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
    对传统抗癫痫药物的耐药性是慢性癫痫治疗的主要挑战。基于microRNA的基因治疗是一种有希望的替代方案,但由于血脑屏障通透性差,已证明疗效有限。细胞摄取,和目标效率。腺苷是由于反应性A1星形胶质细胞中腺苷激酶(ADK)活性升高而在癫痫脑中缺乏的内源性抗癫痫药。我们设计了一种基于四面体骨架核酸(tFNA)的核酸纳米抗癫痫药物(tFNA-ADKASO@AS1),携带反义寡核苷酸靶向ADK(ADKASO)和A1星形胶质细胞靶向肽(AS1)。这种tFNA-ADKASO@AS1构建体有效降低大脑ADK,大脑腺苷增加,缓解异常苔藓纤维发芽,并降低了慢性颞叶癫痫小鼠模型中反复发作的自发性癫痫尖峰频率。Further,治疗未引起任何神经毒性或主要器官损伤.这项工作为新型抗癫痫药物递送策略和内源性腺苷作为基于基因的调节的有希望的靶标提供了概念验证。
    Resistance to traditional antiepileptic drugs is a major challenge in chronic epilepsy treatment. MicroRNA-based gene therapy is a promising alternative but has demonstrated limited efficacy due to poor blood-brain barrier permeability, cellular uptake, and targeting efficiency. Adenosine is an endogenous antiseizure agent deficient in the epileptic brain due to elevated adenosine kinase (ADK) activity in reactive A1 astrocytes. We designed a nucleic acid nanoantiepileptic drug (tFNA-ADKASO@AS1) based on a tetrahedral framework nucleic acid (tFNA), carrying an antisense oligonucleotide targeting ADK (ADKASO) and A1 astrocyte-targeted peptide (AS1). This tFNA-ADKASO@AS1 construct effectively reduced brain ADK, increased brain adenosine, mitigated aberrant mossy fiber sprouting, and reduced the recurrent spontaneous epileptic spike frequency in a mouse model of chronic temporal lobe epilepsy. Further, the treatment did not induce any neurotoxicity or major organ damage. This work provides proof-of-concept for a novel antiepileptic drug delivery strategy and for endogenous adenosine as a promising target for gene-based modulation.
    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
    急性肾损伤(AKI)不仅是一个具有严重医院死亡率的世界性问题,而且是慢性肾脏病的独立危险因素和其进展的促进因素。尽管采取了支持性治疗措施,目前尚无有效的治疗方法。这项研究采用四面体框架核酸(tFNA)作为载体,并结合了伤寒苷(Typ)来开发用于治疗AKI的tFNA-Typ复合物(TTC)。具有对线粒体和肾小管的精确靶向能力,增加抗凋亡和抗氧化作用,促进线粒体和肾功能恢复,TTC代表了一种用于AKI治疗的有前途的纳米药物。总的来说,这项研究开发了一种双靶向纳米颗粒,对AKI的治疗效果增强,未来可能具有关键的临床应用.
    Acute kidney injury (AKI) is not only a worldwide problem with a cruel hospital mortality rate but also an independent risk factor for chronic kidney disease and a promoting factor for its progression. Despite supportive therapeutic measures, there is no effective treatment for AKI. This study employs tetrahedral framework nucleic acid (tFNA) as a vehicle and combines typhaneoside (Typ) to develop the tFNA-Typ complex (TTC) for treating AKI. With the precise targeting ability on mitochondria and renal tubule, increased antiapoptotic and antioxidative effect, and promoted mitochondria and kidney function restoration, the TTC represents a promising nanomedicine for AKI treatment. Overall, this study has developed a dual-targeted nanoparticle with enhanced therapeutic effects on AKI and could have critical clinical applications 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
    三阴性乳腺癌(TNBC)是乳腺癌的一种亚型,与其他乳腺癌亚型相比,它具有侵袭性和更频繁的组织转移。因为TNBC肿瘤细胞的增殖不依赖于雌激素受体(ER),孕激素受体(PR),和Erb-B2受体酪氨酸激酶2(HER2),缺乏准确的药物靶点,常规化疗是具有挑战性的是有效的,不良反应严重。目前,TNBC的治疗策略通常取决于手术的组合,放射治疗,和化疗。常规给药方法对TNBC的影响很小,对正常组织造成严重损害。因此,开发高效、实用的给药方式,开辟TNBC靶向治疗的新视野是当务之急。在我们的工作中,牛血清白蛋白(BSA)作为保护膜,延长了四面体骨架核酸(tFNA)递送系统的循环时间,抵抗了体内免疫清除。tFNA作为负载DOX和AS1411适体的载体用于三阴性乳腺癌的靶向治疗。与现有方法相比,这种优化的系统表现出更强的肿瘤靶向性,因此tFNA可以更集中在肿瘤组织周围,减少DOX对其他器官的毒性。这种仿生递送系统在TNBC小鼠模型中表现出有效的肿瘤生长抑制,提供临床潜力,以促进TNBC的治疗,具有巨大的临床翻译潜力。
