anti‐tumor immunity

  • 文章类型: Journal Article
    这项研究通过使用高通量单细胞转录组测序和先进的生物信息学分析研究TRIM28的作用及其复杂的分子机制,深入研究了去势抵抗前列腺癌(CRPC)的未开发领域。我们的全面检查揭示了动态的TRIM28表达变化,特别是在CRPC内的免疫细胞如巨噬细胞和CD8+T细胞中。与TCGA数据的相关性分析强调了TRIM28与免疫检查点表达之间的联系,并强调了其对免疫细胞数量和功能的关键影响。使用TRIM28基因敲除小鼠模型,我们确定了差异表达的基因和富集的途径,揭示TRIM28在cGAS-STING途径中的潜在调控参与。体外,实验进一步说明,前列腺癌细胞中TRIM28基因敲除通过抑制M2巨噬细胞极化和增强CD8+T细胞活性而诱导了显著的抗肿瘤免疫作用。这一有影响力的发现在原位移植肿瘤模型中得到了验证,其中TRIM28基因敲除表现出肿瘤生长的减速,减少M2巨噬细胞的比例,增强CD8+T细胞的浸润。总之,这项研究阐明了迄今为止未知的TRIM28在CRPC中的抗肿瘤免疫作用,并通过cGAS-STING信号通路揭示了其潜在的调节机制。这些发现为CRPC的免疫前景提供了新的见解,为开发创新的治疗策略提供有希望的方向。
    This study delves into the unexplored realm of castration-resistant prostate cancer (CRPC) by investigating the role of TRIM28 and its intricate molecular mechanisms using high-throughput single-cell transcriptome sequencing and advanced bioinformatics analysis. Our comprehensive examination unveiled dynamic TRIM28 expression changes, particularly in immune cells such as macrophages and CD8+ T cells within CRPC. Correlation analyses with TCGA data highlighted the connection between TRIM28 and immune checkpoint expression and emphasized its pivotal influence on the quantity and functionality of immune cells. Using TRIM28 knockout mouse models, we identified differentially expressed genes and enriched pathways, unraveling the potential regulatory involvement of TRIM28 in the cGAS-STING pathway. In vitro, experiments further illuminated that TRIM28 knockout in prostate cancer cells induced a notable anti-tumor immune effect by inhibiting M2 macrophage polarization and enhancing CD8+ T cell activity. This impactful discovery was validated in an in situ transplant tumor model, where TRIM28 knockout exhibited a deceleration in tumor growth, reduced proportions of M2 macrophages, and enhanced infiltration of CD8+ T cells. In summary, this study elucidates the hitherto unknown anti-tumor immune role of TRIM28 in CRPC and unravels its potential regulatory mechanism via the cGAS-STING signaling pathway. These findings provide novel insights into the immune landscape of CRPC, offering promising directions for developing innovative therapeutic strategies.
    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
    焦亡,炎症细胞死亡,在激活炎症反应中起关键作用,逆转免疫抑制和增强抗肿瘤免疫力。然而,关于如何在肿瘤细胞中有效和精确地诱导焦亡以扩增抗肿瘤免疫疗法仍然存在挑战。在这里,pH响应型聚多巴胺(PDA)纳米簇,全氟化碳(PFC)@八精氨酸(R8)-1-十六胺(He)-卟啉(Por)@PDA-藤黄酸(GA)-cRGD(R-P@PDA-GC),通过两输入程序级联疗法合理设计以增强光疗诱导的焦亡并增强抗肿瘤免疫力。简而言之,氧供体PFC封装在R8连接的光敏剂Por和He胶束作为核心,然后掺入GA和cRGD肽修饰的PDA壳,产生最终的R-P@PDA-GC纳米平台(NPs)。pH响应性NP通过经由PFC递送氧来有效地缓解缺氧,并且通过GA减轻肿瘤细胞中的耐热性。在双输入程序照射时,R-P@PDA-GCNP显着增强肿瘤细胞内活性氧的产生,通过Caspase-1/GSDMD途径触发细胞凋亡,并释放多种炎症因子进入TME。这导致树突状细胞的成熟,细胞毒性CD8+T和NK细胞的强大浸润,免疫抑制Treg细胞的减少,从而增强抗肿瘤免疫力。
