RNA delivery

RNA 递送
  • 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
    脂质纳米颗粒(LNPs)已成功进入临床,用于递送基于mRNA和siRNA的治疗剂,最近作为COVID-19的疫苗。然而,对它们的体内行为缺乏了解,特别是细胞靶向。这种LNP向性的一部分是基于内源性蛋白质对颗粒表面的粘附。这种蛋白质形成所谓的电晕,可以改变,除其他外,循环时间,这些颗粒的生物分布和细胞摄取。这个蛋白质日冕的形成,反过来,取决于纳米粒子的性质(例如,尺寸,charge,表面化学和疏水性)以及衍生它的生物环境。基因疗法有可能针对几乎任何疾病,考虑静脉途径以外的给药部位,导致组织特异性蛋白电晕。对于神经系统疾病,LNP的颅内给药导致脑脊髓液衍生的蛋白质电晕,与静脉内给药相比,可能改变脂质纳米颗粒的性质。这里,在体外研究了临床相关的LNP制剂上血浆和CSF衍生的蛋白冠之间的差异.蛋白质分析显示在人CSF中孵育的LNP(C-LNP)产生与在血浆中孵育的LNP(P-LNP)不同的蛋白质冠组合物。脂蛋白作为一个整体,但特别是载脂蛋白E,C-LNP上的总蛋白冠百分比高于P-LNP。与P-LNP相比,这导致C-LNP的细胞摄取改善,不管细胞起源。重要的是,更高的LNP吸收并没有直接转化为更有效的货物运输,强调有必要进一步评估这种机制。这些发现表明,生物流体特异性蛋白冠改变LNP功能,提示给药部位可能会影响LNP的体内疗效,需要在制剂开发过程中加以考虑。
    Lipid nanoparticles (LNPs) have successfully entered the clinic for the delivery of mRNA- and siRNA-based therapeutics, most recently as vaccines for COVID-19. Nevertheless, there is a lack of understanding regarding their in vivo behavior, in particular cell targeting. Part of this LNP tropism is based on the adherence of endogenous protein to the particle surface. This protein forms a so-called corona that can change, amongst other things, the circulation time, biodistribution and cellular uptake of these particles. The formation of this protein corona, in turn, is dependent on the nanoparticle properties (e.g., size, charge, surface chemistry and hydrophobicity) as well as the biological environment from which it is derived. With the potential of gene therapy to target virtually any disease, administration sites other than intravenous route are considered, resulting in tissue specific protein coronas. For neurological diseases, intracranial administration of LNPs results in a cerebral spinal fluid derived protein corona, possibly changing the properties of the lipid nanoparticle compared to intravenous administration. Here, the differences between plasma and CSF derived protein coronas on a clinically relevant LNP formulation were studied in vitro. Protein analysis showed that LNPs incubated in human CSF (C-LNPs) developed a protein corona composition that differed from that of LNPs incubated in plasma (P-LNPs). Lipoproteins as a whole, but in particular apolipoprotein E, represented a higher percentage of the total protein corona on C-LNPs than on P-LNPs. This resulted in improved cellular uptake of C-LNPs compared to P-LNPs, regardless of cell origin. Importantly, the higher LNP uptake did not directly translate into more efficient cargo delivery, underlining that further assessment of such mechanisms is necessary. These findings show that biofluid specific protein coronas alter LNP functionality, suggesting that the site of administration could affect LNP efficacy in vivo and needs to be considered during the development of the formulation.
