Receptors, Interleukin-7

受体,白细胞介素 - 7
  • 文章类型: Journal Article
    IL-7R调节体内平衡,激活,以及T细胞在外周组织中的分布。尽管已经鉴定了几种调节αβT细胞中IL-7Rα表达的转录增强子,在γδT细胞中活跃的增强子仍然未知。在这篇文章中,我们在IL-7Rα链(IL-7Rα)基因座的内含子2中发现了一个进化上保守的非编码序列(CNS),并将该区域命名为CNS9。CNS9包含保守的视黄酸受体相关的孤儿受体(ROR)响应元件(RORE),并在体外发挥RORγt依赖性增强剂活性。在CNS9(CNS9-RORmut)的RORE中携带点突变的小鼠在产生IL-17的Vγ4γδT细胞中显示IL-7Rα表达降低。此外,CNS9-RORmut小鼠脂肪组织中Vγ4γδT细胞的细胞数量和IL-17A的产生减少。与IL-17A的减少一致,CNS9-RORmut小鼠脂肪组织中IL-33表达降低,导致较少的调节性T细胞和葡萄糖耐受不良。CNS9-ROR基序部分负责RORγt+调节性T细胞中IL-7Rα的表达,而IL-7Rα表达在RORγt表达Vγ2+γδT细胞中不受影响,Th17细胞,3型先天淋巴细胞,和不变的NKT细胞。我们的结果表明,CNS9是RORE依赖的,Vγ4+γδT细胞特异性IL-7Rα增强子,通过调节性T细胞在脂肪组织稳态中起关键作用,提示IL-7Rα内含子2中进化保守的RORE可能影响2型糖尿病的发病率。
    The IL-7R regulates the homeostasis, activation, and distribution of T cells in peripheral tissues. Although several transcriptional enhancers that regulate IL-7Rα expression in αβ T cells have been identified, enhancers active in γδ T cells remain unknown. In this article, we discovered an evolutionarily conserved noncoding sequence (CNS) in intron 2 of the IL-7Rα-chain (IL-7Rα) locus and named this region CNS9. CNS9 contained a conserved retinoic acid receptor-related orphan receptor (ROR)-responsive element (RORE) and exerted RORγt-dependent enhancer activity in vitro. Mice harboring point mutations in the RORE in CNS9 (CNS9-RORmut) showed reduced IL-7Rα expression in IL-17-producing Vγ4+ γδ T cells. In addition, the cell number and IL-17A production of Vγ4+ γδ T cells were reduced in the adipose tissue of CNS9-RORmut mice. Consistent with the reduction in IL-17A, CNS9-RORmut mice exhibited decreased IL-33 expression in the adipose tissue, resulting in fewer regulatory T cells and glucose intolerance. The CNS9-ROR motif was partially responsible for IL-7Rα expression in RORγt+ regulatory T cells, whereas IL-7Rα expression was unaffected in RORγt-expressing Vγ2+ γδ T cells, Th17 cells, type 3 innate lymphoid cells, and invariant NKT cells. Our results indicate that CNS9 is a RORΕ-dependent, Vγ4+ γδ T cell-specific IL-7Rα enhancer that plays a critical role in adipose tissue homeostasis via regulatory T cells, suggesting that the evolutionarily conserved RORΕ in IL-7Rα intron 2 may influence the incidence of type 2 diabetes.
    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
    T细胞受体(TCR)发出的信号,共刺激受体,和细胞因子受体各自影响CD8T细胞命运。了解这些信号对稳态和微环境线索的反应可以揭示治疗性指导T细胞功能的新方法。通过小鼠的正向遗传筛查,我们发现LDL受体相关蛋白10(Lrp10)的功能缺失突变导致幼稚和中枢记忆性CD8T细胞在外周淋巴器官中积累.Lrp10编码免疫功能未知的保守细胞表面蛋白。T细胞活化诱导Lrp10表达,它在翻译后抑制IL7受体(IL7R)水平。因此,Lrp10缺失通过IL7R信号增强T细胞稳态扩增。Lrp10缺陷型小鼠也对同基因肿瘤具有内在抗性。这种表型取决于CD8T细胞的致密肿瘤浸润,显示出增强的存储单元特性,减少终端疲惫,和增强对免疫检查点抑制的反应。这里,我们提出Lrp10作为CD8T细胞稳态的一种新的负调节因子和一种控制肿瘤耐药的宿主因子,对免疫治疗有意义.
