首页 > 最新文献

Cellular &Molecular Immunology最新文献

英文 中文
Immune surveillance of cytomegalovirus in tissues 对组织中巨细胞病毒的免疫监视
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-12 DOI: 10.1038/s41423-024-01186-2
Andrea Mihalić, Jelena Železnjak, Berislav Lisnić, Stipan Jonjić, Vanda Juranić Lisnić, Ilija Brizić
Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.
巨细胞病毒(CMV)是疱疹病毒 Betaherpesvirinae 亚科的代表成员,在人类中很常见,但免疫功能正常的人感染这种病毒后一般不会出现症状。然而,对于免疫力低下的人以及免疫功能不成熟的胎儿和新生儿,CMV 可导致多种病症,通常是长期病症,甚至死亡。CMV 不仅在人群中广泛传播,而且在宿主中也很普遍,几乎可感染所有组织和器官并在其中建立潜伏期。因此,了解这种病毒的发病机制和免疫反应是制定有效预防和治疗策略的先决条件。免疫系统的多个臂膀都参与了遏制感染的工作,目前人们对 CMV 免疫控制的一般概念已经有了相当深入的了解。然而,近年来,组织特异性免疫反应已成为解决 CMV 感染的一个重要因素。由于各组织的生物学特性和功能不同,对 CMV 的免疫反应和感染期间的病理过程也不尽相同。本综述讨论了组织对 CMV 感染的免疫反应的最新知识,特别强调了几个研究较多和最常受影响的器官。
{"title":"Immune surveillance of cytomegalovirus in tissues","authors":"Andrea Mihalić, Jelena Železnjak, Berislav Lisnić, Stipan Jonjić, Vanda Juranić Lisnić, Ilija Brizić","doi":"10.1038/s41423-024-01186-2","DOIUrl":"10.1038/s41423-024-01186-2","url":null,"abstract":"Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01186-2.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141949447","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 immunology of sickness metabolism 疾病代谢免疫学。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-06 DOI: 10.1038/s41423-024-01192-4
Felix M. Wensveen, Marko Šestan, Bojan Polić
Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.
每个人都知道,感染会让人感到不适。虽然我们认为感染引起的新陈代谢变化是一种病理现象,但它们是一个经过精心调节的过程的一部分,这一过程取决于免疫系统与参与调节全身平衡的器官之间的组织特异性相互作用。免疫介导的体内平衡参数变化会导致循环中营养物质的产生和吸收发生改变,从而改变主要器官的新陈代谢率。这就是我们生病的感受。病态新陈代谢的目的是创造一种新陈代谢环境,使机体能够以最佳状态抵抗感染,同时剥夺病原体复制所需的重要营养物质。疾病新陈代谢依赖于组织特异性免疫细胞,它们根据威胁的性质和严重程度调控反应。随着感染的严重程度增加,参与其中的免疫细胞的数量和类型以及器官受影响的程度也会增加,这就决定了我们生病的程度。有趣的是,许多与新陈代谢疾病相关的变化似乎与感染后观察到的免疫介导的变化重叠。因此,针对涉及活化免疫细胞和代谢器官之间组织特异性相互作用的过程,对于治疗严重感染者和代谢疾病患者都具有巨大的潜力。在本综述中,我们将讨论免疫系统如何与参与调节体内平衡的器官进行原位交流,以及这种交流如何受到感染的影响。
{"title":"The immunology of sickness metabolism","authors":"Felix M. Wensveen, Marko Šestan, Bojan Polić","doi":"10.1038/s41423-024-01192-4","DOIUrl":"10.1038/s41423-024-01192-4","url":null,"abstract":"Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01192-4.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896937","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
Author Correction: Genetic and epigenetic influences on the loss of tolerance in autoimmunity 作者更正:遗传和表观遗传对自身免疫丧失耐受性的影响。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-29 DOI: 10.1038/s41423-024-01201-6
Peng Zhang, Qianjin Lu
{"title":"Author Correction: Genetic and epigenetic influences on the loss of tolerance in autoimmunity","authors":"Peng Zhang, Qianjin Lu","doi":"10.1038/s41423-024-01201-6","DOIUrl":"10.1038/s41423-024-01201-6","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01201-6.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792080","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
Targeting the glucocorticoid receptor-CCR8 axis mediated bone marrow T cell sequestration enhances infiltration of anti-tumor T cells in intracranial cancers. 靶向糖皮质激素受体-CCR8轴介导的骨髓T细胞螯合可增强颅内癌中抗肿瘤T细胞的浸润。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-23 DOI: 10.1038/s41423-024-01202-5
Jia Zhang, Yuzhu Shi, Xiaotong Xue, Wenqing Bu, Yanan Li, Tingting Yang, Lijuan Cao, Jiankai Fang, Peishan Li, Yongjing Chen, Zhen Li, Changshun Shao, Yufang Shi

