Incomplete Freund's adjuvant (IFA) has been used for many years to induce autoimmune diseases in animal models, including experimental autoimmune encephalitis and collagen-induced arthritis. However, it remains unclear why it is necessary to emulsify autoantigen and heat-killed Mycobacterium tuberculosis (HKMtb) with IFA to induce experimental autoimmune diseases. Here, we found that immunization with self-antigen and HKMtb was insufficient to induce autoimmune diseases in mice. Furthermore, IFA or one of its components, mineral oil, but not mannide monooleate, was required for the development of experimental autoimmune disease. Immunization with autoantigen and HKMtb emulsified in mineral oil facilitated innate immune activation and promoted the differentiation of pathogenic CD4+ T cells, followed by their accumulation in neuronal tissues. Several water-soluble hydrocarbon compounds were identified in mineral oil. Of these, immunization with HKMtb and autoantigen emulsified with the same amount of hexadecane or tridecylcyclohexane as mineral oil induced the development of experimental autoimmune encephalitis. In contrast, immunization with HKMtb and autoantigen emulsified with tridecylcyclohexane, but not hexadecane, at doses equivalent to those found in mineral oil, resulted in neuronal dysfunction. These data indicate that tridecylcyclohexane in mineral oil is a critical component in the induction of experimental autoimmune disease.
不完全弗氏佐剂(IFA)多年来一直被用于诱导动物模型的自身免疫性疾病,包括实验性自身免疫性脑炎和胶原诱导性关节炎。然而,目前仍不清楚为什么必须用 IFA 乳化自身抗原和热死结核分枝杆菌(HKMtb)才能诱导实验性自身免疫疾病。在这里,我们发现用自身抗原和 HKMtb 进行免疫不足以诱发小鼠自身免疫性疾病。此外,实验性自身免疫疾病的发生需要 IFA 或其成分之一矿物油,但不需要单油酸甘露酯。用矿物油乳化的自身抗原和 HKMtb 进行免疫,可促进先天性免疫激活,促进致病 CD4+ T 细胞的分化,继而在神经元组织中积累。在矿物油中发现了几种水溶性碳氢化合物。其中,用与矿物油等量的十六烷或十三烷基环己烷乳化的香港霉菌和自身抗原进行免疫,会诱发实验性自身免疫性脑炎。相反,用HKMtb和用十三烷基环己烷(而不是十六烷)乳化的自身抗原进行免疫,其剂量与矿物油中的剂量相当,却会导致神经元功能障碍。这些数据表明,矿物油中的十三烷基环己烷是诱导实验性自身免疫疾病的关键成分。
{"title":"Tridecylcyclohexane in incomplete Freund's adjuvant is a critical component in inducing experimental autoimmune diseases","authors":"Norifumi Iijima, Tomoya Hayashi, Masaaki Niino, Yoichi Miyamoto, Masahiro Oka, Ken J Ishii","doi":"10.1002/eji.202350957","DOIUrl":"10.1002/eji.202350957","url":null,"abstract":"<p>Incomplete Freund's adjuvant (IFA) has been used for many years to induce autoimmune diseases in animal models, including experimental autoimmune encephalitis and collagen-induced arthritis. However, it remains unclear why it is necessary to emulsify autoantigen and heat-killed <i>Mycobacterium tuberculosis</i> (HKMtb) with IFA to induce experimental autoimmune diseases. Here, we found that immunization with self-antigen and HKMtb was insufficient to induce autoimmune diseases in mice. Furthermore, IFA or one of its components, mineral oil, but not mannide monooleate, was required for the development of experimental autoimmune disease. Immunization with autoantigen and HKMtb emulsified in mineral oil facilitated innate immune activation and promoted the differentiation of pathogenic CD4<sup>+</sup> T cells, followed by their accumulation in neuronal tissues. Several water-soluble hydrocarbon compounds were identified in mineral oil. Of these, immunization with HKMtb and autoantigen emulsified with the same amount of hexadecane or tridecylcyclohexane as mineral oil induced the development of experimental autoimmune encephalitis. In contrast, immunization with HKMtb and autoantigen emulsified with tridecylcyclohexane, but not hexadecane, at doses equivalent to those found in mineral oil, resulted in neuronal dysfunction. These data indicate that tridecylcyclohexane in mineral oil is a critical component in the induction of experimental autoimmune disease.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202350957","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141730818","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anna Bootz, Nina Reuter, Falk Nimmerjahn, William J. Britt, Michael Mach, Thomas H. Winkler
Human cytomegalovirus is a medically important pathogen. Previously, using murine CMV (MCMV), we provided evidence that both neutralizing and nonneutralizing antibodies can confer protection from viral infection in vivo. In this study, we report that serum derived from infected animals had a greater protective capacity in MCMV-infected RAG−/− mice than serum from animals immunized with purified virus. The protective activity of immune serum was strictly dependent on functional Fcγ receptors (FcγR). Deletion of individual FcγRs or combined deletion of FcγRI and FcγRIV had little impact on the protection afforded by serum. Adoptive transfer of CD115-positive cells from noninfected donors demonstrated that monocytes represent important cellular mediators of the protective activity provided by immune serum. Our studies suggest that Fc–FcγR interactions and monocytic cells are critical for antibody-mediated protection against MCMV infection in vivo. These findings may provide new avenues for the development of novel strategies for more effective CMV vaccines or antiviral immunotherapies.
