Small activating RNA

小活化 RNA
  • 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
    代谢综合征(MetS)是一种以腹部肥胖为特征的病理状态,胰岛素抵抗,高血压,和高脂血症。Sirtuin1(SIRT1),一种高度保守的组蛋白脱乙酰酶,其特征是作为一个关键的代谢调节剂和保护老化相关的病理,包括MetS。在这项研究中,我们研究了使用小激活RNA(saRNA)激活SIRT1的治疗潜力,从而减少炎症样反应并重建正常的脂质代谢。SIRT1saRNA显着增加脂多糖刺激和非刺激巨噬细胞中SIRT1信使RNA(mRNA)和蛋白质水平。SIRT1saRNA显著降低炎症样反应,通过降低关键炎症细胞因子的mRNA水平,如肿瘤坏死因子α,白细胞介素1β(IL-1β),白细胞介素6(IL-6),和趋化因子单核细胞化学引诱物蛋白-1和角质形成细胞化学引诱物。SIRT1过表达也显著降低核因子-κB和c-Jun氨基末端激酶的磷酸化,都是炎症途径的关键信号分子。探讨SIRT1上调的治疗效果,我们使用与转铁蛋白受体适体缀合的SIRT1saRNA治疗高脂饮食模型,用于递送至肝脏和细胞内化.SIRT1saRNA治疗组中的动物表现出显著减少的体重增加,白色脂肪组织显著减少,甘油三酯,空腹血糖水平,和细胞内脂质积累。这些表明治疗诱导的动物中脂质和葡萄糖代谢的变化。这项研究的结果表明,saRNA对SIRT1的靶向激活是逆转MetS的潜在策略。
    Metabolic syndrome (MetS) is a pathological condition characterized by abdominal obesity, insulin resistance, hypertension, and hyperlipidemia. Sirtuin 1 (SIRT1), a highly conserved histone deacetylase, is characterized as a key metabolic regulator and protector against aging-associated pathologies, including MetS. In this study, we investigate the therapeutic potential of activating SIRT1 using small activating RNAs (saRNA), thereby reducing inflammatory-like responses and re-establishing normal lipid metabolism. SIRT1 saRNA significantly increased SIRT1 messenger RNA (mRNA) and protein levels in both lipopolysaccharide-stimulated and nonstimulated macrophages. SIRT1 saRNA significantly decreased inflammatory-like responses, by reducing mRNA levels of key inflammatory cytokines, such as Tumor Necrosis Factor alpha, Interleukin 1 beta (IL-1β), Interleukin 6 (IL-6), and chemokines Monocyte Chemoattractant Protein-1 and keratinocyte chemoattractant. SIRT1 overexpression also significantly reduced phosphorylation of nuclear factor-κB and c-Jun N-terminal kinase, both key signaling molecules for the inflammatory pathway. To investigate the therapeutic effect of SIRT1 upregulation, we treated a high-fat diet model with SIRT1 saRNA conjugated to a transferrin receptor aptamer for delivery to the liver and cellular internalization. Animals in the SIRT1 saRNA treatment arm demonstrated significantly decreased weight gain with a significant reduction in white adipose tissue, triglycerides, fasting glucose levels, and intracellular lipid accumulation. These suggest treatment-induced changes to lipid and glucose metabolism in the animals. The results of this study demonstrate that targeted activation of SIRT1 by saRNAs is a potential strategy to reverse MetS.
