Yang Chen, Jiajia Shen, Xiaoli Zhao, Qiyang He, Juan Zhang
Therapy-induced senescence can regulate both the innate and adaptive immune systems, thereby affecting therapeutic efficacy. Bleomycin is a major component of combined chemotherapy regimens, utilized for the treatment of multiple tumors, whereas pulmonary toxicity severely restricts its clinical benefits. As a member of the bleomycin family, boningmycin (BON) exhibits potent anticancer activity with minimal pulmonary toxicity, making it a potential alternative to bleomycin. Low concentrations of BON can induce senescence, but the impact of BON-induced senescence on anticancer immunity remains unclear. This study investigates the effects of BON-induced senescence on PD-L1 expression and the underlying mechanisms in human cancer cells. Firstly, the elevation of PD-L1 protein during BON-induced senescence was confirmed by a senescence β-galactosidase staining assay, detection of the senescence-associated secretory phenotype (SASP), western blot and flow cytometry in human lung cancer NCI-H460 cells and breast cancer MDA-MB-231 cells. Subsequently, it was shown that the increase in PD-L1 protein is mediated by SASP, as evidenced by the use of conditional media, knockdown of cyclic GMP-AMP synthase and inhibition of stimulator of interferon genes. Ultimately, it was demonstrated that SASP-mediated PD-L1 up-regulation is dependent on the activation of the JAK/STAT pathway through the use of specific inhibitors and siRNAs. These findings clarify the impact of BON-induced senescence on PD-L1 expression and may contribute to the optimization of the therapeutic efficacy of bleomycin-related compounds and the clinical transformation of BON.
{"title":"The up-regulation of PD-L1 during boningmycin-induced senescence in human cancer cells depends on the activation of the JAK/STAT signaling pathway mediated by SASP","authors":"Yang Chen, Jiajia Shen, Xiaoli Zhao, Qiyang He, Juan Zhang","doi":"10.1111/imcb.12812","DOIUrl":"10.1111/imcb.12812","url":null,"abstract":"<p>Therapy-induced senescence can regulate both the innate and adaptive immune systems, thereby affecting therapeutic efficacy. Bleomycin is a major component of combined chemotherapy regimens, utilized for the treatment of multiple tumors, whereas pulmonary toxicity severely restricts its clinical benefits. As a member of the bleomycin family, boningmycin (BON) exhibits potent anticancer activity with minimal pulmonary toxicity, making it a potential alternative to bleomycin. Low concentrations of BON can induce senescence, but the impact of BON-induced senescence on anticancer immunity remains unclear. This study investigates the effects of BON-induced senescence on PD-L1 expression and the underlying mechanisms in human cancer cells. Firstly, the elevation of PD-L1 protein during BON-induced senescence was confirmed by a senescence β-galactosidase staining assay, detection of the senescence-associated secretory phenotype (SASP), western blot and flow cytometry in human lung cancer NCI-H460 cells and breast cancer MDA-MB-231 cells. Subsequently, it was shown that the increase in PD-L1 protein is mediated by SASP, as evidenced by the use of conditional media, knockdown of cyclic GMP-AMP synthase and inhibition of stimulator of interferon genes. Ultimately, it was demonstrated that SASP-mediated PD-L1 up-regulation is dependent on the activation of the JAK/STAT pathway through the use of specific inhibitors and siRNAs. These findings clarify the impact of BON-induced senescence on PD-L1 expression and may contribute to the optimization of the therapeutic efficacy of bleomycin-related compounds and the clinical transformation of BON.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 9","pages":"847-859"},"PeriodicalIF":3.2,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141750658","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Paulina Żelechowska, Magdalena Wiktorska, Elżbieta Kozłowska, Justyna Agier
Adipokines play essential roles in regulating a range of biological processes, but growing evidence indicates that they are also fundamental in immunological mechanisms and, primarily, inflammatory responses. Adipokines mediate their actions through specific receptors. However, although adipokine receptors are widely distributed in many cell and tissue types, limited data are available on their expression in mast cells (MCs) and, consequently, adipokine's significance in the modulation of MC activity within the tissues. In this study, we demonstrate that rat peritoneal MCs constitutively express the leptin receptor (i.e. LEPR), adiponectin receptors (i.e. ADIPOR1 and ADIPOR2) and the chemerin receptor (i.e. CMKLR1). We also found that LEPR, ADIPOR1, ADIPOR2 and CMKLR1 expression in MCs changes in response to stimulation by their specific ligands and some cytokines with potent proinflammatory properties. Furthermore, the involvement of intracellular signaling molecules in leptin-, adiponectin- and chemerin-induced MC response was analyzed. Overall, our findings suggest that adipokines leptin, adiponectin and chemerin can significantly affect the activity of MCs in various processes, especially during inflammation. These observations may contribute significantly to understanding the relationship between adipokines, immune mechanisms and diseases or conditions with an inflammatory component.
