The role of LTA4H-LTB4-BLT1 axis in regulation of neutrophil responses to fungal infection remains poorly understood. Here, we demonstrated that the LTA4H-LTB4-BLT1 axis protects the host against Candida albicans and Aspergillus fumigatus, but not Cryptococcus neoformans infection, by regulating the antifungal activity of neutrophils. Our results show that deleting Lta4h or Blt1 substantially impairs the fungal-specific phagocytic capacity of neutrophils. Moreover, defective activation of the spleen tyrosine kinase (Syk) and extracellular signal-related kinase (ERK1/2) pathways in neutrophils accompanies this impairment. Mechanistically, BLT1 regulates CR3-mediated, β-1,3-glucan-induced neutrophil phagocytosis, while a physical interaction with CR3 with slight influence on its dynamics is observed. Our findings thus demonstrate that the LTA4H-LTB4-BLT1 axis is essential for the phagocytic function of neutrophils in host antifungal immune response against Candida albicans and Aspergillus fumigatus.
侵袭性真菌感染危及生命,而中性粒细胞是先天性免疫系统中抵御真菌感染的重要细胞。人们对 LTA4H-LTB4-BLT1 轴在调节中性粒细胞对真菌感染的反应中的作用仍然知之甚少。在这里,我们证明了 LTA4H-LTB4-BLT1 轴通过调节中性粒细胞的抗真菌活性,保护宿主免受白色念珠菌和烟曲霉感染,但不能保护新生隐球菌感染。我们的研究结果表明,删除 Lta4h 或 Blt1 会大大削弱中性粒细胞对真菌的特异性吞噬能力。此外,中性粒细胞中脾酪氨酸激酶(Syk)和细胞外信号相关激酶(ERK1/2)通路的激活缺陷也伴随着这种损害。从机理上讲,BLT1 可调节 CR3 介导的、β-1,3-葡聚糖诱导的中性粒细胞吞噬作用,同时还可观察到与 CR3 的物理相互作用对其动力学的轻微影响。因此,我们的研究结果表明,LTA4H-LTB4-BLT1 轴对中性粒细胞在宿主抗白色念珠菌和曲霉菌免疫反应中的吞噬功能至关重要。
{"title":"Activation of leukotriene B4 receptor 1 is a prerequisite for complement receptor 3-mediated antifungal responses of neutrophils","authors":"Yan Xin, Sihan Xiong, Linghong Zhou, Xin Lin","doi":"10.1038/s41423-024-01130-4","DOIUrl":"10.1038/s41423-024-01130-4","url":null,"abstract":"Invasive fungal infections are life-threatening, and neutrophils are vital cells of the innate immune system that defend against them. The role of LTA4H-LTB4-BLT1 axis in regulation of neutrophil responses to fungal infection remains poorly understood. Here, we demonstrated that the LTA4H-LTB4-BLT1 axis protects the host against Candida albicans and Aspergillus fumigatus, but not Cryptococcus neoformans infection, by regulating the antifungal activity of neutrophils. Our results show that deleting Lta4h or Blt1 substantially impairs the fungal-specific phagocytic capacity of neutrophils. Moreover, defective activation of the spleen tyrosine kinase (Syk) and extracellular signal-related kinase (ERK1/2) pathways in neutrophils accompanies this impairment. Mechanistically, BLT1 regulates CR3-mediated, β-1,3-glucan-induced neutrophil phagocytosis, while a physical interaction with CR3 with slight influence on its dynamics is observed. Our findings thus demonstrate that the LTA4H-LTB4-BLT1 axis is essential for the phagocytic function of neutrophils in host antifungal immune response against Candida albicans and Aspergillus fumigatus.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139646191","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
Id2 epigenetically controls CD8+ T-cell exhaustion by disrupting the assembly of the Tcf3-LSD1 complex Id2 通过破坏 Tcf3-LSD1 复合物的组装,从表观遗传学上控制 CD8+ T 细胞的衰竭。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-29 DOI: 10.1038/s41423-023-01118-6
Yiming Li, Mingwei Han, Haolin Wei, Wan Huang, Zhinan Chen, Tianjiao Zhang, Meirui Qian, Lin Jing, Gang Nan, Xiuxuan Sun, Shuhui Dai, Kun Wang, Jianli Jiang, Ping Zhu, Liang Chen
