Tetrahedral framework nucleic acid

四面体骨架核酸
  • 文章类型: Journal Article
    胰腺癌,主要是胰腺导管腺癌(PDAC),仍然是一种高度致命的恶性肿瘤,治疗选择有限,预后不佳。通过靶向导致PDAC发育和进展的潜在分子异常,基因治疗为克服常规放疗和化疗带来的挑战提供了一个有希望的策略.这项研究旨在探索专门针对PDAC中CCAAT/增强子结合蛋白α(CEBPA)基因的小激活RNA(saRNA)的治疗潜力。为了克服与saRNA递送相关的挑战,四面体框架核酸(tFNA)被合理地设计为纳米载体。用截短的运铁蛋白受体适体(tTR14)进一步官能化这些tFNA以增强对PDAC细胞的靶向特异性。构建的基于tFNA的saRNA制剂表现出卓越的稳定性,高效的saRNA释放能力,大量的细胞摄取,生物相容性,和无毒。体外实验显示,利用tTR14修饰的tFNA纳米载体成功地在细胞内递送CEBPA-saRNA,导致抑癌基因的显著激活,即,CEBPA及其下游效应子P21,导致PDAC细胞增殖的显著抑制。此外,在PDAC的小鼠模型中,tTR14修饰的tFNA介导的CEBPA-saRNA的递送有效地上调了CEBPA和P21基因的表达,从而抑制肿瘤生长。这些令人信服的发现强调了通过设计的tFNA纳米载体递送的saRNA作为PDAC的创新治疗方法诱导肿瘤抑制基因激活的潜在效用。
    Pancreatic cancer, predominantly pancreatic ductal adenocarcinoma (PDAC), remains a highly lethal malignancy with limited therapeutic options and a dismal prognosis. By targeting the underlying molecular abnormalities responsible for PDAC development and progression, gene therapy offers a promising strategy to overcome the challenges posed by conventional radiotherapy and chemotherapy. This study sought to explore the therapeutic potential of small activating RNAs (saRNAs) specifically targeting the CCAAT/enhancer-binding protein alpha (CEBPA) gene in PDAC. To overcome the challenges associated with saRNA delivery, tetrahedral framework nucleic acids (tFNAs) were rationally engineered as nanocarriers. These tFNAs were further functionalized with a truncated transferrin receptor aptamer (tTR14) to enhance targeting specificity for PDAC cells. The constructed tFNA-based saRNA formulation demonstrated exceptional stability, efficient saRNA release ability, substantial cellular uptake, biocompatibility, and nontoxicity. In vitro experiments revealed successful intracellular delivery of CEBPA-saRNA utilizing tTR14-decorated tFNA nanocarriers, resulting in significant activation of tumor suppressor genes, namely, CEBPA and its downstream effector P21, leading to notable inhibition of PDAC cell proliferation. Moreover, in a mouse model of PDAC, the tTR14-decorated tFNA-mediated delivery of CEBPA-saRNA effectively upregulated the expression of the CEBPA and P21 genes, consequently suppressing tumor growth. These compelling findings highlight the potential utility of saRNA delivered via a designed tFNA nanocarrier to induce the activation of tumor suppressor genes as an innovative therapeutic approach for PDAC.
    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
    脉络膜新生血管(CNV)是多种眼部疾病的共同病理特征,是中老年患者视力损害的重要原因。在以往的研究中,四面体骨架核酸(tFNA)表现出良好的载体性能。在这个实验中,我们开发了配备microRNA-155的tFNA(T-155),并探索了其对CNV的生物学效应。根据体外实验的结果,T-155可以调节巨噬细胞进入抗血管生成M1型。然后,我们将T-155注射到激光诱导的CNV模型小鼠的玻璃体中,发现T-155显着减小了CNV的大小和面积,抑制血管渗漏。总之,我们证明T-155可以通过极化巨噬细胞调节CNV的炎症过程,从而改善CNV的症状。因此,T-155可能成为一种新的基于DNA的药物,具有治疗CNV的巨大潜力。
    Choroidal neovascularization (CNV) is a common pathological feature of various eye diseases and an important cause of visual impairment in middle-aged and elderly patients. In previous studies, tetrahedral framework nucleic acids (tFNAs) showed good carrier performance. In this experiment, we developed microRNA-155-equipped tFNAs (T-155) and explored its biological effects on CNV. Based on the results of in-vitro experiments, T-155 could regulate macrophages into the antiangiogenic M1 type. Then, we injected T-155 into the vitreous of laser-induced CNV model mice and found that T-155 significantly reduced the size and area of CNV, inhibited blood vessel leakage. In summary, we prove that T-155 could regulate the inflammatory process of CNV by polarizing macrophages, thereby improving the symptoms of CNV. Thus, T-155 might become a new DNA-based drug with great potential for treating CNV.