脂肪因子在调节一系列生物过程中发挥着至关重要的作用,但越来越多的证据表明,它们也是免疫机制的基础,主要是炎症反应。脂肪因子通过特定的受体介导其作用。然而,虽然脂肪因子受体广泛分布于许多细胞和组织类型中,但有关它们在肥大细胞(MCs)中的表达以及脂肪因子在调节组织内 MC 活性方面的意义的数据却很有限。在这项研究中,我们证明了大鼠腹膜肥大细胞会连续表达瘦素受体(即 LEPR)、脂肪连通素受体(即 ADIPOR1 和 ADIPOR2)和螯合素受体(即 CMKLR1)。我们还发现,LEPR、ADIPOR1、ADIPOR2 和 CMKLR1 在 MCs 中的表达会随着其特定配体和一些具有强效促炎特性的细胞因子的刺激而发生变化。此外,我们还分析了细胞内信号分子在瘦素、脂肪连通素和螯合素诱导的 MC 反应中的参与情况。总之,我们的研究结果表明,脂肪因子瘦素、脂肪连通素和螯合素能显著影响 MCs 在各种过程中的活性,尤其是在炎症过程中。这些观察结果可能会大大有助于理解脂肪因子、免疫机制和具有炎症成分的疾病或病症之间的关系。
{"title":"Adipokine receptor expression in mast cells is altered by specific ligands and proinflammatory cytokines","authors":"Paulina Żelechowska, Magdalena Wiktorska, Elżbieta Kozłowska, Justyna Agier","doi":"10.1111/imcb.12809","DOIUrl":"10.1111/imcb.12809","url":null,"abstract":"<p>Adipokines play essential roles in regulating a range of biological processes, but growing evidence indicates that they are also fundamental in immunological mechanisms and, primarily, inflammatory responses. Adipokines mediate their actions through specific receptors. However, although adipokine receptors are widely distributed in many cell and tissue types, limited data are available on their expression in mast cells (MCs) and, consequently, adipokine's significance in the modulation of MC activity within the tissues. In this study, we demonstrate that rat peritoneal MCs constitutively express the leptin receptor (i.e. LEPR), adiponectin receptors (i.e. ADIPOR1 and ADIPOR2) and the chemerin receptor (i.e. CMKLR1). We also found that LEPR, ADIPOR1, ADIPOR2 and CMKLR1 expression in MCs changes in response to stimulation by their specific ligands and some cytokines with potent proinflammatory properties. Furthermore, the involvement of intracellular signaling molecules in leptin-, adiponectin- and chemerin-induced MC response was analyzed. Overall, our findings suggest that adipokines leptin, adiponectin and chemerin can significantly affect the activity of MCs in various processes, especially during inflammation. These observations may contribute significantly to understanding the relationship between adipokines, immune mechanisms and diseases or conditions with an inflammatory component.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 9","pages":"817-829"},"PeriodicalIF":3.2,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141625494","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zewen Kelvin Tuong, Rohan van der Merwe, Pablo F Canete, Jonathan A Roco
Diversity is the cornerstone of the adaptive immune system, crucial for its effectiveness against constantly evolving pathogens that pose threats to higher vertebrates. Accurately measuring and interpreting this diversity presents challenges for immunologists, as changes in diversity and clonotype composition can tip the balance between protective immunity and autoimmunity. In this review, we present the current methods commonly used to measure diversity from single-cell T-cell receptor and B-cell receptor sequencing. We also discuss two case studies where single-cell sequencing and diversity estimations have led to breakthroughs in autoimmune disease discovery and therapeutic innovation, and reflect upon the necessity and importance of accurately defining and measuring lymphocyte diversity in these contexts.
