ASOs

ASOS
  • 文章类型: Journal Article
    环状RNA(circularRNAs,circRNAs)是一种非编码RNA,通过真核蛋白质编码基因的反向剪接形成。circRNAs最常报道和特征明确的功能是它们作为分子诱饵的能力,最常见的miRNA和蛋白质海绵。然而,大多数circRNAs的功能仍然需要更好地理解。为了更全面地理解已验证的circRNAs的生物学相关性,可以使用反义寡核苷酸进行敲低功能分析,RNA干扰(RNAi)实验(例如,靶向反向剪接连接位点),成簇的规则间隔短回文重复(CRISPR)-CRISPR相关(Cas)-9系统(例如,产生circRNA特异性敲除),和CRISPR-Cas13技术有效地靶向circRNAs而不影响宿主基因。在这次审查中,我总结了通过反义策略研究circRNA在培养细胞和动物模型中的生物学作用的circRNA敲低的可行性和有效性。
    Circular RNAs (circRNAs) are a type of noncoding RNA that are formed by back-splicing from eukaryotic protein-coding genes. The most frequently reported and well-characterized function of circRNAs is their ability to act as molecular decoys, most often as miRNA and protein sponges. However, the functions of most circRNAs still need to be better understood. To more fully understand the biological relevance of validated circRNAs, knockdown functional analyses can be performed using antisense oligonucleotides, RNA interference (RNAi) experiments (e.g., targeting back-splicing junction sites), the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated (Cas)-9 system (e.g., generating circRNA-specific knockouts), and CRISPR-Cas13 technology to effectively target circRNAs without affecting host genes. In this review, I summarize the feasibility and effectiveness of circRNA knockdown through antisense strategies for investigating the biological roles of circRNAs in cultured cells and animal models.
    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
    孤立性纤维瘤(SFT)是一种罕见的,非遗传性软组织肉瘤被认为起源于成纤维细胞间充质干细胞。SFT的病因被认为是由于NGFI-A结合蛋白2(NAB2)与染色体12上的信号转导和激活蛋白6(STAT6)基因之间的环境染色体内基因融合,其中STAT6的激活域与NAB2的DNA结合域融合,导致SFT的肿瘤发生。在SFT中发现的所有NAB2-STAT6融合变异都含有STAT6转录本的C末端,因此可以作为基于反义寡核苷酸(ASO)的治疗的靶位点。的确,我们的体外研究表明,靶向STAT63非翻译区(UTR)的ASO(ASO993523)能够高效降低多种SFT细胞模型中NAB2-STAT6融合转录本的表达(半数最大抑制浓度:116-300nM).令人鼓舞的是,用ASO993523对SFT患者来源的异种移植小鼠模型进行体内治疗可获得可接受的耐受性,NAB2-STAT6融合转录本在异种移植组织中的表达降低(21.9%),and,重要的是,肿瘤生长减少(与未治疗的对照相比,肿瘤体积减少32.4%)。一起来看,我们的研究将ASO993523确立为治疗SFT的潜在药物。
    Solitary fibrous tumor (SFT) is a rare, non-hereditary soft tissue sarcoma thought to originate from fibroblastic mesenchymal stem cells. The etiology of SFT is thought to be due to an environmental intrachromosomal gene fusion between NGFI-A-binding protein 2 (NAB2) and signal transducer and activator protein 6 (STAT6) genes on chromosome 12, wherein the activation domain of STAT6 is fused with the DNA-binding domain of NAB2 resulting in the oncogenesis of SFT. All NAB2-STAT6 fusion variations discovered in SFTs contain the C-terminal of STAT6 transcript, and thus can serve as target site for antisense oligonucleotides (ASOs)-based therapies. Indeed, our in vitro studies show the STAT6 3\' untranslated region (UTR)-targeting ASO (ASO 993523) was able to reduce expression of NAB2-STAT6 fusion transcripts in multiple SFT cell models with high efficiency (half-maximal inhibitory concentration: 116-300 nM). Encouragingly, in vivo treatment of SFT patient-derived xenograft mouse models with ASO 993523 resulted in acceptable tolerability profiles, reduced expression of NAB2-STAT6 fusion transcripts in xenograft tissues (21.9%), and, importantly, reduced tumor growth (32.4% decrease in tumor volume compared with the untreated control). Taken together, our study established ASO 993523 as a potential agent for the treatment of SFTs.
