首页 > 最新文献

Immunity最新文献

英文 中文
Mast cells control lung type 2 inflammation via prostaglandin E2-driven soluble ST2 肥大细胞通过前列腺素 E2 驱动的可溶性 ST2 控制肺部 2 型炎症
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-30 DOI: 10.1016/j.immuni.2024.05.003
Kinan Alhallak, Jun Nagai, Kendall Zaleski, Sofia Marshall, Tamara Salloum, Tahereh Derakhshan, Hiroaki Hayashi, Chunli Feng, Radomir Kratchmarov, Juying Lai, Virinchi Kuchibhotla, Airi Nishida, Barbara Balestrieri, Tanya Laidlaw, Daniel F. Dwyer, Joshua A. Boyce

Severe asthma and sinus disease are consequences of type 2 inflammation (T2I), mediated by interleukin (IL)-33 signaling through its membrane-bound receptor, ST2. Soluble (s)ST2 reduces available IL-33 and limits T2I, but little is known about its regulation. We demonstrate that prostaglandin E2 (PGE2) drives production of sST2 to limit features of lung T2I. PGE2-deficient mice display diminished sST2. In humans with severe respiratory T2I, urinary PGE2 metabolites correlate with serum sST2. In mice, PGE2 enhanced sST2 secretion by mast cells (MCs). Mice lacking MCs, ST2 expression by MCs, or E prostanoid (EP)2 receptors by MCs showed reduced sST2 lung concentrations and strong T2I. Recombinant sST2 reduced T2I in mice lacking PGE2 or ST2 expression by MCs back to control levels. PGE2 deficiency also reversed the hyperinflammatory phenotype in mice lacking ST2 expression by MCs. PGE2 thus suppresses T2I through MC-derived sST2, explaining the severe T2I observed in low PGE2 states.

严重哮喘和鼻窦疾病是 2 型炎症(T2I)的后果,由白细胞介素(IL)-33 通过其膜结合受体 ST2 发出信号介导。可溶性(s)ST2 可减少可用的 IL-33 并限制 T2I,但人们对其调控知之甚少。我们证明,前列腺素 E2(PGE2)能驱动 sST2 的产生,从而限制肺 T2I 的特征。PGE2 缺乏的小鼠显示出 sST2 的减少。在患有严重呼吸道 T2I 的人类中,尿中 PGE2 代谢物与血清 sST2 相关。在小鼠中,PGE2 可增强肥大细胞(MCs)分泌 sST2。缺乏肥大细胞、肥大细胞 ST2 表达或肥大细胞 E 类前列腺素(EP)2 受体的小鼠显示出 sST2 肺浓度降低和强烈的 T2I。重组 sST2 可使缺乏 PGE2 或 MCs 缺乏 ST2 表达的小鼠的 T2I 降低到控制水平。PGE2 的缺乏也逆转了缺乏由 MCs 表达 ST2 的小鼠的高炎症表型。因此,PGE2 可通过 MC 衍生的 sST2 抑制 T2I,从而解释在低 PGE2 状态下观察到的严重 T2I。
{"title":"Mast cells control lung type 2 inflammation via prostaglandin E2-driven soluble ST2","authors":"Kinan Alhallak, Jun Nagai, Kendall Zaleski, Sofia Marshall, Tamara Salloum, Tahereh Derakhshan, Hiroaki Hayashi, Chunli Feng, Radomir Kratchmarov, Juying Lai, Virinchi Kuchibhotla, Airi Nishida, Barbara Balestrieri, Tanya Laidlaw, Daniel F. Dwyer, Joshua A. Boyce","doi":"10.1016/j.immuni.2024.05.003","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.05.003","url":null,"abstract":"<p>Severe asthma and sinus disease are consequences of type 2 inflammation (T2I), mediated by interleukin (IL)-33 signaling through its membrane-bound receptor, ST2. Soluble (s)ST2 reduces available IL-33 and limits T2I, but little is known about its regulation. We demonstrate that prostaglandin E<sub>2</sub> (PGE<sub>2</sub>) drives production of sST2 to limit features of lung T2I. PGE<sub>2</sub>-deficient mice display diminished sST2. In humans with severe respiratory T2I, urinary PGE<sub>2</sub> metabolites correlate with serum sST2. In mice, PGE<sub>2</sub> enhanced sST2 secretion by mast cells (MCs). Mice lacking MCs, ST2 expression by MCs, or E prostanoid (EP)<sub>2</sub> receptors by MCs showed reduced sST2 lung concentrations and strong T2I. Recombinant sST2 reduced T2I in mice lacking PGE<sub>2</sub> or ST2 expression by MCs back to control levels. PGE<sub>2</sub> deficiency also reversed the hyperinflammatory phenotype in mice lacking ST2 expression by MCs. PGE<sub>2</sub> thus suppresses T2I through MC-derived sST2, explaining the severe T2I observed in low PGE<sub>2</sub> states.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141177924","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
Intestinal CXCR6+ ILC3s migrate to the kidney and exacerbate renal fibrosis via IL-23 receptor signaling enhanced by PD-1 expression 肠道CXCR6+ ILC3迁移到肾脏并通过PD-1表达增强的IL-23受体信号加重肾脏纤维化
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-29 DOI: 10.1016/j.immuni.2024.05.004
Zhou Liang, Ziwen Tang, Changjian Zhu, Feng Li, Shuaijiabin Chen, Xu Han, Ruilin Zheng, Xinrong Hu, Ruoni Lin, Qiaoqiao Pei, Changjun Yin, Ji Wang, Ce Tang, Nan Cao, Jincun Zhao, Rong Wang, Xiaoyan Li, Ning Luo, Qiong Wen, Jianwen Yu, Yi Zhou