多样性是适应性免疫系统的基石,是其有效抵御不断进化的病原体威胁高等脊椎动物的关键。精确测量和解释这种多样性给免疫学家带来了挑战,因为多样性和克隆型组成的变化会影响保护性免疫和自身免疫之间的平衡。在这篇综述中,我们介绍了目前常用于测量单细胞 T 细胞受体和 B 细胞受体测序多样性的方法。我们还讨论了两个案例研究,在这两个案例研究中,单细胞测序和多样性估计为自身免疫性疾病的发现和治疗创新带来了突破性进展,并反思了在这些情况下准确定义和测量淋巴细胞多样性的必要性和重要性。
{"title":"Computational estimation of clonal diversity in autoimmunity","authors":"Zewen Kelvin Tuong, Rohan van der Merwe, Pablo F Canete, Jonathan A Roco","doi":"10.1111/imcb.12801","DOIUrl":"10.1111/imcb.12801","url":null,"abstract":"<p>Diversity is the cornerstone of the adaptive immune system, crucial for its effectiveness against constantly evolving pathogens that pose threats to higher vertebrates. Accurately measuring and interpreting this diversity presents challenges for immunologists, as changes in diversity and clonotype composition can tip the balance between protective immunity and autoimmunity. In this review, we present the current methods commonly used to measure diversity from single-cell T-cell receptor and B-cell receptor sequencing. We also discuss two case studies where single-cell sequencing and diversity estimations have led to breakthroughs in autoimmune disease discovery and therapeutic innovation, and reflect upon the necessity and importance of accurately defining and measuring lymphocyte diversity in these contexts.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 8","pages":"692-701"},"PeriodicalIF":3.2,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12801","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141618749","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Stefanie F Valbon, Marie-Eve Lebel, H Alex Feldman, Stephanie A Condotta, Mengqi Dong, Daniela Giordano, Stephen N Waggoner, Heather J Melichar, Martin J Richer
Chronic viral infections cause thymic involution yet the potential for broader, longer-term impact on thymic composition remains unexplored. Here we show that chronic, but not acute, lymphocytic choriomeningitis virus infection promotes a unique population of immature B cells in the thymus. We show that chronic viral infection promotes signals within the thymus, including the expression of B-cell activating factor (BAFF), that favor the maturation of this population as these cells acquire expression of CD19 and immunoglobulin M. Mechanistically, type I interferon (IFN-I), predominantly IFNβ, signals to thymic hematopoietic cells, strongly delaying T-cell development at the earliest precursor stage. Furthermore, IFN-I signaling to the nonhematopoietic compartment provides a second signal essential to favor B-cell differentiation and maturation within the thymus. Importantly, chronic infection yields changes in the B-cell population for at least 50 days following infection, long after thymic atrophy has subsided. Thus, the inflammatory milieu induced by chronic viral infection has a profound, and long-lasting, effect on thymic composition leading to the generation of a novel population of thymic B cells.
