immunoengineering

免疫工程
  • 文章类型: Journal Article
    IL-1β是多种局部和全身性慢性炎症包括银屑病的主要促炎细胞因子。类风湿性关节炎,炎症性肠病,和2型糖尿病。被动免疫疗法和针对IL-1β的生物药物,在提供显著临床益处的同时,然而有局限性,如显著的无反应率,诱导抗药物抗体,和高成本。这里,描述了一种使用自组装肽纳米纤维引起针对IL-1β的抗体应答的主动免疫疗法。纳米纤维含有确定量的来自IL-1β的B细胞表位和外源T辅助表位,并采用Q11自组装肽平台。没有佐剂,纳米纤维引起持久的抗IL-1β抗体应答,其在体外和体内抑制IL-1β活性。在咪喹莫特诱发的银屑病小鼠模型中,纳米纤维的预防性免疫减少了表皮增厚的症状。这种治疗效果与免疫应答偏向抗炎IgG1/Th2表型和皮肤中促炎基因表达降低有关。Further,抗IL-1β纳米纤维诱导治疗性免疫抑制CD62L+Treg细胞。该技术代表了被动免疫疗法和其他用于治疗慢性炎性病症的生物制剂的潜在替代方案。
    IL-1β is a principal proinflammatory cytokine underlying multiple local and systemic chronic inflammatory conditions including psoriasis, rheumatoid arthritis, inflammatory bowel disease, and type 2 diabetes. Passive immunotherapies and biologic drugs targeting IL-1β, while offering significant clinical benefit, nevertheless have limitations such as significant non-response rates, induction of anti-drug antibodies, and high costs. Here, an active immunotherapy raising antibody responses against IL-1β employing self-assembling peptide nanofibers is described. The nanofibers contain defined quantities of B-cell epitopes from IL-1β and exogenous T helper epitopes and employ the Q11 self-assembling peptide platform. Without adjuvant, the nanofibers raised durable anti-IL-1β antibody responses that inhibit IL-1β activity in vitro and in vivo. In a mouse model of imiquimod-induced psoriasis, prophylactic immunizations with the nanofibers diminished symptoms of epidermal thickening. This therapeutic effect is associated with biasing the immune response toward an anti-inflammatory IgG1/Th2 phenotype and a lowered expression of proinflammatory genes in the skin. Further, anti-IL-1β nanofibers induced therapeutic immunosuppressive CD62L+ Treg cells. This technology represents a potential alternative for passive immunotherapies and other biologics for treating chronic inflammatory conditions.
    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
    对去细胞细胞外基质(ECM)肌肉损伤的免疫应答以Th2T细胞为特征,Tregs,M2样巨噬细胞,和大量的嗜酸性粒细胞.嗜酸性粒细胞先前已被描述为肌肉再生的介质,但抑制皮肤伤口愈合。除了对受伤的反应,大量嗜酸性粒细胞对生物材料治疗的肌肉损伤有反应,特别是响应脱细胞ECM。肌肉伤口的ECM治疗与组织再生的积极结果有关,但是详细的行动机制仍在评估中。这里,这项工作调查了这些嗜酸性粒细胞在免疫表型方面的作用以及随后对局部组织微环境的影响.这些细胞具有混合表型,显示2型和调节基因上调,但不是巨噬细胞极化所必需的。除了局部组织,观察到ECM治疗诱导嗜酸性粒细胞向肺的短暂流动,但在损伤小鼠的肺中预防与创伤相关的嗜中性粒细胞增多症。这项工作认为,这种局部和全身免疫调节有助于材料的再生作用,并且在治疗设计和实施中应考虑这种远端组织作用。
    The immune response to decellularized extracellular matrix (ECM) muscle injury is characterized by Th2 T cells, Tregs, M2-like macrophages, and an abundance of eosinophils. Eosinophils have previously been described as mediators of muscle regeneration but inhibit skin wound healing. In addition to response to wounding, a large number of eosinophils respond to biomaterial-treated muscle injury, specifically in response to decellularized ECM. ECM treatment of muscle wounds has been associated with positive outcomes in tissue regeneration, but the detailed mechanisms of action are still being evaluated. Here, this work investigates the role of these eosinophils in terms of their immunologic phenotype and subsequent effect on the local tissue microenvironment. These cells have a mixed phenotype showing both type-2 and regulatory gene upregulation and but are not required for macrophage polarization. Beyond the local tissue, ECM treatment is seen to induce a transient flux of eosinophils to the lungs but prevented a trauma-associated neutrophilia in the lungs of injured mice. This work believes this local and systemic immunomodulation contributes to the regenerative effects of the material and such distal tissue effects should be considered in therapeutic design and implementation.