{"title":"Functional Fc receptors are crucial in antibody-mediated protection against cytomegalovirus","authors":"Anna Bootz, Nina Reuter, Falk Nimmerjahn, William J. Britt, Michael Mach, Thomas H. Winkler","doi":"10.1002/eji.202451044","DOIUrl":"10.1002/eji.202451044","url":null,"abstract":"<p>Human cytomegalovirus is a medically important pathogen. Previously, using murine CMV (MCMV), we provided evidence that both neutralizing and nonneutralizing antibodies can confer protection from viral infection in vivo. In this study, we report that serum derived from infected animals had a greater protective capacity in MCMV-infected RAG<sup>−/−</sup> mice than serum from animals immunized with purified virus. The protective activity of immune serum was strictly dependent on functional Fcγ receptors (FcγR). Deletion of individual FcγRs or combined deletion of FcγRI and FcγRIV had little impact on the protection afforded by serum. Adoptive transfer of CD115-positive cells from noninfected donors demonstrated that monocytes represent important cellular mediators of the protective activity provided by immune serum. Our studies suggest that Fc–FcγR interactions and monocytic cells are critical for antibody-mediated protection against MCMV infection in vivo. These findings may provide new avenues for the development of novel strategies for more effective CMV vaccines or antiviral immunotherapies.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202451044","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141625492","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Charlotte M. Leane, Caroline E. Sutton, Barry Moran, Kingston H.G. Mills
The PD-1-PD-L1 immune checkpoint helps to maintain self-tolerance and prevent the development of autoimmune diseases. Immune checkpoint inhibitors are successful immunotherapeutics for several cancers, but responding patients can develop immune-mediated adverse events. It is well established that PD-1 regulates CD4 and CD8 T-cell responses, but its role in controlling the activation of pathogenic γδ T cells is less clear. Here we examined the role of PD-1 in regulating γδ T cells in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. We found that PD-1 was highly expressed on CD27− Vγ4 γδ T cells in the lymph node (LN) and CNS of mice with EAE. Treatment of mice with anti-PD-1 significantly augmented IL-17A-producing CD27− Vγ4 γδ T cells in the LN and CNS and enhanced the severity of EAE. The exacerbating effect of anti-PD-1 on EAE was lost in Tcrd−/− mice. Conversely, ligation of PD-1 suppressed Il17a and Rorc gene expression and IL-17A production by purified Vγ4 γδ T cells stimulated via the TCR, but not with IL-1β and IL-23. Our study demonstrates that PD-1 regulates TCR-activated CD27− Vγ4 γδ T cells, but that cytokine-activated IL-17A producing γδ T cells escape the regulatory effects of the PD-1-PD-L1 pathway.