慢性病毒感染会导致胸腺萎缩,但对胸腺组成产生更广泛、更长期影响的可能性仍有待探索。在这里,我们发现慢性淋巴细胞性脉络膜炎病毒感染(而非急性淋巴细胞性脉络膜炎病毒感染)会促进胸腺中一种独特的未成熟 B 细胞群。从机理上讲,I 型干扰素(IFN-I),主要是 IFNβ,向胸腺造血细胞发出信号,在最早的前体阶段强烈延迟 T 细胞的发育。此外,IFN-I 还向非造血细胞区发出信号,提供了有利于胸腺内 B 细胞分化和成熟所必需的第二个信号。重要的是,在胸腺萎缩消退后的很长一段时间内,慢性感染会在感染后至少 50 天内导致 B 细胞群发生变化。因此,慢性病毒感染诱导的炎症环境对胸腺组成产生了深远而持久的影响,导致胸腺B细胞新群体的产生。
{"title":"Type I interferon induced during chronic viral infection favors B-cell development in the thymus","authors":"Stefanie F Valbon, Marie-Eve Lebel, H Alex Feldman, Stephanie A Condotta, Mengqi Dong, Daniela Giordano, Stephen N Waggoner, Heather J Melichar, Martin J Richer","doi":"10.1111/imcb.12808","DOIUrl":"10.1111/imcb.12808","url":null,"abstract":"<p>Chronic viral infections cause thymic involution yet the potential for broader, longer-term impact on thymic composition remains unexplored. Here we show that chronic, but not acute, lymphocytic choriomeningitis virus infection promotes a unique population of immature B cells in the thymus. We show that chronic viral infection promotes signals within the thymus, including the expression of B-cell activating factor (BAFF), that favor the maturation of this population as these cells acquire expression of CD19 and immunoglobulin M. Mechanistically, type I interferon (IFN-I), predominantly IFNβ, signals to thymic hematopoietic cells, strongly delaying T-cell development at the earliest precursor stage. Furthermore, IFN-I signaling to the nonhematopoietic compartment provides a second signal essential to favor B-cell differentiation and maturation within the thymus. Importantly, chronic infection yields changes in the B-cell population for at least 50 days following infection, long after thymic atrophy has subsided. Thus, the inflammatory milieu induced by chronic viral infection has a profound, and long-lasting, effect on thymic composition leading to the generation of a novel population of thymic B cells.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 9","pages":"801-816"},"PeriodicalIF":3.2,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11444890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141618791","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Juliana Silva, Emma P Halmos, Benjamin J Marsland, Richelle Mychasiuk
The Gastroenterology Immunology Neuroscience (GIN) Discovery Program represents a new model for research that overcomes the limitations imposed by traditional “research silos” in science. By uniting these three fields, the GIN Program aims to enhance the understanding and treatment of chronic conditions through a system-wide perspective focusing on the gut–immune–brain axis. Key initiatives include monthly interdisciplinary seminars, an annual symposium, and GINnovate, a commercialization and entrepreneurship event. Additionally, the program offers a seed grant competition for early and mid-career researchers, promoting advancements in gut–immune–brain axis research through the power of collaboration. The GIN Program in a short period of time has facilitated the formation of a vibrant community, captivating attention from both national and international institutions. This effort to break down barriers in research aims to inspire similar models that prioritize open communication, mutual respect and a commitment to impactful science.
{"title":"Breaking down silos: The Gastroenterology Immunology Neuroscience (GIN) Discovery Program – a new model for research","authors":"Juliana Silva, Emma P Halmos, Benjamin J Marsland, Richelle Mychasiuk","doi":"10.1111/imcb.12802","DOIUrl":"10.1111/imcb.12802","url":null,"abstract":"<p>The Gastroenterology Immunology Neuroscience (GIN) Discovery Program represents a new model for research that overcomes the limitations imposed by traditional “research silos” in science. By uniting these three fields, the GIN Program aims to enhance the understanding and treatment of chronic conditions through a system-wide perspective focusing on the gut–immune–brain axis. Key initiatives include monthly interdisciplinary seminars, an annual symposium, and GINnovate, a commercialization and entrepreneurship event. Additionally, the program offers a seed grant competition for early and mid-career researchers, promoting advancements in gut–immune–brain axis research through the power of collaboration. The GIN Program in a short period of time has facilitated the formation of a vibrant community, captivating attention from both national and international institutions. This effort to break down barriers in research aims to inspire similar models that prioritize open communication, mutual respect and a commitment to impactful science.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 8","pages":"658-662"},"PeriodicalIF":3.2,"publicationDate":"2024-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12802","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141615416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pawel Borowicz, Carolyn G King, Michael L Dustin, E John Wherry, Gary A Koretzky, Anne Spurkland
This Future Challenges article summarizes views on future directions in immunological research presented at round-table discussions at the 4th Immunology workshop in the Lofoten Islands in Norway, held in August 2023, and subsequent responses to surveys sent to meeting participants. It also summarizes some of the conversations around the responsibility of scientists to communicate with the non-science community, and the approaches that we may use to meet this obligation.