    Signals emanating from the T-cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T-cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T-cell function. Through forward genetic screening in mice, we discover that loss-of-function mutations in LDL receptor-related protein 10 (Lrp10) cause naive and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. T-cell activation induces Lrp10 expression, which post-translationally suppresses IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhances T-cell homeostatic expansion through IL7R signaling. Lrp10-deficient mice are also intrinsically resistant to syngeneic tumors. This phenotype depends on dense tumor infiltration of CD8 T cells, which display increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T-cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.
    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
    γδT细胞对免疫反应的贡献与干扰素-γ(IFN-γ)的快速分泌有关。这里,我们显示了先天性IFN-γ产生γδT细胞的围产期胸腺波,该细胞表达CD8αβ异二聚体,并在感染和癌症的临床前模型中扩展。最佳⑶8αβ+γδT细胞发育由低T细胞受体信号传导和通过提供白介素(IL)-4和IL-7来指导。该人群在病理上与过度活跃有关,或构成,在两种T细胞瘤形成的小鼠模型中,IL-7R-STAT5B信号传导促进胸腺和外周淋巴器官中CD8αβγδT细胞的超生理积累。同样,CD8αβ+γδT细胞定义了人类T细胞急性淋巴细胞白血病儿科患者的不同子集。这项工作描述了CD8αβ+γδT细胞的正常和恶性发育,这些细胞在生命早期富集,并有助于对感染和癌症的先天IFN-γ反应。
    The contribution of γδ T cells to immune responses is associated with rapid secretion of interferon-γ (IFN-γ). Here, we show a perinatal thymic wave of innate IFN-γ-producing γδ T cells that express CD8αβ heterodimers and expand in preclinical models of infection and cancer. Optimal CD8αβ+ γδ T cell development is directed by low T cell receptor signaling and through provision of interleukin (IL)-4 and IL-7. This population is pathologically relevant as overactive, or constitutive, IL-7R-STAT5B signaling promotes a supraphysiological accumulation of CD8αβ+ γδ T cells in the thymus and peripheral lymphoid organs in two mouse models of T cell neoplasia. Likewise, CD8αβ+ γδ T cells define a distinct subset of human T cell acute lymphoblastic leukemia pediatric patients. This work characterizes the normal and malignant development of CD8αβ+ γδ T cells that are enriched in early life and contribute to innate IFN-γ responses to infection and cancer.
    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
    共同γ受体链(γc)家族的细胞因子对于T细胞分化至关重要,而γc细胞因子途径的失调与自身免疫性疾病的发病机理有关。越来越多的证据表明,相关受体链的γc受体(CD132)的可用性对T细胞功能具有影响。在这里,我们研究了差异γc表达对IL-2Rα(CD25)表达的影响,IL-7Rα(CD127)和活化的初始T细胞的分化。我们使用小发夹(sh)RNA和γccDNA通过慢病毒转导微调了人原代幼稚T细胞中γc表达的调节。然后分析差异γc水平对活化后T细胞表型和功能的影响。差异γc表达显着影响活化的初始T细胞上的IL-2Rα和IL-7Rα表达。高γc表达(γc-high)诱导激活后IL-2Rα的表达和IL-7Rα的再表达。γc的抑制导致较低的IL-2Rα/IL-7Rα表达和活化的初始T细胞的增殖受损。相比之下,γc-高T细胞分泌显著更高浓度的效应细胞因子(即,IFN-γ,IL-6)并在初始阶段显示出较高的细胞因子受体诱导的STAT5磷酸化,以及活化后持续较高的pSTAT1和pSTAT3水平。最后,特别是对于CD4+γc高幼稚T细胞,观察到向表达CD45RO的效应子/记忆表型的加速转变。这些结果表明,γc的高表达可促进活化的原始T细胞上IL-2Rα和IL-7Rα的表达,对分化和效应细胞因子的表达具有显着影响。
    Cytokines of the common-γ receptor chain (γc) family are crucial for T-cell differentiation and dysregulation of γc cytokine pathways is involved in the pathogenesis of autoimmune diseases. There is increasing evidence that the availability of the γc receptor (CD132) for the associated receptor chains has implications for T-cell functions. Here we studied the influence of differential γc expression on the expression of the IL-2Rα (CD25), the IL-7Rα (CD127) and the differentiation of activated naïve T cells. We fine-tuned the regulation of γc expression in human primary naïve T cells by lentiviral transduction using small hairpin (sh)RNAs and γc cDNA. Differential γc levels were then analysed for effects on T-cell phenotype and function after activation. Differential γc expression markedly affected IL-2Rα and IL-7Rα expression on activated naïve T cells. High γc expression (γc-high) induced significantly higher expression of IL-2Rα and re-expression of IL-7Rα after activation. Inhibition of γc caused lower IL-2Rα/IL-7Rα expression and impaired proliferation of activated naïve T cells. In contrast, γc-high T cells secreted significantly higher concentrations of effector cytokines (i.e., IFN-γ, IL-6) and showed higher cytokine-receptor induced STAT5 phosphorylation during initial stages as well as persistently higher pSTAT1 and pSTAT3 levels after activation. Finally, accelerated transition towards a CD45RO expressing effector/memory phenotype was seen especially for CD4+ γc-high naïve T cells. These results suggested that high expression of γc promotes expression of IL-2Rα and IL-7Rα on activated naïve T cells with significant effects on differentiation and effector cytokine expression.