{"title":"PD-1 regulation of pathogenic IL-17-secreting γδ T cells in experimental autoimmune encephalomyelitis","authors":"Charlotte M. Leane, Caroline E. Sutton, Barry Moran, Kingston H.G. Mills","doi":"10.1002/eji.202451212","DOIUrl":"10.1002/eji.202451212","url":null,"abstract":"<p>The PD-1-PD-L1 immune checkpoint helps to maintain self-tolerance and prevent the development of autoimmune diseases. Immune checkpoint inhibitors are successful immunotherapeutics for several cancers, but responding patients can develop immune-mediated adverse events. It is well established that PD-1 regulates CD4 and CD8 T-cell responses, but its role in controlling the activation of pathogenic γδ T cells is less clear. Here we examined the role of PD-1 in regulating γδ T cells in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. We found that PD-1 was highly expressed on CD27<sup>−</sup> Vγ4 γδ T cells in the lymph node (LN) and CNS of mice with EAE. Treatment of mice with anti-PD-1 significantly augmented IL-17A-producing CD27<sup>−</sup> Vγ4 γδ T cells in the LN and CNS and enhanced the severity of EAE. The exacerbating effect of anti-PD-1 on EAE was lost in <i>Tcrd</i><sup>−/−</sup> mice. Conversely, ligation of PD-1 suppressed <i>Il17a</i> and <i>Rorc</i> gene expression and IL-17A production by purified Vγ4 γδ T cells stimulated via the TCR, but not with IL-1β and IL-23. Our study demonstrates that PD-1 regulates TCR-activated CD27<sup>−</sup> Vγ4 γδ T cells, but that cytokine-activated IL-17A producing γδ T cells escape the regulatory effects of the PD-1-PD-L1 pathway.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202451212","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141597911","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Malignant cells are part of a complex network within the tumor microenvironment, where their interaction with host cells and soluble mediators, including complement components, is pivotal. The complement system, known for its role in immune defense and homeostasis, exhibits a dual effect on cancer progression. This dichotomy arises from its antitumoral opsonophagocytosis and cytotoxicity versus its protumoral chronic inflammation mediated by the C5a/C5aR1 axis, influencing antitumor T-cell responses. Recent studies have revealed distinct co-expression patterns of complement genes in various cancer types, correlating with prognosis. Notably, some cancers exhibit co-regulated overexpression of complement genes associated with poor prognosis, while others show favorable outcomes. However, significant intra-patient heterogeneity further complicates this classification. Moreover, the involvement of locally produced and intracellular complement proteins adds complexity to the tumor microenvironment dynamics. This review highlights the unique interplay of complement components within different cancers and patient cohorts, showing that “one size does not fit all”, for complement in cancer. It summarizes the clinical trials for complement targeting in cancer, emphasizing the need for tailored therapeutic approaches. By elucidating the mechanistic basis of complement's context-dependent role, this review aims to facilitate the development of personalized cancer therapies, ultimately improving patient care and outcomes.
恶性细胞是肿瘤微环境中复杂网络的一部分,它们与宿主细胞和包括补体成分在内的可溶性介质的相互作用至关重要。补体系统因其在免疫防御和平衡中的作用而闻名,但它对癌症的发展却有双重影响。这种二分法源于补体系统的抗肿瘤溶血吞噬作用和细胞毒性,以及由 C5a/C5aR1 轴介导的原瘤慢性炎症,后者会影响抗肿瘤 T 细胞的反应。最近的研究发现,补体基因在不同癌症类型中的共表达模式各不相同,并与预后相关。值得注意的是,一些癌症表现出与预后不良相关的补体基因共调过表达,而另一些则表现出良好的预后。然而,患者内部的明显异质性使这一分类更加复杂。此外,局部产生的补体蛋白和细胞内补体蛋白的参与也增加了肿瘤微环境动态的复杂性。本综述强调了补体成分在不同癌症和患者队列中的独特相互作用,说明补体在癌症中的作用并不是 "放之四海而皆准 "的。它总结了针对癌症补体的临床试验,强调了定制治疗方法的必要性。本综述旨在通过阐明补体的作用依赖于环境的机理基础,促进个性化癌症疗法的开发,最终改善患者的护理和预后。
{"title":"The complement system as a target in cancer immunotherapy","authors":"Nicolas S. Merle, Lubka T. Roumenina","doi":"10.1002/eji.202350820","DOIUrl":"10.1002/eji.202350820","url":null,"abstract":"<p>Malignant cells are part of a complex network within the tumor microenvironment, where their interaction with host cells and soluble mediators, including complement components, is pivotal. The complement system, known for its role in immune defense and homeostasis, exhibits a dual effect on cancer progression. This dichotomy arises from its antitumoral opsonophagocytosis and cytotoxicity versus its protumoral chronic inflammation mediated by the C5a/C5aR1 axis, influencing antitumor T-cell responses. Recent studies have revealed distinct co-expression patterns of complement genes in various cancer types, correlating with prognosis. Notably, some cancers exhibit co-regulated overexpression of complement genes associated with poor prognosis, while others show favorable outcomes. However, significant intra-patient heterogeneity further complicates this classification. Moreover, the involvement of locally produced and intracellular complement proteins adds complexity to the tumor microenvironment dynamics. This review highlights the unique interplay of complement components within different cancers and patient cohorts, showing that “one size does not fit all”, for complement in cancer. It summarizes the clinical trials for complement targeting in cancer, emphasizing the need for tailored therapeutic approaches. By elucidating the mechanistic basis of complement's context-dependent role, this review aims to facilitate the development of personalized cancer therapies, ultimately improving patient care and outcomes.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202350820","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141597912","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gina Leisching, Anjali Yennemadi, Karl Gogan, Joseph Keane