Group 3 innate lymphoid cells (ILC3s) regulate inflammation and tissue repair at mucosal sites, but whether these functions pertain to other tissues—like the kidneys—remains unclear. Here, we observed that renal fibrosis in humans was associated with increased ILC3s in the kidneys and blood. In mice, we showed that CXCR6+ ILC3s rapidly migrated from the intestinal mucosa and accumulated in the kidney via CXCL16 released from the injured tubules. Within the fibrotic kidney, ILC3s increased the expression of programmed cell death-1 (PD-1) and subsequent IL-17A production to directly activate myofibroblasts and fibrotic niche formation. ILC3 expression of PD-1 inhibited IL-23R endocytosis and consequently amplified the JAK2/STAT3/RORγt/IL-17A pathway that was essential for the pro-fibrogenic effect of ILC3s. Thus, we reveal a hitherto unrecognized migration pathway of ILC3s from the intestine to the kidney and the PD-1-dependent function of ILC3s in promoting renal fibrosis.

第 3 组先天性淋巴细胞(ILC3s)能调节粘膜部位的炎症和组织修复,但这些功能是否与其他组织(如肾脏)有关尚不清楚。在这里,我们观察到人类肾脏纤维化与肾脏和血液中 ILC3 的增加有关。在小鼠体内,我们发现 CXCR6+ ILC3 从肠粘膜迅速迁移,并通过损伤肾小管释放的 CXCL16 在肾脏中聚集。在纤维化的肾脏中,ILC3 增加了程序性细胞死亡-1(PD-1)的表达,随后产生了 IL-17A,直接激活了肌成纤维细胞和纤维化龛的形成。ILC3表达的PD-1抑制了IL-23R的内吞,从而扩大了JAK2/STAT3/RORγt/IL-17A途径,而该途径是ILC3s促纤维化效应的关键。因此,我们揭示了迄今尚未认识到的 ILC3 从肠道到肾脏的迁移途径,以及 ILC3 在促进肾脏纤维化中的 PD-1 依赖性功能。
{"title":"Intestinal CXCR6+ ILC3s migrate to the kidney and exacerbate renal fibrosis via IL-23 receptor signaling enhanced by PD-1 expression","authors":"Zhou Liang, Ziwen Tang, Changjian Zhu, Feng Li, Shuaijiabin Chen, Xu Han, Ruilin Zheng, Xinrong Hu, Ruoni Lin, Qiaoqiao Pei, Changjun Yin, Ji Wang, Ce Tang, Nan Cao, Jincun Zhao, Rong Wang, Xiaoyan Li, Ning Luo, Qiong Wen, Jianwen Yu, Yi Zhou","doi":"10.1016/j.immuni.2024.05.004","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.05.004","url":null,"abstract":"<p>Group 3 innate lymphoid cells (ILC3s) regulate inflammation and tissue repair at mucosal sites, but whether these functions pertain to other tissues—like the kidneys—remains unclear. Here, we observed that renal fibrosis in humans was associated with increased ILC3s in the kidneys and blood. In mice, we showed that CXCR6<sup>+</sup> ILC3s rapidly migrated from the intestinal mucosa and accumulated in the kidney via CXCL16 released from the injured tubules. Within the fibrotic kidney, ILC3s increased the expression of programmed cell death-1 (PD-1) and subsequent IL-17A production to directly activate myofibroblasts and fibrotic niche formation. ILC3 expression of PD-1 inhibited IL-23R endocytosis and consequently amplified the JAK2/STAT3/RORγt/IL-17A pathway that was essential for the pro-fibrogenic effect of ILC3s. Thus, we reveal a hitherto unrecognized migration pathway of ILC3s from the intestine to the kidney and the PD-1-dependent function of ILC3s in promoting renal fibrosis.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141177980","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
Breadth of Fc-mediated effector function correlates with clinical immunity following human malaria challenge Fc 介导的效应器功能的广度与人类疟疾挑战后的临床免疫力有关
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-23 DOI: 10.1016/j.immuni.2024.05.001
Irene N. Nkumama, Rodney Ogwang, Dennis Odera, Fauzia Musasia, Kennedy Mwai, Lydia Nyamako, Linda Murungi, James Tuju, Kristin Fürle, Micha Rosenkranz, Rinter Kimathi, Patricia Njuguna, Mainga Hamaluba, Melissa C. Kapulu, Roland Frank, , Faith H.A. Osier

Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of Plasmodium falciparum. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.

疟疾是一种威胁生命的疾病,对全球健康具有重要意义,尤其是在撒哈拉以南非洲地区。生长抑素测定 (GIA) 通常用于评估、优先考虑和量化疟疾血期候选疫苗的功效,但不能可靠地预测自然获得或疫苗诱导的保护。在半免疫志愿者中进行的受控人类疟疾挑战研究提供了一个无与伦比的机会,可以有力地确定保护的机理相关因素。我们利用这一平台,对与恶性疟原虫红细胞裂殖阶段免疫相关的七种功能抗体检测进行了正面比较。Fc 介导的效应功能与疟疾临床症状和寄生虫指数繁殖的保护密切相关,而金标准 GIA 则不然。Fc 介导的效应物功能的广度可区分挑战后的临床免疫力。这些发现改变了人们对疟疾免疫机制的认识,对疫苗开发具有重要意义。
{"title":"Breadth of Fc-mediated effector function correlates with clinical immunity following human malaria challenge","authors":"Irene N. Nkumama, Rodney Ogwang, Dennis Odera, Fauzia Musasia, Kennedy Mwai, Lydia Nyamako, Linda Murungi, James Tuju, Kristin Fürle, Micha Rosenkranz, Rinter Kimathi, Patricia Njuguna, Mainga Hamaluba, Melissa C. Kapulu, Roland Frank, , Faith H.A. Osier","doi":"10.1016/j.immuni.2024.05.001","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.05.001","url":null,"abstract":"<p>Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of <em>Plasmodium falciparum</em>. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141085717","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 RIPK1-specific PROTAC degrader achieves potent antitumor activity by enhancing immunogenic cell death RIPK1特异性PROTAC降解剂通过增强免疫原性细胞死亡实现强效抗肿瘤活性
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-23 DOI: 10.1016/j.immuni.2024.04.025
Jonathan Mannion, Valentina Gifford, Benjamin Bellenie, Winnie Fernando, Laura Ramos Garcia, Rebecca Wilson, Sidonie Wicky John, Savita Udainiya, Emmanuel C. Patin, Crescens Tiu, Angel Smith, Maria Goicoechea, Andrew Craxton, Nathalia Moraes de Vasconcelos, Naomi Guppy, Kwai-Ming J. Cheung, Nicholas J. Cundy, Olivier Pierrat, Alfie Brennan, Theodoros I. Roumeliotis, Pascal Meier

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.