    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
    四面体骨架核酸(tFNA),一种特殊的DNA纳米装置,广泛应用于各种生物医学领域。由于其高可编程性,生物相容性,组织通透性及其细胞增殖和分化的能力,tFNA提供了一种强大的工具,可以克服治疗神经系统疾病的潜在障碍。本文综述了基于tFNA的纳米材料在神经系统疾病中的应用和进展的最新研究。
    Tetrahedral framework nucleic acid (tFNA), a special DNA nanodevice, is widely applied in diverse biomedical fields. Due to its high programmability, biocompatibility, tissue permeability as well as its capacity for cell proliferation and differentiation, tFNA presents a powerful tool that could overcome potential barriers in the treatment of neurological disorders. This review evaluates recent studies on the use and progress of tFNA-based nanomaterials in neurological 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
    目的:自身免疫性疾病是一组对自身抗原失去免疫耐受的异质性疾病。众所周知,不规则激发的T细胞参与病理性免疫应答。作为一种具有广阔应用前景的新型纳米材料,研究发现四面体骨架核酸(TFNA)纳米结构对T细胞具有免疫调节作用。
    方法:为了验证TFNA的成功制造,通过原子力显微镜(AFM)和动态光散射观察TFNA的形貌。在与从健康供体分离的CD3+T细胞共培养后,通过流式细胞术评估TFNA的调节作用。此外,研究了相关的信号通路。最后,我们在视神经脊髓炎谱系障碍(NMOSD)患者的T细胞上验证了我们的结果,这是一种典型的由T细胞诱导的自身免疫性疾病。
    结果:我们通过JNK信号通路揭示了TFNA在稳定状态的人原代T细胞中的替代调节功能。此外,通过抑制JNK和ERK磷酸化,TFNA对来自激发T细胞的IFNγ分泌表现出显著的抑制作用,而不影响TNF分泌。在NMOSD患者的自身反应性T细胞中也观察到TFNA的类似免疫调节作用。
    结论:总体而言,我们的结果揭示了TFNA在调节适应性免疫系统中的潜在应用,以及阐明T细胞介导的自身免疫性疾病的治疗。
    OBJECTIVE: Autoimmune diseases are a heterogeneous group of diseases which lose the immunological tolerance to self-antigens. It is well recognized that irregularly provoked T cells participate in the pathological immune responses. As a novel nanomaterial with promising applications, tetrahedral framework nucleic acid (TFNA) nanostructure was found to have immune regulatory effects on T cells in this study.
    METHODS: To verify the successful fabrication of TFNA, the morphology of TFNA was observed by atomic force microscopy (AFM) and dynamic light scattering. The regulatory effect of TFNA was evaluated by flow cytometry after cocultured with CD3+ T cells isolated from healthy donors. Moreover, the associated signaling pathways were investigated. Finally, we verified our results on the T cells from patients with neuromyelitis optica spectrum disorder (NMOSD), which is a typical autoimmune disease induced by T cells.
    RESULTS: We revealed the alternative regulatory functions of TFNA in human primary T cells with steady status via the JNK signaling pathway. Moreover, by inhibiting both JNK and ERK phosphorylation, TFNA exhibited significant suppressive effects on IFNγ secretion from provoking T cells without affecting TNF secretion. Similar immune regulatory effects of TFNA were also observed in autoreactive T cells from patients with NMOSD.