{"title":"The future of immunology: a Lofoten perspective","authors":"Pawel Borowicz, Carolyn G King, Michael L Dustin, E John Wherry, Gary A Koretzky, Anne Spurkland","doi":"10.1111/imcb.12805","DOIUrl":"10.1111/imcb.12805","url":null,"abstract":"<p>This Future Challenges article summarizes views on future directions in immunological research presented at round-table discussions at the 4th Immunology workshop in the Lofoten Islands in Norway, held in August 2023, and subsequent responses to surveys sent to meeting participants. It also summarizes some of the conversations around the responsibility of scientists to communicate with the non-science community, and the approaches that we may use to meet this obligation.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 9","pages":"760-765"},"PeriodicalIF":3.2,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12805","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141589125","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p>In this Special Feature, we bring you the “Highlights of 2023”, a collection of short articles that discuss key research findings published in 2023 that advanced a specific research area of immunology. Booty<span><sup>1</sup></span> discusses recent mechanistic insights in T-cell immunometabolism, highlighting pathways and metabolites that modulate T effector and T regulatory (Treg) cell function in cancer and autoimmunity. Zhang and Chong<span><sup>2</sup></span> review key findings demonstrating the roles of microRNA in T-cell apoptosis and differentiation and Treg proliferation in experimental autoimmune encephalomyelitis and myeloma. Makuyana and Liston<span><sup>3</sup></span> focus on publications revealing new functions for Treg cells in the lung, including in alveolar regeneration. Guo <i>et al</i>.<span><sup>4</sup></span> highlight work that has advanced our understanding of age-related effects on T cells with large-scale analyses showing genetic, transcriptomic and T-cell receptor repertoire changes with age. Pasquin<span><sup>5</sup></span> discusses key findings in the functional characterization and diversity of γδ T cells and mucosal-associated invariant T (MAIT) cells. Cellular therapy was a pivotal theme in 2023. Chinni <i>et al</i>.<span><sup>6</sup></span> highlight findings demonstrating how CD4<sup>+</sup> T cells impact the manufacturing and quality of chimeric antigen receptor (CAR) T-cell products and contribute to long-term tumor control and adverse events such as cytokine release syndrome. Lee and Reed<span><sup>7</sup></span> discuss current clinical trials and basic research studies that are improving the specificity, safety and accessibility of CAR T-cell therapy for autoimmune disease. Bourel and Lesage<span><sup>8</sup></span> focus on publications defining the phenotypic, genetic and functional attributes that influence natural killer (NK) cell–mediated killing of tumor cells for the development of NK cellular therapies. Lam and Souza-Fonseca-Guimaraes<span><sup>9</sup></span> continue the NK cell theme, highlighting technological advances in genomics and proteomics that elucidate key functions of NK cells in cancer and infection. Lombard-Vadnais and Lesage<span><sup>10</sup></span> uncover the role of class switching in thymic B cells for negative selection of CD4<sup>+</sup> thymocytes and Treg generation. Barra and Marshall<span><sup>11</sup></span> highlight key findings on the diversity and function of mast cells and how they integrate with host defense to prevent immune-mediated damage in barrier tissues and the central nervous system. Dashwood and Liston<span><sup>12</sup></span> bring us up to date on microglia biology, with mechanistic insights into cognitive development, synaptic pruning and new approaches to evaluate microglia function. Van Nieuwenhove<span><sup>13</sup></span> highlights major translational advances in the detection and treatment of monogenic and polygenic pediatric immune deficienci