与受体相互作用的丝氨酸/苏氨酸蛋白激酶1(RIPK1)是协调细胞存活、炎症和免疫原性细胞死亡(ICD)的重要应激哨兵。虽然触发细胞死亡需要 RIPK1 的催化功能,但它的非催化支架功能可介导强大的促生存信号。因此,癌细胞可以劫持 RIPK1 来阻断坏死并逃避免疫检测。我们生成了一种小分子蛋白水解靶向嵌合体(PROTAC),它能选择性地降解人和小鼠的RIPK1。PROTAC 介导的 RIPK1 降解会降低 TNFR1 和 TLR3/4 信号转导中枢的功能,从而增强 NF-κB、MAPK 和 IFN 信号的输出。此外,RIPK1 的降解同时促进了 RIPK3 的激活和坏死诱导。我们进一步证实,RIPK1 降解通过使癌细胞对治疗诱导的 TNF 和干扰素敏感,增强了放射治疗和免疫治疗的免疫刺激作用。这促进了ICD、抗肿瘤免疫和持久的治疗反应。因此,用 PROTACs 靶向 RIPK1 是克服放射或免疫疗法耐药性和增强抗癌疗法的一种很有前途的方法。
{"title":"A RIPK1-specific PROTAC degrader achieves potent antitumor activity by enhancing immunogenic cell death","authors":"Jonathan Mannion, Valentina Gifford, Benjamin Bellenie, Winnie Fernando, Laura Ramos Garcia, Rebecca Wilson, Sidonie Wicky John, Savita Udainiya, Emmanuel C. Patin, Crescens Tiu, Angel Smith, Maria Goicoechea, Andrew Craxton, Nathalia Moraes de Vasconcelos, Naomi Guppy, Kwai-Ming J. Cheung, Nicholas J. Cundy, Olivier Pierrat, Alfie Brennan, Theodoros I. Roumeliotis, Pascal Meier","doi":"10.1016/j.immuni.2024.04.025","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.025","url":null,"abstract":"<p>Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141085788","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
Metabolic regulator LKB1 controls adipose tissue ILC2 PD-1 expression and mitochondrial homeostasis to prevent insulin resistance 代谢调节因子 LKB1 控制脂肪组织 ILC2 PD-1 的表达和线粒体稳态,以防止胰岛素抵抗
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-20 DOI: 10.1016/j.immuni.2024.04.024
Jiping Sun, Youqin Zhang, Qingbing Zhang, Lin Hu, Linfeng Zhao, Hongdong Wang, Yue Yuan, Hongshen Niu, Dongdi Wang, Huasheng Zhang, Jianyue Liu, Xujiao Feng, Xiaohui Su, Ju Qiu, Jing Sun, Heping Xu, Catherine Zhang, Kathleen Wang, Yan Bi, Edgar G. Engleman, Lei Shen

Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.

脂肪组织第 2 组先天性淋巴细胞(ILC2)通过维持 2 型免疫和促进脂肪变性来帮助维持代谢平衡。虽然 ILC2 区系的损伤会导致肥胖相关的胰岛素抵抗,但其潜在机制尚未阐明。在这里,我们发现肥胖小鼠和人类的 ILC2 表现出肝脏激酶 B1(LKB1)激活受损。LKB1 的基因消减破坏了 ILC2 的线粒体代谢,抑制了 ILC2 的反应,导致胰岛素抵抗加剧。从机制上讲,LKB1 缺乏会通过激活 NFAT 诱导 PD-1 的异常表达,而 NFAT 又会通过抑制 Bcl-xL 的表达来增强有丝分裂。阻断 PD-1 可恢复 ILC2 的正常功能,并逆转肥胖诱导的小鼠胰岛素抵抗。总之,这些数据表明,LKB1-PD-1 轴是治疗代谢性疾病的一个很有前景的治疗靶点。
{"title":"Metabolic regulator LKB1 controls adipose tissue ILC2 PD-1 expression and mitochondrial homeostasis to prevent insulin resistance","authors":"Jiping Sun, Youqin Zhang, Qingbing Zhang, Lin Hu, Linfeng Zhao, Hongdong Wang, Yue Yuan, Hongshen Niu, Dongdi Wang, Huasheng Zhang, Jianyue Liu, Xujiao Feng, Xiaohui Su, Ju Qiu, Jing Sun, Heping Xu, Catherine Zhang, Kathleen Wang, Yan Bi, Edgar G. Engleman, Lei Shen","doi":"10.1016/j.immuni.2024.04.024","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.024","url":null,"abstract":"<p>Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141069219","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
Metabolic coordination between skin epithelium and type 17 immunity sustains chronic skin inflammation 皮肤上皮与 17 型免疫之间的代谢协调可维持慢性皮肤炎症
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-20 DOI: 10.1016/j.immuni.2024.04.022
Ipsita Subudhi, Piotr Konieczny, Aleksandr Prystupa, Rochelle L. Castillo, Erica Sze-Tu, Yue Xing, Daniel Rosenblum, Ilana Reznikov, Ikjot Sidhu, Cynthia Loomis, Catherine P. Lu, Niroshana Anandasabapathy, Mayte Suárez-Fariñas, Johann E. Gudjonsson, Aristotelis Tsirigos, Jose U. Scher, Shruti Naik