    CONCLUSIONS: Overall, our results revealed a potential application of TFNA in regulating the adaptive immune system, as well as shed a light on the treatment of T cell-mediated autoimmune diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    肥胖诱导的胰岛素抵抗是代谢综合征的标志,慢性,低度组织炎症通过激活组织浸润的免疫细胞将肥胖与胰岛素抵抗联系起来.目前的治疗方法缺乏功效和免疫调节能力。因此,需要一种新的治疗方法来预防慢性炎症和减轻胰岛素抵抗.这里,我们合成了携带白藜芦醇(RSV)的四面体骨架核酸(tFNA)纳米颗粒,以抑制肥胖小鼠的组织炎症并改善胰岛素敏感性.所制备的纳米粒子,即TFNA-RSV,具有简单合成的特点,性能稳定,良好的水溶性,和优越的生物相容性。基于tFNA的递送改善了RSV的不稳定性并增强了其治疗功效。在高脂饮食(HFD)喂养的小鼠中,tFNA-RSV的给药通过减轻炎症状态来改善胰岛素抵抗。tFNAs-RSV可以将组织中M1表型巨噬细胞逆转为M2表型巨噬细胞。至于适应性免疫,制备的纳米粒子可以抑制Th1和Th17的活化,促进Th2和Treg,导致胰岛素抵抗的缓解。此外,这项研究是第一个证明tFNA,核酸物质,具有免疫调节能力。总的来说,我们的研究结果表明,tFNAs-RSV减轻胰岛素抵抗和改善HFD小鼠的炎症,这表明核酸物质或基于核酸的递送系统可能是治疗胰岛素抵抗和肥胖相关代谢性疾病的潜在药物。
    Obesity-induced insulin resistance is the hallmark of metabolic syndrome, and chronic, low-grade tissue inflammation links obesity to insulin resistance through the activation of tissue-infiltrating immune cells. Current therapeutic approaches lack efficacy and immunomodulatory capacity. Thus, a new therapeutic approach is needed to prevent chronic inflammation and alleviate insulin resistance. Here, we synthesized a tetrahedral framework nucleic acid (tFNA) nanoparticle that carried resveratrol (RSV) to inhibit tissue inflammation and improve insulin sensitivity in obese mice. The prepared nanoparticles, namely tFNAs-RSV, possessed the characteristics of simple synthesis, stable properties, good water solubility, and superior biocompatibility. The tFNA-based delivery ameliorated the lability of RSV and enhanced its therapeutic efficacy. In high-fat diet (HFD)-fed mice, the administration of tFNAs-RSV ameliorated insulin resistance by alleviating inflammation status. tFNAs-RSV could reverse M1 phenotype macrophages in tissues to M2 phenotype macrophages. As for adaptive immunity, the prepared nanoparticles could repress the activation of Th1 and Th17 and promote Th2 and Treg, leading to the alleviation of insulin resistance. Furthermore, this study is the first to demonstrate that tFNAs, a nucleic acid material, possess immunomodulatory capacity. Collectively, our findings demonstrate that tFNAs-RSV alleviate insulin resistance and ameliorate inflammation in HFD mice, suggesting that nucleic acid materials or nucleic acid-based delivery systems may be a potential agent for the treatment of insulin resistance and obesity-related metabolic diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    在寻找大面积软组织和硬组织缺陷的解决方案时,是否使用组织再生或组织替代品移植,血管生成的问题迫切需要解决。因此,需要一种新的有效的促血管生成方法.纳米工程系统被认为是最有前途的方法之一。在这项研究中,我们首次用两种不同的血管生成DNA适体修饰四面体框架核酸(tFNA),形成适体-tFNA纳米结构,tFNA-Apt02和tFNA-AptVEGF,并研究了它们在体外和体内对血管生成的影响。我们开发新的纳米材料用于增强血管生成,以解决组织工程血管化和缺血性疾病的问题。我们的研究结果证实,tFNA-Apt02和tFNA-AptVEGF具有更强的加速内皮细胞增殖和迁移的能力,小管形成,球状体发芽,和体内血管生成。我们首先证明了工程化的新型tFNA-Apt02和tFNA-AptVEGF在体外和体内对血管生成的促进作用,并为它们在组织工程血管化和缺血性疾病中的应用提供了理论基础和机会。
    In a search for a solution to large-area soft and hard tissue defects, whether or not tissue regeneration or tissue-substitutes transplantation is used, the problems with angiogenesis need to be solved urgently. Thus, a new and efficient proangiogenic approach is needed. Nanoengineering systems have been considered one of the most promising approaches. In this study, we modify the tetrahedral framework nucleic acid (tFNA) for the first time with two different angiogenic DNA aptamers to form aptamer-tFNA nanostructures, tFNA-Apt02 and tFNA-AptVEGF, and the effects of them on angiogenesis both in vitro and in vivo are investigated. We develop new nanomaterials for enhancing angiogenesis to solve the problem of tissue engineering vascularization and ischemic diseases. The results of our study confirm that tFNA-Apt02 and tFNA-AptVEGF has a stronger ability to accelerate endothelial cell proliferation and migration, tubule formation, spheroid sprouting, and angiogenesis in vivo. We first demonstrate that the engineered novel tFNA-Apt02 and tFNA-AptVEGF have promoting effects on angiogenesis both in vitro and in vivo and provide a theoretical basis and opportunity for their application in tissues engineering vascularization and ischemic diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    化疗药物的穿透性差和非选择性分布是三阴性乳腺癌(TNBC)化疗的主要障碍。