immune responses

免疫应答
  • 文章类型: Journal Article
    髓系来源的抑制细胞(MDSC)是不成熟骨髓细胞的异质群体,其在维持免疫稳态和调节免疫应答中起重要作用。基于其表面标志物和功能特性,MDSCs可分为两个主要子集:粒细胞MDSCs(G-MDSC)和单核细胞MDSCs(M-MDSC)。近年来,多发性硬化症(MS)的先天免疫受到了最大的关注,因此,我们综述的目的是通过讨论可获得的最新数据,概述MS中MDSCs的主要特征及其临床前模型.MDSCs的免疫抑制功能在MS中可能失调,导致自身免疫反应和疾病进展的恶化。抗原特异性肽免疫治疗,旨在恢复耐受性,同时避免使用非特异性免疫抑制药物,是治疗自身免疫性疾病的一种有希望的方法,但成功治疗背后的细胞机制仍然知之甚少。因此,靶向MDSCs可能是一种有前途的MS治疗方法。已经研究了调节MDSCs的各种策略,包括使用药物,生物制剂,和外源MDSCs的过继转移。然而,目前尚不清楚MDSCs是否在MS中显示出任何治疗潜力,以及该疗法如何调节疾病的不同方面.总的来说,所有描述的研究都揭示了MDSCs在调节MS中的关键作用。

    Myeloid-Derived Suppressor Cells (MDSCs) are a heterogeneous population of immature myeloid cells that play important roles in maintaining immune homeostasis and regulating immune responses. MDSCs can be divided into two main subsets based on their surface markers and functional properties: granulocytic MDSCs (G-MDSCs) and monocytic MDSCs (M-MDSCs). Recently greatest attention has been paid to innate immunity in Multiple Sclerosis (MS), so the aim of our review is to provide an overview of the main characteristics of MDSCs in MS and its preclinical model by discussing the most recent data available. The immunosuppressive functions of MDSCs can be dysregulated in MS, leading to an exacerbation of the autoimmune response and disease progression. Antigen-specific peptide immunotherapy, which aims to restore tolerance while avoiding the use of non-specific immunosuppressive drugs, is a promising approach for autoimmune diseases, but the cellular mechanisms behind successful therapy remain poorly understood. Therefore, targeting MDSCs could be a promising therapeutic approach for MS. Various strategies for modulating MDSCs have been investigated, including the use of pharmacological agents, biological agents, and adoptive transfer of exogenous MDSCs. However, it remained unclear whether MDSCs display any therapeutic potential in MS and how this therapy could modulate different aspects of the disease. Collectively, all the described studies revealed a pivotal role for MDSCs in the regulation of MS.

    .
    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
    脂肪组织,一个不可或缺的器官,发挥能量储存和新陈代谢的关键作用,并有助于维持生物体能量和健康的动态平衡。脂肪细胞肥大和脂肪细胞增生(脂肪生成)是脂肪沉积的两个主要机制。通过将间充质干细胞分化成前脂肪细胞并再分化来获得成熟的脂肪细胞。然而,脂肪生成的机制尚不清楚.自噬,一种替代的细胞死亡途径,通过细胞成分的降解来维持细胞内能量稳态,与调节脂肪生成有关。此外,脂肪组织作为一种内分泌器官,产生各种细胞因子,和某些炎症因子,反过来,调节自噬和脂肪生成。此外,自噬通过调节活性氧影响细胞内氧化还原稳态,在脂肪形成中起关键作用。人们对探索自噬的参与越来越感兴趣,炎症,和脂肪形成中的氧化应激。本手稿回顾了自噬的影响,氧化应激,和炎症对脂肪生成的调节,第一次,讨论了它们在脂肪生成过程中的相互作用。自噬作用的综合分析,炎症和氧化应激将有助于阐明脂肪形成的机制,并加快探索治疗肥胖相关代谢紊乱的分子靶标。
    Adipose tissue, an indispensable organ, fulfils the pivotal role of energy storage and metabolism and is instrumental in maintaining the dynamic equilibrium of energy and health of the organism. Adipocyte hypertrophy and adipocyte hyperplasia (adipogenesis) are the two primary mechanisms of fat deposition. Mature adipocytes are obtained by differentiating mesenchymal stem cells into preadipocytes and redifferentiation. However, the mechanisms orchestrating adipogenesis remain unclear. Autophagy, an alternative cell death pathway that sustains intracellular energy homeostasis through the degradation of cellular components, is implicated in regulating adipogenesis. Furthermore, adipose tissue functions as an endocrine organ, producing various cytokines, and certain inflammatory factors, in turn, modulate autophagy and adipogenesis. Additionally, autophagy influences intracellular redox homeostasis by regulating reactive oxygen species, which play pivotal roles in adipogenesis. There is a growing interest in exploring the involvement of autophagy, inflammation, and oxidative stress in adipogenesis. The present manuscript reviews the impact of autophagy, oxidative stress, and inflammation on the regulation of adipogenesis and, for the first time, discusses their interactions during adipogenesis. An integrated analysis of the role of autophagy, inflammation and oxidative stress will contribute to elucidating the mechanisms of adipogenesis and expediting the exploration of molecular targets for treating obesity-related metabolic disorders.