CD8+ T-cell exhaustion is a state of dysfunction that promotes tumor progression and is marked by the generation of Slamf6+ progenitor exhausted (Texprog) and Tim-3+ terminally exhausted (Texterm) subpopulations. Inhibitor of DNA binding protein 2 (Id2) has been shown to play important roles in T-cell development and CD8+ T-cell immunity. However, the role of Id2 in CD8+ T-cell exhaustion is unclear. Here, we found that Id2 transcriptionally and epigenetically regulates the generation of Texprog cells and their conversion to Texterm cells. Genetic deletion of Id2 dampens CD8+ T-cell-mediated immune responses and the maintenance of stem-like CD8+ T-cell subpopulations, suppresses PD-1 blockade and increases tumor susceptibility. Mechanistically, through its HLH domain, Id2 binds and disrupts the assembly of the Tcf3-Tal1 transcriptional regulatory complex, and thus modulates chromatin accessibility at the Slamf6 promoter by preventing the interaction of Tcf3 with the histone lysine demethylase LSD1. Therefore, Id2 increases the abundance of the permissive H3K4me2 mark on the Tcf3-occupied E-boxes in the Slamf6 promoter, modulates chromatin accessibility at the Slamf6 promoter and epigenetically regulates the generation of Slamf6+ Texprog cells. An LSD1 inhibitor GSK2879552 can rescue the Id2 knockout phenotype in tumor-bearing mice. Inhibition of LSD1 increases the abundance of Slamf6+Tim-3− Texprog cells in tumors and the expression level of Tcf1 in Id2-deleted CD8+ T cells. This study demonstrates that Id2-mediated transcriptional and epigenetic modification drives hierarchical CD8+ T-cell exhaustion, and the mechanistic insights gained may have implications for therapeutic intervention with tumor immune evasion.
CD8+ T细胞衰竭是一种促进肿瘤进展的功能障碍状态,其特征是产生Slamf6+祖细胞衰竭(Texprog)和Tim-3+终末衰竭(Texterm)亚群。DNA 结合蛋白 2 抑制剂(Id2)已被证明在 T 细胞发育和 CD8+ T 细胞免疫中发挥重要作用。然而,Id2在CD8+ T细胞衰竭中的作用尚不清楚。在这里,我们发现Id2转录和表观遗传调控Texprog细胞的生成及其向Texterm细胞的转化。基因缺失Id2会抑制CD8+ T细胞介导的免疫反应和干样CD8+ T细胞亚群的维持,抑制PD-1阻断并增加肿瘤易感性。从机理上讲,Id2 通过其 HLH 结构域结合并破坏 Tcf3-Tal1 转录调控复合物的组装,从而通过阻止 Tcf3 与组蛋白赖氨酸去甲基化酶 LSD1 的相互作用来调节 Slamf6 启动子的染色质可及性。因此,Id2增加了Slamf6启动子中Tcf3占据的E-box上允许的H3K4me2标记的丰度,调节了Slamf6启动子的染色质可及性,并从表观遗传学上调控了Slamf6+ Texprog细胞的产生。LSD1抑制剂GSK2879552能挽救肿瘤小鼠的Id2基因敲除表型。抑制 LSD1 会增加肿瘤中 Slamf6+Tim-3- Texprog 细胞的数量,并提高 Id2 基因敲除的 CD8+ T 细胞中 Tcf1 的表达水平。这项研究表明,Id2介导的转录和表观遗传修饰推动了CD8+ T细胞的分层衰竭,所获得的机理认识可能对治疗干预肿瘤免疫逃避有意义。
{"title":"Id2 epigenetically controls CD8+ T-cell exhaustion by disrupting the assembly of the Tcf3-LSD1 complex","authors":"Yiming Li, Mingwei Han, Haolin Wei, Wan Huang, Zhinan Chen, Tianjiao Zhang, Meirui Qian, Lin Jing, Gang Nan, Xiuxuan Sun, Shuhui Dai, Kun Wang, Jianli Jiang, Ping Zhu, Liang Chen","doi":"10.1038/s41423-023-01118-6","DOIUrl":"10.1038/s41423-023-01118-6","url":null,"abstract":"CD8+ T-cell exhaustion is a state of dysfunction that promotes tumor progression and is marked by the generation of Slamf6+ progenitor exhausted (Texprog) and Tim-3+ terminally exhausted (Texterm) subpopulations. Inhibitor of DNA binding protein 2 (Id2) has been shown to play important roles in T-cell development and CD8+ T-cell immunity. However, the role of Id2 in CD8+ T-cell exhaustion is unclear. Here, we found that Id2 transcriptionally and epigenetically regulates the generation of Texprog cells and their conversion to Texterm cells. Genetic deletion of Id2 dampens CD8+ T-cell-mediated immune responses and the maintenance of stem-like CD8+ T-cell subpopulations, suppresses PD-1 blockade and increases tumor susceptibility. Mechanistically, through