    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
    肝病和心脏病是全世界死亡的主要原因。已知导致2型糖尿病(T2D)和非酒精性脂肪肝(NAFLD)的代谢改变与脂质稳态紊乱,可能会加剧肝脏和心血管疾病。一些药物治疗可以减轻器官功能障碍,但是重要的副作用限制了它们的功效,通常导致组织的恶化。它需要开发新的个性化治疗方法,工程化RNA分子的最新进展正变得越来越可行作为替代治疗。这篇综述概述了反义寡核苷酸(ASO)的当前用途,RNA干扰(RNAi)和RNA基因组编辑作为罕见代谢紊乱的治疗。然而,小型非编码RNA作为肝脏和心脏疾病治疗剂的潜力尚未得到充分探索.尽管miRNA被认为是许多疾病的生物标志物,它们也能够作为药物进行医疗干预;一些临床试验正在测试miRNAs作为2型糖尿病的治疗方法,非酒精性脂肪肝以及心脏疾病。基于RNA的疗法的最新进展可能潜在地促进miRNA作为试剂和作为药物靶标的新应用。在这项工作中,我们试图总结miRNA选择性治疗与常规药物相比的进展和优势.特别是,我们试图强调可药用miRNA,超过ASO或其他RNA治疗剂或常规药物。最后,我们试图解决与功效相关的研究问题,副作用,以及RNA疗法的使用范围。此外,我们涵盖了障碍,并检查了基于miRNA的RNA治疗在代谢紊乱如糖尿病中的最新进展,肝脏,还有心脏病.
    Liver and heart disease are major causes of death worldwide. It is known that metabolic alteration causing type 2 diabetes (T2D) and Nonalcoholic fatty liver (NAFLD) coupled with a derangement in lipid homeostasis, may exacerbate hepatic and cardiovascular diseases. Some pharmacological treatments can mitigate organ dysfunctions but the important side effects limit their efficacy leading often to deterioration of the tissues. It needs to develop new personalized treatment approaches and recent progresses of engineered RNA molecules are becoming increasingly viable as alternative treatments. This review outlines the current use of antisense oligonucleotides (ASOs), RNA interference (RNAi) and RNA genome editing as treatment for rare metabolic disorders. However, the potential for small non-coding RNAs to serve as therapeutic agents for liver and heart diseases is yet to be fully explored. Although miRNAs are recognized as biomarkers for many diseases, they are also capable of serving as drugs for medical intervention; several clinical trials are testing miRNAs as therapeutics for type 2 diabetes, nonalcoholic fatty liver as well as cardiac diseases. Recent advances in RNA-based therapeutics may potentially facilitate a novel application of miRNAs as agents and as druggable targets. In this work, we sought to summarize the advancement and advantages of miRNA selective therapy when compared to conventional drugs. In particular, we sought to emphasise druggable miRNAs, over ASOs or other RNA therapeutics or conventional drugs. Finally, we sought to address research questions related to efficacy, side-effects, and range of use of RNA therapeutics. Additionally, we covered hurdles and examined recent advances in the use of miRNA-based RNA therapy in metabolic disorders such as diabetes, liver, and heart 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
    杜氏肌营养不良症(DMD)是最严重和破坏性的神经肌肉遗传性疾病之一,每年男性新生儿发病率为20.000例。由突变引起的疾病(外显子缺失,无意义的突变,外显子内插入或缺失,外显子重复,拼接位置缺陷,和深层内含子突变)在DMD基因中,逐渐导致肌肉萎缩和步行丧失。这种情况对患者和他们的家人来说都是痛苦的,呼吁迫切需要有效的治疗。
    在这篇评论中,作者描述了DMD临床试验中基因治疗方法的现状.这种疗法包括基因置换,基因替换,基于RNA的疗法,通读突变,和CRISPR方法。
    只有少数候选药物已获得有条件批准用于治疗DMD。大多数这些疗法只有适度的恢复肌营养不良蛋白或改善肌肉功能的能力。这表明在DMD疗法的开发中存在重要的未满足的需求。互补基因和细胞疗法都需要探索以恢复肌营养不良蛋白,改善肌肉功能,并在疾病晚期有效地重建肌肉纤维。
    Duchenne muscular dystrophy (DMD) is one of the most severe and devastating neuromuscular hereditary diseases with a male newborn incidence of 20 000 cases each year. The disease caused by mutations (exon deletions, nonsense mutations, intra-exonic insertions or deletions, exon duplications, splice site defects, and deep intronic mutations) in the DMD gene, progressively leads to muscle wasting and loss of ambulation. This situation is painful for both patients and their families, calling for an emergent need for effective treatments.