    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
    糖尿病引起的周围神经病变的治疗主要强调神经营养药物。然而,越来越多的临床研究表明,神经炎症在神经性疼痛的发病机制中起着重要作用。这促使人们积极探索利用纳米医学治疗疾病的治疗策略,旨在获得更好的治疗效果。在这种情况下,我们开发了由聚乳酸-羟基乙酸共聚物制成的可生物降解纳米颗粒,装载雷公藤甲素(pCel),旨在缓解糖尿病引起的体细胞神经性疼痛。用pCel处理显著降低了体外活性氧和细胞凋亡的水平。此外,链脲佐菌素诱导的糖尿病的进展,以肾功能指标升高为特征(血尿素氮,肌酐),肝功能指标(胆红素,碱性磷酸酶)和白蛋白和球蛋白水平降低,pCel给药后缓解。重要的是,pCel口服治疗可显着抑制糖尿病大鼠的机械性异常疼痛和坐骨神经胶质细胞的活化。这些发现表明,这种合成,可生物降解的纳米药物表现出优异的稳定性,生物相容性和催化活性,使其成为治疗与糖尿病神经病变相关的慢性疼痛疾病的一种有前途的创新方法。
    The treatment of peripheral neuropathy resulting from diabetes primarily emphasizes neurotrophic medications. However, a growing body of clinical studies indicates that neuroinflammation plays a significant role in the pathogenesis of neuropathic pain. This has spurred active exploration of treatment strategies leveraging nanomedicine for diseases, aiming for superior therapeutic outcomes. In this context, we have developed biodegradable nanoparticles made of polylactic-co-glycolic acid, loaded with triptolide (pCel), designed to alleviate somatic cell neuropathic pain induced by diabetes. Treatment with pCel notably reduced levels of reactive oxygen species and apoptosis in vitro. Furthermore, the progression of streptozotocin-induced diabetes, characterized by elevated renal function indices (blood urea nitrogen, creatinine), liver function indices (bilirubin, alkaline phosphatase) and decreased levels of albumin and globulin, was mitigated following pCel administration. Importantly, oral treatment with pCel significantly inhibited mechanical allodynia and the activation of the sciatic glial cells in diabetic rats. These findings indicate that this synthetic, biodegradable nanomedicine exhibits excellent stability, biocompatibility and catalytic activity, making it a promising and innovative approach for the management of chronic pain conditions associated with diabetic neuropathy.