its HLH domain, Id2 binds and disrupts the assembly of the Tcf3-Tal1 transcriptional regulatory complex, and thus modulates chromatin accessibility at the Slamf6 promoter by preventing the interaction of Tcf3 with the histone lysine demethylase LSD1. Therefore, Id2 increases the abundance of the permissive H3K4me2 mark on the Tcf3-occupied E-boxes in the Slamf6 promoter, modulates chromatin accessibility at the Slamf6 promoter and epigenetically regulates the generation of Slamf6+ Texprog cells. An LSD1 inhibitor GSK2879552 can rescue the Id2 knockout phenotype in tumor-bearing mice. Inhibition of LSD1 increases the abundance of Slamf6+Tim-3− Texprog cells in tumors and the expression level of Tcf1 in Id2-deleted CD8+ T cells. This study demonstrates that Id2-mediated transcriptional and epigenetic modification drives hierarchical CD8+ T-cell exhaustion, and the mechanistic insights gained may have implications for therapeutic intervention with tumor immune evasion.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-023-01118-6.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139574348","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
COVID-19 vaccines and beyond COVID-19 疫苗及其他
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-26 DOI: 10.1038/s41423-024-01132-2
Yiyuan Liu, Danying Li, Jiahuai Han
{"title":"COVID-19 vaccines and beyond","authors":"Yiyuan Liu, Danying Li, Jiahuai Han","doi":"10.1038/s41423-024-01132-2","DOIUrl":"10.1038/s41423-024-01132-2","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139563810","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
Disulfiram ameliorates STING/MITA-dependent inflammation and autoimmunity by targeting RNF115 二硫仑通过靶向 RNF115 改善 STING/MITA 依赖性炎症和自身免疫。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-24 DOI: 10.1038/s41423-024-01131-3
Zhi-Dong Zhang, Chang-Rui Shi, Fang-Xu Li, Hu Gan, Yanhong Wei, Qianhui Zhang, Xin Shuai, Min Chen, Yu-Lin Lin, Tian-Chen Xiong, Xiaoqi Chen, Bo Zhong, Dandan Lin
STING (also known as MITA) is an adaptor protein that mediates cytoplasmic DNA-triggered signaling, and aberrant activation of STING/MITA by cytosolic self-DNA or gain-of-function mutations causes severe inflammation. Here, we show that STING-mediated inflammation and autoimmunity are promoted by RNF115 and alleviated by the RNF115 inhibitor disulfiram (DSF). Knockout of RNF115 or treatment with DSF significantly inhibit systemic inflammation and autoimmune lethality and restore immune cell development in Trex1–/– mice and STINGN153S/WT bone marrow chimeric mice. In addition, knockdown or pharmacological inhibition of RNF115 substantially downregulate the expression of IFN-α, IFN-γ and proinflammatory cytokines in PBMCs from patients with systemic lupus erythematosus (SLE) who exhibit high concentrations of dsDNA in peripheral blood. Mechanistically, knockout or inhibition of RNF115 impair the oligomerization and Golgi localization of STING in various types of cells transfected with cGAMP and in organs and cells from Trex1–/– mice. Interestingly, knockout of RNF115 inhibits the activation and Golgi localization of STINGN153S as well as the expression of proinflammatory cytokines in myeloid cells but not in endothelial cells or fibroblasts. Taken together, these findings highlight the RNF115-mediated cell type-specific regulation of STING and STINGN153S and provide potential targeted intervention strategies for STING-related autoimmune diseases.
