antigen-specific tolerance

抗原特异性耐受性
  • 文章类型: Journal Article
    氧化铈纳米颗粒(CeNPs)对活性氧的清除能力在催化领域得到了广泛的研究。然而,这些颗粒的免疫学影响尚未得到彻底研究,尽管大量研究表明,活性氧的调节可能潜在地调节细胞命运和适应性免疫反应。在这项研究中,我们检查了当自身抗原肽与CeNP简单混合时,CeNP通过其活性氧清除作用诱导耐受性树突状细胞的内在能力。CeNPs有效降低体外树突状细胞的细胞内活性氧水平,导致共刺激分子的抑制以及NLRP3炎性体的激活,即使在促炎刺激的存在。皮下施用的PEG化的CeNP主要被淋巴结中的抗原呈递细胞摄取,并在体内抑制细胞成熟。聚乙二醇化CeNP和髓鞘少突胶质细胞糖蛋白肽的混合物的给药,一种与抗髓鞘素自身免疫相关的公认的自身抗原,导致小鼠脾脏中产生抗原特异性Foxp3+调节性T细胞。诱导的外周调节性T细胞积极抑制自身反应性T细胞和抗原呈递细胞向中枢神经系统的浸润,当使用模拟人类多发性硬化症的小鼠模型进行测试时,最终保护动物免受实验性自身免疫性脑脊髓炎的影响。总的来说,我们的发现揭示了CeNPs产生抗原特异性免疫耐受以预防多发性硬化症的潜力,通过简单地将已明确鉴定的自身抗原与免疫抑制CeNP混合,为恢复针对特定抗原的免疫耐受开辟了一条途径。
    The scavenging ability of cerium oxide nanoparticles (CeNPs) for reactive oxygen species has been intensively studied in the field of catalysis. However, the immunological impact of these particles has not yet been thoroughly investigated, despite intensive research indicating that modulation of the reactive oxygen species could potentially regulate cell fate and adaptive immune responses. In this study, we examined the intrinsic capability of CeNPs to induce tolerogenic dendritic cells via their reactive oxygen species-scavenging effect when the autoantigenic peptides were simply mixed with CeNPs. CeNPs effectively reduced the intracellular reactive oxygen species levels in dendritic cells in vitro, leading to the suppression of costimulatory molecules as well as NLRP3 inflammasome activation, even in the presence of pro-inflammatory stimuli. Subcutaneously administrated PEGylated CeNPs were predominantly taken up by antigen-presenting cells in lymph nodes and to suppress cell maturation in vivo. The administration of a mixture of PEGylated CeNPs and myelin oligodendrocyte glycoprotein peptides, a well-identified autoantigen associated with antimyelin autoimmunity, resulted in the generation of antigen-specific Foxp3+ regulatory T cells in mouse spleens. The induced peripheral regulatory T cells actively inhibited the infiltration of autoreactive T cells and antigen-presenting cells into the central nervous system, ultimately protecting animals from experimental autoimmune encephalomyelitis when tested using a mouse model mimicking human multiple sclerosis. Overall, our findings reveal the potential of CeNPs for generating antigen-specific immune tolerance to prevent multiple sclerosis, opening an avenue to restore immune tolerance against specific antigens by simply mixing the well-identified autoantigens with the immunosuppressive CeNPs.