    In this review, the authors describe the state of the gene therapy approach in clinical trials for DMD. This therapeutics included gene replacement, gene substitution, RNA-based therapeutics, readthrough mutation, and the CRISPR approach.
    Only a few drug candidates have yet been granted conditional approval for the treatment of DMD. Most of these therapies have only a modest capability to restore the dystrophin or improve muscle function, suggesting an important unmet need in the development of DMD therapeutics. Complementary genes and cellular therapeutics need to be explored to both restore dystrophin, improve muscle function, and efficiently reconstitute the muscle fibers in the advanced stage of the disease.
    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
    越来越多的临床前证据导致了这样的假设,即人类Toll样受体8(hTLR8)在肿瘤微环境(TME)中的激活可能通过其对单核细胞的作用而具有有效的抗癌作用。髓样树突状细胞(mDCs),自然杀伤(NK)细胞这促使在多种癌症中启动化学hTLR8激动剂的若干临床试验。同时,越来越多的合成反义寡核苷酸(ASO)正被开发作为癌症治疗剂。我们最近报道了2'-O-甲基(2'OMe)修饰的ASO可以序列依赖性方式增强hTLR8化学激动剂的感知。这表明选择具有抗癌活性的靶向基因的ASO可以与TME中的低剂量hTLR8激动剂协同作用。这里,我们提供了一个详细的方案来快速筛选和鉴定这种对hTLR8传感具有协同活性的合成双功能寡核苷酸。
    A growing body of preclinical evidence has led to the hypothesis that human Toll-like receptor 8 (hTLR8) activation in the tumor microenvironment (TME) could have potent anticancer effects through its action on monocytes, myeloid dendritic cells (mDCs), and natural killer (NK) cells. This has motivated the initiation of several clinical trials for chemical hTLR8 agonists in a variety of cancers. Concurrently, a growing number of synthetic antisense oligonucleotides (ASOs) are being developed as cancer therapeutics. We have recently reported that 2\'-O-methyl (2\'OMe)-modified ASOs can potentiate sensing of hTLR8 chemical agonists in a sequence-dependent manner. This suggests that select gene-targeting ASOs with anticancer activity may synergize with low-dose hTLR8 agonists in the TME. Here, we provide a detailed protocol to rapidly screen and identify such synthetic bifunctional oligonucleotides with synergistic activity on hTLR8 sensing.