    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
    G蛋白偶联受体(GPCRs)是最常见和最重要的药物靶标。然而,>70%的GPCR是不可用的或难以使用常规化学激动剂/拮抗剂靶向的。小核酸分子,可以序列特异性调节任何基因,提供了一个独特的机会,有效地扩大药物的目标,尤其是那些难以处理或难以处理的问题,比如GPCRs。这里,作者首次报道了小激活RNA(saRNA)有效调节GPCR用于癌症治疗。具体来说,saRNA促进MAS受体(MAS1)的表达,在癌细胞增殖和迁移中抵消经典血管紧张素II途径的GPCR,被识别。这些saRNA,由两亲性树枝状聚合物载体传递,增强MAS1表达,抵消血管紧张素II/血管紧张素II受体1型轴,并导致在肿瘤异种移植小鼠模型和患者来源的肿瘤模型中,肿瘤发生的显着抑制和多种癌症的肿瘤进展的抑制。这项研究不仅为靶向肾素-血管紧张素系统的癌症治疗提供了新的策略,也是通过RNA激活调节GPCR信号的新途径。
    G protein-coupled receptors (GPCRs) are the most common and important drug targets. However, >70% of GPCRs are undruggable or difficult to target using conventional chemical agonists/antagonists. Small nucleic acid molecules, which can sequence-specifically modulate any gene, offer a unique opportunity to effectively expand drug targets, especially those that are undruggable or difficult to address, such as GPCRs. Here, the authors report  for the first time that small activating RNAs (saRNAs) effectively modulate a GPCR for cancer treatment. Specifically, saRNAs promoting the expression of Mas receptor (MAS1), a GPCR that counteracts the classical angiotensin II pathway in cancer cell proliferation and migration, are identified. These saRNAs, delivered by an amphiphilic dendrimer vector, enhance MAS1 expression, counteracting the angiotensin II/angiotensin II Receptor Type 1 axis, and leading to significant suppression of tumorigenesis and the inhibition of tumor progression of multiple cancers in tumor-xenografted mouse models and patient-derived tumor models. This study provides not only a new strategy for cancer therapy by targeting the renin-angiotensin system, but also a new avenue to modulate GPCR signaling by RNA activation.
    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
    RNA激活(RNAa)是由小RNA介导的转录激活的一种未表征的机制,例如microRNAs(miRNAs)。RNAa研究中的一个关键问题是难以区分由转录后调控间接引起的基因表达变化和RNAa对基因表达的直接诱导。因此,在这项研究中,我们试图找出参与RNAa的关键因素,使用miR-34a诱导ZMYND10作为评价RNAa的系统。我们鉴定了阳性转录延伸因子CDK9和DDX21,它们与核AGO和TNRC6A形成复合物,作为RNAa的重要转录激活因子。此外,我们发现DDX21的抑制抑制miR-34a和其他miRNA的RNAa而不抑制转录后调控。我们的发现揭示了RNAa与暂停的PolII释放之间的紧密联系,通过单独分析转录后调控和RNAa来促进RNAa研究。
    RNA activation (RNAa) is an uncharacterized mechanism of transcriptional activation mediated by small RNAs, such as microRNAs (miRNAs). A critical issue in RNAa research is that it is difficult to distinguish between changes in gene expression caused indirectly by post-transcriptional regulation and direct induction of gene expression by RNAa. Therefore, in this study, we seek to identify a key factor involved in RNAa, using the induction of ZMYND10 by miR-34a as a system to evaluate RNAa. We identify the positive transcription elongation factors CDK9 and DDX21, which form a complex with nuclear AGO and TNRC6A, as important transcriptional activators of RNAa. In addition, we find that inhibition of DDX21 suppresses RNAa by miR-34a and other miRNAs without inhibiting post-transcriptional regulation. Our findings reveal a strong connection between RNAa and release of paused Pol II, facilitating RNAa research by making it possible to separately analyze post-transcriptional regulation and RNAa.