    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
    胸腺瘤与重症肌无力(MG)密切相关。然而,由于胸腺瘤的异质性和MG的复杂发病机制,目前尚不清楚为什么一些胸腺瘤患者发展为MG,而另一些患者没有.在这项研究中,我们通过荧光激活细胞分选术(FACS)对患有MG(MG(+)胸腺瘤)和不患有MG(MG(-)胸腺瘤)的B型胸腺瘤患者的胸腺细胞进行了比较表型分析.我们的结果表明,由CD3,CD4和CD8的表达所定义的发育阶段在MG(+)和MG(-)胸腺瘤中基本保持,在B型胸腺瘤中,CD4+CD8+细胞构成了大部分的胸腺细胞,在MG(-)和MG(-)胸腺瘤中,该细胞群之间没有显着差异。我们发现MG()胸腺瘤中的CD4CD8胸腺细胞表达低水平的αβTCR和高水平的IL-7受体α(IL-7Rα),而在MG(-)胸腺瘤中,CD4CD8胸腺细胞表现出相反的αβTCR和IL-7Rα表达模式。这些结果表明,MG()胸腺瘤和MG(-)胸腺瘤之间CD4CD8胸腺细胞的阳性和阴性选择过程可能有所不同。Helios转录因子的表达在阴性选择期间被诱导,并且标记已经经历阴性选择并且可能由于与自身肽/MHC配体的强TCR结合而被缺失的一组T细胞。我们观察到,MG(-)中Helios阳性CD4SPT细胞的百分比高于MG()胸腺瘤。因此,CD4+CD8+胸腺细胞的差异调节选择过程,其中涉及TCR和IL-7/IL-7Rα信号,与B型胸腺瘤中MG的存在有关。
    Thymoma is closely associated with myasthenia gravis (MG). However, due to the heterogeneity of thymoma and the intricate pathogenesis of MG, it remains unclear why some patients with thymoma develop MG and others do not. In this study, we conducted a comparative phenotype analysis of thymocytes in type B thymomas in patients with MG (MG (+) thymomas) and without MG (MG (-) thymomas) via fluorescence-activated cell sorting (FACS). Our results show that the developmental stages defined by the expression of CD3, CD4, and CD8 were largely maintained in both MG (+) and MG (-) thymomas, with CD4+CD8+ cells constituting the majority of thymocytes in type B thymoma, and no significant difference between this cell population was observed in MG (+) and MG (-) thymomas.We discovered that CD4+CD8+ thymocytes in MG (+) thymomas expressed low levels of αβ TCR and high levels of IL-7 receptor α (IL-7Rα), whereas in MG (-) thymomas, CD4+CD8+ thymocytes exhibited the opposite pattern of αβ TCR and IL-7Rα expression. These results suggest that the positive and negative selection processes of CD4+CD8+ thymocytes might differ between MG (+) thymomas and MG (-) thymomas. The expression of the Helios transcription factor is induced during negative selection and marks a group of T cells that have undergone negative selection and are likely to be deleted due to strong TCR binding with self-peptides/MHC ligands. We observed that the percentage of Helios-positive CD4SP T cells was greater in MG (-) than in MG (+) thymomas. Thus, the differentially regulated selection process of CD4+CD8+ thymocytes, which involves TCR and IL-7/IL-7Rα signaling, is associated with the presence of MG in type B thymomas.