    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
    基因组编辑工具,特别是成簇的定期间隔短回文重复(CRISPR)系统(例如,CRISPR/Cas9),以及它们重新用于表观遗传编辑平台,作为安全和可定制的癌症疗法提供了巨大的潜力。具体来说,人类恶性肿瘤中的各种转录异常,如肿瘤抑制基因的沉默和癌基因的异位再表达,使用CRISPR激活和抑制系统已成功靶向,几乎没有脱靶效应。在这些系统中,例如,核酸酶失活的Cas9蛋白(dCas9)融合至诱导靶向基因的选择性激活或抑制的一个或多个结构域。尽管取得了这些进展,这些分子有效的体内递送到靶癌细胞中代表了完成转化为癌症临床治疗环境的关键障碍。主要障碍包括dCas9融合蛋白的大尺寸,蛋白质和gRNA的多模态递送的必要性,以及这些制剂引发有害免疫反应的潜力。在这种情况下,提供CRISPR的病毒方法面临几个限制,例如病毒基因组的包装能力,核酸整合到宿主细胞基因组中的潜力,和病毒蛋白的免疫原性,带来严重的安全问题。响应COVID-19大流行的mRNA疫苗的快速发展重新激发了人们对基于mRNA的CRISPR/dCas9递送方法的兴趣。同时,由于它们的高承载能力,可扩展性,用于细胞靶向的可定制表面修饰,低免疫原性,脂质纳米颗粒(LNP)作为非病毒载体已被广泛研究。在这一章中,我们首先描述了用于表观遗传编辑的优化的dCas9效应子mRNA和gRNA的设计。我们概述了适用于dCas9mRNA递送的LNP的制剂。此外,我们提供了将dCas9效应mRNA和gRNA共封装到这些LNP中的方案,以及将这些制剂递送至乳腺癌细胞系(体外)和小鼠模型(体内)的详细方法。
    Genome editing tools, particularly the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) systems (e.g., CRISPR/Cas9), and their repurposing into epigenetic editing platforms, offer enormous potential as safe and customizable therapies for cancer. Specifically, various transcriptional abnormalities in human malignancies, such as silencing of tumor suppressors and ectopic re-expression of oncogenes, have been successfully targeted with virtually no off-target effects using CRISPR activation and repression systems. In these systems, the nuclease-deactivated Cas9 protein (dCas9) is fused to one or more domains inducing selective activation or repression of the targeted genes. Despite these advances, the efficient in vivo delivery of these molecules into the target cancer cells represents a critical barrier to accomplishing translation into a clinical therapy setting for cancer. Major obstacles include the large size of dCas9 fusion proteins, the necessity of multimodal delivery of protein and gRNAs, and the potential of these formulations to elicit detrimental immune responses.In this context, viral methods for delivering CRISPR face several limitations, such as the packaging capacity of the viral genome, the potential for integration of the nucleic acids into the host cells genome, and immunogenicity of viral proteins, posing serious safety concerns. The rapid development of mRNA vaccines in response to the COVID-19 pandemic has rekindled interest in mRNA-based approaches for CRISPR/dCas9 delivery. Simultaneously, due to their high loading capacity, scalability, customizable surface modification for cell targeting, and low immunogenicity, lipid nanoparticles (LNPs) have been widely explored as nonviral vectors. In this chapter, we first describe the design of optimized dCas9-effector mRNAs and gRNAs for epigenetic editing. We outline formulations of LNPs suitable for dCas9 mRNA delivery. Additionally, we provide a protocol for the co-encapsulation of the dCas9-effector mRNAs and gRNA into these LNPs, along with detailed methods for delivering these formulations to both cell lines (in vitro) and mouse models of breast cancer (in vivo).
    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
    基于核酸的疗法正在改变医学,但依赖于有效的递送载体,例如脂质纳米颗粒(LNP)。关注存在于纳米医学领域,集合中的大部分LNP不包含任何核酸货物,因此不发挥功能作用。然而,如何LNP脂质配方,所采用的LNP制备方法和核酸货物大小与空LNP的比例相关仍未被探索。在这里,我们采用建立良好的基于单颗粒的方法来研究LNP中的核酸负载异质性。我们发现只有一小部分LNP是“空的”,对于装载siRNA的LNP,mRNA和质粒。对于mRNA递送的临床相关LNP,我们从未在集合中检测到超过16%的空纳米粒子。因此,采用标准LNP脂质-货物组合和制备方案导致LNP具有发挥其生物医学功能的潜力。
    Nucleic acid-based therapies are transforming medicine, but rely on an efficient delivery vehicle such as lipid nanoparticles (LNPs). Concerns exists in the nanomedicine field, that a large fraction of the LNPs in the ensemble does not contain any nucleic acid cargo and thus exert no functional effect. Nevertheless, how LNP lipid formulation, the LNP preparation method employed and nucleic acid cargo size correlates with the proportion of empty LNPs remains largely unexplored. Here we employ a well-established single particle based method to study nucleic acid loading heterogeneity in LNPs. We find that only a minor fraction of LNPs are \"empty\", both for LNPs loaded with siRNA, mRNA and plasmids. For clinically relevant LNPs for mRNA delivery, we never detected more than 16% empty nanoparticles in the ensemble. Thus employing standard LNP lipid-cargo combinations and preparation schemes results in LNPs with the potential to serve their biomedical function.
    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
    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)

公众号