    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
    足细胞,作为内在的肾细胞,还可以在炎症条件下表达MHC-II和共刺激分子,提示它们可能作为抗原呈递细胞(APC)激活免疫细胞反应,然后导致免疫介导的肾损伤。它们已经被认为是乙型肝炎病毒(HBV)相关性肾小球肾炎(HBV-GN)致病机制的主要靶标。先前的研究还表明,炎症细胞浸润和免疫介导的组织损伤在HBV-GN患者的肾脏样本中是明显的。然而,足细胞免疫紊乱在HBV-GN致病机制中的作用尚不清楚。
    在HBV转基因(HBV-Tg)小鼠中测量肾功能和炎性细胞浸润。体外,建立足细胞/CD4+T细胞或巨噬细胞共培养体系。然后,HBx的表达,通过免疫组织化学测定CD4和CD68,而MHC-II的表达,通过免疫荧光测定CD40和CD40L。通过流式细胞术检查共刺激分子的表达。ELISA法检测炎症因子水平。
    体内,HBV-Tg小鼠肾功能明显受损。HBV-Tg小鼠肾小球中HBx显著上调,免疫细胞浸润。MHC-II和共刺激分子CD40在HBV-Tg小鼠足细胞中的表达增加;CD4+T细胞在肾小球中表现出增加的CD40L表达。体外,CD40在HBx足细胞中表达显著升高。在共同文化系统中,HBx足细胞刺激CD4+T细胞活化并引起IFN-γ和IL-4之间的失衡。HBx足细胞还增强巨噬细胞的粘附能力并诱导促炎介质的释放。
    放在一起,这些与足细胞相关的免疫紊乱可能与HBV-GN的致病机制有关。
    UNASSIGNED: Podocytes, as intrinsic renal cells, can also express MHC-II and costimulatory molecules under inflammatory conditions, suggesting that they may act as antigen-presenting cells (APCs) to activate immune cell responses and then lead to immune-mediated renal injury. They are already recognized as main targets in the pathogenic mechanism of hepatitis B virus (HBV)-associated glomerulonephritis (HBV-GN). Previous studies also have indicated that inflammatory cells infiltration and immune-mediated tissue injury are evident in the kidney samples of patients with HBV-GN. However, the role of podocytes immune disorder in the pathogenic mechanism of HBV-GN remains unclear.
    UNASSIGNED: Renal function and inflammatory cells infiltration were measured in HBV transgenic (HBV-Tg) mice. In vitro, podocytes/CD4+ T cells or macrophages co-culture system was established. Then, the expression of HBx, CD4, and CD68 was determined by immunohistochemistry, while the expression of MHC-II, CD40, and CD40L was determined by immunofluorescence. Co-stimulatory molecules expression was examined by flow cytometry. The levels of inflammatory factors were detected by ELISA.
    UNASSIGNED: In vivo, renal function was obviously impaired in HBV-Tg mice. HBx was significantly upregulated and immune cells infiltrated in the glomerulus of HBV-Tg mice. Expression of MHC-II and costimulatory molecule CD40 increased in the podocytes of HBV-Tg mice; CD4+ T cells exhibited increased CD40L expression in glomerulus. In vitro, CD40 expression was markedly elevated in HBx-podocytes. In co-culture systems, HBx-podocytes stimulated CD4+ T cells activation and caused the imbalance between IFN-γ and IL-4. HBx-podocytes also enhanced the adhesion ability of macrophages and induced the release of proinflammatory mediators.
    UNASSIGNED: Taken together, these podocyte-related immune disorder may be involved in the pathogenic mechanism of HBV-GN.