    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
    在大多数癌症中,嵌合抗原受体(CAR)工程化T细胞疗法的疗效欠佳。需要进一步改善其治疗作用。然而,增强抗肿瘤T细胞反应不可避免地会增加与单核细胞来源的白介素-6(IL-6)相关的细胞因子释放综合征的风险.因此,同时提高疗效和安全性的方法是必要的.这里,我们开发了一种嵌合细胞因子受体,该受体由GP130和IL6RA的细胞外结构域与IL-7R突变体的跨膜和细胞质结构域连接组成,该突变体组成性激活JAK-STAT途径(G6/7R或G6/7R-M452L).具有G6/7R的CAR-T细胞在体外有效吸收和降解单核细胞来源的IL-6。表达G6/7R的CAR-T细胞在体内表现出优异的扩增和持久性,在液体和实体瘤小鼠模型中产生持久的抗肿瘤反应。我们的策略可以广泛应用于CAR-T细胞治疗,以提高其疗效和安全性,与靶抗原无关。
    The efficacy of chimeric antigen receptor (CAR)-engineered T cell therapy is suboptimal in most cancers, necessitating further improvement in their therapeutic actions. However, enhancing antitumor T cell response inevitably confers an increased risk of cytokine release syndrome associated with monocyte-derived interleukin-6 (IL-6). Thus, an approach to simultaneously enhance therapeutic efficacy and safety is warranted. Here, we develop a chimeric cytokine receptor composed of the extracellular domains of GP130 and IL6RA linked to the transmembrane and cytoplasmic domain of IL-7R mutant that constitutively activates the JAK-STAT pathway (G6/7R or G6/7R-M452L). CAR-T cells with G6/7R efficiently absorb and degrade monocyte-derived IL-6 in vitro. The G6/7R-expressing CAR-T cells show superior expansion and persistence in vivo, resulting in durable antitumor response in both liquid and solid tumor mouse models. Our strategy can be widely applicable to CAR-T cell therapy to enhance its efficacy and safety, irrespective of the target antigen.
    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
    CAR-T细胞疗法在治疗血液恶性肿瘤方面显示出希望,但由于肿瘤微环境中T细胞功能受损,在治疗实体肿瘤方面面临挑战。为了提供最佳的T细胞激活,我们开发了B7同源物3蛋白(B7H3)靶向CAR构建体,由三个激活信号组成:CD3ζ(信号1),41BB(信号2),和白细胞介素7受体α(IL7Rα)细胞质结构域(信号3)。我们产生了具有不同长度的IL7Rα细胞质结构域的B7H3CAR-T细胞,包括全长(IL7R-L),中间长度(IL7R-M),和短长度(IL7R-S)结构域,并在体外和体内评估了它们的功能。所有B7H3-IL7RαCAR-T细胞均表现出较低分化的表型,并在体外有效消除了B7H3阳性胶质母细胞瘤。在包含短长度的IL7Rα细胞质结构域的B7H3CAR-T细胞中发现了优越性。IL7R-S胞质结构域的整合维持了pSTAT5活化并增加了T细胞增殖,同时减少了活化诱导的细胞死亡。此外,与胶质母细胞瘤细胞系共培养后的B7H3-IL7R-SCAR-T细胞的RNA测序分析显示,与第二代B7H3CAR-T细胞相比,促凋亡基因的下调和与T细胞增殖相关的基因的上调。在动物模型中,与传统的CAR-T细胞相比,B7H3-IL7R-SCAR-T细胞抑制肿瘤生长并延长总生存期。我们的研究证明了掺入IL7Rα的CAR-T细胞治疗胶质母细胞瘤的治疗潜力,提出了增强CAR-T细胞疗法有效性的有希望的策略。
    CAR-T-cell therapy has shown promise in treating hematological malignancies but faces challenges in treating solid tumors due to impaired T-cell function in the tumor microenvironment. To provide optimal T-cell activation, we developed a B7 homolog 3 protein (B7H3)-targeting CAR construct consisting of three activation signals: CD3ζ (signal 1), 41BB (signal 2), and the interleukin 7 receptor alpha (IL7Rα) cytoplasmic domain (signal 3). We generated B7H3 CAR-T cells with different lengths of the IL7Rα cytoplasmic domain, including the full length (IL7R-L), intermediate length (IL7R-M), and short length (IL7R-S) domains, and evaluated their functionality in vitro and in vivo. All the B7H3-IL7Rα CAR-T cells exhibited a less differentiated phenotype and effectively eliminated B7H3-positive glioblastoma in vitro. Superiority was found in B7H3 CAR-T cells contained the short length of the IL7Rα cytoplasmic domain. Integration of the IL7R-S cytoplasmic domain maintained pSTAT5 activation and increased T-cell proliferation while reducing activation-induced cell death. Moreover, RNA-sequencing analysis of B7H3-IL7R-S CAR-T cells after coculture with a glioblastoma cell line revealed downregulation of proapoptotic genes and upregulation of genes associated with T-cell proliferation compared with those in 2nd generation B7H3 CAR-T cells. In animal models, compared with conventional CAR-T cells, B7H3-IL7R-S CAR-T cells suppressed tumor growth and prolonged overall survival. Our study demonstrated the therapeutic potential of IL7Rα-incorporating CAR-T cells for glioblastoma treatment, suggesting a promising strategy for augmenting the effectiveness of CAR-T cell therapy.