The impact of chronic exposure to type I interferons (IFN)-α2a, 2b, and β on macrophage metabolism, intimately linked to macrophage function, is not well understood. This study assesses the nuanced host responses induced by type I IFN cytokines, offering insights into potential therapeutic approaches in diseases associated with these cytokines. Employing a combination of transcriptional profiling and real-time functional analysis, we delineated metabolic reprogramming in response to chronic IFN exposure. Our results reveal distinct transcriptional metabolic profiles between macrophages chronically exposed to IFN-α and IFN-β. IFN-β significantly diminishes the oxygen consumption rate and glycolytic proton extrusion rate in macrophages. Conversely, IFN-α2b decreased parameters of mitochondrial fitness and induced a shift toward glutamine oxidation. Assessing the ability of macrophages to induce glycolysis in response to antigenic stimuli (LPS and iH37Rv), we found that chronic exposure to all IFN subtypes limited glycolytic induction. This study addresses a critical oversight in the literature, where individual roles of IFN subtypes are frequently amalgamated and lack distinction. These findings not only provide novel insights into the divergent effects of IFN-α2a, α2b, and β on macrophage metabolism but also highlight their potential implications for developing targeted therapeutic strategies.
{"title":"Interferon α and β induce differential transcriptional and functional metabolic phenotypes in human macrophages and blunt glycolysis in response to antigenic stimuli","authors":"Gina Leisching, Anjali Yennemadi, Karl Gogan, Joseph Keane","doi":"10.1002/eji.202451032","DOIUrl":"10.1002/eji.202451032","url":null,"abstract":"<p>The impact of chronic exposure to type I interferons (IFN)-α2a, 2b, and β on macrophage metabolism, intimately linked to macrophage function, is not well understood. This study assesses the nuanced host responses induced by type I IFN cytokines, offering insights into potential therapeutic approaches in diseases associated with these cytokines. Employing a combination of transcriptional profiling and real-time functional analysis, we delineated metabolic reprogramming in response to chronic IFN exposure. Our results reveal distinct transcriptional metabolic profiles between macrophages chronically exposed to IFN-α and IFN-β. IFN-β significantly diminishes the oxygen consumption rate and glycolytic proton extrusion rate in macrophages. Conversely, IFN-α2b decreased parameters of mitochondrial fitness and induced a shift toward glutamine oxidation. Assessing the ability of macrophages to induce glycolysis in response to antigenic stimuli (LPS and iH37Rv), we found that chronic exposure to all IFN subtypes limited glycolytic induction. This study addresses a critical oversight in the literature, where individual roles of IFN subtypes are frequently amalgamated and lack distinction. These findings not only provide novel insights into the divergent effects of IFN-α2a, α2b, and β on macrophage metabolism but also highlight their potential implications for developing targeted therapeutic strategies.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 9","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202451032","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141589121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Daniel Vasek, Peter Holicek, Frantisek Galatik, Anna Kratochvilova, Bianka Porubska, Veronika Somova, Natalie Fikarova, Michaela Hajkova, Martin Prevorovsky, Jitka M Zurmanova, Magdalena Krulova
The mammalian body possesses remarkable adaptability to cold exposure, involving intricate adjustments in cellular metabolism, ultimately leading to thermogenesis. However, cold-induced stress can impact immune response, primarily through noradrenaline-mediated pathways. In our study, we utilized a rat model subjected to short-term or long-term mild cold exposure to investigate systemic immune response during the cold acclimation. To provide human relevance, we included a group of regular cold swimmers in our study. Our research revealed complex relationship between cold exposure, neural signaling, immune response, and thermogenic regulation. One-day cold exposure triggered stress response, including cytokine production in white adipose tissue, subsequently activating brown adipose tissue, and inducing thermogenesis. We further studied systemic immune response, including the proportion of leukocytes and cytokines production. Interestingly, γδ T cells emerged as possible regulators in the broader systemic response, suggesting their possible contribution in the dynamic process of cold adaptation. We employed RNA-seq to gain further insights into the mechanisms by which γδ T cells participate in the response to cold. Additionally, we challenged rats exposed to cold with the Toll-like receptor 2 agonist, showing significant modulation of immune response. These findings significantly contribute to understanding of the physiological acclimation that occur in response to cold exposure.
