Oligodeoxyribonucleotides

寡脱氧核糖核苷酸
  • 文章类型: Journal Article
    在实验室设置中,近交小鼠品系如BALB/c,C57BL/6J,和C57BL/6N常用。免疫学和感染性疾病的研究表明,它们的Th1和Th2免疫应答不同。然而,对疫苗接种的免疫反应的具体差异仍需要调查。在这项研究中,卵清蛋白(OVA)用作抗原,富含CpG的重组质粒(pUC18-CpG)用作免疫佐剂。间接ELISA法检测血清特异性抗体IgG水平。在35dpi,使用MILLIPLEX®测量血清细胞因子水平。使用流式细胞术检查小鼠脾脏中的T淋巴细胞簇,以研究CPG-OVA疫苗对三种不同类型小鼠的免疫作用。结果表明,pUC18-CpG作为佐剂可以成功增强免疫应答。BALB/c的IgG抗体水平最高。在OVA-only组中,三种小鼠的CD4+/CD8+比值普遍升高,BALB/c组比例最高。接种CpG-OVA后,3种小鼠的CD4+/CD8+比值均低于OVA组,C57BL/6J最低。与CpG-OVA组的三种小鼠比拟,与C57BL/6J和C57BL/6N相比,BALB/c中Th2细胞因子IL-6和IL-10的水平升高。OVA之后,C57BL/6J分泌的6种细胞因子高于C57BL/6NOVA组。因此,C57是检查疫苗在细胞免疫中的功能的更好的模型,而BALB/c小鼠更容易发生体液免疫。除了突出CpG质粒成功激活Th1和Th2的免疫应答以及体内IgG表达和促进T细胞免疫分型的能力外,这项研究为免疫学和传染病小鼠模型的选择提供了有价值的见解,为将来设计更有效的疫苗提供了宝贵的资源。
    In lab settings, inbred mouse strains like BALB/c, C57BL/6J, and C57BL/6N are commonly used. Research in immunology and infectious diseases indicates that their Th1 and Th2 immune responses differ. However, the specific differences in the immune response to the vaccination still require investigation. In this study, ovalbumin (OVA) was used as an antigen and CpG-enriched recombinant plasmid (pUC18-CpG) as an adjuvant for immunisation. The level of serum-specific antibody IgG was detected by indirect ELISA. At 35dpi, serum cytokine levels were measured using MILLIPLEX®. T lymphocyte clusters from mouse spleen were examined using flow cytometry to investigate the immunological effects of the CPG-OVA vaccine on three different types of mice. The results showed that pUC18-CpG as an adjuvant could successfully enhance the immune response. BALB/c had the highest level of IgG antibody. In the OVA-only group, the CD4+/CD8+ ratio of the three types of mice was generally increased, and the BALB/c group had the highest ratio. After inoculation with CpG-OVA, the CD4+/CD8+ ratio of the three types of mice was lower than that of the OVA-only group, and C57BL/6J was the lowest. Compared with the CpG-OVA group of the three kinds of mice, the levels of Th2 cytokines IL-6 and IL-10 in BALB/c were increased compared with C57BL/6J and C57BL/6N. After OVA, the six cytokines secreted in C57BL/6J were higher than those in the C57BL/6N OVA group. Therefore, C57 is a better model for examining the function of the vaccine in cellular immunity, whereas BALB/c mice are more prone to humoral immunity. In addition to highlighting the CpG plasmid\'s ability to successfully activate the immune response of Th1 and Th2, as well as the expression of IgG in vivo and promote T cell immune typing, this study provides valuable insights into immunology and the selection of mouse models for infectious diseases, providing a valuable resource for designing more effective vaccines in the future.
