首页> 外文会议>World biomaterials congress >Multivalent nanomaterial arrays leverage antigen specificity to modulate immune response in autoimmune disease
【24h】

Multivalent nanomaterial arrays leverage antigen specificity to modulate immune response in autoimmune disease

机译:多价纳米材料阵列利用抗原特异性调节自身免疫性疾病的免疫反应

获取原文

摘要

Introduction: Many current therapies for autoimmune diseases such as multiple sclerosis (MS) act through nonspecific targeting of the immune response, rendering global immunosuppression, poor efficacy, and adverse side effects. To address a pressing need for safer and more effective therapies, multivalent soluble antigen arrays (SAgAs) have been developed to induce antigen-specific tolerance in MS. SAgAs consist of a flexible hyaluronic acid (HA) linear polymer cografted with multiple copies of autoantigen (proteolipid protein peptide, PLP_(139-151)) and secondary inhibitory signal (ICAM-1 inhibitor peptide, LABL). These peptides are intended to target SAgA molecules to the immunological synapse, interrupting signaling between T cells and antigen presenting cells (APCs) to halt propagation of an autoimmune attack. Previous in vivo studies showed SAgAs containing both signals were therapeutically effective against EAE (murine model of MS). We hypothesize SAgAs enact a therapeutic effect due to enhanced, antigen-specific binding with APCs. To investigate this hypothesis, in vitro studies were performed in a model APC system to investigate antigen-specific binding and receptor response; ex vivo studies were performed in murine splenocytes to investigate targeting of EAE immune cells. Methods: 16 kDa HA was labeled with fluorescein isothiocyanate (FITC) to form 'fHA, then conjugated with aminooxy-LABL and/or -PLP to achieve a valency of -10 peptides per HA backbone. Homopolymers (fHA_(PLP) and fHA_(LABL)) were grafted with one peptide and a heteropolymer (fSAgA_(PLP:LABL)) was co-grafted with both peptides. Conjugation was determined by RP-HPLC. in vitro binding studies were performed in Raji B cells (APCs) using flow cytometry. Cells were primed and activated with TNF-α and PLP for 24 hours. To observe maximum binding, cells were mixed with labeled arrays immediately before injection on the cytometer and allowed to reach maximum steady state (SS). Specific binding was then determined through competitive dissociation with excess addition of unlabeled HA, PLP, or LABL. Cell binding and receptor clustering were observed in real-time by fluorescence microscopy using the CellASIC ONIX Microfluidics Platform and M04S switching plate. Ex vivo binding studies were performed in splenocytes isolated from the spleens of both healthy and sick SJL/J mice, harvested at peak of disease following in vivo EAE induction. Isolated splenocytes were stained with CD3, CD22, and CD11c antibodies to identify T cells, B cells, and dendritic cells, respectively. Binding was evaluated by flow cytometry and fluorescence microscopy, as detailed above. Results and Discussion: fSAgA_(PLP:LABL) exhibited significantly greater binding in vitro than fHA, fHA_(PLP), and fHA_(LABL). Specific binding determined by competitive dissociation was driven by PLP. Receptor clustering, observed using real-time fluorescence microscopy, is an important precursor for signaling and activation of an antigen-specific cellular response. fSAgA PLP:LABL. induced mature receptor clustering in Raji B cells to a greater extent than fHA, fHA_(PLP), or fHA_(LABL). fSAgA_(PLP:LABL) exhibited greater binding ex vivo than other arrays and greater binding in EAE splenocytes than healthy splenocytes. Conclusions: SAgAp_(PLP:LABL) exhibited enhanced cell binding due to the presence of both PLP and LABL signals, primarily driven by the antigen. SAgA_(PLP:LABL) binding was accompanied by progressive receptor clustering which may contribute to modulation of the immune response. Additionally, SAgA_(PLP:LABL) exhibited greater binding with EAE splenocytes over healthy splenocytes. Leveraging enhanced, antigen-specific binding and receptor modulation, SAgAs offer a promising option for antigen-specific immunotherapy to repress autoimmune disease.
机译:简介:当前针对自身免疫性疾病(例如多发性硬化症(MS))的许多疗法都是通过非特异性地靶向免疫反应而起作用的,从而导致整体免疫抑制,疗效差和不良副作用。为了满足对更安全,更有效治疗的紧迫需求,已开发出多价可溶性抗原阵列(SAgAs)来诱导MS中的抗原特异性耐受性。 SAgA由柔性透明质酸(HA)线性聚合物与多拷贝的自身抗原(蛋白脂质蛋白肽,PLP_(139-151))和次级抑制信号(ICAM-1抑制剂肽,LABL)共同移植而成。这些肽旨在将SAgA分子靶向免疫突触,中断T细胞和抗原呈递细胞(APC)之间的信号传导,以阻止自身免疫攻击的传播。先前的体内研究表明,包含这两种信号的SAgA对EAE(MS的小鼠模型)具有治疗效果。我们推测SAgAs会由于与APC的抗原特异性结合增强而发挥治疗作用。为了研究该假设,在模型APC系统中进行了体外研究,以研究抗原特异性结合和受体反应。在鼠脾细胞中进行了离体研究,以研究EAE免疫细胞的靶向性。方法:用异硫氰酸荧光素(FITC)标记16 kDa HA形成'fHA,然后与氨氧基-LABL和/或-PLP偶联,每个HA主链的化合价为-10个肽。用一种肽接枝均聚物(fHA_(PLP)和fHA_(LABL)),将杂聚物(fSAgA_(PLP:LABL))与两种肽共接枝。通过RP-HPLC确定缀合。使用流式细胞仪在Raji B细胞(APC)中进行了体外结合研究。灌注细胞并用TNF-α和PLP激活24小时。为了观察最大的结合,将细胞与标记的阵列混合,然后立即注射到细胞仪上,使其达到最大稳态(SS)。然后通过竞争性解离与过量添加未标记的HA,PLP或LABL来确定特异性结合。使用CellASIC ONIX Microfluidics Platform和M04S交换板通过荧光显微镜实时观察细胞结合和受体聚集。在从健康和患病的SJL / J小鼠的脾脏分离的脾细胞中进行了离体结合研究,这些脾细胞是在体内EAE诱导后疾病高峰时收获的。分离的脾细胞用CD3,CD22和CD11c抗体染色,分别鉴定T细胞,B细胞和树突状细胞。如上所述,通过流式细胞术和荧光显微镜术评估结合。结果与讨论:与fHA,fHA_(PLP)和fHA_(LABL)相比,fSAgA_(PLP:LABL)在体外表现出明显更大的结合。通过竞争解离确定的特异性结合是由PLP驱动的。使用实时荧光显微镜观察到的受体簇是信号传导和激活抗原特异性细胞应答的重要前体。 fSAgA PLP:LABL。与fHA,fHA_(PLP)或fHA_(LABL)相比,Raji B细胞中诱导的成熟受体簇化程度更大。与健康的脾细胞相比,fSAgA_(PLP:LABL)与其他阵列相比,离体结合率更高,在EAE脾细胞中的结合率更高。结论:SAgAp_(PLP:LABL)由于主要由抗原驱动的PLP和LABL信号的存在而显示出增强的细胞结合。 SAgA_(PLP:LABL)结合伴随着进行性受体簇集,这可能有助于调节免疫应答。另外,与健康的脾细胞相比,SAgA_(PLP:LABL)与EAE脾细胞显示出更大的结合。利用增强的抗原特异性结合和受体调节,SAgA为抗原特异性免疫疗法抑制自身免疫性疾病提供了有希望的选择。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号