哺乳动物的机体对寒冷具有极强的适应能力,这涉及到细胞代谢的复杂调整,最终导致产热。然而,寒冷诱导的压力会影响免疫反应,主要是通过去甲肾上腺素介导的途径。在我们的研究中,我们利用短期或长期轻度寒冷暴露的大鼠模型来研究寒冷适应过程中的全身免疫反应。为了提供与人类的相关性,我们在研究中加入了一组经常进行冷水游泳的人。我们的研究揭示了寒冷暴露、神经信号传导、免疫反应和产热调节之间的复杂关系。为期一天的寒冷暴露会引发应激反应,包括白色脂肪组织产生细胞因子,随后激活棕色脂肪组织并诱导产热。我们进一步研究了全身免疫反应,包括白细胞比例和细胞因子的产生。有趣的是,γδ T 细胞在更广泛的系统反应中成为可能的调节因子,这表明它们可能在寒冷适应的动态过程中做出了贡献。我们利用 RNA-seq进一步了解了γδT 细胞参与寒冷反应的机制。此外,我们还用 Toll 样受体 2 激动剂挑战了暴露于寒冷中的大鼠,结果显示免疫反应受到了显著的调节。这些发现大大有助于人们理解寒冷暴露下的生理适应。
{"title":"Immune response to cold exposure: Role of γδ T cells and TLR2-mediated inflammation","authors":"Daniel Vasek, Peter Holicek, Frantisek Galatik, Anna Kratochvilova, Bianka Porubska, Veronika Somova, Natalie Fikarova, Michaela Hajkova, Martin Prevorovsky, Jitka M Zurmanova, Magdalena Krulova","doi":"10.1002/eji.202350897","DOIUrl":"10.1002/eji.202350897","url":null,"abstract":"<p>The mammalian body possesses remarkable adaptability to cold exposure, involving intricate adjustments in cellular metabolism, ultimately leading to thermogenesis. However, cold-induced stress can impact immune response, primarily through noradrenaline-mediated pathways. In our study, we utilized a rat model subjected to short-term or long-term mild cold exposure to investigate systemic immune response during the cold acclimation. To provide human relevance, we included a group of regular cold swimmers in our study. Our research revealed complex relationship between cold exposure, neural signaling, immune response, and thermogenic regulation. One-day cold exposure triggered stress response, including cytokine production in white adipose tissue, subsequently activating brown adipose tissue, and inducing thermogenesis. We further studied systemic immune response, including the proportion of leukocytes and cytokines production. Interestingly, γδ T cells emerged as possible regulators in the broader systemic response, suggesting their possible contribution in the dynamic process of cold adaptation. We employed RNA-seq to gain further insights into the mechanisms by which γδ T cells participate in the response to cold. Additionally, we challenged rats exposed to cold with the Toll-like receptor 2 agonist, showing significant modulation of immune response. These findings significantly contribute to understanding of the physiological acclimation that occur in response to cold exposure.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202350897","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141578423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The antiallergic effects of gut microbiota have been attracting attention in recent years, but the underlying cellular and molecular mechanisms have not yet been fully understood. In this study, we aimed to investigate these mechanisms specifically focusing on mast cells. Mast cells retain intracellular granules containing various inflammatory mediators such as histamine, which are released outside the cells upon IgE and allergen stimulation. We previously reported that increased expression of the transcription factor, CCAAT/enhancer-binding protein α (C/EBPα), suppresses granule formation in mast cells and that Lacticaseibacillus casei JCM1134T (LC) upregulates C/EBPα levels. Here, granule formation in mouse bone marrow-derived mast cells was suppressed in a MyD88-dependent manner after LC treatment due to C/EBPα-dependent downregulation of the genes encoding serglycin (SRGN) and mast cell protease 4 (Mcpt4). Furthermore, C/EBPα expression was regulated by DNA methylation in the 5′ region far upstream of the transcription start site. LC suppressed DNA methylation of specific CpG motifs in the 5′ region of the C/EBPα gene. These results conclude that specific gut microbial components, such as those from LC, suppress granule formation in mast cells by inhibiting SRGN and Mcpt4 expression via reduced C/EBPα gene methylation.
