Germinal center response

  • 文章类型: Journal Article
    γ疱疹病毒无处不在,与多种癌症相关的终生病原体感染超过95%的成年人。病毒再激活的增加,由于压力和其他未知因素影响免疫反应,经常先于淋巴发生。一种可能促进病毒再激活和增加病毒潜伏期的潜在应激源将是我们一生中经历的来自细菌和病毒病原体的无数感染。使用鼠γ疱疹病毒68(MHV68),γ疱疹病毒感染的小鼠模型,我们研究了细菌攻击对γ疱疹病毒感染的影响。我们在用不可分型的流感嗜血杆菌(NTHi)建立潜伏期期间对MHV68感染的小鼠进行了挑战,以确定细菌感染对病毒再激活和潜伏期的影响。小鼠感染了MHV68,然后用NTHi攻击,病毒再激活和病毒潜伏期增加。这些数据支持以下假设:细菌攻击可以促进γ疱疹病毒的再激活和潜伏期的建立,对病毒淋巴发生有可能的后果。
    Gammaherpesviruses are ubiquitous, lifelong pathogens associated with multiple cancers that infect over 95% of the adult population. Increases in viral reactivation, due to stress and other unknown factors impacting the immune response, frequently precedes lymphomagenesis. One potential stressor that could promote viral reactivation and increase viral latency would be the myriad of infections from bacterial and viral pathogens that we experience throughout our lives. Using murine gammaherpesvirus 68 (MHV68), a mouse model of gammaherpesvirus infection, we examined the impact of bacterial challenge on gammaherpesvirus infection. We challenged MHV68 infected mice during the establishment of latency with nontypeable Haemophilus influenzae (NTHi) to determine the impact of bacterial infection on viral reactivation and latency. Mice infected with MHV68 and then challenged with NTHi, saw increases in viral reactivation and viral latency. These data support the hypothesis that bacterial challenge can promote gammaherpesvirus reactivation and latency establishment, with possible consequences for viral lymphomagenesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    迫切需要开发安全有效的疫苗,以诱导针对SARS-CoV-2的有效细胞和体液免疫反应,以结束全球COVID-19的流行。这里,我们开发了一种基于甲病毒来源的自我复制RNA(repRNA)的疫苗平台,该平台编码SARS-CoV-2刺突糖蛋白的受体结合域(RBD).由于repRNA的复制机制,与常规mRNA相比,repRNA触发延长的抗原表达。为了提高repRNA的递送和疫苗功效,我们开发了一种自组装脂质体-鱼精蛋白-RNA(LPR)纳米颗粒,具有高效的包封和转染repRNA。LPR-repRNA疫苗显著激活I型干扰素应答和先天免疫信号通路。LPR-repRNA-RBD的皮下免疫导致抗原表达延长,刺激先天免疫细胞,并诱导引流淋巴结的生发中心反应。LPR-repRNA-RBD诱导抗原特异性T细胞反应,并使细胞免疫偏向效应记忆CD8+T细胞反应。用LPR-repRNA-RBD免疫引发了抗RBDIgG抗体的产生,并诱导了针对SARS-CoV-2假病毒的中和抗体反应。LPR-repRNA-RBD疫苗减少小鼠SARS-CoV-2感染和肺部炎症。总之,这些数据表明,LPR-repRNA平台可能是COVID-19疫苗开发的一个有希望的途径.