    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: Review
    人类朊病毒疾病是异质性的,通常是快速进步的,与错误折叠的朊病毒蛋白(PrP)聚集和自我繁殖相关的传染性神经退行性疾病。尽管它们很少,朊病毒疾病包括在分子水平上由错误折叠的PrP的不同构象异构体和宿主基因型变异性确定的广泛表型变体。此外,它们独特地发生在特发性,基因决定的,和具有不同病因的后天形式。
    这篇综述提供了朊病毒疾病的潜在治疗靶标以及在细胞和动物模型以及人体试验中获得的主要结果的最新概述。还讨论了与开发有效疗法和信息丰富的临床试验相关的开放问题和挑战。
    目前测试的治疗策略靶向细胞PrP以防止形成错误折叠的PrP或有利于其消除。其中,被动免疫和基因治疗反义寡核苷酸抗朊病毒蛋白mRNA是最有前途的。然而,这种疾病的稀有性,异质性,快速进展严重阻碍了在发生重大脑损伤之前的无症状或早期阶段成功开展有力的治疗试验和患者识别。因此,迄今为止最有希望的治疗目标是通过降低朊病毒蛋白表达来预防或延缓致病突变携带者的表型转化。
    Human prion diseases are heterogeneous, and often rapidly progressive, transmissible neurodegenerative disorders associated with misfolded prion protein (PrP) aggregation and self-propagation. Despite their rarity, prion diseases comprise a broad spectrum of phenotypic variants determined at the molecular level by different conformers of misfolded PrP and host genotype variability. Moreover, they uniquely occur in idiopathic, genetically determined, and acquired forms with distinct etiologies.
    This review provides an up-to-date overview of potential therapeutic targets in prion diseases and the main results obtained in cell and animal models and human trials. The open issues and challenges associated with developing effective therapies and informative clinical trials are also discussed.
    Currently tested therapeutic strategies target the cellular PrP to prevent the formation of misfolded PrP or to favor its elimination. Among them, passive immunization and gene therapy with antisense oligonucleotides against prion protein mRNA are the most promising. However, the disease\'s rarity, heterogeneity, and rapid progression profoundly frustrate the successful undertaking of well-powered therapeutic trials and patient identification in the asymptomatic or early stage before the development of significant brain damage. Thus, the most promising therapeutic goal to date is preventing or delaying phenoconversion in carriers of pathogenic mutations by lowering prion protein expression.
    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
    乳腺癌(BC)导致女性死亡的最多。化疗-,内分泌-,靶向治疗是临床上治疗BC的主要药物。然而,耐药性是BC患者的主要障碍,会导致预后不良.越来越多的证据表明,非编码RNA(ncRNAs)与广泛的病理过程密切相关,包括耐药性。直到日期,在BC中,耐药性与ncRNAs之间的相关性尚未完全了解。在这里,我们全面总结了一个失调的ncRNAs景观,它在化疗中促进或抑制耐药性,内分泌-,和有针对性的BC疗法。我们的综述将为通过靶向致癌ncRNAs来有效管理耐药性铺平道路。which,反过来又会促进未来BC的药物敏感性。
    Breast cancer (BC) leads to the most amounts of deaths among women. Chemo-, endocrine-, and targeted therapies are the mainstay drug treatments for BC in the clinic. However, drug resistance is a major obstacle for BC patients, and it leads to poor prognosis. Accumulating evidences suggested that noncoding RNAs (ncRNAs) are intricately linked to a wide range of pathological processes, including drug resistance. Till date, the correlation between drug resistance and ncRNAs is not completely understood in BC. Herein, we comprehensively summarized a dysregulated ncRNAs landscape that promotes or inhibits drug resistance in chemo-, endocrine-, and targeted BC therapies. Our review will pave way for the effective management of drug resistance by targeting oncogenic ncRNAs, which, in turn will promote drug sensitivity of BC in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    Tau病变代表tau神经胶质和/或神经元包涵体增加和异常的临床病理实体,一种微管结合蛋白。反义寡核苷酸(ASO)是治疗tau蛋白病变的有前途的治疗方法,因为它们可以靶向taumRNA以减少总的人tau表达或tau外显子10表达和4Rtau。此外,用肽特异性靶向tau可能是一种独特的药理学方法,在小分子和蛋白质之间。
    本综述研究了设计目前已知用于治疗tau蛋白病变的ASO和肽的化学基础。在靶向tau表达的ASOs中,BIIB080是第一个进入临床试验开发的治疗轻度阿尔茨海默病(AD)。此外,基于临床前研究讨论了肽021(P021,Ac-DGGLAG-NH2)在tau蛋白病中的治疗潜力。
    ASO是一种很有前途的tau病治疗方法,特别是因为ASO可以抑制有害基因的表达并直接传递到大脑,几乎没有全身副作用.然而,全身性脑tau蛋白降低是否会产生积极的临床效果尚不清楚.BIIB080的II期试验正在轻度AD中进行。神经营养性和神经源性肽模拟化合物也显示出作为AD和其他tau蛋白病变的治疗选择的潜力。
    Tauopathies represent clinicopathological entities with increased and abnormal glial and/or neuronal inclusions of tau, a microtubule-binding protein. Antisense oligonucleotides (ASOs) are a promising therapeutic approach for treating tauopathies as they can target tau mRNA to reduce total human tau expression or tau exon 10 expression and 4 R tau. Additionally, targeting the tau specifically with peptides may be a unique pharmacological approach, between small molecules and proteins.