在本特辑中,我们将为您带来 "2023 年亮点",这是一组讨论 2023 年发表的、推动免疫学特定研究领域发展的重要研究成果的短文集。Booty1讨论了T细胞免疫代谢的最新机理见解,重点介绍了在癌症和自身免疫中调节T效应细胞和T调节(Treg)细胞功能的途径和代谢产物。Zhang和Chong2回顾了在实验性自身免疫性脑脊髓炎和骨髓瘤中证明微RNA在T细胞凋亡和分化以及Treg增殖中作用的重要发现。Makuyana 和 Liston3 重点介绍了揭示 Treg 细胞在肺部(包括肺泡再生)新功能的论文。Guo 等人4 重点介绍了通过大规模分析显示基因、转录组和 T 细胞受体组随着年龄的增长而发生变化,从而加深了我们对年龄对 T 细胞影响的理解。Pasquin5 讨论了γδ T 细胞和粘膜相关不变 T 细胞(MAIT)的功能特征和多样性方面的重要发现。细胞疗法是2023年的一个重要主题。Chinni等人6重点介绍了CD4+ T细胞如何影响嵌合抗原受体(CAR)T细胞产品的生产和质量,以及如何促进长期肿瘤控制和细胞因子释放综合征等不良反应的研究结果。Lee 和 Reed7 讨论了当前的临床试验和基础研究,这些研究正在提高 CAR T 细胞疗法治疗自身免疫性疾病的特异性、安全性和可及性。Bourel 和 Lesage8 重点介绍了有关影响自然杀伤(NK)细胞介导的肿瘤细胞杀伤的表型、遗传和功能特性的出版物,以开发 NK 细胞疗法。Lam和Souza-Fonseca-Guimaraes9继续以NK细胞为主题,重点介绍了基因组学和蛋白质组学方面的技术进展,这些进展阐明了NK细胞在癌症和感染中的关键功能。Lombard-Vadnais和Lesage10揭示了胸腺B细胞在CD4+胸腺细胞负向选择和Treg生成中的类别转换作用。Barra 和 Marshall11 重点介绍了肥大细胞的多样性和功能,以及肥大细胞如何与宿主防御相结合,防止屏障组织和中枢神经系统中免疫介导的损伤。Dashwood 和 Liston12 为我们介绍了小胶质细胞生物学的最新进展,包括对认知发展、突触修剪和评估小胶质细胞功能的新方法的机理认识。Van Nieuwenhove13 重点介绍了在检测和治疗单基因和多基因小儿免疫缺陷、自身免疫和自身炎症方面取得的重大转化进展。Pankhurst 和 Linterman14 总结了生殖中心领域的许多重要发现以及对长效体液免疫的影响。本研究亮点集旨在更新和庆祝 2023 年免疫学领域的重要发现。
{"title":"ICB Special Feature: Highlights of 2023","authors":"Joanne H Reed","doi":"10.1111/imcb.12792","DOIUrl":"10.1111/imcb.12792","url":null,"abstract":"<p>In this Special Feature, we bring you the “Highlights of 2023”, a collection of short articles that discuss key research findings published in 2023 that advanced a specific research area of immunology. Booty<span><sup>1</sup></span> discusses recent mechanistic insights in T-cell immunometabolism, highlighting pathways and metabolites that modulate T effector and T regulatory (Treg) cell function in cancer and autoimmunity. Zhang and Chong<span><sup>2</sup></span> review key findings demonstrating the roles of microRNA in T-cell apoptosis and differentiation and Treg proliferation in experimental autoimmune encephalomyelitis and myeloma. Makuyana and Liston<span><sup>3</sup></span> focus on publications revealing new functions for Treg cells in the lung, including in alveolar regeneration. Guo <i>et al</i>.<span><sup>4</sup></span> highlight work that has advanced our understanding of age-related effects on T cells with large-scale analyses showing genetic, transcriptomic and T-cell receptor repertoire changes with age. Pasquin<span><sup>5</sup></span> discusses key findings in the functional characterization and diversity of γδ T cells and mucosal-associated invariant T (MAIT) cells. Cellular therapy was a pivotal theme in 2023. Chinni <i>et al</i>.<span><sup>6</sup></span> highlight findings demonstrating how CD4<sup>+</sup> T cells impact the manufacturing and quality of chimeric antigen receptor (CAR) T-cell products and contribute to long-term tumor control and adverse events such as cytokine release syndrome. Lee and Reed<span><sup>7</sup></span> discuss current clinical trials and basic research studies that are improving the specificity, safety and accessibility of CAR T-cell therapy for autoimmune disease. Bourel and Lesage<span><sup>8</sup></span> focus on publications defining the phenotypic, genetic and functional attributes that influence natural killer (NK) cell–mediated killing of tumor cells for the development of NK cellular therapies. Lam and Souza-Fonseca-Guimaraes<span><sup>9</sup></span> continue the NK cell theme, highlighting technological advances in genomics and proteomics that elucidate key functions of NK cells in cancer and infection. Lombard-Vadnais and Lesage<span><sup>10</sup></span> uncover the role of class switching in thymic B cells for negative selection of CD4<sup>+</sup> thymocytes and Treg