Inflammatory epithelial diseases are spurred by the concomitant dysregulation of immune and epithelial cells. How these two dysregulated cellular compartments simultaneously sustain their heightened metabolic demands is unclear. Single-cell and spatial transcriptomics (ST), along with immunofluorescence, revealed that hypoxia-inducible factor 1α (HIF1α), downstream of IL-17 signaling, drove psoriatic epithelial remodeling. Blocking HIF1α in human psoriatic lesions ex vivo impaired glycolysis and phenocopied anti-IL-17 therapy. In a murine model of skin inflammation, epidermal-specific loss of HIF1α or its target gene, glucose transporter 1, ameliorated epidermal, immune, vascular, and neuronal pathology. Mechanistically, glycolysis autonomously fueled epithelial pathology and enhanced lactate production, which augmented the γδ T17 cell response. RORγt-driven genetic deletion or pharmacological inhibition of either lactate-producing enzymes or lactate transporters attenuated epithelial pathology and IL-17A expression in vivo. Our findings identify a metabolic hierarchy between epithelial and immune compartments and the consequent coordination of metabolic processes that sustain inflammatory disease.

免疫细胞和上皮细胞同时失调会引发炎症性上皮疾病。目前还不清楚这两个失调的细胞区如何同时维持它们的高代谢需求。单细胞和空间转录组学(ST)以及免疫荧光发现,IL-17信号下游的缺氧诱导因子1α(HIF1α)驱动了银屑病上皮的重塑。体内阻断人类银屑病皮损中的 HIF1α 会损害糖酵解,并与抗 IL-17 疗法相仿。在小鼠皮肤炎症模型中,表皮特异性缺失 HIF1α 或其靶基因葡萄糖转运体 1 可改善表皮、免疫、血管和神经元病理学。从机理上讲,糖酵解自发地助长了上皮病理变化并增强了乳酸的产生,从而增强了γδ T17 细胞的反应。RORγt驱动的基因缺失或药物抑制乳酸生成酶或乳酸转运体可减轻上皮病理变化和体内IL-17A的表达。我们的研究结果确定了上皮细胞和免疫细胞之间的代谢层次,以及由此产生的维持炎症性疾病的代谢过程的协调。
{"title":"Metabolic coordination between skin epithelium and type 17 immunity sustains chronic skin inflammation","authors":"Ipsita Subudhi, Piotr Konieczny, Aleksandr Prystupa, Rochelle L. Castillo, Erica Sze-Tu, Yue Xing, Daniel Rosenblum, Ilana Reznikov, Ikjot Sidhu, Cynthia Loomis, Catherine P. Lu, Niroshana Anandasabapathy, Mayte Suárez-Fariñas, Johann E. Gudjonsson, Aristotelis Tsirigos, Jose U. Scher, Shruti Naik","doi":"10.1016/j.immuni.2024.04.022","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.022","url":null,"abstract":"<p>Inflammatory epithelial diseases are spurred by the concomitant dysregulation of immune and epithelial cells. How these two dysregulated cellular compartments simultaneously sustain their heightened metabolic demands is unclear. Single-cell and spatial transcriptomics (ST), along with immunofluorescence, revealed that hypoxia-inducible factor 1α (HIF1α), downstream of IL-17 signaling, drove psoriatic epithelial remodeling. Blocking HIF1α in human psoriatic lesions <em>ex vivo</em> impaired glycolysis and phenocopied anti-IL-17 therapy. In a murine model of skin inflammation, epidermal-specific loss of HIF1α or its target gene, glucose transporter 1, ameliorated epidermal, immune, vascular, and neuronal pathology. Mechanistically, glycolysis autonomously fueled epithelial pathology and enhanced lactate production, which augmented the γδ T17 cell response. RORγt-driven genetic deletion or pharmacological inhibition of either lactate-producing enzymes or lactate transporters attenuated epithelial pathology and IL-17A expression <em>in vivo</em>. Our findings identify a metabolic hierarchy between epithelial and immune compartments and the consequent coordination of metabolic processes that sustain inflammatory disease.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141069217","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
Deficiency of CBL and CBLB ubiquitin ligases leads to hyper T follicular helper cell responses and lupus by reducing BCL6 degradation CBL 和 CBLB 泛素连接酶的缺乏会减少 BCL6 的降解,从而导致 T 滤泡辅助细胞反应亢进和红斑狼疮
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-17 DOI: 10.1016/j.immuni.2024.04.023
Xin Li, Weili Sun, Mengxing Huang, Liying Gong, Xiaochen Zhang, Li Zhong, Virginie Calderon, Zhenhua Bian, Yi He, Woong-Kyung Suh, Yang Li, Tengfei Song, Yongrui Zou, Zhe-Xiong Lian, Hua Gu