STING(又称 MITA)是一种适配蛋白,可介导细胞质 DNA 触发的信号转导,细胞质自 DNA 或功能增益突变对 STING/MITA 的异常激活会导致严重的炎症。在这里,我们发现 RNF115 能促进 STING 介导的炎症和自身免疫,而 RNF115 抑制剂双硫仑(DSF)能减轻炎症和自身免疫。在 Trex1-/- 小鼠和 STINGN153S/WT 骨髓嵌合小鼠中,敲除 RNF115 或用 DSF 治疗可显著抑制全身炎症和自身免疫致死,并恢复免疫细胞的发育。此外,系统性红斑狼疮(SLE)患者的外周血中dsDNA浓度较高,敲除或药物抑制RNF115可大幅下调这些患者的PBMC中IFN-α、IFN-γ和促炎细胞因子的表达。从机理上讲,在转染了 cGAMP 的各类细胞中以及在 Trex1-/- 小鼠的器官和细胞中,敲除或抑制 RNF115 会损害 STING 的寡聚化和高尔基体定位。有趣的是,敲除 RNF115 可抑制 STINGN153S 的活化和高尔基体定位以及促炎细胞因子在骨髓细胞中的表达,但不能抑制内皮细胞或成纤维细胞中的表达。综上所述,这些发现突显了 RNF115 介导的 STING 和 STINGN153S 的细胞类型特异性调控,并为 STING 相关的自身免疫性疾病提供了潜在的靶向干预策略。
{"title":"Disulfiram ameliorates STING/MITA-dependent inflammation and autoimmunity by targeting RNF115","authors":"Zhi-Dong Zhang, Chang-Rui Shi, Fang-Xu Li, Hu Gan, Yanhong Wei, Qianhui Zhang, Xin Shuai, Min Chen, Yu-Lin Lin, Tian-Chen Xiong, Xiaoqi Chen, Bo Zhong, Dandan Lin","doi":"10.1038/s41423-024-01131-3","DOIUrl":"10.1038/s41423-024-01131-3","url":null,"abstract":"STING (also known as MITA) is an adaptor protein that mediates cytoplasmic DNA-triggered signaling, and aberrant activation of STING/MITA by cytosolic self-DNA or gain-of-function mutations causes severe inflammation. Here, we show that STING-mediated inflammation and autoimmunity are promoted by RNF115 and alleviated by the RNF115 inhibitor disulfiram (DSF). Knockout of RNF115 or treatment with DSF significantly inhibit systemic inflammation and autoimmune lethality and restore immune cell development in Trex1–/– mice and STINGN153S/WT bone marrow chimeric mice. In addition, knockdown or pharmacological inhibition of RNF115 substantially downregulate the expression of IFN-α, IFN-γ and proinflammatory cytokines in PBMCs from patients with systemic lupus erythematosus (SLE) who exhibit high concentrations of dsDNA in peripheral blood. Mechanistically, knockout or inhibition of RNF115 impair the oligomerization and Golgi localization of STING in various types of cells transfected with cGAMP and in organs and cells from Trex1–/– mice. Interestingly, knockout of RNF115 inhibits the activation and Golgi localization of STINGN153S as well as the expression of proinflammatory cytokines in myeloid cells but not in endothelial cells or fibroblasts. Taken together, these findings highlight the RNF115-mediated cell type-specific regulation of STING and STINGN153S and provide potential targeted intervention strategies for STING-related autoimmune diseases.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139545825","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
SARS-CoV-2 immunity SARS-CoV-2 免疫
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-18 DOI: 10.1038/s41423-024-01128-y
Antonio Bertoletti
{"title":"SARS-CoV-2 immunity","authors":"Antonio Bertoletti","doi":"10.1038/s41423-024-01128-y","DOIUrl":"10.1038/s41423-024-01128-y","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139490130","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
SARS-CoV-2 immunity in animal models 动物模型中的 SARS-CoV-2 免疫力。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-18 DOI: 10.1038/s41423-023-01122-w
