RNA delivery

RNA 递送
  • 文章类型: Journal Article
    工程化环状RNA(circRNAs)是一类具有头对尾共价连接的结构的单链RNA,其整合了开放阅读框(ORF)和内部核糖体进入位点(IRES),具有编码和表达蛋白质的功能。与mRNA疫苗相比,circRNA疫苗提供了一种更安全的改进方法,稳定,制造简单。随着circRNA生物学功能的快速揭示和严重急性呼吸道冠状病毒II型(SARS-CoV-2)mRNA疫苗的成功,全球的生物制药公司和研究人员正试图开发更稳定的circRNA疫苗来预防和治疗疾病。然而,对circRNA疫苗的研究仍处于起步阶段,它们的综合需要更多的工作和评估,delivery,和使用。在这次审查中,基于目前对circRNA的分子生物学特性和免疫治疗机制的理解,我们总结了目前circRNA疫苗的制备方法,包括设计,合成,净化,和识别。我们讨论了它们的递送策略,并总结了circRNA临床应用面临的挑战,为circRNA疫苗相关研究提供参考。
    Engineered circular RNAs (circRNAs) are a class of single-stranded RNAs with head-to-tail covalently linked structures that integrate open reading frames (ORFs) and internal ribosome entry sites (IRESs) with the function of coding and expressing proteins. Compared to mRNA vaccines, circRNA vaccines offer a more improved method that is safe, stable, and simple to manufacture. With the rapid revelation of the biological functions of circRNA and the success of Severe Acute Respiratory Coronavirus Type II (SARS-CoV-2) mRNA vaccines, biopharmaceutical companies and researchers around the globe are attempting to develop more stable circRNA vaccines for illness prevention and treatment. Nevertheless, research on circRNA vaccines is still in its infancy, and more work and assessment are needed for their synthesis, delivery, and use. In this review, based on the current understanding of the molecular biological properties and immunotherapeutic mechanisms of circRNA, we summarize the current preparation methods of circRNA vaccines, including design, synthesis, purification, and identification. We discuss their delivery strategies and summarize the challenges facing the clinical application of circRNAs to provide references for circRNA vaccine-related research.
    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
    糖尿病伤口的特点是基本愈合阶段的中断和停止,其中包括止血,炎症,扩散,和重塑。然而,糖尿病伤口的传统治疗集中在愈合过程的各个阶段。在这里,本研究利用掩模介导的顺序聚合和各种交联技术来开发具有快速和慢速模块的双模块微针(MN),表现出针对糖尿病伤口愈合的全谱定制的不同降解速率。首先,掺入钙离子和多巴胺的MN协同促进快速止血。第二,快速模块物理交联的MN快速D-甘露糖/多巴胺增强的三聚磷酸盐季铵化壳聚糖(mDTC)纳米颗粒(NP)负载有microRNA-147(miRNA-147),以管理糖尿病伤口中的炎症和氧化应激。此外,这些NPs中的多巴胺增强其内在化并保护miRNA-147免受氧化应激和RNase降解。最后,慢模块化学交联的MN促进去铁胺(DFO)和多巴胺的连续释放,在增殖和重塑阶段加速血管生成和组织再生。MNs内的锰/多巴胺增强的过氧化钙NPs引发类似爆炸的氧气气泡生成,不仅增强miRNA-mDTCNP和DFO的递送,而且减轻组织缺氧。因此,双模块MNs有助于通过愈合的所有阶段促进糖尿病伤口的快速和完全愈合。
    Diabetic wounds are characterized by the disruption and cessation of essential healing stages, which include hemostasis, inflammation, proliferation, and remodeling. However, traditional treatments for diabetic wounds concentrate on individual stages of the healing process. Herein, this study utilizes mask-mediated sequential polymerization and varied cross-linking techniques to develop dual-modular microneedles (MNs) with fast- and slow-module, exhibiting varying degradation rates tailored for the full spectrum of diabetic wound healing. First, MNs incorporating calcium ions and dopamine synergistically promote rapid hemostasis. Second, fast-module physically cross-linked MNs rapidly D-mannose/dopamine-enhanced tripolyphosphate-quaternized chitosan (mDTC) nanoparticles (NPs) loaded with microRNA-147 (miRNA-147) to manage inflammation and oxidative stress in diabetic wounds. Additionally, dopamine in these NPs enhances their internalization and safeguards miRNA-147 from oxidative stress and RNase degradation. Finally, slow-module chemically cross-linked MNs facilitate the continuous release of deferoxamine (DFO) and dopamine, accelerating angiogenesis and tissue regeneration during the proliferation and remodeling stages. Manganese/dopamine-enhanced calcium peroxide NPs within the MNs initiate a blast-like generation of oxygen bubbles, not only enhancing the delivery of miRNA-mDTC NPs and DFO but also alleviating tissue hypoxia. Consequently, dual-modular MNs are instrumental in promoting rapid and complete healing of diabetic wounds through all stages of healing.