    The creation of safe and effective vaccines that induce potent cellular and humoral immune responses against SARS-CoV-2 is urgently needed to end the global COVID-19 epidemic. Here, we developed an alphavirus-derived self-replicating RNA (repRNA)-based vaccine platform encoding the receptor-binding domain (RBD) of SARS-CoV-2 spike glycoprotein. The repRNA triggers prolonged antigen expression compared with conventional mRNA due to the replication machinery of repRNA. To improve the delivery and vaccine efficacy of repRNA, we developed a self-assembling liposome-protamine-RNA (LPR) nanoparticle with highly efficient encapsulation and transfection of repRNA. LPR-repRNA vaccines substantially activated type I interferon response and innate immune signaling pathways. Subcutaneous immunization of LPR-repRNA-RBD led to prolonged antigen expression, stimulation of innate immune cells, and induction of germinal center response in draining lymph nodes. LPR-repRNA-RBD induced antigen-specific T cell responses and skewed cellular immunity toward an effector memory CD8+ T cell response. Immunizations with LPR-repRNA-RBD triggered the production of anti-RBD IgG antibodies and induced neutralizing antibody response against SARS-CoV-2 pseudovirus. LPR-repRNA-RBD vaccines reduced SARS-CoV-2 infection and lung inflammation in mice. Altogether, these data suggest that the LPR-repRNA platform can be a promising avenue for COVID-19 vaccine development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    我们先前已经表明,由RNA酶Drosha和DiGeorge临界区(DGCR)8蛋白组成的microRNA(miRNA)处理器复合物对于B细胞成熟至关重要。为了确定是否需要miRNA加工来启动T细胞介导的抗体反应,我们通过将具有loxP侧翼DGCR8等位基因的小鼠与CD23-Cre小鼠杂交,在成熟的B2细胞中删除了DGCR8。不出所料,未免疫的小鼠显示成熟B2细胞和IgG分泌细胞的数量减少,血清IgG滴度降低。InAccording,用T依赖性抗原免疫的小鼠中不存在生发中心和抗原特异性IgG分泌细胞。因此,需要DGCR8来建立有效的T依赖性抗体应答。然而,B1细胞中DGCR8缺失不完整,在DGCR8敲除小鼠中产生未改变的B1细胞数量和正常的IgM和IgA滴度。因此,该小鼠模型可用于分析缺乏功能性B2细胞时的B1反应.
    We have previously shown that the microRNA (miRNA) processor complex consisting of the RNAse Drosha and the DiGeorge Critical Region (DGCR) 8 protein is essential for B cell maturation. To determine whether miRNA processing is required to initiate T cell-mediated antibody responses, we deleted DGCR8 in maturing B2 cells by crossing a mouse with loxP-flanked DGCR8 alleles with a CD23-Cre mouse. As expected, non-immunized mice showed reduced numbers of mature B2 cells and IgG-secreting cells and diminished serum IgG titers. In accordance, germinal centers and antigen-specific IgG-secreting cells were absent in mice immunized with T-dependent antigens. Therefore, DGCR8 is required to mount an efficient T-dependent antibody response. However, DGCR8 deletion in B1 cells was incomplete, resulting in unaltered B1 cell numbers and normal IgM and IgA titers in DGCR8-knock-out mice. Therefore, this mouse model could be used to analyze B1 responses in the absence of functional B2 cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    γ疱疹病毒感染大多数脊椎动物物种,并与B细胞淋巴瘤有关。操纵B细胞分化对于由γ疱疹病毒驱动的自然感染和淋巴生成至关重要。具体来说,人类爱泼斯坦-巴尔病毒(EBV)和鼠γ疱疹病毒68(MHV68)驱动感染的幼稚B细胞分化为生发中心,以在慢性感染建立过程中实现潜伏病毒储库的指数增加。感染的生发中心B细胞也是病毒淋巴发生的目标,因为大多数EBV阳性B细胞淋巴瘤具有生发中心反应的特征。所有γ疱疹病毒都编码一种蛋白激酶,which,在卡波西肉瘤相关疱疹病毒(KSHV)和MHV68的情况下,是足够和必要的,分别,驱动体内B细胞分化。在这项研究中,我们使用高度可处理的慢性γ疱疹病毒感染的MHV68模型揭示了MHV68蛋白激酶与干扰素调节因子1(IRF-1)之间的拮抗关系.IRF-1缺乏对体内无激酶MHV68的裂解复制减弱的影响最小。相比之下,在IRF-1-/-小鼠中,激酶无效的MHV68的减弱的潜伏储库被部分完全挽救,以及完全抢救MHV68驱动的生发中心反应。因此,新型病毒蛋白激酶-IRF-1拮抗作用主要限于以病毒潜伏期为主的慢性感染,与急性感染期间和体外裂解复制的相关性较小.鉴于病毒和宿主蛋白的保守性,两者之间的对立,根据这项研究的定义,可以调节跨物种的γ疱疹病毒感染。重要性γ疱疹病毒是操纵生理B细胞分化以建立终身感染的流行病原体。这种操作也涉及γ疱疹病毒驱动的B细胞淋巴瘤,作为潜伏感染的B细胞的分化,通过生发中心反应靶向这些转化。在这项研究中,我们定义了一种保守的γ疱疹病毒蛋白激酶与宿主抗病毒和肿瘤抑制转录因子之间的新型拮抗相互作用。在这项研究中揭示的病毒-宿主拮抗作用对于塑造γ疱疹病毒驱动的生发中心反应的程度至关重要。相比之下,病毒-宿主拮抗作用与体外和体内急性感染期间的裂解病毒复制关系不大,强调了新兴的概念,即非重叠机制决定了急性和慢性γ疱疹病毒感染的参数。