generation. Barra and Marshall<span><sup>11</sup></span> highlight key findings on the diversity and function of mast cells and how they integrate with host defense to prevent immune-mediated damage in barrier tissues and the central nervous system. Dashwood and Liston<span><sup>12</sup></span> bring us up to date on microglia biology, with mechanistic insights into cognitive development, synaptic pruning and new approaches to evaluate microglia function. Van Nieuwenhove<span><sup>13</sup></span> highlights major translational advances in the detection and treatment of monogenic and polygenic pediatric immune deficienci","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 6","pages":"412-413"},"PeriodicalIF":3.2,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12792","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141562149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bangyan Xu, Bethany M Anderson, Justine D Mintern, Laura E Edgington-Mitchell
Cysteine cathepsins are lysosomal proteases subject to dynamic regulation within antigen-presenting cells during the immune response and associated diseases. To investigate the regulation of cathepsin X, a carboxy-mono-exopeptidase, during maturation of dendritic cells (DCs), we exposed immortalized mouse DCs to various Toll-like receptor agonists. Using a cathepsin X-selective activity-based probe, sCy5-Nle-SY, we observed a significant increase in cathepsin X activation upon TLR-9 agonism with CpG, and to a lesser extent with Pam3 (TLR1/2), FSL-1 (TLR2/6) and LPS (TLR4). Despite clear maturation of DCs in response to Poly I:C (TLR3), cathepsin X activity was only slightly increased by this agonist, suggesting differential regulation of cathepsin X downstream of TLR activation. We demonstrated that cathepsin X was upregulated at the transcriptional level in response to CpG. This occurred at late time points and was not dampened by NF-κB inhibition. Factors secreted from CpG-treated cells were able to provoke cathepsin X upregulation when applied to naïve cells. Among these factors was IL-6, which on its own was sufficient to induce transcriptional upregulation and activation of cathepsin X. IL-6 is highly secreted by DCs in response to CpG but much less so in response to poly I:C, and inhibition of the IL-6 receptor subunit glycoprotein 130 prevented CpG-mediated cathepsin X upregulation. Collectively, these results demonstrate that cathepsin X is differentially transcribed during DC maturation in response to diverse stimuli, and that secreted IL-6 is critical for its dynamic regulation.
半胱氨酸酪蛋白是一种溶酶体蛋白酶,在免疫反应和相关疾病过程中会受到抗原递呈细胞内的动态调控。为了研究在树突状细胞(DCs)成熟过程中对羧基单外肽酶 X 的调控,我们将永生化小鼠 DCs 暴露于各种 Toll 样受体激动剂。通过使用基于活性的酪蛋白酶 X 选择性探针 sCy5-Nle-SY,我们观察到当 TLR-9 与 CpG(TLR1/2)、Pam3(TLR2/6)、FSL-1(TLR2/6)和 LPS(TLR4)激动时,酪蛋白酶 X 的活化显著增加。尽管直流细胞对 Poly I:C(TLR3)有明显的成熟反应,但这种激动剂只略微提高了酪蛋白酶 X 的活性,这表明酪蛋白酶 X 在 TLR 激活的下游有不同的调节作用。我们证实,CpG 在转录水平上上调了 cathepsin X 的活性。这种上调发生在晚期时间点,并且不会受到 NF-κB 抑制的抑制。将 CpG 处理过的细胞分泌的因子应用于天真细胞时,能够引起酪蛋白酶 X 的上调。这些因子中包括 IL-6,它本身就足以诱导转录上调和激活 cathepsin X。IL-6 在对 CpG 作出反应时由 DC 分泌得很高,但在对 poly I:C 作出反应时分泌得少得多,抑制 IL-6 受体亚基糖蛋白 130 能阻止 CpG 介导的 cathepsin X 上调。总之,这些结果表明,在 DC 成熟过程中,针对不同的刺激,酪蛋白酶 X 的转录是不同的,而分泌的 IL-6 对其动态调节至关重要。
{"title":"TLR9-dependent dendritic cell maturation promotes IL-6-mediated upregulation of cathepsin X","authors":"Bangyan Xu, Bethany M Anderson, Justine D Mintern, Laura E Edgington-Mitchell","doi":"10.1111/imcb.12806","DOIUrl":"10.1111/imcb.12806","url":null,"abstract":"<p>Cysteine cathepsins are lysosomal proteases subject to dynamic regulation within antigen-presenting cells during the immune response and associated diseases. To investigate the regulation of cathepsin X, a carboxy-mono-exopeptidase, during maturation of dendritic cells (DCs), we exposed immortalized