{"title":"CCAAT/enhancer-binding protein α-dependent regulation of granule formation in mast cells by intestinal bacteria","authors":"Ayaka Iketani, Mai Takano, Kazumi Kasakura, Miono Iwatsuki, Ayu Tsuji, Kou Matsuda, Remina Minegishi, Akira Hosono, Yusuke Nakanishi, Kyoko Takahashi","doi":"10.1002/eji.202451094","DOIUrl":"10.1002/eji.202451094","url":null,"abstract":"<p>The antiallergic effects of gut microbiota have been attracting attention in recent years, but the underlying cellular and molecular mechanisms have not yet been fully understood. In this study, we aimed to investigate these mechanisms specifically focusing on mast cells. Mast cells retain intracellular granules containing various inflammatory mediators such as histamine, which are released outside the cells upon IgE and allergen stimulation. We previously reported that increased expression of the transcription factor, CCAAT/enhancer-binding protein α (C/EBPα), suppresses granule formation in mast cells and that <i>Lacticaseibacillus casei</i> JCM1134<sup>T</sup> (LC) upregulates C/EBPα levels. Here, granule formation in mouse bone marrow-derived mast cells was suppressed in a MyD88-dependent manner after LC treatment due to C/EBPα-dependent downregulation of the genes encoding serglycin (SRGN) and mast cell protease 4 (Mcpt4). Furthermore, C/EBPα expression was regulated by DNA methylation in the 5′ region far upstream of the transcription start site. LC suppressed DNA methylation of specific CpG motifs in the 5′ region of the C/EBPα gene. These results conclude that specific gut microbial components, such as those from LC, suppress granule formation in mast cells by inhibiting SRGN and Mcpt4 expression via reduced C/EBPα gene methylation.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141557642","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The emergence of 16S rRNA and metagenomic sequencing has gradually revealed the close relationship between dysbiosis and colorectal cancer (CRC). Recent studies have confirmed that intestinal dysbiosis plays various roles in the occurrence, development, and therapeutic response of CRC. Perturbation of host immunity is one of the key mechanisms involved. The intestinal microbiota, or specific bacteria and their metabolites, can modulate the progression of CRC through pathogen recognition receptor signaling or via the recruitment, polarization, and activation of both innate and adaptive immune cells to reshape the protumor/antitumor microenvironment. Therefore, the administration of gut bacteria to enhance immune homeostasis represents a new strategy for the treatment of CRC. In this review, we cover recent studies that illuminate the role of gut bacteria in the progression and treatment of CRC through orchestrating the immune response, which potentially offers insights for subsequent transformative research.