Brain tumors such as glioblastomas are resistant to immune checkpoint blockade therapy, largely due to limited T cell infiltration in the tumors. Here, we show that mice bearing intracranial tumors exhibit systemic immunosuppression and T cell sequestration in bone marrow, leading to reduced T cell infiltration in brain tumors. Elevated plasma corticosterone drives the T cell sequestration via glucocorticoid receptors in tumor-bearing mice. Immunosuppression mediated by glucocorticoid-induced T cell dynamics and the subsequent tumor growth promotion can be abrogated by adrenalectomy, the administration of glucocorticoid activation inhibitors or glucocorticoid receptor antagonists, and in mice with T cell-specific deletion of glucocorticoid receptor. CCR8 expression in T cells is increased in tumor-bearing mice in a glucocorticoid receptor-dependent manner. Additionally, chemokines CCL1 and CCL8, the ligands for CCR8, are highly expressed in bone marrow immune cells in tumor-bearing mice to recruit T cells. These findings suggested that brain tumor-induced glucocorticoid surge and CCR8 upregulation in T cells lead to T cell sequestration in bone marrow, impairing the anti-tumor immune response. Targeting the glucocorticoid receptor-CCR8 axis may offer a promising immunotherapeutic approach for the treatment of intracranial tumors.

胶质母细胞瘤等脑肿瘤对免疫检查点阻断疗法具有抗药性,这主要是由于肿瘤中的 T 细胞浸润有限。在这里,我们发现颅内肿瘤小鼠表现出全身免疫抑制和骨髓中的T细胞封存,导致脑肿瘤中的T细胞浸润减少。血浆中皮质酮的升高通过糖皮质激素受体驱动肿瘤小鼠的T细胞封存。肾上腺切除术、糖皮质激素活化抑制剂或糖皮质激素受体拮抗剂以及T细胞特异性糖皮质激素受体缺失的小鼠均可减轻糖皮质激素诱导的T细胞动力学介导的免疫抑制以及随后的肿瘤生长促进作用。肿瘤小鼠 T 细胞中 CCR8 的表达以糖皮质激素受体依赖的方式增加。此外,CCR8 的配体趋化因子 CCL1 和 CCL8 在肿瘤小鼠骨髓免疫细胞中高表达,以招募 T 细胞。这些研究结果表明,脑肿瘤诱导的糖皮质激素激增和T细胞中CCR8的上调会导致T细胞在骨髓中固着,从而损害抗肿瘤免疫反应。以糖皮质激素受体-CCR8轴为靶点可能为治疗颅内肿瘤提供一种前景广阔的免疫治疗方法。
{"title":"Targeting the glucocorticoid receptor-CCR8 axis mediated bone marrow T cell sequestration enhances infiltration of anti-tumor T cells in intracranial cancers.","authors":"Jia Zhang, Yuzhu Shi, Xiaotong Xue, Wenqing Bu, Yanan Li, Tingting Yang, Lijuan Cao, Jiankai Fang, Peishan Li, Yongjing Chen, Zhen Li, Changshun Shao, Yufang Shi","doi":"10.1038/s41423-024-01202-5","DOIUrl":"https://doi.org/10.1038/s41423-024-01202-5","url":null,"abstract":"<p><p>Brain tumors such as glioblastomas are resistant to immune checkpoint blockade therapy, largely due to limited T cell infiltration in the tumors. Here, we show that mice bearing intracranial tumors exhibit systemic immunosuppression and T cell sequestration in bone marrow, leading to reduced T cell infiltration in brain tumors. Elevated plasma corticosterone drives the T cell sequestration via glucocorticoid receptors in tumor-bearing mice. Immunosuppression mediated by glucocorticoid-induced T cell dynamics and the subsequent tumor growth promotion can be abrogated by adrenalectomy, the administration of glucocorticoid activation inhibitors or glucocorticoid receptor antagonists, and in mice with T cell-specific deletion of glucocorticoid receptor. CCR8 expression in T cells is increased in tumor-bearing mice in a glucocorticoid receptor-dependent manner. Additionally, chemokines CCL1 and CCL8, the ligands for CCR8, are highly expressed in bone marrow immune cells in tumor-bearing mice to recruit T cells. These findings suggested that brain tumor-induced glucocorticoid surge and CCR8 upregulation in T cells lead to T cell sequestration in bone marrow, impairing the anti-tumor immune response. Targeting the glucocorticoid receptor-CCR8 axis may offer a promising immunotherapeutic approach for the treatment of intracranial tumors.</p>","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141751217","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
GPR34 senses demyelination to promote neuroinflammation and pathologies. GPR34 可感知脱髓鞘,从而促进神经炎症和病变。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-19 DOI: 10.1038/s41423-024-01204-3