Recent evidence reveals hyper T follicular helper (Tfh) cell responses in systemic lupus erythematosus (SLE); however, molecular mechanisms responsible for hyper Tfh cell responses and whether they cause SLE are unclear. We found that SLE patients downregulated both ubiquitin ligases, casitas B-lineage lymphoma (CBL) and CBLB (CBLs), in CD4+ T cells. T cell-specific CBLs-deficient mice developed hyper Tfh cell responses and SLE, whereas blockade of Tfh cell development in the mutant mice was sufficient to prevent SLE. ICOS was upregulated in SLE Tfh cells, whose signaling increased BCL6 by attenuating BCL6 degradation via chaperone-mediated autophagy (CMA). Conversely, CBLs restrained BCL6 expression by ubiquitinating ICOS. Blockade of BCL6 degradation was sufficient to enhance Tfh cell responses. Thus, the compromised expression of CBLs is a prevalent risk trait shared by SLE patients and causative to hyper Tfh cell responses and SLE. The ICOS-CBLs axis may be a target to treat SLE.

最近的证据显示,系统性红斑狼疮(SLE)患者的T滤泡辅助细胞(Tfh)反应亢进;然而,导致Tfh细胞反应亢进的分子机制以及它们是否会引起系统性红斑狼疮尚不清楚。我们发现,系统性红斑狼疮患者的 CD4+ T 细胞中,泛素连接酶 Casitas B lineage lymphoma(CBL)和 CBLB(CBLs)都出现了下调。T细胞特异性CBLs缺陷小鼠会出现Tfh细胞反应亢进和系统性红斑狼疮,而阻断突变小鼠的Tfh细胞发育足以预防系统性红斑狼疮。ICOS 在系统性红斑狼疮 Tfh 细胞中上调,其信号通过伴侣介导的自噬(CMA)抑制 BCL6 降解,从而增加了 BCL6。相反,CBLs 则通过泛素化 ICOS 来抑制 BCL6 的表达。阻断 BCL6 降解足以增强 Tfh 细胞的反应。因此,CBLs表达受损是系统性红斑狼疮患者共有的普遍风险特征,也是Tfh细胞反应亢进和系统性红斑狼疮的致病因素。ICOS-CBLs轴可能是治疗系统性红斑狼疮的靶点。
{"title":"Deficiency of CBL and CBLB ubiquitin ligases leads to hyper T follicular helper cell responses and lupus by reducing BCL6 degradation","authors":"Xin Li, Weili Sun, Mengxing Huang, Liying Gong, Xiaochen Zhang, Li Zhong, Virginie Calderon, Zhenhua Bian, Yi He, Woong-Kyung Suh, Yang Li, Tengfei Song, Yongrui Zou, Zhe-Xiong Lian, Hua Gu","doi":"10.1016/j.immuni.2024.04.023","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.023","url":null,"abstract":"<p>Recent evidence reveals hyper T follicular helper (Tfh) cell responses in systemic lupus erythematosus (SLE); however, molecular mechanisms responsible for hyper Tfh cell responses and whether they cause SLE are unclear. We found that SLE patients downregulated both ubiquitin ligases, casitas B-lineage lymphoma (CBL) and CBLB (CBLs), in CD4<sup>+</sup> T cells. T cell-specific CBLs-deficient mice developed hyper Tfh cell responses and SLE, whereas blockade of Tfh cell development in the mutant mice was sufficient to prevent SLE. ICOS was upregulated in SLE Tfh cells, whose signaling increased BCL6 by attenuating BCL6 degradation via chaperone-mediated autophagy (CMA). Conversely, CBLs restrained BCL6 expression by ubiquitinating ICOS. Blockade of BCL6 degradation was sufficient to enhance Tfh cell responses. Thus, the compromised expression of CBLs is a prevalent risk trait shared by SLE patients and causative to hyper Tfh cell responses and SLE. The ICOS-CBLs axis may be a target to treat SLE.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140954524","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
CXCL16-dependent scavenging of oxidized lipids by islet macrophages promotes differentiation of pathogenic CD8+ T cells in diabetic autoimmunity 依赖于 CXCL16 的胰岛巨噬细胞清除氧化脂质的作用可促进糖尿病自身免疫中致病性 CD8+ T 细胞的分化
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-15 DOI: 10.1016/j.immuni.2024.04.017
Neetu Srivastava, Hao Hu, Orion J. Peterson, Anthony N. Vomund, Marta Stremska, Mohammad Zaman, Shilpi Giri, Tiandao Li, Cheryl F. Lichti, Pavel N. Zakharov, Bo Zhang, Nada A. Abumrad, Yi-Guang Chen, Kodi S. Ravichandran, Emil R. Unanue, Xiaoxiao Wan