    The present review investigates the chemical basis of designing ASOs and peptides currently known to treat tauopathies. Among ASOs targeting tau expression, BIIB080 was the first to enter clinical trial development for treating mild Alzheimer\'s disease (AD). Furthermore, the therapeutic potential of peptide 021 (P021, Ac-DGGLAG-NH2) in tauopathies is discussed based on preclinical studies.
    ASOs are a promising therapeutic approach for tauopathies, particularly because ASOs may suppress the expression of harmful genes and are directly delivered to the brain, showing little systemic side effects. However, whether a generalized brain tau decrease will produce positive clinical effects remains unclear. A Phase II trial of BIIB080 is ongoing in mild AD. Neurotrophic and neurogenic peptide mimetic compounds have also shown potential as treatment options for AD and other tauopathies.
    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: Review
    黄病毒是新出现或重新出现的病原体,已在世界各地引起多次暴发,对人类健康和经济发展构成严重威胁。基于RNA的疗法正在迅速发展,并在对抗黄病毒的斗争中充满希望。然而,开发有效和安全的黄病毒疗法,许多挑战仍未解决。
    在这篇评论中,作者简要介绍了黄病毒的生物学以及基于RNA的治疗方法的最新进展。此外,作者列出了该领域的挑战和可能的解决方案。最后,作者对基于RNA的黄病毒疗法的发展和未来发表了他们的意见。
    随着结构生物学的迅速发展,黄病毒蛋白的晶体结构为今后合理的药物设计奠定了基础。关于黄病毒与宿主之间的相互作用的研究对于抑制剂设计也是非常有价值的。研究人员应保持目前的势头,通过学术界的共同努力,使安全有效的抗黄病毒药物获得许可,政府,和工业。
    Flaviviruses are emerging or reemerging pathogens that have caused several outbreaks throughout the world and pose serious threats on human health and economic development. RNA-based therapeutics are developing rapidly, and hold promise in the fight against flaviviruses. However, to develop efficient and safe therapeutics for flaviviruses, many challenges remain unsolved.
    In this review, the authors briefly introduced the biology of flaviviruses and the current advances in RNA-based therapeutics for them. Furthermore, the authors list the challenges and possible solutions in this area. Finally, the authors give their opinion on the development and future of RNA-based therapeutics for flaviviruses.
    With the rapid development of structural biology, the crystal structures of flavivirus proteins may lay the foundation for future rational drug design. Studies regarding the interactions between the flavivirus and the host will also be invaluable to inhibitor design. Researchers should maintain the current momentum to bring about safe and effective anti-flavivirus drugs to licensure through joint efforts of academia, government, and industry.