    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
    疫苗是预防传染病的有效干预措施。目前,许多疫苗策略旨在通过控制抗原释放来提高疫苗效力。通常涉及在注射部位的各种方法。然而,疫苗中抗原在细胞内缓慢释放的策略仍未探索。我们的研究表明,控制树突状细胞中抗原的降解并减缓其从早期内体到溶酶体的转运显着增强了抗原特异性T细胞免疫反应和生发中心B细胞反应。这导致了持续的体液和细胞免疫的建立,体内成像和流式细胞术表明,这种方法不仅延长了抗原在注射部位的滞留,而且提高了淋巴结中的抗原浓度。超越传统的铝(明矾)佐剂。此外,我们证明缓慢的抗原降解诱导更强的滤泡辅助性T细胞应答,并增加了长寿命浆细胞和记忆B细胞的比例.总的来说,这些发现表明,控制树突状细胞中抗原转运的速度可以显著提高疫苗的效力,为开发高免疫原性的下一代疫苗提供了创新的途径。
    Vaccines are an effective intervention for preventing infectious diseases. Currently many vaccine strategies are designed to improve vaccine efficacy by controlling antigen release, typically involving various approaches at the injection site. Yet, strategies for intracellular slow-release of antigens in vaccines are still unexplored. Our study showed that controlling the degradation of antigens in dendritic cells and slowing their transport from early endosomes to lysosomes markedly enhances both antigen-specific T-cell immune responses and germinal center B cell responses. This leads to the establishment of sustained humoral and cellular immunity in vivo imaging and flow cytometry indicated this method not only prolongs antigen retention at the injection site but also enhances antigen concentration in lymph nodes, surpassing traditional Aluminium (Alum) adjuvants. Additionally, we demonstrated that the slow antigen degradation induces stronger follicular helper T cell responses and increases proportions of long-lived plasma cells and memory B cells. Overall, these findings propose that controlling the speed of antigens transport in dendritic cells can significantly boost vaccine efficacy, offering an innovative avenue for developing highly immunogenic next-generation vaccines.
    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
    反应性星形胶质细胞是重要的病理生理和合成神经甾体。我们观察到LPS增加了大鼠皮质星形胶质细胞中的免疫反应性TLR4和关键类固醇生成酶,并研究了皮质类固醇是否产生并介导星形胶质细胞TLR4依赖性先天免疫反应。我们发现LPS增加了纯化的星形胶质细胞中类固醇生成的急性调节蛋白(StAR)和StAR依赖性醛固酮的产生。两种增加都被TLR4拮抗剂TAK242阻断。LPS还增加了11β-羟基类固醇脱氢酶1型(11β-HSD1)和皮质酮的产生,两者都被TAK242和针对11β-HSD1,StAR,或醛固酮合酶(CYP11B2)。11β-HSD1,StAR的击倒,或CYP11B2或阻断盐皮质激素受体(MR)或糖皮质激素受体(GR)可防止p-Ser9GSK-3β的去磷酸化,NF-κB的激活,以及C3、IL-1β的GSK-3β依赖性增加,和LPS引起的TNF-α。外源性醛固酮模拟了LPS在星形胶质细胞中的MR和GSK-3β依赖性促炎作用,但皮质酮没有。用LPS处理的星形胶质细胞的上清液降低了MAP2和培养的神经元的活力,除非星形胶质细胞StAR或MR受到抑制。在肾上腺切除的大鼠中,脑室内注射LPS增加星形胶质细胞TLR4,StAR,CYP11B2和11β-HSD1,NF-κB,C3和IL-1β,皮质中星形胶质细胞p-Ser9GSK-3β减少,具有神经毒性,除非同时注射螺内酯,与体外结果一致。LPS还激活了皮质中一些NeuN+和CD11b+细胞中的NF-κB,螺内酯可以预防这些影响。我们得出的结论是,内分泌醛固酮可能参与星形胶质细胞的TLR4依赖性先天免疫反应,并可以通过激活星形细胞MR/GSK-3β/NF-κB信号传导来触发旁分泌效应。
    Reactive astrocytes are important pathophysiologically and synthesize neurosteroids. We observed that LPS increased immunoreactive TLR4 and key steroidogenic enzymes in cortical astrocytes of rats and investigated whether corticosteroids are produced and mediate astrocytic TLR4-dependent innate immune responses. We found that LPS increased steroidogenic acute regulatory protein (StAR) and StAR-dependent aldosterone production in purified astrocytes. Both increases were blocked by the TLR4 antagonist TAK242. LPS also increased 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) and corticosterone production, and both were prevented by TAK242 and by siRNAs