    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
    背景:三阴性乳腺癌(TNBC)是一种复发性乳腺癌,异质,和浸润性乳腺癌。以顺序方式用紫杉醇和氟尿嘧啶治疗TNBC患者已显示出有希望的结果。然而,将这些化学治疗剂依次递送至TNBC肿瘤具有挑战性.我们旨在通过紫杉醇和氟尿嘧啶的序贯给药探索TNBC的精确治疗策略。
    方法:我们开发了一种双重化学负载的适体,该适体具有对氧化还原敏感的笼式紫杉醇,用于快速释放,而不可裂解的笼式氟尿嘧啶用于缓慢释放。使用酶连接的寡核苷酸测定和表面等离子体共振测定验证对靶蛋白的结合亲和力。使用流式细胞术测定和共聚焦显微镜测定证实了进入肿瘤的靶向和内化能力。通过体外和体内药理学研究评估了对TNBC进展的抑制作用。
    结果:合成了各种氧化还原响应性适体-紫杉醇缀合物。其中,具有硫醚接头(ASP)的AS1411-紫杉醇缀合物对TNBC细胞具有较高的抗增殖能力,并通过氟尿嘧啶修饰进一步提高了其靶向能力。具有硫醚接头(FASP)的氟尿嘧啶修饰的AS1411-紫杉醇缀合物表现出对TNBC细胞的有效靶向,并且在体外和体内显著改善对TNBC进展的抑制作用。
    结论:本研究成功开发了具有硫醚接头的氟尿嘧啶修饰的AS1411-紫杉醇偶联物,用于TNBC的靶向联合化疗。这些缀合物证明了对TNBC细胞的有效识别,实现紫杉醇和氟尿嘧啶的靶向递送和控释。这种方法导致协同抗肿瘤作用和降低的体内毒性。然而,与稳定有关的挑战,免疫原性,和可扩展性需要进一步研究未来的翻译应用程序。
    BACKGROUND: Triple-negative breast cancer (TNBC) is a recurrent, heterogeneous, and invasive form of breast cancer. The treatment of TNBC patients with paclitaxel and fluorouracil in a sequential manner has shown promising outcomes. However, it is challenging to deliver these chemotherapeutic agents sequentially to TNBC tumors. We aim to explore a precision therapy strategy for TNBC through the sequential delivery of paclitaxel and fluorouracil.
    METHODS: We developed a dual chemo-loaded aptamer with redox-sensitive caged paclitaxel for rapid release and non-cleavable caged fluorouracil for slow release. The binding affinity to the target protein was validated using Enzyme-linked oligonucleotide assays and Surface plasmon resonance assays. The targeting and internalization abilities into tumors were confirmed using Flow cytometry assays and Confocal microscopy assays. The inhibitory effects on TNBC progression were evaluated by pharmacological studies in vitro and in vivo.
    RESULTS: Various redox-responsive aptamer-paclitaxel conjugates were synthesized. Among them, AS1411-paclitaxel conjugate with a thioether linker (ASP) exhibited high anti-proliferation ability against TNBC cells, and its targeting ability was further improved through fluorouracil modification. The fluorouracil modified AS1411-paclitaxel conjugate with a thioether linker (FASP) exhibited effective targeting of TNBC cells and significantly improved the inhibitory effects on TNBC progression in vitro and in vivo.
    CONCLUSIONS: This study successfully developed fluorouracil-modified AS1411-paclitaxel conjugates with a thioether linker for targeted combination chemotherapy in TNBC. These conjugates demonstrated efficient recognition of TNBC cells, enabling targeted delivery and controlled release of paclitaxel and fluorouracil. This approach resulted in synergistic antitumor effects and reduced toxicity in vivo. However, challenges related to stability, immunogenicity, and scalability need to be further investigated for future translational applications.