    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
    通过向免疫细胞内递送核苷修饰的mRNA进行免疫调节是体内免疫工程的一种有吸引力的方法。在传染病中的应用,癌症免疫疗法,和超越。脂质纳米颗粒(LNP)已成为有前途的核酸递送平台,但LNP设计标准仍然定义不清,使LNP发现的限速步骤成为筛选过程。在这项研究中,我们采用基于分子条形码的高通量体内LNP筛选方法,研究了LNP组合物在疫苗和全身免疫疗法中的应用对免疫嗜性的影响.在肌内(i.m.)和静脉(i.v.)注射下筛选大型LNP库,我们观察到两种给药途径中免疫群体对LNP摄取的不同影响,了解体内免疫工程的LNP设计标准。在验证研究中,与用临床标准可电离脂质DLin-MC3-DMA(MC3)配制的LNP相比,用于i.m.给药的前导LNP制剂在脾脏和引流淋巴结中显示出大量mRNA翻译,具有更有利的生物分布特征。用于静脉内给药的前导LNP制剂在脾脏和外周血中显示出有效的免疫转染,其中一个前导LNP证明了脾树突状细胞的大量转染,另一个则诱导了循环单核细胞的大量转染。总之,通过高通量体内筛选鉴定的免疫型LNP对局部和系统递送的mRNA显示出显著的希望,并证实了从我们的筛选过程中收集的LNP设计标准的价值。这可能为mRNA疫苗和免疫疗法应用的未来努力提供信息。
    Immune modulation through the intracellular delivery of nucleoside-modified mRNA to immune cells is an attractive approach for in vivo immunoengineering, with applications in infectious disease, cancer immunotherapy, and beyond. Lipid nanoparticles (LNPs) have come to the fore as a promising nucleic acid delivery platform, but LNP design criteria remain poorly defined, making the rate-limiting step for LNP discovery the screening process. In this study, we employed high-throughput in vivo LNP screening based on molecular barcoding to investigate the influence of LNP composition on immune tropism with applications in vaccines and systemic immunotherapies. Screening a large LNP library under both intramuscular (i.m.) and intravenous (i.v.) injection, we observed differential influences on LNP uptake by immune populations across the two administration routes, gleaning insight into LNP design criteria for in vivo immunoengineering. In validation studies, the lead LNP formulation for i.m. administration demonstrated substantial mRNA translation in the spleen and draining lymph nodes with a more favorable biodistribution profile than LNPs formulated with the clinical standard ionizable lipid DLin-MC3-DMA (MC3). The lead LNP formulations for i.v. administration displayed potent immune transfection in the spleen and peripheral blood, with one lead LNP demonstrating substantial transfection of splenic dendritic cells and another inducing substantial transfection of circulating monocytes. Altogether, the immunotropic LNPs identified by high-throughput in vivo screening demonstrated significant promise for both locally- and systemically-delivered mRNA and confirmed the value of the LNP design criteria gleaned from our screening process, which could potentially inform future endeavors in mRNA vaccine and immunotherapy applications.
    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
    对于因受伤或自身免疫性疾病而导致器官衰竭的患者,慢性免疫抑制的同种异体器官移植被认为是临床治疗的金标准。然而,移植免疫学真正的“圣杯”是手术耐受性,其中受体对供体移植物呈递的未遇到的抗原表现出持续缺乏同种异体反应性。该结果是由响应于可溶性和机械敏感性线索的特定信号传导途径的激活所预测的免疫库的表型和基因型的关键变化引起的。生物材料已经成为与这些内源性途径进行接口和重新编程以实现精确的耐受性的媒介,微创,和时空定义的方式。通过观察通过生物材料介导的免疫调节策略(包括内在材料免疫原性)在移植耐受诱导方面的开创性和当代突破,仓库效应,接枝涂层,诱导和递送耐受性免疫细胞,耐受性免疫细胞的仿生,和原位重新编程-这篇评论强调了该领域方法的惊人多样性,并为未来的研究指明了令人兴奋的未来方向。本文受版权保护。保留所有权利。
    For patients suffering from organ failure due to injury or autoimmune disease, allogeneic organ transplantation with chronic immunosuppression is considered the god standard in terms of clinical treatment. However, the true \"holy grail\" of transplant immunology is operational tolerance, in which the recipient exhibits a sustained lack of alloreactivity toward unencountered antigen presented by the donor graft. This outcome is resultant from critical changes to the phenotype and genotype of the immune repertoire predicated by the activation of specific signaling pathways responsive to soluble and mechanosensitive cues. Biomaterials have emerged as a medium for interfacing with and reprogramming these endogenous pathways toward tolerance in precise, minimally invasive, and spatiotemporally defined manners. By viewing seminal and contemporary breakthroughs in transplant tolerance induction through the lens of biomaterials-mediated immunomodulation strategies-which include intrinsic material immunogenicity, the depot effect, graft coatings, induction and delivery of tolerogenic immune cells, biomimicry of tolerogenic immune cells, and in situ reprogramming-this review emphasizes the stunning diversity of approaches in the field and spotlights exciting future directions for research to come.