Zhao Chen, Yaochang Yuan, Qingtao Hu, Airu Zhu, Fenghua Chen, Shu Li, Xin Guan, Chao Lv, Tian Tang, Yiyun He, Jinling Cheng, Jie Zheng, Xiaoyu Hu, Jingxian Zhao, Jincun Zhao, Jing Sun
The COVID-19 pandemic, which was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a worldwide health crisis due to its transmissibility. SARS-CoV-2 infection results in severe respiratory illness and can lead to significant complications in affected individuals. These complications encompass symptoms such as coughing, respiratory distress, fever, infectious shock, acute respiratory distress syndrome (ARDS), and even multiple-organ failure. Animal models serve as crucial tools for investigating pathogenic mechanisms, immune responses, immune escape mechanisms, antiviral drug development, and vaccines against SARS-CoV-2. Currently, various animal models for SARS-CoV-2 infection, such as nonhuman primates (NHPs), ferrets, hamsters, and many different mouse models, have been developed. Each model possesses distinctive features and applications. In this review, we elucidate the immune response elicited by SARS-CoV-2 infection in patients and provide an overview of the characteristics of various animal models mainly used for SARS-CoV-2 infection, as well as the corresponding immune responses and applications of these models. A comparative analysis of transcriptomic alterations in the lungs from different animal models revealed that the K18-hACE2 and mouse-adapted virus mouse models exhibited the highest similarity with the deceased COVID-19 patients. Finally, we highlighted the current gaps in related research between animal model studies and clinical investigations, underscoring lingering scientific questions that demand further clarification.
由严重急性呼吸系统综合症冠状病毒 2(SARS-CoV-2)引起的 COVID-19 大流行因其传播性而成为世界性的健康危机。SARS-CoV-2 感染会导致严重的呼吸道疾病,并可能给患者带来严重的并发症。这些并发症包括咳嗽、呼吸窘迫、发热、感染性休克、急性呼吸窘迫综合征(ARDS)甚至多器官衰竭等症状。动物模型是研究致病机制、免疫反应、免疫逃逸机制、抗病毒药物开发和 SARS-CoV-2 疫苗的重要工具。目前,已开发出多种感染 SARS-CoV-2 的动物模型,如非人灵长类动物(NHPs)、雪貂、仓鼠和许多不同的小鼠模型。每种模型都具有不同的特点和应用。在这篇综述中,我们阐明了患者感染 SARS-CoV-2 所引起的免疫反应,并概述了主要用于 SARS-CoV-2 感染的各种动物模型的特点,以及这些模型的相应免疫反应和应用。对不同动物模型肺部转录组变化的比较分析表明,K18-hACE2和小鼠适应病毒小鼠模型与COVID-19死亡患者的相似度最高。最后,我们强调了目前相关研究在动物模型研究与临床研究之间存在的差距,强调了有待进一步澄清的科学问题。
{"title":"SARS-CoV-2 immunity in animal models","authors":"Zhao Chen, Yaochang Yuan, Qingtao Hu, Airu Zhu, Fenghua Chen, Shu Li, Xin Guan, Chao Lv, Tian Tang, Yiyun He, Jinling Cheng, Jie Zheng, Xiaoyu Hu, Jingxian Zhao, Jincun Zhao, Jing Sun","doi":"10.1038/s41423-023-01122-w","DOIUrl":"10.1038/s41423-023-01122-w","url":null,"abstract":"The COVID-19 pandemic, which was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a worldwide health crisis due to its transmissibility. SARS-CoV-2 infection results in severe respiratory illness and can lead to significant complications in affected individuals. These complications encompass symptoms such as coughing, respiratory distress, fever, infectious shock, acute respiratory distress syndrome (ARDS), and even multiple-organ failure. Animal models serve as crucial tools for investigating pathogenic mechanisms, immune responses, immune escape mechanisms, antiviral drug development, and vaccines against SARS-CoV-2. Currently, various animal models for SARS-CoV-2 infection, such as nonhuman primates (NHPs), ferrets, hamsters, and many different mouse models, have been developed. Each model possesses distinctive features and applications. In this review, we elucidate the immune response elicited by SARS-CoV-2 infection in patients and provide an overview of the characteristics of various animal models mainly used for SARS-CoV-2 infection, as well as the corresponding immune responses and applications of these models. A comparative analysis of transcriptomic alterations in the lungs from different animal models revealed that the K18-hACE2 and mouse-adapted virus mouse models exhibited the highest similarity with the deceased COVID-19 patients. Finally, we highlighted the current gaps in related research between animal model studies and clinical investigations, underscoring lingering scientific questions that demand further clarification.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10806257/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139490019","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
CD8+ T cell metabolic flexibility elicited by CD28-ARS2 axis-driven alternative splicing of PKM supports antitumor immunity CD28-ARS2 轴驱动的 PKM 替代剪接激发的 CD8+ T 细胞代谢灵活性支持抗肿瘤免疫。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-18 DOI: 10.1038/s41423-024-01124-2
G. Aaron Holling, Colin A. Chavel, Anand P. Sharda, Mackenzie M. Lieberman, Caitlin M. James, Shivana M. Lightman, Jason H. Tong, Guanxi Qiao, Tiffany R. Emmons, Thejaswini Giridharan, Shengqi Hou, Andrew M. Intlekofer, Richard M. Higashi, Teresa W. M. Fan, Andrew N. Lane, Kevin H. Eng, Brahm H. Segal, Elizabeth A. Repasky, Kelvin P. Lee, Scott H. Olejniczak
Metabolic flexibility has emerged as a critical determinant of CD8+ T-cell antitumor activity, yet the mechanisms driving the metabolic flexibility of T cells have not been determined. In this study, we investigated the influence of the nuclear cap-binding complex (CBC) adaptor protein ARS2 on mature T cells. In doing so, we discovered a novel signaling axis that endows activated CD8+ T cells with flexibility of glucose catabolism. ARS2 upregulation driven by CD28 signaling reinforced splicing factor recruitment to pre-mRNAs and affected approximately one-third of T-cell activation-induced alternative splicing events. Among these effects, the CD28-ARS2 axis suppressed the expression of the M1 isoform of pyruvate kinase in favor of PKM2, a key determinant of CD8+ T-cell glucose utilization, interferon gamma production, and antitumor effector function. Importantly, PKM alternative splicing occurred independently of CD28-driven PI3K pathway activation, revealing a novel means by which costimulation reprograms glucose metabolism in CD8+ T cells.