mouse DCs to various Toll-like receptor agonists. Using a cathepsin X-selective activity-based probe, sCy5-Nle-SY, we observed a significant increase in cathepsin X activation upon TLR-9 agonism with CpG, and to a lesser extent with Pam3 (TLR1/2), FSL-1 (TLR2/6) and LPS (TLR4). Despite clear maturation of DCs in response to Poly I:C (TLR3), cathepsin X activity was only slightly increased by this agonist, suggesting differential regulation of cathepsin X downstream of TLR activation. We demonstrated that cathepsin X was upregulated at the transcriptional level in response to CpG. This occurred at late time points and was not dampened by NF-κB inhibition. Factors secreted from CpG-treated cells were able to provoke cathepsin X upregulation when applied to naïve cells. Among these factors was IL-6, which on its own was sufficient to induce transcriptional upregulation and activation of cathepsin X. IL-6 is highly secreted by DCs in response to CpG but much less so in response to poly I:C, and inhibition of the IL-6 receptor subunit glycoprotein 130 prevented CpG-mediated cathepsin X upregulation. Collectively, these results demonstrate that cathepsin X is differentially transcribed during DC maturation in response to diverse stimuli, and that secreted IL-6 is critical for its dynamic regulation.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 9","pages":"787-800"},"PeriodicalIF":3.2,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12806","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141557651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
UNC93B1 is essential for the stability and endosomal trafficking of nucleic-acid sensing Toll-like receptors (TLRs) including TLR7 and TLR8. Increased TLR7 responses are associated with lupus autoimmunity in both mice and humans. In a recent article, Al-Azab et al. demonstrate the role of a variant of UNC93B1 (p.V117L) in the induction of pediatric systemic lupus erythematosus in patients and in mice through TLR7/8 hyperresponsiveness. They also highlight a potential role for the pharmacological inhibition of interleukin-1 receptor–associated kinase (IRAK) 1 and/or 4 in ameliorating disease.
{"title":"UNCovering new causes of monogenic systemic lupus erythematosus","authors":"Julia I Ellyard, Michael P Gantier","doi":"10.1111/imcb.12807","DOIUrl":"10.1111/imcb.12807","url":null,"abstract":"<p>UNC93B1 is essential for the stability and endosomal trafficking of nucleic-acid sensing Toll-like receptors (TLRs) including TLR7 and TLR8. Increased TLR7 responses are associated with lupus autoimmunity in both mice and humans. In a recent article, Al-Azab <i>et al.</i> demonstrate the role of a variant of <i>UNC93B1</i> (p.V117L) in the induction of pediatric systemic lupus erythematosus in patients and in mice through TLR7/8 hyperresponsiveness. They also highlight a potential role for the pharmacological inhibition of interleukin-1 receptor–associated kinase (IRAK) 1 and/or 4 in ameliorating disease.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"102 8","pages":"651-654"},"PeriodicalIF":3.2,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12807","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141562148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In this article, we discuss new insights into the distinct mechanisms for V(D)J recombination for different immunoglobulin loci. This follows the recent revelation that recombination signal sequences (RSS) within the IGKV locus have evolved to be more efficient mediators of recombination activating gene (RAG) recombination compared to the same elements in the IGH locus. This difference in RSS strength is proposed to be driven by different molecular mechanisms for RAG-mediated recombination between the two loci.