    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
    自身免疫性疾病,过敏,移植排斥,产生抗药物抗体,和慢性炎症性疾病影响了全球的一大群人。常规治疗和疗法通常使用全身性或广泛免疫抑制,具有严重的功效和安全性问题。耐受性疫苗代表的概念已从其传统的免疫调节功能扩展到通过产生调节性T细胞诱导抗原特异性耐受性。在不损害免疫稳态的情况下,耐受性疫苗抑制炎症并诱导耐受性调节。然而,实现耐受原性疫苗作为预防和治疗方式所需的效力,需要精确操纵免疫微环境并控制针对自身抗原的耐受原性反应,过敏原,和/或同种异体抗原。工程化的纳米/微粒具有期望的设计特征,其可以支持靶向免疫调节并增强抗原特异性耐受性的诱导。因此,基于颗粒的耐受性疫苗在未来治疗上述免疫疾病的临床转化中具有巨大的前景.在这次审查中,我们强调了使用颗粒作为令人兴奋的耐受性疫苗的主要策略,重点关注颗粒在促进抗原特异性耐受诱导中的作用。我们描述了促进其使用的颗粒设计特征,并讨论了设计下一代基于颗粒的耐受性疫苗的挑战和机遇,这些疫苗具有强大的功效以促进免疫疗法的抗原特异性耐受性。
    Autoimmune diseases, allergies, transplant rejections, generation of antidrug antibodies, and chronic inflammatory diseases have impacted a large group of people across the globe. Conventional treatments and therapies often use systemic or broad immunosuppression with serious efficacy and safety issues. Tolerogenic vaccines represent a concept that has been extended from their traditional immune-modulating function to induction of antigen-specific tolerance through the generation of regulatory T cells. Without impairing immune homeostasis, tolerogenic vaccines dampen inflammation and induce tolerogenic regulation. However, achieving the desired potency of tolerogenic vaccines as preventive and therapeutic modalities calls for precise manipulation of the immune microenvironment and control over the tolerogenic responses against the autoantigens, allergens, and/or alloantigens. Engineered nano-/microparticles possess desirable design features that can bolster targeted immune regulation and enhance the induction of antigen-specific tolerance. Thus, particle-based tolerogenic vaccines hold great promise in clinical translation for future treatment of aforementioned immune disorders. In this review, we highlight the main strategies to employ particles as exciting tolerogenic vaccines, with a focus on the particles\' role in facilitating the induction of antigen-specific tolerance. We describe the particle design features that facilitate their usage and discuss the challenges and opportunities for designing next-generation particle-based tolerogenic vaccines with robust efficacy to promote antigen-specific tolerance for immunotherapy.
    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
    多发性硬化症是由自身抗原反应性免疫细胞引起的疾病,其破坏中枢神经系统(CNS)中的髓磷脂。虽然免疫抑制药物用于抑制症状,没有明确的治疗方法.与多发性硬化症的实验性自身免疫性脑炎(EAE)模型一样,髓鞘少突胶质细胞糖蛋白(MOG35-55)的部分序列被鉴定为致病自身抗原.这表明对MOG35-55特异性的免疫耐受的诱导将是EAE的基本治疗。我们以前报道过含有阴离子磷脂的脂质纳米颗粒(LNPs),磷脂酰丝氨酸(PS),在它们的脂质组成中,可用于递送mRNA,并且这导致感兴趣的蛋白质在脾脏中表达。除了PS的瞄准能力,PS分子避免激活免疫系统。生理学上,PS对凋亡细胞的识别通过释放细胞因子来抑制针对这些细胞的免疫激活,如白细胞介素-10(IL-10)和负调节免疫的转化生长因子(TGF)-β。在这项研究中,我们测试了通过PS-LNP将自身抗原的mRNA递送到脾脏是否在最小的免疫刺激下导致MOG35-55抗原的表达,以及这是否可以通过诱导免疫耐受来治疗EAE模型.
    Multiple sclerosis is a disease caused by autoantigen-responsive immune cells that disrupt the myelin in the central nervous system (CNS). Although immunosuppressive drugs are used to suppress symptoms, no definitive therapy exists. As in the experimental autoimmune encephalitis (EAE) model of multiple sclerosis, a partial sequence of the myelin oligodendrocyte glycoprotein (MOG35-55) was identified as a causative autoantigen. This suggests that the induction of immune tolerance that is specific to MOG35-55 would be a fundamental treatment for EAE. We previously reported that lipid nanoparticles (LNPs) containing an anionic phospholipid, phosphatidylserine (PS), in their lipid composition, can be used to deliver mRNA and that this leads to proteins of interest to be expressed in the spleen. In addition to the targeting capability of PS, PS molecules avoid activating the immune system. Physiologically, the recognition of PS on apoptotic cells suppresses immune activation against these cells by releasing cytokines, such as interleukin-10 (IL-10) and transforming growth factor (TGF)-β that negatively regulate immunity. In this study, we tested whether mRNA delivery of autoantigens to the spleen by PS-LNPs causes the expression of MOG35-55 antigens with minimal immune stimulation and whether this could be used to treat an EAE model by inducing immune tolerance.