在我们的工作中,我们开发了基于DNA的药物递送系统来克服这些障碍。此外,采用四面体框架核酸(tFNA)负载具有iRGD修饰的多柔比星(DOX)以形成新型纳米颗粒(tFNA/DOX@iRGD)。iRGD中的RGD序列和CendR基序用于肿瘤靶向和组织穿透,分别。基于DOX的持续血清稳定性和pH敏感释放行为,tFNA/DOX@iRGD在生物医学应用中表现出优越性。此外,与DOX和tFNA/DOX相比,tFNA/DOX@iRGD在三维(3D)多细胞肿瘤球体中显示出出色的深度渗透和药物积累。此外,在4T1皮下肿瘤模型中验证了治疗效果,复合物显示出优异的抗肿瘤和抗血管生成效率,而附带损害较少。因此,这些研究结果表明,tFNA/DOX@iRGD可能是一种更有效的给药和TNBC治疗模式.
    Poor penetrability and nonselective distribution of chemotherapeutic drugs are the main obstacles for chemotherapy for triple-negative breast cancer (TNBC). In our work, we developed a DNA-based drug delivery system to surmount these barriers. In addition, a tetrahedral framework nucleic acid (tFNA) was employed to load doxorubicin (DOX) with iRGD decoration to form a novel nanoparticle (tFNA/DOX@iRGD). The RGD sequence and the CendR motif in iRGD are used in tumor targeting and tissue penetration, respectively. Based on the sustained serum stability and pH-sensitive release behavior of DOX, tFNA/DOX@iRGD exhibited superiority for biomedical application. Moreover, tFNA/DOX@iRGD showed excellent deep penetration and drug accumulation in three-dimensional (3D) multicellular tumor spheroids compared to DOX and tFNA/DOX. Additionally, the therapeutic effect was verified in a 4T1 subcutaneous tumor model, and the complexes displayed a superior antitumor and antiangiogenic efficiency with fewer collateral damages. Therefore, these findings suggested that tFNA/DOX@iRGD might be a more effective pattern for drug delivery and TNBC therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    骨关节炎(OA)是伴随着软骨细胞过度凋亡的退行性关节软骨致病过程。软骨细胞死亡和OA的发生与自噬减少有关。四面体骨架核酸(TFNA),一种有效的生物活性DNA纳米材料,在各种疾病中发挥抗凋亡和抗氧化作用,导致自噬促进和抑制Wnt/β-catenin信号通路。这里,我们旨在阐明TFNA对OA的治疗作用及其潜在的分子作用机制。合成了TFNA,并通过建立的方法进行了表征。建立白细胞介素(IL)-1β刺激的OA细胞模型并用TFNA处理。通过免疫荧光和流式细胞术检查TFNA的细胞摄取和细胞内活性氧水平。通过细胞计数试剂盒-8(CCK8)测定和流式细胞术记录凋亡细胞死亡。应用透射电子显微镜观察自噬体。BCL2、BAX、caspase-3,Nrf2,HO-1,LC3-II,Beclin1,Atg7,β-连环蛋白,通过免疫荧光和蛋白质印迹检测Lef-1和CyclinD1。在不存在或存在IL-1β的情况下,TFNA成功合成并有效进入软骨细胞,而无需转染剂的帮助。TFNA处理IL-1β诱导的软骨细胞通过激活BCL2/BAX/caspase-3通路减少凋亡,通过调节Nrf2/HO-1信号通路抑制氧化应激,并通过上调LC3-II增强自噬,Beclin1和Atg7.此外,TFNA通过调节Wnt/β-catenin信号通路显示软骨保护作用。总的来说,TFNA可用作OA的治疗性纳米药物。
    Osteoarthritis (OA) is a degenerative articular cartilage pathogenic process that is accompanied by excessive chondrocyte apoptosis. The occurrence of chondrocyte death and OA is related to decreased autophagy. Tetrahedral framework nucleic acid (TFNA), a potent bioactive DNA nanomaterial, exerts antiapoptotic and antioxidative effects in various diseases, resulting in autophagy promotion and inhibition of the Wnt/β-catenin-signaling pathway. Here, we aimed to elucidate the therapeutic effects of TFNA on OA and its potential molecular mechanism of action. TFNA was synthesized and characterized by established methods. An interleukin (IL)-1β stimulated OA cell model was established and treated with TFNA. Cellular uptake of TFNA and intracellular reactive oxygen species levels were examined via immunofluorescence and flow cytometry. Apoptotic cell death was documented by the Cell Counting Kit-8 (CCK8) assay and flow cytometry. Transmission electron microscopy was applied to view the autophagosomes. The expression of BCL2, BAX, caspase-3, Nrf2, HO-1, LC3-II, Beclin1, Atg7, β-catenin, Lef-1, and CyclinD1 was detected by immunofluorescence and western blotting. TFNA was