    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
    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
    背景:前列腺癌(PCa)在全球男性中发病率很高,几乎所有PCa患者都进展到雄激素非依赖性阶段,缺乏有效的治疗措施。PTENP1,一种长非编码RNA,已显示通过竞争性内源性RNA(ceRNA)机制挽救PTEN表达来抑制肿瘤生长。然而,PTENP1由于酶的快速降解而被限制在PCa的处理中,细胞内摄取差,和过长的碱基序列要合成。考虑到人工纳米材料在药物装载和运输方面的独特优势,本研究采用黑磷(BP)纳米片作为基因药物载体。
    结果:以PTENP1序列为模板,随机分成4个长度约1000个核苷酸碱基的片段,合成4个不同的RNA片段作为基因药物,并加载到聚乙烯亚胺(PEI)修饰的BP纳米片上以构建BP-PEI@RNA递送平台。RNA可以通过BP-PEI纳米片有效地递送到PC3细胞中,并通过靶向PTENmRNA的竞争性结合microRNA(miRNA)提高PTEN表达,最终发挥抗肿瘤作用。
    结论:因此,这项研究表明,BP-PEI@RNA是PCa治疗的一个有前途的基因治疗平台。
    BACKGROUND: Prostate cancer (PCa) has a high incidence in men worldwide, and almost all PCa patients progress to the androgen-independent stage which lacks effective treatment measures. PTENP1, a long non-coding RNA, has been shown to suppress tumor growth through the rescuing of PTEN expression via a competitive endogenous RNA (ceRNA) mechanism. However, PTENP1 was limited to be applied in the treatment of PCa for the reason of rapid enzymatic degradation, poor intracellular uptake, and excessively long base sequence to be synthesized. Considering the unique advantages of artificial nanomaterials in drug loading and transport, black phosphorus (BP) nanosheet was employed as a gene-drug carrier in this study.
    RESULTS: The sequence of PTENP1 was adopted as a template which was randomly divided into four segments with a length of about 1000 nucleotide bases to synthesize four different RNA fragments as gene drugs, and loaded onto polyethyleneimine (PEI)-modified BP nanosheets to construct BP-PEI@RNA delivery platforms. The RNAs could be effectively delivered into PC3 cells by BP-PEI nanosheets and elevating PTEN expression by competitive binding microRNAs (miRNAs) which target PTEN mRNA, ultimately exerting anti-tumor effects.
    CONCLUSIONS: Therefore, this study demonstrated that BP-PEI@RNAs is a promising gene therapeutic platform for PCa treatment.
    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
    mRNA治疗的想法在新冠肺炎大流行期间成为现实之前已经构思了几十年。mRNA疫苗成为对抗新的病毒感染的强大而通用的工具,很大程度上是由于其多功能性和快速发展。除了预防性疫苗,mRNA技术也为作为通用药物形式的新应用提供了巨大的希望。然而,实现概念潜力面临相当大的挑战,比如最小的免疫刺激,高和长期表达,并有效递送到靶细胞和组织。在这里,我们回顾了基于mRNA的疗法的应用,强调创新设计和未来的挑战/解决方案。此外,我们还讨论了下一代mRNA治疗,包括环状mRNA和自扩增RNA。我们旨在提供预防性疫苗以外的mRNA治疗的概念概述和展望。
    The idea of mRNA therapy had been conceived for decades before it came into reality during the Covid-19 pandemic. The mRNA vaccine emerges as a powerful and general tool against new viral infections, largely due to its versatility and rapid development. In addition to prophylactic vaccines, mRNA technology also offers great promise for new applications as a versatile drug modality. However, realizing the conceptual potential faces considerable challenges, such as minimal immune stimulation, high and long-term expression, and efficient delivery to target cells and tissues. Here we review the applications of mRNA-based therapeutics, with emphasis on the innovative design and future challenges/solutions. In addition, we also discuss the next generation of mRNA therapy, including circular mRNA and self-amplifying RNAs. We aim to provide a conceptual overview and outlook on mRNA therapeutics beyond prophylactic vaccines.