    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
    涂覆有单特异性自身免疫性疾病相关肽-主要组织相容性复合物II类(pMHCII)分子的纳米颗粒(NP)的系统递送可以以疾病特异性方式解决各种疾病模型中的器官炎症而不损害正常免疫力。这些化合物总是触发同源pMHCII特异性T调节型1(TR1)细胞的形成和系统扩增。通过关注1型糖尿病(T1D)相关的pMHCII-NP类型,该类型显示胰岛素B链的表位与三个不同寄存器上的相同MHCII分子(IAg7)结合,我们显示pMHCII-NP诱导的TR1细胞总是与同源T-卵泡辅助细胞(TFH)样细胞共存,具有准相同的克隆型组成,并且是寡克隆的,然而转录同质。此外,尽管这三种不同的TR1特异性对显示在NP上的肽MHCII结合寄存器具有独特的反应性,但在体内具有相似的糖尿病逆转特性.因此,使用显示不同表位特异性的纳米药物的pMHCII-NP处理导致多个抗原特异性TFH样细胞克隆同时分化为TR1样细胞,所述TR1样细胞继承其前体的优良抗原特异性,同时获得确定的转录免疫调节程序。
    Systemic delivery of nanoparticles (NPs) coated with mono-specific autoimmune disease-relevant peptide-major histocompatibility complex class II (pMHCII) molecules can resolve organ inflammation in various disease models in a disease-specific manner without impairing normal immunity. These compounds invariably trigger the formation and systemic expansion of cognate pMHCII-specific T-regulatory type 1 (TR1) cells. By focusing on type 1 diabetes (T1D)-relevant pMHCII-NP types that display an epitope from the insulin B-chain bound to the same MHCII molecule (IAg7) on three different registers, we show that pMHCII-NP-induced TR1 cells invariably co-exist with cognate T-Follicular Helper (TFH)-like cells of quasi-identical clonotypic composition and are oligoclonal, yet transcriptionally homogeneous. Furthermore, these three different TR1 specificities have similar diabetes reversal properties in vivo despite being uniquely reactive against the peptide MHCII-binding register displayed on the NPs. Thus, pMHCII-NP treatment using nanomedicines displaying different epitope specificities results in the simultaneous differentiation of multiple antigen-specific TFH-like cell clones into TR1-like cells that inherit the fine antigenic specificity of their precursors while acquiring a defined transcriptional immunoregulatory program.
    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
    抗原特异性免疫疗法(ASIT)解决了治疗自身免疫性疾病的重要临床需求。然而,1型糖尿病是一种异质性疾病,其中患者特征影响对ASIT的反应性。不仅针对疾病相关的T细胞群,而且,使用精准医学的特定患者群体也是实现有效治疗的新目标。HLA限制肽提供优于蛋白质的抗原,然而,抗原特异性T细胞谱分析的方法需要提高灵敏度,深度,和通量以促进表位选择。交付方式非常多样化,说明了相关的抗原呈递细胞群体和解剖位置可以被靶向诱导耐受的许多方式。抗原呈递的持久性在促进持久的抗原特异性耐受性中的作用需要进一步研究。根据ASIT试验的结果,该领域正在朝着使用患者特定的变化来提高疗效的方向发展,但在为T1D患者群体提供更有效和更安全的治疗方面仍存在挑战.
    Antigen-specific immunotherapies (ASITs) address important clinical needs in treating autoimmune diseases. However, Type 1 diabetes is a heterogeneous disease wherein patient characteristics influence responsiveness to ASITs. Targeting not only disease-relevant T cell populations, but also specific groups of patients using precision medicine is a new goal toward achieving effective treatment. HLA-restricted peptides provide advantages over protein as antigens, however, methods for profiling antigen-specific T cells need to improve in sensitivity, depth, and throughput to facilitate epitope selection. Delivery approaches are highly diverse, illustrating the many ways relevant antigen-presenting cell populations and anatomical locations can be targeted for tolerance induction. The role of persistence of antigen presentation in promoting durable antigen-specific tolerance requires further investigation. Based on the outcome of ASIT trials, the field is moving toward using patient-specific variations to improve efficacy, but challenges still lie on the path to delivering more effective and safer treatment to the T1D patient population.