    Pyroptosis, an inflammatory cell death, plays a pivotal role in activating inflammatory response, reversing immunosuppression and enhancing anti-tumor immunity. However, challenges remain regarding how to induce pyroptosis efficiently and precisely in tumor cells to amplify anti-tumor immunotherapy. Herein, a pH-responsive polydopamine (PDA) nanocluster, perfluorocarbon (PFC)@octo-arginine (R8)-1-Hexadecylamine (He)-porphyrin (Por)@PDA-gambogic acid (GA)-cRGD (R-P@PDA-GC), is rationally design to augment phototherapy-induced pyroptosis and boost anti-tumor immunity through a two-input programmed cascade therapy. Briefly, oxygen doner PFC is encapsulated within R8 linked photosensitizer Por and He micelles as the core, followed by incorporation of GA and cRGD peptides modified PDA shell, yielding the ultimate R-P@PDA-GC nanoplatforms (NPs). The pH-responsive NPs effectively alleviate hypoxia by delivering oxygen via PFC and mitigate heat resistance in tumor cells through GA. Upon two-input programmed irradiation, R-P@PDA-GC NPs significantly enhance reactive oxygen species production within tumor cells, triggering pyroptosis via the Caspase-1/GSDMD pathway and releasing numerous inflammatory factors into the TME. This leads to the maturation of dendritic cells, robust infiltration of cytotoxic CD8+ T and NK cells, and diminution of immune suppressor Treg cells, thereby amplifying anti-tumor immunity.
    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
    肿瘤微环境(TME)诱导的纳米催化疗法是一种有前途的癌症治疗策略。但低催化效率限制了其治疗效果。单原子催化剂(SAC)是一种新型的纳米酶,具有令人难以置信的催化效率。这里,构建了单原子锰(Mn)-N/C纳米酶。Mn-N/C通过类Fenton反应催化细胞H2O2转化为OH,并能够充分产生活性氧(ROS),它诱导肿瘤细胞的免疫原性细胞死亡(ICD)并显着促进CD8T抗肿瘤免疫。此外,RNA测序分析显示,Mn-N/C处理激活I型干扰素(IFN)信号,这对于Mn-N/C介导的抗肿瘤免疫应答至关重要。机械上,Mn-N/C触发的胞浆DNA和Mn2+的释放共同激活了cGAS-STING途径,随后刺激I型IFN诱导。一种高效的单原子纳米酶,Mn-N/C,与抗PD-L1阻断联合增强抗肿瘤免疫反应并表现出协同治疗效果,是提议的。
    Tumor microenvironment (TME)-induced nanocatalytic therapy is a promising strategy for cancer treatment, but the low catalytic efficiency limits its therapeutic efficacy. Single-atom catalysts (SACs) are a new type of nanozyme with incredible catalytic efficiency. Here, a single-atom manganese (Mn)-N/C nanozyme is constructed. Mn-N/C catalyzes the conversion of cellular H2O2 to ∙OH through a Fenton-like reaction and enables the sufficient generation of reactive oxygen species (ROS), which induces immunogenic cell death (ICD) of tumor cells and significantly promotes CD8+T anti-tumor immunity. Moreover, RNA sequencing analysis reveals that Mn-N/C treatment activates type I interferon (IFN) signaling, which is critical for Mn-N/C-mediated anti-tumor immune response. Mechanistically, the release of cytosolic DNA and Mn2+ triggered by Mn-N/C collectively activates the cGAS-STING pathway, subsequently stimulating type I IFN induction. A highly efficient single-atom nanozyme, Mn-N/C, which enhances anti-tumor immune response and exhibits synergistic therapeutic effects when combined with the anti-PD-L1 blockade, is proposed.