{"title":"Gut bacteria, host immunity, and colorectal cancer: From pathogenesis to therapy","authors":"Yuyi Li, Jinjin Peng, Xiangjun Meng","doi":"10.1002/eji.202451022","DOIUrl":"10.1002/eji.202451022","url":null,"abstract":"<p>The emergence of 16S rRNA and metagenomic sequencing has gradually revealed the close relationship between dysbiosis and colorectal cancer (CRC). Recent studies have confirmed that intestinal dysbiosis plays various roles in the occurrence, development, and therapeutic response of CRC. Perturbation of host immunity is one of the key mechanisms involved. The intestinal microbiota, or specific bacteria and their metabolites, can modulate the progression of CRC through pathogen recognition receptor signaling or via the recruitment, polarization, and activation of both innate and adaptive immune cells to reshape the protumor/antitumor microenvironment. Therefore, the administration of gut bacteria to enhance immune homeostasis represents a new strategy for the treatment of CRC. In this review, we cover recent studies that illuminate the role of gut bacteria in the progression and treatment of CRC through orchestrating the immune response, which potentially offers insights for subsequent transformative research.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202451022","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141557643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anna Laura Calvanese, Virginia Cecconi, Severin Stäheli, Daniel Schnepf, Marc Nater, Paulo Pereira, Julia Gschwend, Mathias Heikenwälder, Christoph Schneider, Burkhard Ludewig, Karina Silina, Maries van den Broek
Tertiary lymphoid structures (TLS) resemble follicles of secondary lymphoid organs and develop in nonlymphoid tissues during inflammation and cancer. Which cell types and signals drive the development of TLS is largely unknown. To investigate early events of TLS development in the lungs, we repeatedly instilled p(I:C) plus ovalbumin (Ova) intranasally. This induced TLS ranging from lymphocytic aggregates to organized and functional structures containing germinal centers. We found that TLS development is independent of FAP+ fibroblasts, alveolar macrophages, or CCL19 but crucially depends on type I interferon (IFN-I). Mechanistically, IFN-I initiates two synergistic pathways that culminate in the development of TLS. On the one hand, IFN-I induces lymphotoxin (LT)α in lymphoid cells, which stimulate stromal cells to produce the B-cell-attracting chemokine CXCL13 through LTβR-signaling. On the other hand, IFN-I is sensed by stromal cells that produce the T-cell-attracting chemokines CXCL9, CXCL10 as well as CCL19 and CCL21 independently of LTβR. Consequently, B-cell aggregates develop within a week, whereas follicular dendritic cells and germinal centers appear after 3 weeks. Thus, sustained production of IFN-I together with an antigen is essential for the induction of functional TLS in the lungs.
{"title":"Sustained innate interferon is an essential inducer of tertiary lymphoid structures","authors":"Anna Laura Calvanese, Virginia Cecconi, Severin Stäheli, Daniel Schnepf, Marc Nater, Paulo Pereira, Julia Gschwend, Mathias Heikenwälder, Christoph Schneider, Burkhard Ludewig, Karina Silina, Maries van den Broek","doi":"10.1002/eji.202451207","DOIUrl":"10.1002/eji.202451207","url":null,"abstract":"<p>Tertiary lymphoid structures (TLS) resemble follicles of secondary lymphoid organs and develop in nonlymphoid tissues during inflammation and cancer. Which cell types and signals drive the development of TLS is largely unknown. To investigate early events of TLS development in the lungs, we repeatedly instilled p(I:C) plus ovalbumin (Ova) intranasally. This induced TLS ranging from lymphocytic aggregates to organized and functional structures containing germinal centers. We found that TLS development is independent of FAP<sup>+</sup> fibroblasts, alveolar macrophages, or CCL19 but crucially depends on type I interferon (IFN-I). Mechanistically, IFN-I initiates two synergistic pathways that culminate in the development of TLS. On the one hand, IFN-I induces lymphotoxin (LT)α in lymphoid cells, which stimulate stromal cells to produce the B-cell-attracting chemokine CXCL13 through LTβR-signaling. On the other hand, IFN-I is sensed by stromal cells that produce the T-cell-attracting chemokines CXCL9, CXCL10 as well as CCL19 and CCL21 independently of LTβR. Consequently, B-cell aggregates develop within a week, whereas follicular dendritic cells and germinal centers appear after 3 weeks. Thus, sustained production of IFN-I together with an antigen is essential for the induction of functional TLS in the lungs.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202451207","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141557645","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nordin D. Zandhuis, Aurélie Guislain, Abeera Popalzij, Sander Engels, Branka Popović, Martin Turner, Monika C. Wolkers
CD8+ T cells kill target cells by releasing cytotoxic molecules and proinflammatory cytokines, such as TNF and IFN-γ. The magnitude and duration of cytokine production are defined by posttranscriptional regulation, and critical regulator herein are RNA-binding proteins (RBPs). Although the functional importance of RBPs in regulating cytokine production is established, the kinetics and mode of action through which RBPs control cytokine production are not well understood. Previously, we showed that the RBP ZFP36L2 blocks the translation of preformed cytokine encoding mRNA in quiescent memory T cells. Here, we uncover that ZFP36L2 regulates cytokine production in a time-dependent manner. T cell-specific deletion of ZFP36L2 (CD4-cre) had no effect on T-cell development or cytokine production during early time points (2–6 h) of T-cell activation. In contrast, ZFP36L2 specifically dampened the production of IFN-γ during prolonged T-cell activation (20–48 h). ZFP36L2 deficiency also resulted in increased production of IFN-γ production in tumor-infiltrating T cells that are chronically exposed to antigens. Mechanistically, ZFP36L2 regulates IFN-γ production at late time points of activation by destabilizing Ifng mRNA in an AU-rich element-dependent manner. Together, our results reveal that ZFP36L2 employs different regulatory nodules in effector and memory T cells to regulate cytokine production.