    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
    桥本氏甲状腺炎(HT)和Graves病(GD)是儿童常见的自身免疫性内分泌疾病。研究表明,除了环境因素,遗传背景显著有助于这些疾病的发展。这项研究旨在评估Il7R的选定单核苷酸多态性(SNP)的患病率,CD226,CAPSL,和CLEC16A基因在患有自身免疫性甲状腺疾病的儿童中的作用。我们分析了IL7R的rs3194051,rs6897932位点的SNP,CD226的rs763361,CAPSL的rs1010601,56例HT患者的CLEC16A基因rs725613,124例GD患者,156个健康的孩子我们观察到HT男性和对照组之间的等位基因IL7R(rs6897932)存在显着差异(C>T,p=0.028)以及所有GD患者和健康儿童之间(C>T,p=0.035)以及GD女性和对照组(C>T,p=0.018)。此外,GD患者rs6897932位点和HT男性rs1010601位点的C/T基因型较低。IL7R(rs6897932)基因座中T等位基因的存在似乎对男性的HT和所有儿童的GD具有保护作用。同样,CAPSL基因座中T等位基因的存在(rs1010601)似乎可降低所有患者发生HT的风险.
    Hashimoto\'s thyroiditis (HT) and Graves\' disease (GD) are common autoimmune endocrine disorders in children. Studies indicate that apart from environmental factors, genetic background significantly contributes to the development of these diseases. This study aimed to assess the prevalence of selected single-nucleotide polymorphisms (SNPs) of Il7R, CD226, CAPSL, and CLEC16A genes in children with autoimmune thyroid diseases. We analyzed SNPs at the locus rs3194051, rs6897932 of IL7R, rs763361 of CD226, rs1010601 of CAPSL, and rs725613 of CLEC16A gene in 56 HT patients, 124 GD patients, and 156 healthy children. We observed significant differences in alleles IL7R (rs6897932) between HT males and the control group (C > T, p = 0.028) and between all GD patients and healthy children (C > T, p = 0.035) as well as GD females and controls (C > T, p = 0.018). Moreover, the C/T genotype was less frequent in GD patients at rs6897932 locus and in HT males at rs1010601 locus. The presence of the T allele in the IL7R (rs6897932) locus appears to have a protective effect against HT in males and GD in all children. Similarly, the presence of the T allele in the CAPSL locus (rs1010601) seems to reduce the risk of HT development in all patients.