代谢灵活性已成为 CD8+ T 细胞抗肿瘤活性的一个关键决定因素,但驱动 T 细胞代谢灵活性的机制尚未确定。在这项研究中,我们研究了核帽结合复合体(CBC)适配蛋白 ARS2 对成熟 T 细胞的影响。在此过程中,我们发现了一个新的信号轴,它赋予活化的 CD8+ T 细胞葡萄糖分解代谢的灵活性。CD28 信号驱动的 ARS2 上调加强了剪接因子对前核糖核酸的招募,并影响了大约三分之一的 T 细胞活化诱导的替代剪接事件。在这些影响中,CD28-ARS2 轴抑制了丙酮酸激酶 M1 异构体的表达,而有利于 PKM2 的表达,PKM2 是 CD8+ T 细胞葡萄糖利用、γ 干扰素产生和抗肿瘤效应功能的关键决定因素。重要的是,PKM 替代剪接的发生与 CD28 驱动的 PI3K 通路激活无关,揭示了成本刺激重新规划 CD8+ T 细胞葡萄糖代谢的一种新方法。
{"title":"CD8+ T cell metabolic flexibility elicited by CD28-ARS2 axis-driven alternative splicing of PKM supports antitumor immunity","authors":"G. Aaron Holling, Colin A. Chavel, Anand P. Sharda, Mackenzie M. Lieberman, Caitlin M. James, Shivana M. Lightman, Jason H. Tong, Guanxi Qiao, Tiffany R. Emmons, Thejaswini Giridharan, Shengqi Hou, Andrew M. Intlekofer, Richard M. Higashi, Teresa W. M. Fan, Andrew N. Lane, Kevin H. Eng, Brahm H. Segal, Elizabeth A. Repasky, Kelvin P. Lee, Scott H. Olejniczak","doi":"10.1038/s41423-024-01124-2","DOIUrl":"10.1038/s41423-024-01124-2","url":null,"abstract":"Metabolic flexibility has emerged as a critical determinant of CD8+ T-cell antitumor activity, yet the mechanisms driving the metabolic flexibility of T cells have not been determined. In this study, we investigated the influence of the nuclear cap-binding complex (CBC) adaptor protein ARS2 on mature T cells. In doing so, we discovered a novel signaling axis that endows activated CD8+ T cells with flexibility of glucose catabolism. ARS2 upregulation driven by CD28 signaling reinforced splicing factor recruitment to pre-mRNAs and affected approximately one-third of T-cell activation-induced alternative splicing events. Among these effects, the CD28-ARS2 axis suppressed the expression of the M1 isoform of pyruvate kinase in favor of PKM2, a key determinant of CD8+ T-cell glucose utilization, interferon gamma production, and antitumor effector function. Importantly, PKM alternative splicing occurred independently of CD28-driven PI3K pathway activation, revealing a novel means by which costimulation reprograms glucose metabolism in CD8+ T cells.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01124-2.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139485135","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
Tissue-resident macrophages exacerbate lung injury after remote sterile damage 组织驻留的巨噬细胞会加重远端无菌损伤后的肺损伤。
IF 24.1 1区 医学 Q1 Medicine Pub Date : 2024-01-17 DOI: 10.1038/s41423-024-01125-1
Hanhui Zhong, Jingjing Ji, Jinling Zhuang, Ziying Xiong, Pengyun Xie, Xiaolei Liu, Jundi Zheng, Wangli Tian, Xiaoyang Hong, Jing Tang
Remote organ injury, which is a common secondary complication of sterile tissue damage, is a major cause of poor prognosis and is difficult to manage. Here, we report the critical role of tissue-resident macrophages in lung injury after trauma or stroke through the inflammatory response. We found that depleting tissue-resident macrophages rather than disrupting the recruitment of monocyte-derived macrophages attenuated lung injury after trauma or stroke. Our findings revealed that the release of circulating alarmins from sites of distant sterile tissue damage triggered an inflammatory response in lung-resident macrophages by binding to receptor for advanced glycation end products (RAGE) on the membrane, which activated epidermal growth factor receptor (EGFR). Mechanistically, ligand-activated RAGE triggered EGFR activation through an interaction, leading to Rab5-mediated RAGE internalization and EGFR phosphorylation, which subsequently recruited and activated P38; this, in turn, promoted RAGE translation and trafficking to the plasma membrane to increase the cellular response to RAGE ligands, consequently exacerbating inflammation. Our study also showed that the loss of RAGE or EGFR expression by adoptive transfer of macrophages, blocking the function of RAGE with a neutralizing antibody, or pharmacological inhibition of EGFR activation in macrophages could protect against trauma- or stroke-induced remote lung injury. Therefore, our study revealed that targeting the RAGE-EGFR signaling pathway in tissue-resident macrophages is a potential therapeutic approach for treating secondary complications of sterile damage.