    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
    活化B细胞样弥漫性大B细胞淋巴瘤(ABC-DLBCL)是一种与不良生存结果相关的亚型。尽管通过分子表征确定治疗靶点,靶向治疗的成功有限。需要使用免疫活性组织模型的新策略来了解DLBCL细胞如何逃避治疗。这里,合成的基于水凝胶的淋巴瘤类器官用于证明淋巴肿瘤微环境(Ly-TME)中的信号如何改变B细胞受体(BCR)信号和特定的组蛋白修饰,组蛋白3在赖氨酸9(H3K9me3)的三甲基化,抑制BCR通路的抑制作用。使用成像模式,T细胞增加DNA甲基转移酶3A的表达和近端ABC-DLCBL细胞的细胞骨架形成,受H3K9me3监管。淋巴瘤类器官的扩增显微镜显示T细胞增加了ABC-DLBCL细胞中分离的H3K9me3簇的大小和数量。我们的发现表明,高阶染色质结构的重新组织可能有助于通过新转录状态的出现逃避或抵抗治疗。用G9α组蛋白甲基转移酶抑制剂处理ABC-DLBCL细胞逆转了T细胞介导的对H3K9me3的调节,并克服了T细胞介导的对BCR途径抑制的治疗反应的减弱。这项研究强调了Ly-TME在改变DLBCL命运中的作用,并建议靶向异常信号和微环境串扰可以使高风险患者受益。本文受版权保护。保留所有权利。
    Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL) is a subtype associated with poor survival outcomes. Despite identifying therapeutic targets through molecular characterization, targeted therapies have limited success. New strategies using immune-competent tissue models are needed to understand how DLBCL cells evade treatment. Here, synthetic hydrogel-based lymphoma organoids are used to demonstrate how signals in the lymphoid tumor microenvironment (Ly-TME) can alter B cell receptor (BCR) signaling and specific histone modifications, tri-methylation of histone 3 at lysine 9 (H3K9me3), dampening the effects of BCR pathway inhibition. Using imaging modalities, T cells increase DNA methyltransferase 3A expression and cytoskeleton formation in proximal ABC-DLBCL cells, regulated by H3K9me3. Expansion microscopy on lymphoma organoids reveals T cells increase the size and quantity of segregated H3K9me3 clusters in ABC-DLBCL cells. Findings suggest the re-organization of higher-order chromatin structures that may contribute to evasion or resistance to therapy via the emergence of novel transcriptional states. Treating ABC-DLBCL cells with a G9α histone methyltransferase inhibitor reverses T cell-mediated modulation of H3K9me3 and overcomes T cell-mediated attenuation of treatment response to BCR pathway inhibition. This study emphasizes the Ly-TME\'s role in altering DLBCL fate and suggests targeting aberrant signaling and microenvironmental cross-talk that can benefit high-risk patients.