    Triple-negative breast cancer (TNBC) is a subtype of breast cancer, and it has aggressive and more frequent tissue metastases than other breast cancer subtypes. Because the proliferation of TNBC tumor cells does not depend on estrogen receptor (ER), progesterone receptor (PR), and Erb-B2 receptor tyrosine kinase 2 (HER2) and lacks accurate drug targets, conventional chemotherapy is challenging to be effective, and adverse reactions are severe. At present, the treatment strategy for TNBC generally depends on a combination of surgery, radiotherapy, and chemotherapy. Conventional administration methods have minimal effects on TNBC and cause severe damage to normal tissues. Therefore, it is an urgent task to develop an efficient and practical way of drug delivery and open up a new horizon of targeted therapy for TNBC. In our work, bovine serum albumin (BSA) acted as the protective film to prolong the circulation time of the tetrahedral framework nucleic acid (tFNA) delivery system and resist immune clearance in vivo. tFNA was used as a carrier loaded with DOX and AS1411 aptamers for the targeted treatment of triple-negative breast cancer. Compared with existing approaches, this optimized system exhibits stronger tumor-targeting so that tFNAs can be more concentrated around the tumor tissue, reducing DOX toxicity to other organs. This bionic delivery system exhibited effective tumor growth inhibition in the TNBC mice model, offering the clinical potential to promote the treatment of TNBC with great potential for clinical translation.
    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
    皮肤是人体的第一道防线,容易受伤。各种局部或全身性疾病促进皮肤炎症,当皮肤损伤的强度或持续时间超过组织修复能力时,纤维化,组织修复反应失调的结果,开始主导修复过程。然而,现有的减少皮肤纤维化的方法不足并引起副作用,强调需要有效抑制皮肤纤维化和降低免疫原性的药物,炎症,凋亡,和焦亡。四面体骨架核酸(tFNA)是具有独特空间结构的DNA纳米材料,表现出良好的生物安全性,并促进抗炎,抗氧化,抗纤维化,血管生成,和皮肤伤口愈合活性几乎没有毒性。这里,我们探讨了tFNA在体外和体内皮肤纤维化治疗中的潜力。在孵育细胞或给小鼠注射促纤维化分子和tFNA后,我们发现tFNA抑制上皮-间质转化,降低炎症因子水平,皮肤胶原蛋白含量降低,并抑制焦亡途径。这些发现表明tFNA在治疗焦亡相关疾病中的潜力。
    The skin is the first line of defense for the human body and is vulnerable to injury. Various topical or systemic diseases facilitate skin inflammation, and when the intensity or duration of skin injury exceeds the ability of tissue repair, fibrosis, an outcome of a dysregulated tissue-repair response, begins to dominate the repair process. However, existing methods for reducing skin fibrosis are insufficient and cause side effects, highlighting the need for drugs that effectively inhibit skin fibrosis and reduce immunogenicity, inflammation, apoptosis, and pyroptosis. Tetrahedral framework nucleic acids (tFNAs) are DNA nanomaterials that have a unique spatial structure, demonstrate excellent biosecurity, and promote anti-inflammatory, antioxidative, antifibrotic, angiogenic, and skin-wound-healing activities with almost no toxicity. Here, we explored the potential of tFNAs in skin fibrosis therapy in vitro and in vivo. After incubating cells or injecting mice with profibrogenic molecules and tFNAs, we found that the tFNAs inhibited the epithelial-mesenchymal transition, reduced inflammatory factor levels, decreased skin collagen content, and inhibited the pyroptosis pathway. These findings suggest the potential of tFNAs in treating pyroptosis-related diseases.