    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
    基于RNA的疗法已成为治疗各种疾病的有希望的方法,包括癌症,遗传性疾病,和传染病。然而,由于RNA分子易于降解和细胞摄取效率低下,因此将RNA分子递送到靶细胞中一直是主要挑战。为了克服这些障碍,基于DNA的纳米技术作为RNA疗法的潜在递送平台提供了前所未有的机会。由于其优异的特性,如可编程性和生物相容性,这些基于DNA的纳米结构,由组装成精确可编程结构的DNA分子组成,作为保护RNA有效载荷并将其递送到所需细胞目的地的理想建筑材料,已经引起了极大的关注。在这次审查中,我们重点介绍了三种基于DNA的纳米结构的设计和应用的当前进展:DNA折纸,与框架引导组装(FGA)相关的脂质纳米颗粒(LNP)技术,和用于递送RNA分子的DNA水凝胶。简要讨论了它们的生物医学应用,并强调了该领域的挑战和未来前景。
    RNA-based therapeutics have emerged as a promising approach for the treatment of various diseases, including cancer, genetic disorders, and infectious diseases. However, the delivery of RNA molecules into target cells has been a major challenge due to their susceptibility to degradation and inefficient cellular uptake. To overcome these hurdles, DNA-based nano technology offers an unprecedented opportunity as a potential delivery platform for RNA therapeutics. Due to its excellent characteristics such as programmability and biocompatibility, these DNA-based nanostructures, composed of DNA molecules assembled into precise and programmable structures, have garnered significant attention as ideal building materials for protecting and delivering RNA payloads to the desired cellular destinations. In this review, we highlight the current progress in the design and application of three DNA-based nanostructures: DNA origami, lipid-nanoparticle (LNP) technology related to frame guided assembly (FGA), and DNA hydrogel for the delivery of RNA molecules. Their biomedical applications are briefly discussed and the challenges and future perspectives in this field are also highlighted.
    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
    核糖核酸(RNA)疗法在癌症治疗中提供了广阔的前景。然而,它们的成功应用需要克服各种生理障碍以有效地将RNA递送到靶位点。目前,开发了许多基于聚合物纳米颗粒的RNA递送系统来克服RNA递送中的这些障碍。这项工作概述了用于癌症基因治疗的现有RNA疗法,并特别总结了那些正在进入临床阶段的人。然后,本文讨论了基于肿瘤微环境的病理特征设计的基于肿瘤微环境响应性聚合物的RNA递送载体的核心特征和最新研究进展。最后,这项工作还提出了在临床应用中将RNA疗法转化为癌症免疫治疗方法的机会.
    Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
    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
    生物活性核酸的跨膜递送是细胞中的重要过程,并激发了人们发展先进的药物递送技术。在这一贡献中,据报道,分子水平的单链核酸跨膜载体基于3.2nm长的Huc折叠器(AornQ3Q3)8和(mQ3Q2)8,带有线性和螺旋排列的正电荷,分别。这两个折叠体不仅通过静电相互作用显示出非常强的DNA亲和力,而且还可以区别地结合单链DNA(ss-DNA)和双链DNA(ds-DNA)。证实了静电相互作用中精确电荷排列的重要性。更重要的是,这两个折叠器能够有效地运输SS-DNA穿过脂质膜,并且具有线性排列电荷的(AOrnQ3Q3)8的ss-DNA转运活性高于具有螺旋排列电荷的(mQ3Q2)8。因此,引入了一种基于带正电荷的螺旋折叠器的新型单链核酸跨膜分子载体。Further,EGFP-mRNA转染实验中有效和增强的表达强烈证明了带正电荷的foldamer用于RNA跨膜转运和治疗的潜力。
    Transmembrane delivery of biologically active nucleic acids is an important process in cells and has inspired one to develop advanced drug delivery techniques. In this contribution, molecular-level single-stranded nucleic acid transmembrane carriers are reported based on 3.2 nm long Huc\'s foldamers (AOrnQ3Q3)8 and (mQ3Q2)8 with linearly and helically aligned positive charges, respectively. These two foldamers not only show very strong DNA affinity via electrostatic interactions but also discriminatively bind single-stranded DNA (ss-DNA) and double-stranded DNA (ds-DNA), corroborating the importance of precise charge arrangement in the electrostatic interactions. More importantly, these two foldamers are capable of efficiently transporting ss-DNA across the lipid membranes, and the ss-DNA transport activity of (AOrnQ3Q3)8 with linearly aligned charges is higher than that of (mQ3Q2)8 with helically aligned charges. Thus a type of novel single-stranded nucleic acid transmembrane molecular carriers based on positively charged helical foldamers are introduced. Further, effective and enhanced expression in EGFP-mRNA transfection experiments strongly demonstrates the potential of positively charged foldamers for RNA transmembrane transport and therapy.