The pancreatic islet microenvironment is highly oxidative, rendering β cells vulnerable to autoinflammatory insults. Here, we examined the role of islet resident macrophages in the autoimmune attack that initiates type 1 diabetes. Islet macrophages highly expressed CXCL16, a chemokine and scavenger receptor for oxidized low-density lipoproteins (OxLDLs), regardless of autoimmune predisposition. Deletion of Cxcl16 in nonobese diabetic (NOD) mice suppressed the development of autoimmune diabetes. Mechanistically, Cxcl16 deficiency impaired clearance of OxLDL by islet macrophages, leading to OxLDL accumulation in pancreatic islets and a substantial reduction in intra-islet transitory (Texint) CD8+ T cells displaying proliferative and effector signatures. Texint cells were vulnerable to oxidative stress and diminished by ferroptosis; PD-1 blockade rescued this population and reversed diabetes resistance in NOD.Cxcl16−/− mice. Thus, OxLDL scavenging in pancreatic islets inadvertently promotes differentiation of pathogenic CD8+ T cells, presenting a paradigm wherein tissue homeostasis processes can facilitate autoimmune pathogenesis in predisposed individuals.

胰岛微环境具有高度氧化性,使β细胞容易受到自身炎症的损伤。在此,我们研究了胰岛巨噬细胞在引发1型糖尿病的自身免疫攻击中的作用。胰岛巨噬细胞高度表达CXCL16,这是一种趋化因子,也是氧化低密度脂蛋白(OxLDLs)的清除受体,与自身免疫倾向无关。在非肥胖糖尿病(NOD)小鼠体内删除 Cxcl16 可抑制自身免疫性糖尿病的发生。从机理上讲,Cxcl16的缺失损害了胰岛巨噬细胞对OxLDL的清除,导致OxLDL在胰岛中积聚,并使胰岛内具有增殖和效应特征的CD8+T细胞大幅减少。Texint细胞容易受到氧化应激的影响,并因铁蛋白沉积而减少;PD-1阻断可挽救这一细胞群,并逆转NOD.Cxcl16-/-小鼠的糖尿病抵抗。因此,胰岛中的氧化低密度脂蛋白清除无意中促进了致病性 CD8+ T 细胞的分化,从而提出了一种范例,即组织稳态过程可促进易感个体的自身免疫发病机制。
{"title":"CXCL16-dependent scavenging of oxidized lipids by islet macrophages promotes differentiation of pathogenic CD8+ T cells in diabetic autoimmunity","authors":"Neetu Srivastava, Hao Hu, Orion J. Peterson, Anthony N. Vomund, Marta Stremska, Mohammad Zaman, Shilpi Giri, Tiandao Li, Cheryl F. Lichti, Pavel N. Zakharov, Bo Zhang, Nada A. Abumrad, Yi-Guang Chen, Kodi S. Ravichandran, Emil R. Unanue, Xiaoxiao Wan","doi":"10.1016/j.immuni.2024.04.017","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.017","url":null,"abstract":"<p>The pancreatic islet microenvironment is highly oxidative, rendering β cells vulnerable to autoinflammatory insults. Here, we examined the role of islet resident macrophages in the autoimmune attack that initiates type 1 diabetes. Islet macrophages highly expressed CXCL16, a chemokine and scavenger receptor for oxidized low-density lipoproteins (OxLDLs), regardless of autoimmune predisposition. Deletion of <em>Cxcl16</em> in nonobese diabetic (NOD) mice suppressed the development of autoimmune diabetes. Mechanistically, <em>Cxcl16</em> deficiency impaired clearance of OxLDL by islet macrophages, leading to OxLDL accumulation in pancreatic islets and a substantial reduction in intra-islet transitory (Tex<sup>int</sup>) CD8<sup>+</sup> T cells displaying proliferative and effector signatures. Tex<sup>int</sup> cells were vulnerable to oxidative stress and diminished by ferroptosis; PD-1 blockade rescued this population and reversed diabetes resistance in NOD.<em>Cxcl16</em><sup>−/−</sup> mice. Thus, OxLDL scavenging in pancreatic islets inadvertently promotes differentiation of pathogenic CD8<sup>+</sup> T cells, presenting a paradigm wherein tissue homeostasis processes can facilitate autoimmune pathogenesis in predisposed individuals.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140942983","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
Hair care: Stem cells control immune response during wound repair 头发护理:干细胞控制伤口修复过程中的免疫反应
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-14 DOI: 10.1016/j.immuni.2024.04.013
Shannon McCarthy, Judith Agudo