CD8+ T 细胞通过释放细胞毒性分子和促炎细胞因子(如 TNF 和 IFN-γ)杀死靶细胞。细胞因子产生的程度和持续时间由转录后调控决定,其中关键的调控因子是 RNA 结合蛋白(RBPs)。虽然 RBPs 在调控细胞因子产生方面的重要功能已被证实,但人们对 RBPs 控制细胞因子产生的动力学和作用模式还不十分清楚。此前,我们发现 RBP ZFP36L2 可阻断静止记忆 T 细胞中已形成的细胞因子编码 mRNA 的翻译。在这里,我们发现 ZFP36L2 以时间依赖性的方式调节细胞因子的产生。在T细胞活化的早期时间点(2-6小时),T细胞特异性缺失ZFP36L2(CD4-cre)对T细胞的发育或细胞因子的产生没有影响。与此相反,ZFP36L2 在 T 细胞长期活化(20-48 小时)过程中特异性地抑制了 IFN-γ 的产生。ZFP36L2 缺乏也会导致长期暴露于抗原的肿瘤浸润 T 细胞产生更多的 IFN-γ。从机理上讲,ZFP36L2 以一种依赖富含 AU 元素的方式破坏 Ifng mRNA 的稳定性,从而调节激活晚期 IFN-γ 的产生。总之,我们的研究结果揭示了 ZFP36L2 在效应 T 细胞和记忆 T 细胞中利用不同的调节节点来调节细胞因子的产生。
{"title":"Regulation of IFN-γ production by ZFP36L2 in T cells is time-dependent","authors":"Nordin D. Zandhuis, Aurélie Guislain, Abeera Popalzij, Sander Engels, Branka Popović, Martin Turner, Monika C. Wolkers","doi":"10.1002/eji.202451018","DOIUrl":"10.1002/eji.202451018","url":null,"abstract":"<p>CD8<sup>+</sup> T cells kill target cells by releasing cytotoxic molecules and proinflammatory cytokines, such as TNF and IFN-γ. The magnitude and duration of cytokine production are defined by posttranscriptional regulation, and critical regulator herein are RNA-binding proteins (RBPs). Although the functional importance of RBPs in regulating cytokine production is established, the kinetics and mode of action through which RBPs control cytokine production are not well understood. Previously, we showed that the RBP ZFP36L2 blocks the translation of preformed cytokine encoding mRNA in quiescent memory T cells. Here, we uncover that ZFP36L2 regulates cytokine production in a time-dependent manner. T cell-specific deletion of ZFP36L2 (CD4-cre) had no effect on T-cell development or cytokine production during early time points (2–6 h) of T-cell activation. In contrast, ZFP36L2 specifically dampened the production of IFN-γ during prolonged T-cell activation (20–48 h). ZFP36L2 deficiency also resulted in increased production of IFN-γ production in tumor-infiltrating T cells that are chronically exposed to antigens. Mechanistically, ZFP36L2 regulates IFN-γ production at late time points of activation by destabilizing <i>Ifng</i> mRNA in an AU-rich element-dependent manner. Together, our results reveal that ZFP36L2 employs different regulatory nodules in effector and memory T cells to regulate cytokine production.</p>","PeriodicalId":165,"journal":{"name":"European Journal of Immunology","volume":"54 10","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/eji.202451018","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141557644","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}