    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
    1型糖尿病(T1D)存在两个治疗挑战:需要纠正潜在的自身免疫和恢复β细胞群。我们利用调节性T细胞(Tregs)和T细胞受体(TCR)的独特能力来指导耐受性诱导以及治疗剂的组织定位递送以恢复内源性β细胞功能。具体来说,我们设计了一种组合疗法,涉及基于生物材料的聚(乳酸-共-乙醇酸)纳米颗粒与Treg生长因子共负载,IL-2和β细胞再生剂,harmine(酪氨酸调节激酶1A[DYRK1A]抑制剂),共轭到Tregs的表面。我们观察到IL-2和harmine从纳米颗粒中连续洗脱至少7天。当与初级人类Treg缀合时,IL-2纳米颗粒提供足够的IL-2受体信号传导以支持STAT5磷酸化,以在培养物中实现持续的表型稳定性和活力。在纳米颗粒-细胞偶联过程中包含聚-L-赖氨酸(PLL)显着提高了缀合效率,提供足够的IL-2以支持IL-2依赖性CTLL-2细胞和原代鼠Treg的体外增殖。在12周龄的雌性非肥胖糖尿病小鼠中,IL-2/harmine纳米颗粒缀合的NOD的过继转移。BDC2.5Tregs,表达胰岛抗原特异性TCR,显着预防糖尿病,证明保持体内活力。这些数据为开发生物材料优化的细胞疗法提供了临床前基础,以恢复免疫耐受并通过胰岛内的受体靶向药物递送促进T1D中的β细胞增殖。
    Type 1 diabetes (T1D) presents with two therapeutic challenges: the need to correct underlying autoimmunity and restore β-cell mass. We harnessed the unique capacity of regulatory T cells (Tregs) and the T cell receptor (TCR) to direct tolerance induction along with tissue-localized delivery of therapeutic agents to restore endogenous β-cell function. Specifically, we designed a combinatorial therapy involving biomaterials-based poly(lactic-co-glycolic acid) nanoparticles co-loaded with the Treg growth factor, IL-2, and the β-cell regenerative agent, harmine (a tyrosine-regulated kinase 1A [DYRK1A] inhibitor), conjugated to the surface of Tregs. We observed continuous elution of IL-2 and harmine from nanoparticles for at least 7 days in vitro. When conjugated to primary human Tregs, IL-2 nanoparticles provided sufficient IL-2 receptor signaling to support STAT5 phosphorylation for sustained phenotypic stability and viability in culture. Inclusion of poly-L-lysine (PLL) during nanoparticle-cell coupling dramatically increased conjugation efficiency, providing sufficient IL-2 to support in vitro proliferation of IL-2-dependent CTLL-2 cells and primary murine Tregs. In 12-week-old female non-obese diabetic mice, adoptive transfer of IL-2/harmine nanoparticle-conjugated NOD.BDC2.5 Tregs, which express an islet antigen-specific TCR, significantly prevented diabetes demonstrating preserved in vivo viability. These data provide the preclinical basis to develop a biomaterials-optimized cellular therapy to restore immune tolerance and promote β-cell proliferation in T1D through receptor-targeted drug delivery within pancreatic islets.
    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
    抗原特异性调节性T细胞(Treg)的治疗潜力已被广泛探索,导致了几种耐受性疫苗的开发。地塞米松-抗原缀合物代表一类突出的耐受原性疫苗,其使得抗原和地塞米松能够协调递送至靶免疫细胞。非特异性白蛋白缔合对抗原-佐剂缀合物的生物分布的重要性已获得越来越多的关注。疏水和静电相互作用控制缔合能力。使用计算和实验技术的集合,我们评估了地塞米松-抗原缀合物中药物缀合位点附近的带电残基的影响(Dex-K/E4-OVA323,K:赖氨酸,E:谷氨酸)朝向它们的白蛋白缔合能力和抗原特异性Treg的诱导。我们发现Dex-K4-OVA323具有比Dex-E4-OVA323更高的白蛋白缔合能力,从而导致增强的肝脏分布和抗原呈递细胞摄取。此外,使用OVA323特异性过继转移小鼠模型,我们显示Dex-K4-OVA323选择性上调OVA323特异性Treg细胞,而Dex-E4-OVA323对Treg细胞无显著影响。我们的发现可作为优化地塞米松-抗原缀合物的功能在切换疫苗表位序列中的指导。此外,我们的研究表明,调节缀合位点附近的残基可以作为未来肽-药物缀合物开发的工程方法。
    The therapeutic potential of antigen-specific regulatory T cells (Treg) has been extensively explored, leading to the development of several tolerogenic vaccines. Dexamethasone-antigen conjugates represent a prominent class of tolerogenic vaccines that enable coordinated delivery of antigen and dexamethasone to target immune cells. The importance of nonspecific albumin association towards the biodistribution of antigen-adjuvant conjugates has gained increasing attention, by which hydrophobic and electrostatic interactions govern the association capacity. Using an ensemble of computational and experimental techniques, we evaluate the impact of charged residues adjacent to the drug conjugation site in dexamethasone-antigen conjugates (Dex-K/E4-OVA323, K: lysine, E: glutamate) towards their albumin association capacity and induction of antigen-specific Treg. We find that Dex-K4-OVA323 possesses a higher albumin association capacity than Dex-E4-OVA323, leading to enhanced liver distribution and antigen-presenting cell uptake. Furthermore, using an OVA323-specific adoptive-transfer mouse model, we show that Dex-K4-OVA323 selectively upregulated OVA323-specific Treg cells, whereas Dex-E4-OVA323 exerted no significant effect on Treg cells. Our findings serve as a guide to optimize the functionality of dexamethasone-antigen conjugate amid switching vaccine epitope sequences. Moreover, our study demonstrates that moderating the residues adjacent to the conjugation sites can serve as an engineering approach for future peptide-drug conjugate development.