    Gammaherpesviruses infect most vertebrate species and are associated with B cell lymphomas. Manipulation of B cell differentiation is critical for natural infection and lymphomagenesis driven by gammaherpesviruses. Specifically, human Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) drive differentiation of infected naive B cells into the germinal center to achieve exponential increase in the latent viral reservoir during the establishment of chronic infection. Infected germinal center B cells are also the target of viral lymphomagenesis, as most EBV-positive B cell lymphomas bear the signature of the germinal center response. All gammaherpesviruses encode a protein kinase, which, in the case of Kaposi\'s sarcoma-associated herpesvirus (KSHV) and MHV68, is sufficient and necessary, respectively, to drive B cell differentiation in vivo. In this study, we used the highly tractable MHV68 model of chronic gammaherpesvirus infection to unveil an antagonistic relationship between MHV68 protein kinase and interferon regulatory factor 1 (IRF-1). IRF-1 deficiency had minimal effect on the attenuated lytic replication of the kinase-null MHV68 in vivo. In contrast, the attenuated latent reservoir of the kinase-null MHV68 was partially to fully rescued in IRF-1-/- mice, along with complete rescue of the MHV68-driven germinal center response. Thus, the novel viral protein kinase-IRF-1 antagonism was largely limited to chronic infection dominated by viral latency and was less relevant for lytic replication during acute infection and in vitro. Given the conserved nature of the viral and host protein, the antagonism between the two, as defined in this study, may regulate gammaherpesvirus infection across species. IMPORTANCE Gammaherpesviruses are prevalent pathogens that manipulate physiological B cell differentiation to establish lifelong infection. This manipulation is also involved in gammaherpesvirus-driven B cell lymphomas, as differentiation of latently infected B cells through the germinal center response targets these for transformation. In this study, we define a novel antagonistic interaction between a conserved gammaherpesvirus protein kinase and a host antiviral and tumor suppressor transcription factor. The virus-host antagonism unveiled in this study was critically important to shape the magnitude of gammaherpesvirus-driven germinal center response. In contrast, the virus-host antagonism was far less relevant for lytic viral replication in vitro and during acute infection in vivo, highlighting the emerging concept that nonoverlapping mechanisms shape the parameters of acute and chronic gammaherpesvirus infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    γ疱疹病毒,如人EB病毒(EBV)和鼠γ疱疹病毒68(MHV68),是物种特异性的,与多种癌症相关的无处不在的病原体,包括B细胞淋巴瘤.这些病毒对B细胞具有天然的嗜性,并篡夺B细胞分化,以驱动独特而强大的多克隆生发中心反应,从而在记忆B细胞中建立长期的潜伏库。由γ疱疹病毒感染驱动的强大的多克隆生发中心反应增加了B细胞转化的风险。毫不奇怪,许多γ疱疹病毒癌症来源于生发中心或生发后中心B细胞。影响γ疱疹病毒驱动的生发中心反应的病毒和宿主因素尚未明确定义。我们先前表明,宿主白介素17受体A(IL-17RA)信号促进慢性MHV68感染的建立和MHV68驱动的生发中心反应。在这项研究中,我们发现T细胞固有的IL-17RA信号在MHV68感染期间概括了总体IL-17RA信号的一些前病毒方面.具体来说,我们发现T细胞固有的IL-17RA信号支持MHV68驱动的生发中心反应,脾脏潜伏期的建立,以及病毒在脾脏和腹膜腔的再激活。我们的研究揭示了一个意想不到的发现,即T细胞特异性IL-17RA信号支持建立B细胞嗜性γ疱疹病毒的潜伏库。重要性伽玛疱疹病毒,比如人类EBV,在>95%的成年人中建立终身感染,并与B细胞淋巴瘤相关。γ疱疹病毒篡夺生发中心反应以建立潜伏感染,生发中心B细胞被认为是病毒转化的目标。我们先前发现IL-17RA的整体表达促进慢性MHV68感染的建立和MHV68驱动的生发中心反应。在这项研究中,我们表明,T细胞固有IL-17RA信号传导是通过支持CD4+T滤泡辅助细胞扩增促进MHV68驱动的生发中心反应所必需的.我们还发现,T细胞固有的IL-17RA信号有助于但不完全负责IL-17RA信号的全身前病毒作用,强调MHV68感染期间IL-17RA信号传导的多方面功能。
    Gammaherpesviruses, such as human Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68), are species-specific, ubiquitous pathogens that are associated with multiple cancers, including B cell lymphomas. These viruses have a natural tropism for B cells and usurp B cell differentiation to drive a unique and robust polyclonal germinal center response to establish a long-term latent reservoir in memory B cells. The robust polyclonal germinal center response driven by gammaherpesvirus infection increases the risk for B cell transformation. Unsurprisingly, many gammaherpesvirus cancers are derived from germinal center or post-germinal center B cells. The viral and host factors that influence the gammaherpesvirus-driven germinal center response are not clearly defined. We previously showed that host interleukin 17 receptor A (IL-17RA) signaling promotes the establishment of chronic MHV68 infection and the MHV68-driven germinal center response. In this study, we found that T cell-intrinsic IL-17RA signaling recapitulates some proviral aspects of global IL-17RA signaling during MHV68 