successfully synthesized and effectively entered chondrocytes in the absence or presence of IL-1β without the help of transfection agents. TFNA treatment in IL-1β-induced chondrocytes reduced apoptosis by activating the BCL2/BAX/caspase-3 pathway, inhibited oxidative stress by regulating the Nrf2/HO-1-signaling pathway, and enhanced autophagy through upregulated LC3-II, Beclin1, and Atg7. Moreover, TFNA showed chondroprotective effects by regulating the Wnt/β-catenin-signaling pathway. Overall, TFNA may have utility as a therapeutic nanomedicine for OA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The past decades have witnessed the development of DNA nanotechnology and the emergence of various spatial DNA nanostructures, from two-dimensions to three-dimensions. The typical example is the tetrahedral framework nucleic acid (tFNA). In this review, we summarize the progress in fabrication, modification of tFNA-based functional systems and their potentials in biomedical applications. Through a one-step assembly process, tFNA is synthesized via four single stranded DNAs with three short sequences complementary to the other sequence of another single strand. Characterizations including polyacrylamide gel electrophoresis, atomic force microscopy, and dynamic light scattering measurement show tFNA as a pyramid-like nanostructure with the size of around 10 nm. Feathered with intrinsic biocompatibility and satisfactory cellular membrane permeability, the first generation of tFNA shows promising capacities in regulating cell biological behavior, promoting tissue regeneration, and immunomodulation. Along with excellent editability and relative biostability in complicated conditions, tFNA could be modified via hanging functional domains on the vertex or side arm and incorporating small-molecular-weight drugs to form the second generation, for reversing multidrug resistance in tumor cells or microorganisms, target therapy, anticancer and antibacterial treatments. The third generation of tFNA is currently tried via a multistep assembly process for stimuli-response and precise drug release. Although tFNAs show promising potentials in cargo delivery, massive efforts still need to be made to improve biostability, maximal load, and structural controllability.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Bisphosphonates are often used to treat osteoporosis, malignant bone metastases, and hypercalcemia. However, it can cause serious adverse reactions, bisphosphonate-related osteonecrosis of the jaw (BRONJ), which seriously affects the quality of life of patients. At present, the treatment of BRONJ is still difficult to reach an agreement, and there is no effective treatment. Therefore, it is very important to find effective treatments. Many studies have shown that the occurrence of BRONJ may be due to unbalanced bone turnover, anti-angiogenesis, bacterial infection, direct tissue toxicity, and abnormal immune function. The previous research results show that tetrahedral framework nucleic acids (tFNAs), a new type of nanomaterial, can promote various biological activities of cells, such as cell proliferation, migration, anti-inflammation and anti-oxidation, and angiogenesis. Therefore, we intend to explore the potential of tFNAs in the treatment of BRONJ through this study. The results show that tFNAs can promote the treatment of BRONJ by promoting angiogenesis and promoting M2 polarization in macrophages and inhibiting M1 polarization both in vitro and in vivo. These results provide a theoretical basis for the application of tFNAs in the treatment of BRONJ and also provide new ideas and methods for the treatment of other diseases based on ischemia and immune disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号