    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
    自身免疫性疾病是由免疫调节障碍引起的慢性炎症性疾病。抗原特异性免疫疗法具有抑制炎性T细胞自身反应性和诱导抗原特异性免疫抑制而不损害正常免疫功能的潜力。为自身免疫性疾病的治疗提供了理想的策略。具有免疫调节功能的致耐受性树突状细胞(TolDCs)在诱导免疫耐受中起重要作用。然而,在体内有效产生致耐受性DC仍然是一个巨大的挑战。基于纳米颗粒的药物递送系统在自身免疫性疾病治疗中的应用可以提高体内诱导抗原特异性耐受的效率。在这次审查中,我们讨论了多种纳米粒子,专注于它们在治疗自身免疫性疾病方面的潜力。我们还讨论了纳米颗粒的物理性质如何影响其治疗功效。
    Autoimmune disease is a chronic inflammatory disease caused by disorders of immune regulation. Antigen-specific immunotherapy has the potential to inhibit the autoreactivity of inflammatory T cells and induce antigen-specific immune suppression without impairing normal immune function, offering an ideal strategy for autoimmune disease treatment. Tolerogenic dendritic cells (Tol DCs) with immunoregulatory functions play important roles in inducing immune tolerance. However, the effective generation of tolerogenic DCs in vivo remains a great challenge. The application of nanoparticle-based drug delivery systems in autoimmune disease treatment can increase the efficiency of inducing antigen-specific tolerance in vivo. In this review, we discuss multiple nanoparticles, with a focus on their potential in treatment of autoimmune diseases. We also discuss how the physical properties of nanoparticles influence their therapeutic efficacy.
    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
    Immunotolerance induction in an antigen-specific manner is the long-term goal of immunotherapy to treat autoimmune diseases. Nanocarriers (NCs) can be designed as a new generation of delivery systems to modulate the immune responses through targeted delivery of antigens and immunomodulators to antigen presenting cells (APCs). In this manuscript, several formulation factors in the preparation of NCs which affect their uptake using APCs and generation of tolerance have been reviewed. The physicochemical properties and composition of NCs have been shown to play essential roles in achieving the desired immunological outcome. Also, targeting of dendritic cells and macrophages as APCs and direct targeting of the autoreactive lymphocytes have been presented as two main ways for induction of antigen-specific tolerance by these tolerogenic nanocarriers (tNCs). These particles herald a promising approach to treat or even prevent unwanted immune reactions in humans specifically.
    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
    自身免疫性疾病影响大约5-10%的总人口,女性比男性受影响更大。自身免疫性或自身炎症性疾病的标准治疗一直是免疫抑制剂,直到免疫调节生物药物出现,旨在阻断炎症介质,包括促炎细胞因子。这些生物药物的前沿是TNF-α阻滞剂。这些疗法抑制TNF-α在常见自身免疫性疾病如类风湿性关节炎中的促炎作用,牛皮癣,溃疡性结肠炎,和克罗恩病。由于与广谱免疫抑制剂相比,TNF-α阻断剂在控制疾病和降低患者不良风险方面的有效性,因此迅速成为这些自身免疫性疾病的“护理标准”。然而,抗TNF-α治疗有局限性,包括已知的不良安全风险,由于耐药性而失去治疗功效,在许多自身免疫性疾病中缺乏疗效,包括多发性硬化症.下一波真正的变革性疗法应该渴望通过选择性地抑制致病性自身抗原特异性免疫应答同时保持免疫系统的其余部分完整以控制传染病和恶性肿瘤来提供治愈。在这次审查中,我们将集中在三个主要领域积极研究免疫耐受。首先,旨在强健的致耐受性疫苗,持久的自身抗原特异性免疫耐受。第二,使用Tregs的T细胞疗法(无论是多克隆,抗原特异性,或基因工程以表达嵌合抗原受体)以建立活跃的显性免疫耐受或T细胞(工程化以表达嵌合抗原受体)以删除致病性免疫细胞。第三,旨在在体内扩增免疫抑制调节性T细胞的IL-2疗法。
    Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn\'s disease. TNF-α blockade quickly became the \"standard of care\" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient\'s adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号