    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
    开发了一种用于细胞内miRNA成像和自给自足饥饿疗法(ST)和化学动力学疗法(CDT)的联合治疗的癌症靶向谷胱甘肽(GSH)门控治疗探针(CGT探针)。CGT探针是使用MnO2纳米片(MS)作为载体材料构建的,以吸附精心设计的功能DNA。它可以通过AS1411适体和核仁素之间的特异性识别被癌细胞内化。CGT探针进入癌细胞后,过度表达的GSH,作为栅极控制,可以通过类Fenton反应将MS降解为可用于CDT的Mn2。同时,Mn2+介导的CDT可以进一步与CGT探针的酶样活性(过氧化氢酶样活性和葡萄糖氧化酶样活性)级联,实现自给自足的ST/CDT协同治疗。同时,锚定的DNA被释放,通过miR-21的双取代催化发夹组装(DCHA)和FRET(荧光共振能量转移)成像实现原位信号放大。体外和体内实验表明,使用CGT探针可以实现准确和灵敏的miRNA检测。总的来说,巧妙的CGT探针为早期临床诊断和癌症治疗的发展开辟了新途径。
    A cancer-targeted glutathione (GSH)-gated theranostic probe (CGT probe) for intracellular miRNA imaging and combined treatment of self-sufficient starvation therapy (ST) and chemodynamic therapy (CDT) was developed. The CGT probe is constructed using MnO2 nanosheet (MS) as carrier material to adsorb the elaborately designed functional DNAs. It can be internalized by cancer cells via specific recognition between the AS1411 aptamer and nucleolin. After CGT probe entering the cancer cells, the overexpressed GSH, as gate-control, can degrade MS to Mn2+ which can be used for CDT by Fenton-like reaction. Simultaneously, Mn2+-mediated CDT can further cascade with the enzyme-like activities (catalase-like activity and glucose oxidase-like activity) of CGT probe, achieving self-sufficient ST/CDT synergistic therapy. Meanwhile, the anchored DNAs are released, achieving in situ signal amplification via disubstituted-catalytic hairpin assembly (DCHA) and FRET (fluorescence resonance energy transfer) imaging of miR-21. The in vitro and in vivo experiments demonstrated that accurate and sensitive miRNA detection can be achieved using the CGT probe. Overall, the ingenious CGT probe opens a new avenue for the development of early clinical diagnosis and cancer therapy.
    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
    近几十年来,肿瘤免疫治疗取得了显著进展。然而,治疗干预的临床结果仍然不可预测,很大程度上是因为低效的免疫反应。为了应对这一挑战并优化免疫刺激,我们提出了一种提高免疫治疗药物生物利用度的新给药途径。我们的方法涉及利用源自乙型肝炎病毒核心(HBc)抗原的病毒样颗粒开发口腔肿瘤疫苗。这些颗粒的外表面经过工程改造以显示模型肿瘤抗原OVA,而内部装有胞嘧啶磷酸鸟苷寡脱氧核苷酸(CpGODN),产生称为CpG@OVAHBc的构建体,具有增强的抗原性和免疫应答。对于口服递送,CpG@OVAHBc封装在称为CpG@OVAHBc@Dex的交联葡聚糖水凝胶中。外部水凝胶屏蔽保护仿生病毒颗粒免受胃酸和蛋白酶的降解。暴露于肠道菌群后,水凝胶崩解,在肠粘膜部位释放CpG@OVAHBc。由于它的病毒样结构,CpG@OVAHBc表现出对粘膜表面的粘附增强,促进微折叠细胞(M细胞)的摄取和随后传递给抗原呈递细胞。这种口服水凝胶的酶触发释放确保了肿瘤疫苗在消化道内的完整性,允许靶向释放并显着提高生物利用度。除了它的功效,这种口服水凝胶疫苗简化了药物管理,减轻患者的不适,并提高治疗依从性,而不需要专门的注射方法。因此,我们的方法拓展了口服药物领域疫苗开发的视野.