    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
    内耳毛细胞的丧失导致哺乳动物不可逆的声损伤,和再生内耳毛细胞恢复听力损失是具有挑战性的。ATOH1是毛细胞发育和再生的症结基因。小激活RNA(saRNA)可以靶向基因以特异性上调其表达。本研究旨在探讨小活化RNA是否能在体外联合生长因子诱导人脂肪间充质干细胞分化为毛细胞样细胞,从而为毛细胞再生和感音神经性耳聋的治疗提供新的策略。在293T和人脂肪间充质干细胞中设计并筛选了15种靶向人ATOH1基因的小活化RNA,发现这些候选物中的3个能够有效且稳定地激活ATOH1基因表达。然后将选择的小活化RNA转染到毛细胞祖细胞中,转染后10天检查毛细胞标记物。转染选定的小活化RNA后,内耳毛细胞特征性标志物的表达,POU类4homeobox3(POU4F3)和肌球蛋白VIIA(MYO7A),被检测到。人脂肪间充质干细胞具有向人毛细胞祖细胞分化的潜能。体外,小活化RNA能够诱导毛细胞祖细胞分化为毛细胞样细胞.因此,RNA激活技术有潜力为毛细胞的再生提供新的策略。
    The loss of inner ear hair cells leads to irreversible acoustic injury in mammals, and regeneration of inner ear hair cells to restore hearing loss is challenging. ATOH1 is a key gene in the development and regeneration of hair cells. Small activating RNAs (saRNAs) can target a gene to specifically upregulate its expression. This study aimed to explore whether small activating RNAs could induce the differentiation of human adipose-derived mesenchymal stem cells into hair cell-like cells with a combination of growth factors in vitro and thus provide a new strategy for hair cell regeneration and the treatment of sensorineural hearing loss. Fifteen small activating RNAs targeting the human ATOH1 gene were designed and screened in 293 T and human adipose-derived mesenchymal stem cells, and 3 of these candidates were found to be capable of effectively and stably activating ATOH1 gene expression. The selected small activating RNAs were then transfected into hair cell progenitor cells, and hair cell markers were examined 10 days after transfection. After transfection of the selected small activating RNAs, the expression of the characteristic markers of inner ear hair cells, POU class 4 homeobox 3 (POU4F3) and myosin VIIA (MYO7A), was detected. Human adipose-derived mesenchymal stem cells have the potential to differentiate into human hair cell progenitor cells. In vitro, small activating RNAs were able to induce the differentiation of hair cell progenitor cells into hair cell-like cells. Therefore, RNA activation technology has the potential to provide a new strategy for the regeneration of hair cells.
    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
    在首次证明使用小核酸调节基因表达的概念之后,长时间的成熟导致,在上个世纪末,基于寡核苷酸的疗法的第一个上市授权。从那以后,12更多的化合物进入市场,更多的是在临床开发后期。许多公司的成立是为了利用他们的治疗潜力,大型制药公司很快就确信寡核苷酸可以代表蛋白质靶向产品的可靠替代品。已经开发了许多技术来改善寡核苷酸药代动力学和药效学。最初针对罕见疾病和利基市场,寡核苷酸现在能够使大量患者群体受益。然而,寡核苷酸仍有改进的空间,未来几年可能会有进一步的突破。在这篇综述中,我们提供了治疗性寡核苷酸的概述。我们特别介绍了不同类型的寡核苷酸及其作用方式,它们的目标组织和给患者施用它们的途径,以及使用它们的治疗领域。此外,我们提出了寡核苷酸的不同方法。我们最后讨论了这个药物发现平台未来的挑战和机遇。
    Following the first proof of concept of using small nucleic acids to modulate gene expression, a long period of maturation led, at the end of the last century, to the first marketing authorization of an oligonucleotide-based therapy. Since then, 12 more compounds have hit the market and many more are in late clinical development. Many companies were founded to exploit their therapeutic potential and Big Pharma was quickly convinced that oligonucleotides could represent credible alternatives to protein-targeting products. Many technologies have been developed to improve oligonucleotide pharmacokinetics and pharmacodynamics. Initially targeting rare diseases and niche markets, oligonucleotides are now able to benefit large patient populations. However, there is still room for oligonucleotide improvement and further breakthroughs are likely to emerge in the coming years. In this review we provide an overview of therapeutic oligonucleotides. We present in particular the different types of oligonucleotides and their modes of action, the tissues they target and the routes by which they are administered to patients, and the therapeutic areas in which they are used. In addition, we present the different ways of patenting oligonucleotides. We finally discuss future challenges and opportunities for this drug-discovery platform.