    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
    核糖核酸(RNA)能够抑制疾病相关基因,表达式,和编辑,并代表各种疾病的有前途的治疗方法。RNA的功效在很大程度上依赖于安全有效的递送系统的存在。在这里,我们发现RNA可以通过阳离子脂质和可电离脂质疏水化,并方便地与两亲性聚合物共组装以获得胶束样纳米颗粒(MNP)。研究结果表明MNP在递送RNA方面表现出高水平的效率。此外,在黑色素瘤肿瘤模型中,封装靶向CD47和PD-L1的siRNA的MNP显著阻断了这些免疫检查点,并引发了强烈的免疫应答.此外,包裹OVAmRNA的MNP实现了抗原翻译和呈递,导致针对表达OVA的黑色素瘤模型的有效抗肿瘤免疫预防结果。我们的研究结果表明,RNA疏水化可以作为递送RNA的可行方法,从而促进RNA疗法在疾病治疗中的探索。
    Ribonucleic acids (RNAs) enable disease-related gene inhibition, expression, and editing and represent promising therapeutics in various diseases. The efficacy of RNA relies heavily on the presence of a secure and effective delivery system. Herein, we found that RNA could be hydrophobized by cationic lipid and ionizable lipid and conveniently coassemble with amphiphilic polymer to achieve micelle-like nanoparticles (MNP). The results of the study indicate that MNP exhibits a high level of efficiency in delivering RNA. Besides, the MNP encapsulating siRNA that targets CD47 and PD-L1 remarkably blocked these immune checkpoints in a melanoma tumor model and elicited a robust immune response. Moreover, the MNP encapsulating the mRNA of OVA achieved antigen translation and presentation, leading to an effective antitumor immunoprophylaxis outcome against OVA-expressing melanoma model. Our findings suggest that RNA hydrophobization could serve as a viable approach for delivering RNA, thereby facilitating the exploration of RNA therapy in disease treatment.
    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
    核糖核酸(RNA)药物在临床和临床前研究中对各种疾病显示出有希望的治疗效果,由于它们能够调节目的基因的表达或控制蛋白质合成。不同的策略,如化学改性,配体缀合,和纳米技术,为RNA医学的成功临床翻译做出了贡献,包括用于基因沉默的小干扰RNA(siRNA)和用于疫苗开发的信使RNA(mRNA)。其中,纳米技术可以保护RNA免受酶降解,增加细胞摄取和胞质运输,延长体循环,并改善组织/细胞靶向。这里,集中概述了用于RNA递送的刺激响应纳米技术,它们在促进RNA生物活性和细胞/器官选择性方面显示出独特的益处,提供。许多组织/细胞特异性微环境特征,如pH值,酶,缺氧,和氧化还原,用于设计内部刺激响应性RNA纳米颗粒(NP)。此外,外部刺激,如光,磁场,还有超声波,还用于控制RNA的释放和运输。这篇综述总结了用于RNA递送的各种刺激响应性NP系统,这可能会促进下一代RNA药物的开发。
    Ribonucleic acid (RNA) drugs have shown promising therapeutic effects for various diseases in clinical and preclinical studies, owing to their capability to regulate the expression of genes of interest or control protein synthesis. Different strategies, such as chemical modification, ligand conjugation, and nanotechnology, have contributed to the successful clinical translation of RNA medicine, including small interfering RNA (siRNA) for gene silencing and messenger RNA (mRNA) for vaccine development. Among these, nanotechnology can protect RNAs from enzymatic degradation, increase cellular uptake and cytosolic transportation, prolong systemic circulation, and improve tissue/cell targeting. Here, a focused overview of stimuli-responsive nanotechnologies for RNA delivery, which have shown unique benefits in promoting RNA bioactivity and cell/organ selectivity, is provided. Many tissue/cell-specific microenvironmental features, such as pH, enzyme, hypoxia, and redox, are utilized in designing internal stimuli-responsive RNA nanoparticles (NPs). In addition, external stimuli, such as light, magnetic field, and ultrasound, have also been used for controlling RNA release and transportation. This review summarizes a wide range of stimuli-responsive NP systems for RNA delivery, which may facilitate the development of next-generation RNA medicines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号