    Remarkable progress has been made in tumour immunotherapy in recent decades. However, the clinical outcomes of therapeutic interventions remain unpredictable, largely because of inefficient immune responses. To address this challenge and optimise immune stimulation, we present a novel administration route for enhancing the bioavailability of immunotherapeutic drugs. Our approach involves the development of an oral tumour vaccine utilising virus-like particles derived from the Hepatitis B virus core (HBc) antigen. The external surfaces of these particles are engineered to display the model tumour antigen OVA, whereas the interiors are loaded with cytosine phosphoguanosine oligodeoxynucleotide (CpG ODN), resulting in a construct called CpG@OVAHBc with enhanced antigenicity and immune response. For oral delivery, CpG@OVAHBc is encapsulated in a crosslinked dextran hydrogel called CpG@OVAHBc@Dex. The external hydrogel shield safeguards the biomimetic virus particles from degradation by gastric acid and proteases. Upon exposure to intestinal flora, the hydrogel disintegrates, releasing CpG@OVAHBc at the intestinal mucosal site. Owing to its virus-like structure, CpG@OVAHBc exhibits enhanced adhesion to the mucosal surface, facilitating uptake by microfold cells (M cells) and subsequent transmission to antigen-presenting cells. The enzyme-triggered release of this oral hydrogel ensures the integrity of the tumour vaccine within the digestive tract, allowing targeted release and significantly improving bioavailability. Beyond its efficacy, this oral hydrogel vaccine streamlines drug administration, alleviates patient discomfort, and enhances treatment compliance without the need for specialised injection methods. Consequently, our approach expands the horizons of vaccine development in the field of oral drug administration.
    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
    肝细胞生长因子受体(c-Met)是肿瘤靶向治疗的合适分子靶点。由于常规的c-Met小分子抑制剂和抗体具有一定的局限性,需要开发新的c-Met靶向药物。为了合成这些药物,我们开发了一种双特异性DNA纳米连接器(STPA)来抑制c-Met功能。STPA是通过使用DNA三角棱镜作为支架和适体作为结合分子来构建的。在c-Met特异性SL1和核仁素特异性AS1411适体与STPA整合后,STPA可以与细胞膜上的c-Met和核仁素结合。这导致了c-Met/STPA/核仁素复合物的形成,这反过来又阻止了c-Met激活。体外实验表明,STPA不仅可以抑制c-Met信号通路,而且可以通过溶酶体促进c-Met降解。STPA还抑制c-Met促进细胞迁移,入侵,和扩散。体内实验结果表明,在异种移植小鼠模型中,STPA能特异性靶向肿瘤部位,并通过下调c-Met途径以低毒性抑制肿瘤生长。本研究为开发用于癌症靶向治疗的c-Met靶向药物提供了一种新颖而简单的策略。
    Hepatocyte growth factor receptor (c-Met) is a suitable molecular target for the targeted therapy of cancer. Novel c-Met-targeting drugs need to be developed because conventional small-molecule inhibitors and antibodies of c-Met have some limitations. To synthesize such drugs, we developed a bispecific DNA nanoconnector (STPA) to inhibit c-Met function. STPA was constructed by using DNA triangular prism as a scaffold and aptamers as binding molecules. After c-Met-specific SL1 and nucleolin-specific AS1411 aptamers were integrated with STPA, STPA could bind to c-Met and nucleolin on the cell membrane. This led to the formation of the c-Met/STPA/nucleolin complex, which in turn blocked c-Met activation. In vitro experiments showed that STPA could not only inhibit the c-Met signaling pathways but also facilitate c-Met degradation through lysosomes. STPA also inhibited c-Met-promoted cell migration, invasion, and proliferation. The results of in vivo experiments showed that STPA could specifically target to tumor site in xenograft mouse model, and inhibit tumor growth with low toxicity by downregulating c-Met pathways. This study provided a novel and simple strategy to develop c-Met-targeting drugs for the targeted therapy of 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
    同时递送CpG寡核苷酸与短干扰RNA(siRNA)具有显著增强siRNA药物的抗癌影响的潜力。我们先前的研究表明,用腺苷官能化的Curdlan纳米颗粒能够通过腺苷受体介导的内吞作用选择性地将治疗性siRNA递送至癌细胞。在这里,我们合成了双配体官能化的Curdlan聚合物(由CuMAN表示)以同时靶向肿瘤细胞和肿瘤相关巨噬细胞(TAMs)。含有CpG和siRNA的CuMAN纳米颗粒显示B16F10肿瘤细胞和骨髓源性巨噬细胞的摄取增强,由肿瘤细胞上的AR和巨噬细胞上的甘露糖受体促进。这导致促炎细胞因子在体外和体内设置中的释放增加。CuMAN纳米颗粒介导的CpG对TAM和RNAi对肿瘤细胞的协同作用不仅抑制了肿瘤的生长,而且强烈抑制了肺转移。我们的发现表明CuMAN纳米颗粒具有作为核酸治疗剂的有效双靶向递送系统的潜力。