    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
    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
    全球7000万患者中有1500多万对可用的抗癫痫药物(AED)没有反应。随着纳米医学的出现,纳米材料越来越多地用于治疗许多疾病。这里,我们报道了四面体框架核酸(tFNA),组装好的核酸纳米颗粒,表现出优异的跨血脑屏障(BBB)抑制癫痫持续状态后小鼠海马中M1小胶质细胞活化和A1反应性星形胶质细胞增生的能力。此外,tFNA通过减轻反应性星形胶质细胞的氧化应激来抑制谷氨酰胺合成酶的下调,并随后减少谷氨酸的积累和谷氨酸介导的神经元过度兴奋。同时,tFNA通过调节AMPAR内吞作用促进突触后膜中的α-氨基-3-羟基-5-甲基-4-异恶唑丙酸受体(AMPAR)内化,这有助于减少钙流入,并最终降低过度兴奋和自发性癫痫持续发作频率。这些发现证明了tFNA是一种潜在的AED,核酸物质可能是治疗癫痫的新方向。
    More than 15 million out of 70 million patients worldwide do not respond to available antiepilepticus drugs (AEDs). With the emergence of nanomedicine, nanomaterials are increasingly being used to treat many diseases. Here, we report that tetrahedral framework nucleic acid (tFNA), an assembled nucleic acid nanoparticle, showed an excellent ability to the cross blood-brain barrier (BBB) to inhibit M1 microglial activation and A1 reactive astrogliosis in the hippocampus of mice after status epilepticus. Furthermore, tFNA inhibited the downregulation of glutamine synthetase by alleviating oxidative stress in reactive astrocytes and subsequently reduced glutamate accumulation and glutamate-mediated neuronal hyperexcitability. Meanwhile, tFNA promotes α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) internalization in the postsynaptic membrane by regulating AMPAR endocytosis, which contributed to reduced calcium influx and ultimately reduced hyperexcitability and spontaneous epilepticus spike frequencies. These findings demonstrated tFNA as a potential AED and that nucleic acid material may be a new direction for the treatment of epilepsy.
    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
    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
    Strategies for functionalizing diverse tetrahedral framework nucleic acids (tFNAs) have been extensively explored since the first successful fabrication of tFNA by Turberfield. One-pot annealing of at least four DNA single strands is the most common method to prepare tFNA, as it optimizes the cost, yield, and speed of assembly. This review concentrates on four key merits of tFNAs and their potential for biomedical applications. The natural ability of tFNA to scavenge reactive oxygen species, along with remarkable enhancement in cellular endocytosis and tissue permeability based on its appropriate size and geometry, promotes cell-material interaction to direct or probe cell behavior, especially to treat inflammatory and degenerative diseases. Moreover, the structural programmability of tFNA enables the development of static tFNA-based nanomaterials via engineering of functional oligonucleotides or therapeutic molecules, and dynamic tFNAs via attachment of stimuli-responsive DNA apparatuses, leading to potentially applications in targeted therapies, tissue regeneration, antitumor strategies, and antibacterial treatment. Although there are impressive performance and significant progress, challenges and prospects of functionalizing tFNA-based nanostructures are still indicated in this review. This article is protected by copyright. All rights reserved.
    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
    Articular cartilage (AC) injury repair has always been a difficult problem for clinicians and researchers. Recently, a promising therapy based on mesenchymal stem cells (MSCs) has been developed for the regeneration of cartilage defects. As endogenous articular stem cells, synovial MSCs (SMSCs) possess strong chondrogenic differentiation ability and articular specificity. In this study, a cartilage regenerative system was developed based on a chitosan (CS) hydrogel/3D-printed poly(ε-caprolactone) (PCL) hybrid containing SMSCs and recruiting tetrahedral framework nucleic acid (TFNA) injected into the articular cavity. TFNA, which is a promising DNA nanomaterial for improving the regenerative microenvironment, could be taken up into SMSCs and promoted the proliferation and chondrogenic differentiation of SMSCs. CS, as a cationic polysaccharide, can bind to DNA through electrostatic action and recruit free TFNA after articular cavity injection in vivo. The 3D-printed PCL scaffold provided basic mechanical support, and TFNA provided a good microenvironment for the proliferation and chondrogenic differentiation of the delivered SMSCs and promoted cartilage regeneration, thus greatly improving the repair of cartilage defects. In conclusion, this study confirmed that a CS hydrogel/3D-printed PCL hybrid scaffold containing SMSCs could be a promising strategy for cartilage regeneration based on chitosan-directed TFNA recruitment and TFNA-enhanced cell proliferation and chondrogenesis.
    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