    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
    无细胞组织工程血管移植物为治疗心血管疾病提供了有希望的替代方法,但及时内皮化对确保植入后的通畅和正常功能至关重要。我们实验室的最新研究表明,在小型和大型动物模型中,单核细胞(MC)和巨噬细胞(Mφ)等血细胞可能直接促进生物工程动脉的细胞化和再生。虽然MC和Mφ是白细胞,是先天免疫反应的一部分,它们与内皮细胞(EC)有着共同的发育起源,并且已知在炎症/损伤后的血管形成(血管生成)和血管修复过程中发挥关键作用.它们是高度可塑性的细胞,在暴露于细胞因子后分化为促炎和抗炎表型;并分化为其他细胞类型,包括EC样细胞,在存在适当的化学和机械刺激的情况下。本文重点介绍了MC和EC的发育起源;MC和Mφ在炎症/损伤过程中血管修复/再生中的作用;以及化学信号和机械力在Mφ炎症中介导血管移植物再生的作用。我们推测,对这些机制的全面了解将更好地指导策略的发展,以哄骗MC/Mφ使内腔内皮化,并再生无细胞生物工程动脉和静脉的平滑肌层,这些动脉和静脉旨在治疗心血管疾病,也许也是天然脉管系统。
    Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
    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
    增加效力,疫苗的质量和耐久性是一个重大的公共卫生挑战。形成疫苗免疫的关键参数是免疫细胞与抗原和佐剂相互作用的时空背景。虽然各种基于材料的策略已经证明延长的抗原释放增强了细胞和体液免疫,佐剂动力学对疫苗介导的免疫的影响尚不完全清楚.这里,我们使用先前表征的介孔硅棒(MPS)生物材料疫苗来开发一种简单的,静电驱动的方法来调整TLR9激动剂CpG的体内动力学。我们证明了从MPS疫苗中快速释放CpG,由调节表面电荷的MPS化学改变介导,产生有效的细胞毒性T细胞反应和强大的,Th1偏斜的IgG2a/c抗体滴度。用缓慢或快速的CpG释放动力学接种MPS后淋巴器官的免疫表型分析表明,迁移树突状细胞和自然杀伤细胞的差异参与可能有助于观察到快速佐剂释放的更有效的反应。一起来看,这些发现表明,将抗原的持续释放与具有与CpG相似特征的佐剂的快速释放配对的疫苗方法可能会驱动特别有效的Th1应答.本文受版权保护。保留所有权利。
    Increasing the potency, quality, and durability of vaccines represents a major public health challenge. A critical parameter that shapes vaccine immunity is the spatiotemporal context in which immune cells interact with antigen and adjuvant. While various material-based strategies demonstrate that extended antigen release enhances both cellular and humoral immunity, the effect of adjuvant kinetics on vaccine-mediated immunity remains incompletely understood. Here, a previously characterized mesoporous silica rod (MPS) biomaterial vaccine is used to develop a facile, electrostatics-driven approach to tune in vivo kinetics of the TLR9 agonist cytosine phosphoguanosine oligodeoxynucleotide (CpG). It is demonstrated that rapid release of CpG from MPS vaccines, mediated by alterations in MPS chemistry that tune surface charge, generates potent cytotoxic T cell responses and robust, T helper type 1 (Th1)-skewed IgG2a/c antibody titers. Immunophenotyping of lymphoid organs after MPS vaccination with slow or fast CpG release kinetics suggests that differential engagement of migratory dendritic cells and natural killer cells may contribute to the more potent responses observed with rapid adjuvant release. Taken together, these findings suggest that vaccine approaches that pair sustained release of antigen with rapid release of adjuvants with similar characteristics to CpG may drive particularly potent Th1 responses.
    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
    创伤性脊髓损伤(SCI),通常是由外部物理创伤造成的,启动一系列复杂的病理生理级联,严重病例导致瘫痪,并提出了重大的临床挑战。传统的诊断和治疗方法,特别是X射线成像,在临床实践中普遍存在,然而,损伤部位的药物治疗的有限疗效和显著副作用仍然构成重大障碍。应对这些挑战,多功能纳米技术和协同疗法的发展取得了最新进展,提高射线照相技术的有效性和安全性。在这种情况下,我们开发了一种创新的神经再生和神经保护纳米平台,利用X射线触发,用于SCI回收的按需RuFe金属-有机骨架(P-RuFe)。该平台旨在模拟过氧化氢酶和超氧化物歧化酶的酶活性,有效减少活性氧的产生,去除自由基和活性氮,从而保护细胞免受氧化应激诱导的损伤。体内研究表明,P-RuFe和X射线治疗的组合可显着降低SCI小鼠模型的死亡率,并通过抑制神经胶质细胞增殖和神经炎症来促进脊髓修复。P-RuFe显示出优异的潜力作为一种安全,活性氧和氮的有效清除剂,提供良好的稳定性,生物相容性,和高催化活性,因此有望治疗炎症相关疾病。