    The simultaneous delivery of CpG oligonucleotide along with short interfering RNA (siRNA) has the potential to significantly boost the anticancer impact of siRNA medications. Our previous research demonstrated that Curdlan nanoparticles functionalized with adenosine are capable of selectively delivering therapeutic siRNA to cancerous cells through endocytosis mediated by adenosine receptors. Herein, we synthesized a dual-ligand-functionalized Curdlan polymer (denoted by CuMAN) to simultaneously target tumor cells and tumor-associated macrophages (TAMs). CuMAN nanoparticles containing CpG and siRNA demonstrated enhanced uptake by B16F10 tumor cells and bone marrow-derived macrophages, which are facilitated by AR on tumor cells and mannose receptor on macrophages. This led to increased release of pro-inflammatory cytokines in both in vitro and in vivo settings. The synergistic effect of CpG on TAMs and RNAi on tumor cells mediated by the CuMAN nanoparticle not only suppressed the tumor growth but also strongly inhibited the lung metastasis. Our findings indicate that the CuMAN nanoparticle has potential as an effective dual-targeting delivery system for nucleic acid therapeutics.
    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
    结肠癌的侵袭性和当前不精确的治疗方案模拟了精确有效的治疗策略的发展。为了实现这一点,肿瘤环境激活的光敏化仿生纳米平台(PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM)是通过将负载有开发的光敏剂PEG2000-SiNcTI-Ph和免疫佐剂CpG寡脱氧核苷酸的金属有机框架封装在表达程序性死亡蛋白1(PD-1)和分化簇47(CD47)的融合细胞膜内。通过绊倒,系统评价,用量子化学计算破译,通过PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM验证了肿瘤环境的独特属性(低pH加上高浓度的5'-三磷酸腺苷(ATP))激活的由长波长光子敏感的光动力效应,提高癌症治疗的精确度。此外,PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM逃避了CD47/信号调节蛋白α(SIRPα)相互作用和PD-1/程序性死亡配体1(PD-L1)相互作用介导的小鼠靶向CT26结肠肿瘤的免疫监视,分别。PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM实现的肿瘤环境激活光动力疗法诱导免疫原性细胞死亡(ICD)引发抗肿瘤免疫反应,这是由增强的树突状细胞(DC)摄取CpG和PD-L1阻断由纳米平台贡献。光动力免疫疗法可有效对抗原发性和远处CT26肿瘤,并产生免疫记忆以抑制肿瘤复发和转移。这里开发的纳米平台为开发精确的癌症治疗策略提供了见解。
    Aggressive nature of colon cancer and current imprecise therapeutic scenarios simulate the development of precise and effective treatment strategies. To achieve this, a tumor environment-activated photosensitized biomimetic nanoplatform (PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM) is fabricated by encapsulating metal-organic framework loaded with developed photosensitizer PEG2000-SiNcTI-Ph and immunoadjuvant CpG oligodeoxynucleotide within fusion cell membrane expressing programmed death protein 1 (PD-1) and cluster of differentiation 47 (CD47). By stumbling across, systematic evaluation, and deciphering with quantum chemical calculations, a unique attribute of tumor environment (low pH plus high concentrations of adenosine 5\'-triphosphate (ATP))-activated photodynamic effect sensitized by long-wavelength photons is validated for PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM, advancing the precision of cancer therapy. Moreover, PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM evades immune surveillance to target CT26 colon tumors in mice mediated by CD47/signal regulatory proteins α (SIRPα) interaction and PD-1/programmed death ligand 1 (PD-L1) interaction, respectively. Tumor environment-activated photodynamic therapy realized by PEG2000-SiNcTI-Ph/CpG-ZIF-8@CM induces immunogenic cell death (ICD) to elicit anti-tumor immune response, which is empowered by enhanced dendritic cells (DC) uptake of CpG and PD-L1 blockade contributed by the nanoplatform. The photodynamic immunotherapy efficiently combats primary and distant CT26 tumors, and additionally generates immune memory to inhibit tumor recurrence and metastasis. The nanoplatform developed here provides insights for the development of precise cancer therapeutic strategies.