Stem cells heal wounds. In this issue of Immunity, Luan et al. demonstrate that epidermal stem cells orchestrate the recruitment of regulatory T (Treg) cells and neutrophils during wound healing. Treg cells facilitate a tolerogenic environment to protect epithelial regeneration while neutrophils promote inflammation to ward off infection.

干细胞能愈合伤口在本期《免疫》(Immunity)杂志上,Luan等人证明,表皮干细胞能在伤口愈合过程中协调调节性T(Treg)细胞和中性粒细胞的招募。Treg细胞促进耐受性环境,保护上皮再生,而中性粒细胞则促进炎症,抵御感染。
{"title":"Hair care: Stem cells control immune response during wound repair","authors":"Shannon McCarthy, Judith Agudo","doi":"10.1016/j.immuni.2024.04.013","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.013","url":null,"abstract":"<p>Stem cells heal wounds. In this issue of <em>Immunity</em>, Luan et al. demonstrate that epidermal stem cells orchestrate the recruitment of regulatory T (Treg) cells and neutrophils during wound healing. Treg cells facilitate a tolerogenic environment to protect epithelial regeneration while neutrophils promote inflammation to ward off infection.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140920011","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
Platelets: Orchestrators of immunity in host defense and beyond 血小板:血小板:宿主防御及其他免疫的协调者
IF 32.4 1区 医学 Q1 Medicine Pub Date : 2024-05-14 DOI: 10.1016/j.immuni.2024.04.008
Leo Nicolai, Kami Pekayvaz, Steffen Massberg

Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.

血小板可防止血管损伤时的血液流失,并有助于心血管疾病中血栓的形成。除了这些传统作用外,血小板对宿主免疫反应也至关重要。它们通过专门的受体、细胞内信号级联和效应器功能保护血管免受病原体侵袭。血小板还能通过指示先天性免疫细胞、支持适应性免疫监视、影响抗体产生和 T 细胞极化来歪曲炎症反应。同时,血小板还有助于组织重建,并在炎症挑战后维持血管功能。然而,这些多才多艺细胞的失调激活会加剧免疫病理,导致微血管凝血、过度炎症和大血管血栓风险升高。这种对立强调了精确定义和潜在调节免疫中血小板功能的极端重要性。
{"title":"Platelets: Orchestrators of immunity in host defense and beyond","authors":"Leo Nicolai, Kami Pekayvaz, Steffen Massberg","doi":"10.1016/j.immuni.2024.04.008","DOIUrl":"https://doi.org/10.1016/j.immuni.2024.04.008","url":null,"abstract":"<p>Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.</p>","PeriodicalId":13269,"journal":{"name":"Immunity","volume":null,"pages":null},"PeriodicalIF":32.4,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140920048","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
Immunity
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