Bolong Lin, Yubo Zhou, Zonghui Huang, Ming Ma, Minghui Qi, Zhongjun Jiang, Guoyang Li, Yueli Xu, Jiaxian Yan, Di Wang, Xiaqiong Wang, Wei Jiang, Rongbin Zhou

Sterile neuroinflammation is a major driver of multiple neurological diseases. Myelin debris can act as an inflammatory stimulus to promote inflammation and pathologies, but the mechanism is poorly understood. Here, we showed that lysophosphatidylserine (LysoPS)-GPR34 axis played a critical role in microglia-mediated myelin debris sensing and the subsequent neuroinflammation. Myelin debris-induced microglia activation and proinflammatory cytokine expression relied on its lipid component LysoPS. Both myelin debris and LysoPS promoted microglia activation and the production of proinflammatory cytokines via GPR34 and its downstream PI3K-AKT and ERK signaling. In vivo, reducing the content of LysoPS in myelin or inhibition of GPR34 with genetic or pharmacological approaches reduced neuroinflammation and pathologies in the mouse models of multiple sclerosis and stroke. Thus, our results identify GPR34 as a key receptor to sense demyelination and CNS damage and promote neuroinflammation, and suggest it as a potential therapeutic target for demyelination-associated diseases.

无菌性神经炎症是多种神经系统疾病的主要驱动因素。髓鞘碎片可作为炎症刺激物促进炎症和病变,但其机制尚不清楚。在这里,我们发现溶血磷脂酰丝氨酸(LysoPS)-GPR34 轴在小胶质细胞介导的髓鞘碎片感应和随后的神经炎症中发挥了关键作用。髓鞘碎片诱导的小胶质细胞活化和促炎细胞因子表达依赖于其脂质成分LysoPS。髓鞘碎屑和LysoPS都能通过GPR34及其下游的PI3K-AKT和ERK信号促进小胶质细胞的活化和促炎细胞因子的产生。在体内,通过基因或药物方法减少髓鞘中溶血磷脂的含量或抑制 GPR34 可减轻多发性硬化和中风小鼠模型的神经炎症和病理变化。因此,我们的研究结果确定 GPR34 是感知脱髓鞘和中枢神经系统损伤并促进神经炎症的关键受体,并建议将其作为脱髓鞘相关疾病的潜在治疗靶点。
{"title":"GPR34 senses demyelination to promote neuroinflammation and pathologies.","authors":"Bolong Lin, Yubo Zhou, Zonghui Huang, Ming Ma, Minghui Qi, Zhongjun Jiang, Guoyang Li, Yueli Xu, Jiaxian Yan, Di Wang, Xiaqiong Wang, Wei Jiang, Rongbin Zhou","doi":"10.1038/s41423-024-01204-3","DOIUrl":"https://doi.org/10.1038/s41423-024-01204-3","url":null,"abstract":"<p><p>Sterile neuroinflammation is a major driver of multiple neurological diseases. Myelin debris can act as an inflammatory stimulus to promote inflammation and pathologies, but the mechanism is poorly understood. Here, we showed that lysophosphatidylserine (LysoPS)-GPR34 axis played a critical role in microglia-mediated myelin debris sensing and the subsequent neuroinflammation. Myelin debris-induced microglia activation and proinflammatory cytokine expression relied on its lipid component LysoPS. Both myelin debris and LysoPS promoted microglia activation and the production of proinflammatory cytokines via GPR34 and its downstream PI3K-AKT and ERK signaling. In vivo, reducing the content of LysoPS in myelin or inhibition of GPR34 with genetic or pharmacological approaches reduced neuroinflammation and pathologies in the mouse models of multiple sclerosis and stroke. Thus, our results identify GPR34 as a key receptor to sense demyelination and CNS damage and promote neuroinflammation, and suggest it as a potential therapeutic target for demyelination-associated diseases.</p>","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141726972","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
Orchestration of antiviral responses within the infected central nervous system 在受感染的中枢神经系统内协调抗病毒反应。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-12 DOI: 10.1038/s41423-024-01181-7
Andreas Pavlou, Felix Mulenge, Olivia Luise Gern, Lena Mareike Busker, Elisabeth Greimel, Inken Waltl, Ulrich Kalinke
Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.