    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
    作为一种高度传染性的机会病原体,铜绿假单胞菌(P.铜绿假单胞菌)是医疗保健相关感染的主要原因之一。铜绿假单胞菌的耐药性可使抗生素治疗无效,导致高发病率和死亡率。更高的特异性和更低的毒性是免疫治疗的特征。这可以产生强大的免疫反应,并保持长期的免疫记忆,以彻底根除感染。在这项研究中,我们开发了一种基于金属有机框架的铜绿假单胞菌疫苗。具体来说,合成了MIL-101-Al纳米颗粒,以封装源自PAO1细菌裂解物(BL)的抗原,PAO1是一种耐药的铜绿假单胞菌,和佐剂非甲基化胞嘧啶-磷酸-鸟嘌呤寡核苷酸(CpG),然后用棕榈酸(PAA)修饰得到MIL-BC@PAA。PAA封端的稳定性和生物相容性显著提高。此外,MIL-BC@PAA显示抗原呈递细胞(APC)的摄取显着增强,促进了他们的成熟。重要的是,免疫研究揭示了极大地引发的抗原特异性体液和细胞反应,在铜绿假单胞菌攻击的小鼠中观察到约70%的保护率。总的来说,这些结果证明了MIL-BC@PAA作为用于铜绿假单胞菌疫苗接种的理想纳米疫苗的有希望的潜力。
    As a highly contagious opportunistic pathogen, Pseudomonas aeruginosa (P. aeruginosa) is one of the main causes of healthcare-associated infections. The drug-resistant nature of P. aeruginosa can render antibiotic treatments ineffective, leading to a high morbidity and mortality. Higher specificity and reduced toxicity are features of immunotherapy, which can generate robust immune responses and preserve long-term immunological memory to completely eradicate infections. In this study, we developed a type of P. aeruginosa vaccine based on a metal-organic framework. Specifically, MIL-101-Al nanoparticles were synthesized to encapsulate antigens derived from the bacterial lysate (BL) of PAO1, a drug-resistant P. aeruginosa, and the adjuvant unmethylated cytosine-phosphate-guanine oligonucleotide (CpG), which were then modified with palmitic acid (PAA) to obtain MIL-BC@PAA. The stability and biocompatibility were significantly increased by capping with PAA. Moreover, MIL-BC@PAA showed significantly enhanced uptake by antigen presenting cells (APCs), and promoted their maturation. Importantly, immunity studies revealed the greatly elicited antigen-specific humoral and cellular responses, and a protection rate of about 70% was observed in P. aeruginosa-challenged mice. Overall, these results demonstrate the promising potential of MIL-BC@PAA as an ideal nanovaccine for P. aeruginosa vaccination.