远端器官损伤是无菌组织损伤的常见继发性并发症,是导致预后不良的主要原因,而且难以控制。在此,我们报告了组织驻留巨噬细胞通过炎症反应在创伤或中风后肺损伤中的关键作用。我们发现,耗竭组织驻留的巨噬细胞而不是破坏单核细胞衍生的巨噬细胞的招募可减轻创伤或中风后的肺损伤。我们的研究结果表明,远处无菌组织损伤部位释放的循环alarmins通过与膜上的高级糖化终产物受体(RAGE)结合,激活表皮生长因子受体(EGFR),从而引发肺驻留巨噬细胞的炎症反应。从机理上讲,配体激活的 RAGE 通过相互作用触发表皮生长因子受体活化,导致 Rab5 介导的 RAGE 内化和表皮生长因子受体磷酸化,进而招募和激活 P38;这反过来又促进了 RAGE 翻译和向质膜的迁移,增加了细胞对 RAGE 配体的反应,从而加剧了炎症。我们的研究还表明,通过收养性转移巨噬细胞、用中和抗体阻断 RAGE 的功能或药物抑制巨噬细胞中表皮生长因子受体的活化来减少 RAGE 或表皮生长因子受体的表达,可以防止创伤或中风引起的远端肺损伤。因此,我们的研究表明,靶向组织驻留巨噬细胞中的RAGE-EGFR信号通路是治疗无菌性损伤继发性并发症的一种潜在治疗方法。
{"title":"Tissue-resident macrophages exacerbate lung injury after remote sterile damage","authors":"Hanhui Zhong, Jingjing Ji, Jinling Zhuang, Ziying Xiong, Pengyun Xie, Xiaolei Liu, Jundi Zheng, Wangli Tian, Xiaoyang Hong, Jing Tang","doi":"10.1038/s41423-024-01125-1","DOIUrl":"10.1038/s41423-024-01125-1","url":null,"abstract":"Remote organ injury, which is a common secondary complication of sterile tissue damage, is a major cause of poor prognosis and is difficult to manage. Here, we report the critical role of tissue-resident macrophages in lung injury after trauma or stroke through the inflammatory response. We found that depleting tissue-resident macrophages rather than disrupting the recruitment of monocyte-derived macrophages attenuated lung injury after trauma or stroke. Our findings revealed that the release of circulating alarmins from sites of distant sterile tissue damage triggered an inflammatory response in lung-resident macrophages by binding to receptor for advanced glycation end products (RAGE) on the membrane, which activated epidermal growth factor receptor (EGFR). Mechanistically, ligand-activated RAGE triggered EGFR activation through an interaction, leading to Rab5-mediated RAGE internalization and EGFR phosphorylation, which subsequently recruited and activated P38; this, in turn, promoted RAGE translation and trafficking to the plasma membrane to increase the cellular response to RAGE ligands, consequently exacerbating inflammation. Our study also showed that the loss of RAGE or EGFR expression by adoptive transfer of macrophages, blocking the function of RAGE with a neutralizing antibody, or pharmacological inhibition of EGFR activation in macrophages could protect against trauma- or stroke-induced remote lung injury. Therefore, our study revealed that targeting the RAGE-EGFR signaling pathway in tissue-resident macrophages is a potential therapeutic approach for treating secondary complications of sterile damage.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":24.1,"publicationDate":"2024-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139477531","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