许多新出现和再次出现的病毒都有可能侵入神经系统,因此病毒性脑炎成为全球研究的重点。病毒进入中枢神经系统后,可能会出现严重的神经系统症状,危及生命,发病率和死亡率都很高。目前针对病毒性脑炎的治疗手段非常有限,因此需要更好地了解受感染的中枢神经系统内局部抗病毒免疫的条件。在这篇综述中,我们讨论了对病毒性脑炎病理生理学的新认识,重点是骨髓细胞和 CD8+ T 细胞,它们对保护中枢神经系统免受病毒感染起着至关重要的作用。通过阐明髓系细胞和 T 细胞活化的先决条件、讨论有关其转录特征的新发现以及剖析它们被招募到中枢神经系统内病毒复制位点的机制,我们旨在进一步阐明受感染的中枢神经系统内抗病毒反应的复杂性。此外,我们还总结了病毒感染和神经变性领域的现有知识,并讨论了一些神经滋养病毒与神经变性中观察到的某些病理特征之间的潜在联系。
{"title":"Orchestration of antiviral responses within the infected central nervous system","authors":"Andreas Pavlou,&nbsp;Felix Mulenge,&nbsp;Olivia Luise Gern,&nbsp;Lena Mareike Busker,&nbsp;Elisabeth Greimel,&nbsp;Inken Waltl,&nbsp;Ulrich Kalinke","doi":"10.1038/s41423-024-01181-7","DOIUrl":"10.1038/s41423-024-01181-7","url":null,"abstract":"Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01181-7.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141598744","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
Extramedullary neutrophil progenitors: Quo Vadis? 髓外中性粒细胞祖细胞:何去何从?
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-08 DOI: 10.1038/s41423-024-01191-5
Leo Koenderman, Nienke Vrisekoop
{"title":"Extramedullary neutrophil progenitors: Quo Vadis?","authors":"Leo Koenderman,&nbsp;Nienke Vrisekoop","doi":"10.1038/s41423-024-01191-5","DOIUrl":"10.1038/s41423-024-01191-5","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141558176","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
Targeting pro-fibrotic macrophages with bioactive self-assembly peptides to retard kidney fibrosis – know thyself 用生物活性自组装肽靶向促纤维化巨噬细胞,延缓肾脏纤维化--了解你自己。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-08 DOI: 10.1038/s41423-024-01194-2
Steven O’Reilly
{"title":"Targeting pro-fibrotic macrophages with bioactive self-assembly peptides to retard kidney fibrosis – know thyself","authors":"Steven O’Reilly","doi":"10.1038/s41423-024-01194-2","DOIUrl":"10.1038/s41423-024-01194-2","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141558177","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
Food restriction heals darkness-related second brain inflammation 食物限制能治愈与黑暗有关的第二脑部炎症。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-05 DOI: 10.1038/s41423-024-01196-0
Vinicius Kannen
{"title":"Food restriction heals darkness-related second brain inflammation","authors":"Vinicius Kannen","doi":"10.1038/s41423-024-01196-0","DOIUrl":"10.1038/s41423-024-01196-0","url":null,"abstract":"","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141537605","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
Tissue-resident memory T cells break tolerance to renal autoantigens and orchestrate immune-mediated nephritis 组织驻留记忆 T 细胞打破对肾脏自身抗原的耐受,并协调免疫介导的肾炎。
IF 21.8 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-03 DOI: 10.1038/s41423-024-01197-z
Frederic Arnold, Laurence Kupferschmid, Philipp Weissenborn, Lukas Heldmann, Jonas F. Hummel, Paulina Zareba,  Sagar, Manuel Rogg, Christoph Schell, Yakup Tanriver
Immune-mediated nephritis is a leading cause of acute kidney injury and chronic kidney disease. While the role of B cells and antibodies has been extensively investigated in the past, the advent of immune-checkpoint inhibitors has led to a reappraisal of the role of T cells in renal immunology. However, it remains elusive how T cells with specificity for renal autoantigens are activated and participate in immune-mediated nephritis. Here, we followed the fate and function of pathogen-activated autoreactive CD8 T cells that are specific for a renal autoantigen. We demonstrate that recently activated splenic CD8 T cells developed a hybrid phenotype in the context of renal autoantigen cross-presentation, combining hallmarks of activation and T cell dysfunction. While circulating memory T cells rapidly disappeared, tissue-resident memory T cells emerged and persisted within the kidney, orchestrating immune-mediated nephritis. Notably, T cells infiltrating kidneys of patients with interstitial nephritis also expressed key markers of tissue residency. This study unveils how a tissue-specific immune response can dissociate from its systemic counterpart driving a compartmentalized immune response in the kidneys of mice and man. Consequently, targeting tissue-resident memory T cells emerges as a promising strategy to control immune-mediated kidney disease.