against 11β-HSD1, StAR, or aldosterone synthase (CYP11B2). Knockdown of 11β-HSD1, StAR, or CYP11B2 or blocking either mineralocorticoid receptors (MR) or glucocorticoid receptors (GR) prevented dephosphorylation of p-Ser9GSK-3β, activation of NF-κB, and the GSK-3β-dependent increases of C3, IL-1β, and TNF-α caused by LPS. Exogenous aldosterone mimicked the MR- and GSK-3β-dependent pro-inflammatory effects of LPS in astrocytes, but corticosterone did not. Supernatants from astrocytes treated with LPS reduced MAP2 and viability of cultured neurons except when astrocytic StAR or MR was inhibited. In adrenalectomized rats, intracerebroventricular injection of LPS increased astrocytic TLR4, StAR, CYP11B2, and 11β-HSD1, NF-κB, C3 and IL-1β, decreased astrocytic p-Ser9GSK-3β in the cortex and was neurotoxic, except when spironolactone was co-injected, consistent with the in vitro results. LPS also activated NF-κB in some NeuN+ and CD11b+ cells in the cortex, and these effects were prevented by spironolactone. We conclude that intracrine aldosterone may be involved in the TLR4-dependent innate immune responses of astrocytes and can trigger paracrine effects by activating astrocytic MR/GSK-3β/NF-κB signaling.
    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
    本研究旨在阐明以下假设:螺旋发射器125I颗粒与PARP抑制剂Olaparib组合可通过促进抗肿瘤免疫反应来抑制胰腺癌进展。胰腺癌细胞系(Panc02)和皮下接种Panc02细胞的小鼠用于体外和体内实验。分别,其次是125I和Olaparib管理。流式细胞术检测Panc02细胞的凋亡和CRT暴露。QRT-PCR,免疫荧光,免疫组织化学分析,采用Westernblot检测mRNA和蛋白表达。实验结果表明,125I联合奥拉帕尼诱导胰腺癌免疫原性细胞死亡并影响抗原呈递。125I联合奥拉帕尼通过上调CD4,CD8,CD69,Caspase3,CD86,颗粒酶B影响T细胞和树突状细胞,CD80和I型干扰素(IFN)-γ在体内下调Ki67。该组合还激活了Panc02细胞中IFN基因(Sting)途径的环状GMP-AMP合酶刺激物。此外,Sting敲除减轻了125I和Olaparib的组合对胰腺癌进展的影响。总之,125I联合奥拉帕尼通过促进抗肿瘤免疫反应抑制胰腺癌进展,这可能为胰腺癌提供潜在的治疗方法。
    The present study aimed to clarify the hypothesis that auger emitter 125I particles in combination with PARP inhibitor Olaparib could inhibit pancreatic cancer progression by promoting antitumor immune response. Pancreatic cancer cell line (Panc02) and mice subcutaneously inoculated with Panc02 cells were employed for the in vitro and in vivo experiments, respectively, followed by 125I and Olaparib administrations. The apoptosis and CRT exposure of Panc02 cells were detected using flow cytometry assay. QRT-PCR, immunofluorescence, immunohistochemical analysis, and western blot were employed to examine mRNA and protein expression. Experimental results showed that 125I combined with Olaparib induced immunogenic cell death and affected antigen presentation in pancreatic cancer. 125I in combination with Olaparib influenced T cells and dendritic cells by up-regulating CD4, CD8, CD69, Caspase3, CD86, granzyme B, CD80, and type I interferon (IFN)-γ and down-regulating Ki67 in vivo. The combination also activated the cyclic GMP-AMP synthase stimulator of IFN genes (Sting) pathway in Panc02 cells. Moreover, Sting knockdown alleviated the effect of the combination of 125I and Olaparib on pancreatic cancer progression. In summary, 125I in combination with Olaparib inhibited pancreatic cancer progression through promoting antitumor immune responses, which may provide a potential treatment for pancreatic cancer.