    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
    (1)背景:常染色体隐性遗传性多囊肾病(ARPKD)是一种罕见的纤毛病,其特征是肾脏逐渐增大,集合管梭形扩张。PKHD1基因的功能缺失突变,编码纤维囊素/聚果胶,引起ARPKD;然而,目前尚未找到有效的ARPKD治疗方法和药物。反义寡核苷酸(ASO)是短的特殊寡核苷酸,其功能是调节基因表达和改变mRNA剪接。一些ASO已被FDA批准用于治疗遗传性疾病,许多人目前正在进步。我们设计了ASO来验证ASO是否介导剪接的校正以进一步治疗由剪接缺陷引起的ARPKD,并探索它们作为潜在的治疗选择。(2)方法:采用全外显子组测序(WES)和靶向下一代测序法筛选38例多囊肾病患儿进行基因检测。对他们的临床信息进行了调查和随访。对PKHD1变体进行了总结和分析,并进行关联分析,分析基因型与表型的关系。使用各种生物信息学工具来预测致病性。进行杂合小基因分析作为功能剪接分析的一部分。此外,选择从头蛋白合成抑制剂环己酰亚胺来验证异常前mRNA的降解途径。ASO被设计来拯救异常剪接,这被证实了。(3)结果:11例PKHD1变异患者中,所有患者均表现出不同程度的肝脏和肾脏并发症。我们发现在某些区域具有截短变体和变体的患者具有更严重的表型。通过杂交小基因测定研究了PKHD1基因型的两种剪接变体:变体c.2141-3T>C和c.111745G>A。这些导致异常剪接,并证实了它们的强致病性。我们证明,使用从头蛋白合成抑制剂环己酰亚胺,从变体产生的异常pre-mRNA从NMD途径逃脱。此外,我们发现拼接缺陷是通过使用ASO来拯救的,这有效地诱导了假外显子的排斥。(4)结论:具有截短变异体和某些区域变异体的患者具有更严重的表型。ASO是通过纠正剪接缺陷和增加正常PKHD1基因的表达来治疗具有PKHD1基因剪接突变的ARPKD患者的潜在药物。
    (1) Background: Autosomal recessive polycystic kidney disease (ARPKD) is a rare ciliopathy characterized by progressively enlarged kidneys with fusiform dilatation of the collecting ducts. Loss-of-function mutations in the PKHD1 gene, which encodes fibrocystin/polyductin, cause ARPKD; however, an efficient treatment method and drug for ARPKD have yet to be found. Antisense oligonucleotides (ASOs) are short special oligonucleotides which function to regulate gene expression and alter mRNA splicing. Several ASOs have been approved by the FDA for the treatment of genetic disorders, and many are progressing at present. We designed ASOs to verify whether ASOs mediate the correction of splicing further to treat ARPKD arising from splicing defects and explored them as a potential treatment option. (2) Methods: We screened 38 children with polycystic kidney disease for gene detection using whole-exome sequencing (WES) and targeted next-generation sequencing. Their clinical information was investigated and followed up. The PKHD1 variants were summarized and analyzed, and association analysis was carried out to analyze the relationship between genotype and phenotype. Various bioinformatics tools were used to predict pathogenicity. Hybrid minigene analysis was performed as part of the functional splicing analysis. Moreover, the de novo protein synthesis inhibitor cycloheximide was selected to verify the degraded pathway of abnormal pre-mRNAs. ASOs were designed to rescue aberrant splicing, and this was verified. (3) Results: Of the 11 patients with PKHD1 variants, all of them exhibited variable levels of complications of the liver and kidneys. We found that patients with truncating variants and variants in certain regions had a more severe phenotype. Two splicing variants of the PKHD1 genotypes were studied via the hybrid minigene assay: variants c.2141-3T>C and c.11174+5G>A. These cause aberrant splicing, and their strong pathogenicity was confirmed. We demonstrated that the abnormal pre-mRNAs produced from the variants escaped from the NMD pathway with the use of the de novo protein synthesis inhibitor cycloheximide. Moreover, we found that the splicing defects were rescued by using ASOs, which efficiently induced the exclusion of pseudoexons. (4) Conclusion: Patients with truncating variants and variants in certain regions had a more severe phenotype. ASOs are a potential drug for treating ARPKD patients harboring splicing mutations of the PKHD1 gene by correcting the splicing defects and increasing the expression of the normal PKHD1 gene.
    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