免疫介导的肾炎是急性肾损伤和慢性肾病的主要病因。虽然过去对 B 细胞和抗体的作用进行了广泛研究,但免疫检查点抑制剂的出现促使人们重新评估 T 细胞在肾脏免疫学中的作用。然而,对肾脏自身抗原具有特异性的 T 细胞是如何被激活并参与免疫介导的肾炎的仍是个谜。在这里,我们跟踪了病原体激活的对肾脏自身抗原特异的自身反应性 CD8 T 细胞的命运和功能。我们证明,最近激活的脾脏 CD8 T 细胞在肾脏自身抗原交叉呈递的背景下形成了一种混合表型,结合了激活和 T 细胞功能障碍的特征。在循环记忆 T 细胞迅速消失的同时,组织驻留记忆 T 细胞出现并持续存在于肾脏中,协调免疫介导的肾炎。值得注意的是,浸润间质性肾炎患者肾脏的 T 细胞也表达了组织驻留的关键标记。这项研究揭示了组织特异性免疫反应如何与全身性免疫反应相分离,从而在小鼠和人类肾脏中产生分区免疫反应。因此,靶向组织驻留记忆 T 细胞是控制免疫介导的肾脏疾病的一种有前途的策略。
{"title":"Tissue-resident memory T cells break tolerance to renal autoantigens and orchestrate immune-mediated nephritis","authors":"Frederic Arnold,&nbsp;Laurence Kupferschmid,&nbsp;Philipp Weissenborn,&nbsp;Lukas Heldmann,&nbsp;Jonas F. Hummel,&nbsp;Paulina Zareba,&nbsp; Sagar,&nbsp;Manuel Rogg,&nbsp;Christoph Schell,&nbsp;Yakup Tanriver","doi":"10.1038/s41423-024-01197-z","DOIUrl":"10.1038/s41423-024-01197-z","url":null,"abstract":"Immune-mediated nephritis is a leading cause of acute kidney injury and chronic kidney disease. While the role of B cells and antibodies has been extensively investigated in the past, the advent of immune-checkpoint inhibitors has led to a reappraisal of the role of T cells in renal immunology. However, it remains elusive how T cells with specificity for renal autoantigens are activated and participate in immune-mediated nephritis. Here, we followed the fate and function of pathogen-activated autoreactive CD8 T cells that are specific for a renal autoantigen. We demonstrate that recently activated splenic CD8 T cells developed a hybrid phenotype in the context of renal autoantigen cross-presentation, combining hallmarks of activation and T cell dysfunction. While circulating memory T cells rapidly disappeared, tissue-resident memory T cells emerged and persisted within the kidney, orchestrating immune-mediated nephritis. Notably, T cells infiltrating kidneys of patients with interstitial nephritis also expressed key markers of tissue residency. This study unveils how a tissue-specific immune response can dissociate from its systemic counterpart driving a compartmentalized immune response in the kidneys of mice and man. Consequently, targeting tissue-resident memory T cells emerges as a promising strategy to control immune-mediated kidney disease.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41423-024-01197-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141497262","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
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