    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
    佐剂在疫苗诱导有效免疫应答中起关键作用。这里,制备将佐剂功能整合到递送载体中的自组装纳米疫苗平台。将阳离子香菇多糖(CLNT)与卵清蛋白(OVA)混合以获得自组装纳米疫苗(CLNTO纳米疫苗),通过toll样受体2/4(TLR2/4)诱导骨髓树突状细胞(BMDC)的摄取和成熟,以产生有效的抗原交叉呈递。CLNTO纳米疫苗靶向淋巴结(LN),并通过TLR和肿瘤坏死因子(TNF)信号通路诱导强大的OVA特异性免疫反应,视黄酸诱导基因I(RIG-I)受体,和细胞因子-细胞因子受体相互作用。此外,发现CLNTO纳米疫苗可促进滤泡辅助性T(Tfh)细胞的激活,并诱导生发中心(GC)B细胞分化为记忆B细胞和浆细胞,从而增强免疫反应。用CLNTO纳米疫苗接种显着抑制表达卵清蛋白(OVA)的B16黑色素瘤细胞(B16-OVA)肿瘤的生长,表明其在癌症免疫疗法方面的巨大潜力。因此,这项研究提出了一个简单的,安全,和有效的自组装纳米疫苗,诱导辅助性T细胞1(Th1)和辅助性T细胞(Th2)免疫反应,使其成为有效的疫苗输送系统。
    Adjuvants play a critical role in the induction of effective immune responses by vaccines. Here, a self-assembling nanovaccine platform that integrates adjuvant functions into the delivery vehicle is prepared. Cationic Lentinan (CLNT) is mixed with ovalbumin (OVA) to obtain a self-assembling nanovaccine (CLNTO nanovaccine), which induces the uptake and maturation of bone marrow dendritic cells (BMDCs) via the toll-like receptors 2/4 (TLR2/4) to produce effective antigen cross-presentation. CLNTO nanovaccines target lymph nodes (LNs) and induce a robust OVA-specific immune response via TLR and tumor necrosis factor (TNF) signaling pathways, retinoic acid-inducible gene I (RIG-I) receptor, and cytokine-cytokine receptor interactions. In addition, CLNTO nanovaccines are found that promote the activation of follicular helper T (Tfh) cells and induce the differentiation of germinal center (GC) B cells into memory B cells and plasma cells, thereby enhancing the immune response. Vaccination with CLNTO nanovaccine significantly inhibits the growth of ovalbumin (OVA)-expressing B16 melanoma cell (B16-OVA) tumors, indicating its great potential for cancer immunotherapy. Therefore, this study presents a simple, safe, and effective self-assembling nanovaccine that induces helper T cell 1 (Th1) and helper T cell (Th2) immune responses, making it an effective vaccine delivery system.
    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
    由于在临床前模型和早期临床试验中证明的安全性和有效性,细胞外囊泡(EV)是有前途的下一代治疗剂和药物递送系统。迫切需要解决EV的免疫原性(除了明显缺乏免疫毒性之外)以推进临床开发。迄今为止,很少有研究评估了电动汽车的非预期免疫识别。深入了解EV诱导的免疫原性和清除对于制定有效的治疗策略是必要的。包括在不希望时减轻免疫识别的方法。本文总结了与EV潜在免疫原性有关的各种因素,以及降低免疫学识别以提高治疗效果的策略。
    Extracellular vesicles (EVs) are promising next-generation therapeutics and drug delivery systems due to demonstrated safety and efficacy in preclinical models and early-stage clinical trials. There is an urgent need to address the immunogenicity of EVs (beyond the apparent lack of immunotoxicity) to advance clinical development. To date, few studies have assessed unintended immunological recognition of EVs. An in-depth understanding of EV-induced immunogenicity and clearance is necessary to develop effective therapeutic strategies, including approaches to mitigate immunological recognition when undesired. This article summarizes various factors involved in the potential immunogenicity of EVs and strategies to reduce immunological recognition for improved therapeutic benefit.