    Traumatic spinal cord injury (SCI), often resulting from external physical trauma, initiates a series of complex pathophysiological cascades, with severe cases leading to paralysis and presenting significant clinical challenges. Traditional diagnostic and therapeutic approaches, particularly X-ray imaging, are prevalent in clinical practice, yet the limited efficacy and notable side effects of pharmacological treatments at the injury site continue to pose substantial hurdles. Addressing these challenges, recent advancements have been made in the development of multifunctional nanotechnology and synergistic therapies, enhancing both the efficacy and safety of radiographic techniques. In this context, we have developed an innovative nerve regeneration and neuroprotection nanoplatform utilizing an X-ray-triggered, on-demand RuFe metal-organic framework (P-RuFe) for SCI recovery. This platform is designed to simulate the enzymatic activities of catalase and superoxide dismutase, effectively reducing the production of reactive oxygen species, and to remove free radicals and reactive nitrogen species, thereby protecting cells from oxidative stress-induced damage. In vivo studies have shown that the combination of P-RuFe and X-ray treatment significantly reduces mortality in SCI mouse models and promotes spinal cord repair by inhibiting glial cell proliferation and neuroinflammation. P-RuFe demonstrates excellent potential as a safe, effective scavenger of reactive oxygen and nitrogen species, offering good stability, biocompatibility, and high catalytic activity, and thus holds promise for the treatment of inflammation-related diseases.
    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
    除了SARS-CoV2疫苗,正在探索mRNA药物,以克服当今最大的医疗保健负担,包括癌症和心血管疾病。合成mRNA在转染细胞中触发免疫反应,它可以被化学修饰的核苷酸还原。然而,mRNA触发的免疫激活对细胞功能的副作用以及不同的核苷酸,例如SARS-CoV2疫苗中使用的N1-甲基假尿苷(m1kW),能否调节细胞反应还没有完全理解。这里,在原代人细胞中研究了对尿苷修饰的mRNA文库的细胞反应。靶向蛋白质组学分析表明,未修饰的mRNA诱导以趋化因子分泌为标志的促炎旁分泌模式,向转染细胞募集T和B淋巴细胞。重要的是,mRNA诱导的细胞功能变化的幅度在未修饰之间定量变化,kW-,m1kW-,和5moU修饰的mRNA,可以逐渐定制,这意味着在mRNA药物设计中故意利用这种作用。的确,基质细胞对T细胞增殖的免疫抑制作用,适当的尿苷修饰可增强IL-10mRNA的抗炎作用。这些结果为mRNA药物对细胞功能和细胞间通讯的影响提供了新的见解,并为定制mRNA触发的免疫激活以适应所需的促炎或抗炎应用开辟了新的可能性。
    Beyond SARS-CoV2 vaccines, mRNA drugs are being explored to overcome today\'s greatest healthcare burdens, including cancer and cardiovascular disease. Synthetic mRNA triggers immune responses in transfected cells, which can be reduced by chemically modified nucleotides. However, the side effects of mRNA-triggered immune activation on cell function and how different nucleotides, such as the N1-methylpseudouridine (m1Ψ) used in SARS-CoV2 vaccines, can modulate cellular responses is not fully understood. Here, cellular responses toward a library of uridine-modified mRNAs are investigated in primary human cells. Targeted proteomics analyses reveal that unmodified mRNA induces a pro-inflammatory paracrine pattern marked by the secretion of chemokines, which recruit T and B lymphocytes toward transfected cells. Importantly, the magnitude of mRNA-induced changes in cell function varies quantitatively between unmodified, Ψ-, m1Ψ-, and 5moU-modified mRNA and can be gradually tailored, with implications for deliberately exploiting this effect in mRNA drug design. Indeed, both the immunosuppressive effect of stromal cells on T-cell proliferation, and the anti-inflammatory effect of IL-10 mRNA are enhanced by appropriate uridine modification. The results provide new insights into the effects of mRNA drugs on cell function and cell-cell communication and open new possibilities to tailor mRNA-triggered immune activation to the desired pro- or anti-inflammatory application.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号