infection. Specifically, we found that T cell-intrinsic IL-17RA signaling supports the MHV68-driven germinal center response, the establishment of latency in the spleen, and viral reactivation in the spleen and peritoneal cavity. Our study unveils an unexpected finding where the T cell-specific IL-17RA signaling supports the establishment of a latent reservoir of a B cell-tropic gammaherpesvirus. IMPORTANCE Gammaherpesviruses, such as human EBV, establish lifelong infection in >95% of adults and are associated with B cell lymphomas. Gammaherpesviruses usurp the germinal center response to establish latent infection, and the germinal center B cells are thought to be the target of viral transformation. We previously found that global expression of IL-17RA promotes the establishment of chronic MHV68 infection and the MHV68-driven germinal center response. In this study, we showed that T cell-intrinsic IL-17RA signaling is necessary to promote the MHV68-driven germinal center response by supporting CD4+ T follicular helper cell expansion. We also found that T cell-intrinsic IL-17RA signaling contributes to but is not solely responsible for the systemic proviral role of IL-17RA signaling, highlighting the multifaceted function of IL-17RA signaling during MHV68 infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在对T细胞依赖性抗原的反应中,在与CD4+T细胞同源相互作用后,有抗原经历的B细胞迁移到B细胞卵泡的中心,以种子生发中心(GC)反应。这些GCB细胞最终成熟为记忆和长寿命的抗体分泌浆细胞,从而产生长寿的体液免疫。在GC内,B细胞经历其B细胞受体(BCR)的体细胞超突变和阳性选择,以出现高亲和力抗原特异性B细胞克隆。然而,这个过程可能很危险,由于异常突变的积累可能导致GCB细胞的恶性转化或产生可引起自身免疫的自身反应性B细胞克隆。正因为如此,更好地了解GC的发展为潜在的病理过程提供了诊断和治疗线索。生产性GC响应由多种机制协调。GC反应的一个新兴的重要调节剂是表观遗传调节,具有关键的转录调控特性。在这次审查中,我们总结了目前有关GC反应调节中表观遗传机制生物学的知识,并概述了其在鉴定免疫治疗决策中的重要性。
    In response to T-cell-dependent antigens, antigen-experienced B cells migrate to the center of the B-cell follicle to seed the germinal center (GC) response after cognate interactions with CD4+ T cells. These GC B cells eventually mature into memory and long-lived antibody-secreting plasma cells, thus generating long-lived humoral immunity. Within GC, B cells undergo somatic hypermutation of their B cell receptors (BCR) and positive selection for the emergence of high-affinity antigen-specific B-cell clones. However, this process may be dangerous, as the accumulation of aberrant mutations could result in malignant transformation of GC B cells or give rise to autoreactive B cell clones that can cause autoimmunity. Because of this, better understanding of GC development provides diagnostic and therapeutic clues to the underlying pathologic process. A productive GC response is orchestrated by multiple mechanisms. An emerging important regulator of GC reaction is epigenetic modulation, which has key transcriptional regulatory properties. In this review, we summarize the current knowledge on the biology of epigenetic mechanisms in the regulation of GC reaction and outline its importance in identification of immunotherapy decision making.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Gammaherpesviruses establish lifelong infection and are associated with a variety of cancers, including B cell lymphomas. These viruses manipulate the B cell differentiation process to establish lifelong infection in memory B cells. Specifically, gammaherpesviruses infect naive B cells and promote entry of both infected and uninfected naive B cells into germinal centers, where the virus usurps rapid proliferation of germinal center B cells to exponentially increase its cellular latent reservoir. In addition to facilitating the establishment of latent infection, germinal center B cells are thought to be the target of viral transformation. In this study, we have uncovered a novel proviral role of host interleukin 17A (IL-17A), a well-established antibacterial and antifungal factor. Loss of IL-17A signaling attenuated the establishment of chronic gammaherpesvirus infection and gammaherpesvirus-driven germinal center response in a route of inoculation-dependent manner. Further, IL-17A treatment directly supported gammaherpesvirus reactivation and de novo lytic infection. This study is the first demonstration of a multifaceted proviral