    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
    胶质瘤细胞与星形胶质细胞之间的相互作用促进胶质瘤的增殖。星形胶质细胞外泌体(exos)携带的micro-RNAs(miRNAs)可能参与这一过程,但机制尚不清楚。寡核苷酸AS1411由26个碱基组成,具有G-四链体结构,是一种靶向核仁素的适体。在这项研究中,我们证明了外泌体-miRNA-27a介导的星形胶质细胞和胶质母细胞瘤之间的交叉激活,并表明AS1411降低了星形胶质细胞前胶质瘤的活性。AS1411对核仁素的亲和力增强归因于其G-四链体结构。与核仁素结合后,AS1411抑制NF-κB通路转录因子P65进入细胞核,然后下调胶质瘤周围星形胶质细胞中miRNA-27a的表达。然后,AS1411下调星形胶质细胞外泌体-miRNA-27a并上调INPP4B的表达,胶质瘤中miRNA-27a的靶基因,从而抑制PI3K/AKT途径并抑制神经胶质瘤增殖。这些结果在小鼠原位神经胶质瘤异种移植物和人神经胶质瘤样品中得到证实。总之,AS1411的平行结构使其能够与核仁素结合并破坏外泌体-miRNA-27a介导的神经胶质瘤细胞和星形胶质细胞之间的相互激活环.我们的结果可能有助于开发一种治疗性调节神经胶质瘤微环境的新方法。
    The interaction between glioma cells and astrocytes promotes the proliferation of gliomas. Micro-RNAs (miRNAs) carried by astrocyte exosomes (exos) may be involved in this process, but the mechanism remains unclear. The oligonucleotide AS1411, which consists of 26 bases and has a G-quadruplex structure, is an aptamer that targets nucleolin. In this study, we demonstrate exosome-miRNA-27a-mediated cross-activation between astrocytes and glioblastoma and show that AS1411 reduces astrocytes\' pro-glioma activity. The enhanced affinity of AS1411 toward nucleolin is attributed to its G-quadruplex structure. After binding to nucleolin, AS1411 inhibits the entry of the NF-κB pathway transcription factor P65 into the nucleus, then downregulates the expression of miRNA-27a in astrocytes surrounding gliomas. Then, AS1411 downregulates astrocyte exosome-miRNA-27a and upregulates the expression of INPP4B, the target gene of miRNA-27a in gliomas, thereby inhibiting the PI3K/AKT pathway and inhibiting glioma proliferation. These results were verified in mouse orthotopic glioma xenografts and human glioma samples. In conclusion, the parallel structure of AS1411 allows it to bind to nucleolin and disrupt the exosome-miRNA-27a-mediated reciprocal activation loop between glioma cells and astrocytes. Our results may help in the development of a novel approach to therapeutic modulation of the glioma microenvironment.
    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
    蛋白水解靶向嵌合体(PROTACs)是由两个通过接头连接的配体组成的异双功能分子,使它们能够同时与E3连接酶和目的蛋白(POI)结合,并引发POI的蛋白酶体降解。PROTAC的局限性包括缺乏有效的E3配体,细胞选择性差,和低渗透性。AS1411是特异性识别膜核穿梭核仁素(NCL)的抗肿瘤适体。这里,我们通过锚定NCL-MDM2复合物,将AS1411重新用作E3连接酶小鼠双分2同源物(MDM2)的配体。然后,我们构建了一个基于AS1411-NCL-MDM2的PROTAC(ANM-PROTAC)通过将AS1411与大分子量配体结合,用于“不可药用的”致癌STAT3,c-Myc,p53-R175H,AR-V7我们证明ANM-PROTAC有效地穿透肿瘤细胞,招募MDM2并降低POI。ANM-PROTAC实现了肿瘤选择性分布并表现出优异的抗肿瘤活性而没有全身毒性。这是一种具有内置肿瘤靶向和细胞穿透能力的PROTAC。
    Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules consisting of two ligands joined by a linker, enabling them to simultaneously bind with an E3 ligase and a protein of interest (POI) and trigger proteasomal degradation of the POI. Limitations of PROTAC include lack of potent E3 ligands, poor cell selectivity, and low permeability. AS1411 is an antitumor aptamer specifically recognizing a membrane-nucleus shuttling nucleolin (NCL). Here, we repurpose AS1411 as a ligand for an E3 ligase mouse double minute 2 homolog (MDM2) via anchoring the NCL-MDM2 complex. Then, we construct an AS1411-NCL-MDM2-based PROTAC (ANM-PROTAC) by conjugating AS1411 with large-molecular-weight ligands for \"undruggable\" oncogenic STAT3, c-Myc, p53-R175H, and AR-V7. We show that the ANM-PROTAC efficiently penetrates tumor cells, recruits MDM2 and degrades the POIs. The ANM-PROTAC achieves tumor-selective distribution and exhibits excellent antitumor activity with no systemic toxicity. This is a PROTAC with built-in tumor-targeting and cell-penetrating capacities.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号