    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
    L-精氨酸代谢与对分枝杆菌的免疫力密切相关,主要通过一氧化氮(NO)的抗菌活性。通过靶向L-精氨酸途径的干预来调节结核病(TB)结局的潜力受到对机制的不完全理解和体内建模不足的限制。这些知识差距在艾滋病毒和Mtb共感染方面更加复杂,其中由于HIV感染引起的精氨酸酶-1的激活可能促进Mtb和HIV的存活和复制。我们利用体外和体内系统来确定使用Nω-羟基-nor-L-精氨酸(nor-NOHA)的精氨酸酶抑制相对于Mtb感染后的免疫应答和疾病结果如何改变L-精氨酸途径代谢。用nor-NOHA极化鼠巨噬细胞(RAW264.7)治疗M1表型,增加NO,并降低RAW巨噬细胞中的Mtb。在Balb/c小鼠中,nor-NOHA降低了肺精氨酸酶并增加了抗微生物代谢物精胺,与肺中MtbCFU降低的趋势有关。在人源化免疫系统(HIS)小鼠中,HIV感染增加了血浆精氨酸酶,并增强了肺精氨酸酶对Mtb的反应。nor-NOHA治疗增加了肺组织中对Mtb和Mtb/HIV的细胞因子应答,但没有显著改变细菌负荷或病毒载量。我们的结果表明,L-精氨酸途径调节剂可能具有作为宿主导向疗法的潜力,可以增强结核病化疗中的抗生素。
    L-arginine metabolism is strongly linked with immunity to mycobacteria, primarily through the antimicrobial activity of nitric oxide (NO). The potential to modulate tuberculosis (TB) outcomes through interventions that target L-arginine pathways are limited by an incomplete understanding of mechanisms and inadequate in vivo modeling. These gaps in knowledge are compounded for HIV and Mtb co-infections, where activation of arginase-1 due to HIV infection may promote survival and replication of both Mtb and HIV. We utilized in vitro and in vivo systems to determine how arginase inhibition using Nω-hydroxy-nor-L-arginine (nor-NOHA) alters L-arginine pathway metabolism relative to immune responses and disease outcomes following Mtb infection. Treatment with nor-NOHA polarized murine macrophages (RAW 264.7) towards M1 phenotype, increased NO, and reduced Mtb in RAW macrophages. In Balb/c mice, nor-NOHA reduced pulmonary arginase and increased the antimicrobial metabolite spermine in association with a trend towards reduced Mtb CFU in lung. In humanized immune system (HIS) mice, HIV infection increased plasma arginase and heightened the pulmonary arginase response to Mtb. Treatment with nor-NOHA increased cytokine responses to Mtb and Mtb/HIV in lung tissue but did not significantly alter bacterial burden or viral load. Our results suggest that L-arginine pathway modulators may have potential as host-directed therapies to augment antibiotics in TB chemotherapy.
    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
    尼帕病毒(NiV)一种高度致命的蝙蝠传播的副粘病毒,由于特定地区的反复爆发,构成了重大威胁,引起严重的呼吸系统和神经系统疾病,发病率高。两种不同的菌株,NiV-马来西亚(NiV-M)和NiV-孟加拉国(NiV-B),导致不同地理区域的疫情爆发。目前,没有商业许可的疫苗或药物可用于预防或治疗。为了应对这种针对NiV和相关亨尼帕病毒感染的保护的迫切需求,我们开发了一种新型的同型病毒样纳米颗粒(VLP)疫苗,共同展示来自两种菌株的NiV附着糖蛋白(G),利用铁蛋白蛋白的自组装特性。与NiVG亚单位疫苗相比,我们的纳米颗粒疫苗在叙利亚仓鼠中引发了显著更高水平的中和抗体,并提供了针对NiV感染致死性攻击的完全保护.值得注意的是,纳米颗粒疫苗刺激了抗体的产生,这些抗体表现出与同源或异源乙型肝炎病毒的优异交叉反应性。这些发现强调了基于铁蛋白的纳米颗粒疫苗在提供广谱和长期保护以抵抗NiV和新出现的人畜共患乙型肝炎病毒挑战方面的潜在效用。
    The Nipah virus (NiV), a highly deadly bat-borne paramyxovirus, poses a substantial threat due to recurrent outbreaks in specific regions, causing severe respiratory and neurological diseases with high morbidity. Two distinct strains, NiV-Malaysia (NiV-M) and NiV-Bangladesh (NiV-B), contribute to outbreaks in different geographical areas. Currently, there are no commercially licensed vaccines or drugs available for prevention or treatment. In response to this urgent need for protection against NiV and related henipaviruses infections, we developed a novel homotypic virus-like nanoparticle (VLP) vaccine co-displaying NiV attachment glycoproteins (G) from both strains, utilizing the self-assembling properties of ferritin protein. In comparison to the NiV G subunit vaccine, our nanoparticle vaccine elicited significantly higher levels of neutralizing antibodies and provided complete protection against a lethal challenge with NiV infection in Syrian hamsters. Remarkably, the nanoparticle vaccine stimulated the production of antibodies that exhibited superior cross-reactivity to homologous or heterologous henipavirus. These findings underscore the potential utility of ferritin-based nanoparticle vaccines in providing both broad-spectrum and long-term protection against NiV and emerging zoonotic henipaviruses challenges.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号