role of IL-17 signaling.IMPORTANCE Gammaherpesviruses establish lifelong infections in a majority of humans and are associated with B cell lymphomas. IL-17A is a host cytokine that plays a well-established role in the clearance of bacterial and fungal infections; however, the role of IL-17A in viral infections is poorly understood. In this study, we show that IL-17A signaling promoted the establishment of chronic gammaherpesvirus infection following the mucosal route of infection, viral lytic replication, and reactivation from latency. Thus, our study unveils a novel proviral role of IL-17A signaling in gammaherpesvirus infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    免疫系统在保护宿主免受传染病和癌症的侵害中起着至关重要的作用。值得注意的是,来自免疫系统的适应性臂的B和T淋巴细胞可以协作以形成长寿命的抗体应答,并且因此是疫苗接种方法中的主要靶标。然而,必须严格调节保护性免疫反应,以避免高反应性和可导致自身免疫的针对自身的反应。核受体(NRs)非常适合快速改变转录细胞对改变的环境设置的反应。它们的功能作用与免疫缺陷和自身免疫有关。尽管核受体功能与特定的CD4T辅助亚群有广泛的联系,关于CD4滤泡辅助性T细胞(Tfh)和生发中心(GC)B细胞在生发中心反应过程中的功能作用和机制的研究刚刚兴起。我们回顾了我们对GC反应的特定细胞类型中NR调节的理解的最新进展,并讨论了它们对自身免疫性疾病如系统性红斑狼疮(SLE)的影响。
    The immune system plays an essential role in protecting the host from infectious diseases and cancer. Notably, B and T lymphocytes from the adaptive arm of the immune system can co-operate to form long-lived antibody responses and are therefore the main target in vaccination approaches. Nevertheless, protective immune responses must be tightly regulated to avoid hyper-responsiveness and responses against self that can result in autoimmunity. Nuclear receptors (NRs) are perfectly adapted to rapidly alter transcriptional cellular responses to altered environmental settings. Their functional role is associated with both immune deficiencies and autoimmunity. Despite extensive linking of nuclear receptor function with specific CD4 T helper subsets, research on the functional roles and mechanisms of specific NRs in CD4 follicular T helper cells (Tfh) and germinal center (GC) B cells during the germinal center reaction is just emerging. We review recent advances in our understanding of NR regulation in specific cell types of the GC response and discuss their implications for autoimmune diseases such as systemic lupus erythematosus (SLE).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Patients with Down syndrome (DS) are characterized by increased susceptibility to autoimmunity and respiratory tract infections that are suggestive of humoral immunity impairment. Here, we sought to determine the follicular helper (Tfh) and follicular regulatory (Tfr) T cell profile in the blood of children with DS. Blood was collected from 24 children with DS, nine of which had autoimmune diseases. Children with DS showed skewed Tfh differentiation towards the CXCR3+ phenotype: Tfh1 and Tfh1/17 subsets were increased, while Tfh2 and Tfh17 subsets were reduced. While no differences in the percentage of Tfr cells were seen, the ratio of Tfh1 and CXCR3+PD-1+ subsets to Tfr cells was significantly increased in the affected children. The excessive polarization towards a CXCR3+ phenotype in children with DS suggests that re-calibration of Tfh subset skewing could potentially offer new therapeutic opportunities for these patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The germinal center (GC) response is critical for generating high-affinity humoral immunity and immunological memory, which forms the basis of successful immunization. Control of the GC response is thought to require follicular regulatory T (Tfr) cells, a subset of suppressive Foxp3+ regulatory T cells located within GCs. Relatively little is known about the exact role of Tfr cells within the GC and how they exert their suppressive function. A unique feature of Tfr cells is their reported CXCR5-dependent localization to the GC. Here, we show that the lack of CXCR5 on Foxp3+ regulatory T cells results in a reduced frequency, but not an absence, of GC-localized Tfr cells. This reduction in Tfr cells is not sufficient to alter the magnitude or output of the GC response. This demonstrates that additional, CXCR5-independent mechanisms facilitate Treg cell homing to the GC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号