    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
    急性淋巴细胞白血病(ALL)起因于B细胞前体(BCP-ALL)或T细胞(T-ALL)的不受控制的增殖。目前的治疗方案在儿童中获得了高治愈率,但基于毒性的多化学疗法。迫切需要新的治疗方法,特别是在复发/难治性(R/R)疾病中,高风险(HR)白血病和T-ALL,免疫治疗方法仍然很少。尽管白细胞介素-7受体(IL-7R)在ALL发育中起着关键作用,IL-7R靶向免疫疗法尚未在ALL中达到临床应用。IL-7Rα链(CD127)靶向IgG4抗体lusvertikimab(LUSV;以前是OSE-127)是IL-7R途径的完全拮抗剂,在健康志愿者中显示出良好的安全性。这里,我们显示,85%的所有病例都表达表面CD127。我们证明了LUSV免疫疗法在微小残留病(MRD)和明显白血病模型中BCP和T-ALL患者来源的异种移植物(PDX)的异质性队列中的显着体内疗效。包括R/R和HR白血病。重要的是,在CD127高样品的2期PDX研究中,LUSV与多化学疗法联合特别有效,导致>50%的联合治疗小鼠MRD阴性。机械上,LUSV通过双重作用模式靶向ALL细胞,所述双重作用模式包括直接IL-7R拮抗活性和诱导巨噬细胞介导的抗体依赖性细胞吞噬作用(ADCP)。LUSV介导的体外ADCP水平与CD127表达水平和体内治疗PDX动物时白血病负担的降低显着相关。总之,通过其双重作用模式和良好的安全性,LUSV可能代表任何CD127+ALL的新型免疫疗法选择。特别是与标准的综合化疗。
    UNASSIGNED: Acute lymphoblastic leukemia (ALL) arises from the uncontrolled proliferation of B-cell precursors (BCP-ALL) or T cells (T-ALL). Current treatment protocols obtain high cure rates in children but are based on toxic polychemotherapy. Novel therapies are urgently needed, especially in relapsed/refractory (R/R) disease, high-risk (HR) leukemias and T-ALL, in which immunotherapy approaches remain scarce. Although the interleukin-7 receptor (IL-7R) plays a pivotal role in ALL development, no IL-7R-targeting immunotherapy has yet reached clinical application in ALL. The IL-7Rα chain (CD127)-targeting IgG4 antibody lusvertikimab (LUSV; formerly OSE-127) is a full antagonist of the IL-7R pathway, showing a good safety profile in healthy volunteers. Here, we show that ∼85% of ALL cases express surface CD127. We demonstrate significant in vivo efficacy of LUSV immunotherapy in a heterogeneous cohort of BCP- and T-ALL patient-derived xenografts (PDX) in minimal residual disease (MRD) and overt leukemia models, including R/R and HR leukemias. Importantly, LUSV was particularly effective when combined with polychemotherapy in a phase 2-like PDX study with CD127high samples leading to MRD-negativity in >50% of mice treated with combination therapy. Mechanistically, LUSV targeted ALL cells via a dual mode of action comprising direct IL-7R antagonistic activity and induction of macrophage-mediated antibody-dependent cellular phagocytosis (ADCP). LUSV-mediated in vitro ADCP levels significantly correlated with CD127 expression levels and the reduction of leukemia burden upon treatment of PDX animals in vivo. Altogether, through its dual mode of action and good safety profile, LUSV may represent a novel immunotherapy option for any CD127+ ALL, particularly in combination with standard-of-care polychemotherapy.
    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
    导致建立免疫记忆的分子机制还没有被充分理解,限制开发有效的疫苗和持久的抗肿瘤免疫疗法。这里,我们表明异位OCA-B表达足以改善抗病毒记忆回忆反应,而对主要效应反应的影响最小。在病毒反应高峰期,短寿命效应T细胞群体扩大,但显示增加的Gadd45b和Socs2表达,而记忆前体效应细胞显示Bcl2、Il7r、和Tcf7在每个细胞的基础上。使用OCA-BmCherry记者鼠标线,我们观察到CD4+中枢记忆T细胞中OCA-B的高表达。我们表明,在病毒感染的早期,内源性升高的OCA-B表达前瞻性地鉴定了具有增加的生存能力和记忆回忆潜力的记忆前体细胞。累计,结果证明OCA-B对于体内促进CD4T细胞记忆是必需和充分的,并且可用于前瞻性地鉴定记忆前体细胞.
    The molecular mechanisms leading to the establishment of immunological memory are inadequately understood, limiting the development of effective vaccines and durable antitumor immune therapies. Here, we show that ectopic OCA-B expression is sufficient to improve antiviral memory recall responses, while having minimal effects on primary effector responses. At peak viral response, short-lived effector T cell populations are expanded but show increased Gadd45b and Socs2 expression, while memory precursor effector cells show increased expression of Bcl2, Il7r, and Tcf7 on a per-cell basis. Using an OCA-B mCherry reporter mouse line, we observe high OCA-B expression in CD4+ central memory T cells. We show that early in viral infection, endogenously elevated OCA-B expression prospectively identifies memory precursor cells with increased survival capability and memory recall potential. Cumulatively, the results demonstrate that OCA-B is both necessary and sufficient to promote CD4 T cell memory in vivo and can be used to prospectively identify memory precursor cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号