    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
    PDL1阻断疗法在癌症免疫疗法中具有巨大的前景。肿瘤中PDL1表达的超声成像对预测治疗效果具有重要意义。作为一项概念验证研究,一种新的超声造影剂已经在这里被创新成像和阻断肿瘤组织中的PDL1。简而言之,细胞外囊泡(EV)被设计为在表面上显示截短的PD1(tPD1),以通过与EV丰富的跨膜蛋白PTGFRN融合而以高亲和力结合PDL1。然后通过电穿孔用Ca(HCO3)2封装工程EV,并命名为Gp-EVtPD1,它将识别PDL1高表达的细胞并在内体和溶酶体中产生气体。一方面,回声信号强度与肿瘤中PDL1的表达和免疫反应抑制密切相关。另一方面,在Gp-EVtPD1在受体细胞中的轨迹过程中,EV上的tPD1结合PDL1,并以顺序方式触发内体/溶酶体中的PDL1内吞和降解,从而增强细胞毒性T细胞的抗肿瘤免疫力。总之,Gp-EVtPD1作为一种新型的超声造影剂和PDL1阻断剂,在成像PDL1表达和克服免疫检查点阻断剂抵抗方面可能具有很大的优势。
    PDL1 blockade therapy holds great promise in cancer immunotherapy. Ultrasound imaging of PDL1 expression in the tumor is of great importance in predicting the therapeutic efficacy. As a proof-of-concept study, a novel ultrasound contrast agent has been innovated here to image and block PDL1 in the tumor tissue. Briefly, extracellular vesicles (EVs) are engineered to display truncated PD1 (tPD1) on the surface to bind PDL1 with high affinity by fusion to EV-abundant transmembrane protein PTGFRN. The engineered EVs are then encapsulated with Ca(HCO3)2 via electroporation and designated as Gp-EVtPD1, which would recognize PDL1 highly expressed cells and produce gas in the endosomes and lysosomes. On the one hand, the echogenic signal intensity correlates well with the PDL1 expression and immune response inhibition in the tumor. On the other hand, during the trajectory of Gp-EVtPD1 in the recipient cells, tPD1 on the EV binds PDL1 and triggers the PDL1 endocytosis and degradation in endosomes/lysosomes in a sequential manner, and thus boosts the anti-tumor immunity of cytotoxic T cells. In summary, Gp-EVtPD1 serves as a novel ultrasound contrast agent and blocker of PDL1, which might be of great advantage in imaging PDL1 expression and conquering immune checkpoint blocker resistance.
    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
    细胞外囊泡(EV)由几乎所有细胞分泌。它们含有蛋白质,脂质,和从亲本细胞递送到受体细胞的核酸。因此,它们充当细胞间通讯和分子转移的介质。最近的证据表明外泌体,一小部分电动汽车,参与许多生理和病理过程,甚至在癌症发生和转移之前,在重塑肿瘤免疫微环境中起着至关重要的作用。来源于肿瘤细胞和宿主细胞的外泌体在局部或远程介导它们的相互调节,从而确定癌症治疗的反应性。因此,肿瘤来源的循环外泌体被认为是肿瘤早期检测和诊断的非侵入性生物标志物.基于外泌体的疗法也正在成为可以应用于抑制肿瘤进展或增强抗肿瘤免疫力的尖端和有前途的策略。在这里,目前对外泌体及其在调节免疫反应中的关键作用的理解,以及其潜在的治疗应用概述。还介绍了当前研究的局限性,并描述了未来研究的方向。
    Extracellular vesicles (EVs) are secreted by almost all cells. They contain proteins, lipids, and nucleic acids which are delivered from the parent cells to the recipient cells. Thereby, they function as mediators of intercellular communication and molecular transfer. Recent evidences suggest that exosomes, a small subset of EVs, are involved in numerous physiological and pathological processes and play essential roles in remodeling the tumor immune microenvironment even before the occurrence and metastasis of cancer. Exosomes derived from tumor cells and host cells mediate their mutual regulation locally or remotely, thereby determining the responsiveness of cancer therapies. As such, tumor-derived circulating exosomes are considered as noninvasive biomarkers for early detection and diagnosis of tumor. Exosome-based therapies are also emerging as cutting-edge and promising strategies that could be applied to suppress tumor progression or enhance anti-tumor immunity. Herein, the current understanding of exosomes and their key roles in modulating immune responses, as well as their potential therapeutic applications are outlined. The limitations of current studies are also presented and directions for future research are described.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号