    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
    RNA activation (RNAa) is a mechanism whereby RNA oligos complementary to genomic sequences around the promoter region of genes increase the transcription output of their target gene. Small activating RNA (saRNA) mediate RNAa through interaction with protein co-factors to facilitate RNA polymerase II activity and nucleosome remodeling. As saRNA are small, versatile and safe, they represent a new class of therapeutics that can rescue the downregulation of critical genes in disease settings. This review highlights our current understanding of saRNA biology and describes various examples of how saRNA are successfully used to treat various oncological, neurological and monogenic diseases. MTL-CEBPA, a first-in-class compound that reverses CEBPA downregulation in oncogenic processes using CEBPA-51 saRNA has entered clinical trial for the treatment of hepatocellular carcinoma (HCC). Preclinical models demonstrate that MTL-CEBPA reverses the immunosuppressive effects of myeloid cells and allows for the synergistic enhancement of other anticancer drugs. Encouraging results led to the initiation of a clinical trial combining MTL-CEBPA with a PD-1 inhibitor for treatment of solid tumors.
    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
    Small double-stranded RNAs (dsRNAs) have been proved to effectively up-regulate the expression of particular genes by targeting their promoters. These small dsRNAs were also termed small activating RNAs (saRNAs). We previously reported that several small double-stranded RNAs (dsRNAs) targeting the PRKC apoptosis WT1 regulator (PAWR) promoter can up-regulate PAWR gene expression effectively in human cancer cells. The present study was conducted to evaluate the antitumor potential of PAWR gene induction by these saRNAs in bladder cancer. Promisingly, we found that up-regulation of PAWR by saRNA inhibited the growth of bladder cancer cells by inducing cell apoptosis and cell cycle arrest which was related to inhibition of anti‑apoptotic protein Bcl-2 and inactivation of the NF-κB and Akt pathways. The activation of the caspase cascade and the regulation of cell cycle related proteins also supported the efficacy of the treatment. Moreover, our study also showed that these saRNAs cooperated with cisplatin in the inhibition of bladder cancer cells. Overall, these data suggest that activation of PAWR by saRNA may have a therapeutic benefit for bladder cancer.
    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
    Non-alcoholic fatty liver disease (NAFLD) culminates in insulin resistance and metabolic syndrome. Because there are no approved pharmacological treatment agents for non-alcoholic steatohepatitis (NASH) and NAFLD, different signaling pathways are under investigation for drug development with the focus on metabolic pathways. Hepatocyte nuclear factor 4-alpha (HNF4A) is at the center of a complex transcriptional network where its disruption is directly linked to glucose and lipid metabolism. Resetting HNF4A expression in NAFLD is therefore crucial for re-establishing normal liver function. Here, small activating RNA (saRNA) specific for upregulating HNF4A was injected into rats fed a high-fat diet for 16 weeks. Intravenous delivery was carried out using 5-(G5)-triethanolamine-core polyamidoamine (PAMAM) dendrimers. We observed a significant reduction in liver triglyceride, increased high-density lipoprotein/low-density lipoprotein (HDL/LDL) ratio, and decreased white adipose tissue/body weight ratio, all parameters to suggest that HNF4A-saRNA treatment induced a favorable metabolic profile. Proteomic analysis showed significant regulation of genes involved in sphingolipid metabolism, fatty acid β-oxidation, ketogenesis, detoxification of reactive oxygen species, and lipid transport. We demonstrate that HNF4A activation by oligonucleotide therapy may represent a novel single agent for the treatment of NAFLD and insulin resistance.
    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: Editorial
    暂无摘要。
    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