endosome escape

内体逃逸
  • 文章类型: Journal Article
    免疫毒素(IT)是重组嵌合蛋白,其将蛋白毒素与靶向部分组合以促进毒素向癌细胞的选择性递送。这里,我们提出了一种新的策略,通过在内吞途径的还原条件下促进IT与靶受体的解离来增强IT的细胞溶质进入。我们设计了单体SS,具有含二硫键(SS)的互补位的基于人纤连蛋白III型结构域的单体,靶向受体如EGFR,EpCAM,Her2和FAP。单体SS表现出SS依赖性靶受体结合,在还原条件下结合显著降低。然后,我们创建了基于单体SS的ITs,携带假单胞菌外毒素A(PE25)的25kDa片段,称为单体SS-PE25。这些IT显示针对表达靶受体的癌细胞的剂量依赖性细胞毒性和更宽的治疗窗口,这是由于与SS减少被抑制的对照相比在更低剂量下更高的功效。ERSS/28-PE25,EGFR的KD为28nM,与缺乏SS键的ER/21-PE25相比,显示出更高的肿瘤杀伤能力,在等效和较低剂量。在体内,在EGFR过表达的异种移植小鼠模型中,ERSS/28-PE25在抑制肿瘤生长方面优于ER/21-PE25。这项研究提出了一种使用含SS的互补体开发靶向实体瘤的IT的策略,以增强胞浆递送和抗肿瘤功效。
    Immunotoxins (ITs) are recombinant chimeric proteins that combine a protein toxin with a targeting moiety to facilitate the selective delivery of the toxin to cancer cells. Here, we present a novel strategy to enhance the cytosolic access of ITs by promoting their dissociation from target receptors under the reducing conditions of the endocytic pathway. We engineered monobodySS, a human fibronectin type III domain-based monobody with disulfide bond (SS)-containing paratopes, targeting receptors such as EGFR, EpCAM, Her2, and FAP. MonobodySS exhibited SS-dependent target receptor binding with a significant reduction in binding under reducing conditions. We then created monobodySS-based ITs carrying a 25 kDa fragment of Pseudomonas exotoxin A (PE25), termed monobodySS-PE25. These ITs showed dose-dependent cytotoxicity against target receptor-expressing cancer cells and a wider therapeutic window due to higher efficacy at lower doses compared to controls with SS reduction inhibited. ERSS/28-PE25, with a KD of 28 nM for EGFR, demonstrated superior tumor-killing potency compared to ER/21-PE25, which lacks an SS bond, at equivalent and lower doses. In vivo, ERSS/28-PE25 outperformed ER/21-PE25 in suppressing tumor growth in EGFR-overexpressing xenograft mouse models. This study presents a strategy for developing solid tumor-targeting ITs using SS-containing paratopes to enhance cytosolic delivery and antitumor efficacy.
    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(LNP)代表了RNA分子的先进和高效的递送系统,展示了卓越的生物相容性和显着的输送效率。三种LNP制剂的临床授权证明了这一点:Patisiran,BNT162b2和mRNA-1273。为了进一步最大化基于RNA的治疗的功效,必须开发更有效的LNP递送系统,该系统可以有效地保护固有的不稳定和带负电荷的RNA分子免受核酸酶的降解,同时促进它们的细胞摄取到靶细胞中。因此,这篇综述提出了开发高效LNP传输系统常用的可行策略。这些策略包括组合化学,合理的设计方法,功能分子的衍生策略,LNP配方的优化,以及LNP的粒径和电荷性质的调整。在介绍这些发展战略之前,LNP的体内递送过程,影响LNP制剂临床翻译的关键决定因素,描述是为了更好地理解如何开发LNP交付系统。
    Lipid nanoparticles (LNPs) represent an advanced and highly efficient delivery system for RNA molecules, demonstrating exceptional biocompatibility and remarkable delivery efficiency. This is evidenced by the clinical authorization of three LNP formulations: Patisiran, BNT162b2, and mRNA-1273. To further maximize the efficacy of RNA-based therapy, it is imperative to develop more potent LNP delivery systems that can effectively protect inherently unstable and negatively charged RNA molecules from degradation by nucleases, while facilitating their cellular uptake into target cells. Therefore, this review presents feasible strategies commonly employed for the development of efficient LNP delivery systems. The strategies encompass combinatorial chemistry, a rational design approach, the derivatization strategy of functional molecules, the optimization of LNP formulations, and adjustment of particle size and charge property of LNPs. Prior to introducing these developing strategies, in vivo delivery processes of LNPs, a crucial determinant influencing the clinical translation of LNP formulations, is described to better understand how to develop LNP delivery systems.
    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
    在这项研究中,我们开发了组氨酸寡聚体(oHis;10mer)-掺入LNP(H10LNP)作为高效siRNA递送的新型载体。值得注意的是,未修饰的oHis(10mer)通过与siRNA的离子相互作用大量掺入LNP中,作为内体逃逸增强剂。H10LNP的大小约为65nm,表现出显著增强的内体逃逸程度,正如钙黄绿素测定和细胞内荧光的共聚焦显微镜图像所证明的那样,超越传统的LNP。此外,在H10LNP处理的细胞中,人内源性球形神经酰胺合酶(Gb3合酶)基因的半抑制浓度(IC50)显着降低了三倍,与LNP处理的细胞相比。值得注意的是,H10LNP在体外和体内均保持相当的生物相容性和生物分布。考虑到制造的siRNAH10LNP表现出优异的生物相容性和优于常规LNP的基因沉默活性,这些颗粒可用于安全递送治疗性siRNA.此外,这项研究引入了有希望的,可行,简单,以及用于将未修饰的功能性阳离子肽整合到LNP中以增强宽范围的基于核酸的药物的递送效率的替代配制方法。本文受版权保护。保留所有权利。
    In this study, histidine oligomer (oHis; 10mer)-incorporating LNPs (H10LNPs) are developed as a novel carrier for efficient siRNA delivery. Notably, the unmodified oHis (10mer) is greatly incorporated within LNPs through ionic interaction with siRNAs, which serves as an endosome escape enhancer. H10LNPs with a size of ≈65 nm demonstrate a significantly enhanced extent of endosomal escape, as evidenced by calcein assay and confocal microscopy images of intracellular fluorescence, surpassing conventional LNPs. Furthermore, the half inhibitory concentration (IC50) of the human endogenous globotriaosylceramide synthase (Gb3 synthase) gene in H10LNPs-treated cells exhibits a significant threefold decrease, compared to that in LNP-treated cells. Notably, H10LNPs maintain comparable biocompatibility and biodistribution both in vitro and in vivo. Considering that the fabricated siRNA H10LNPs exhibit excellent biocompatibility and superior gene silencing activity over conventional LNPs, these particles can be harnessed for the safe delivery of therapeutic siRNAs. Additionally, this study introduces promising, feasible, simple, and alternative formulation processes for integrating unmodified functional cationic peptides into LNPs to enhance the delivery efficiency of a wide range of nucleic acid-based drugs.
    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
    遗传药物的药用特性高度依赖于递送系统的设计。可电离的阳离子脂质被认为是递送系统中的核心材料。然而,关于脂质结构设计的广泛多样性与基因传递效率之间的关系,尚未达成广泛共识。研究工作的目的是合成可电离的胆固醇衍生物(iChol-lipides)并评估其作为基因递送载体的潜在应用。用氨基甲酸酯键间隔基合成了一系列具有不同头基的iChol-脂质。化学结构经1HNMR表征,MS,熔化范围,和pKa。通过分子动力学模拟研究了iChol-脂质与MALAT1-siRNA之间的相互作用,并与市场上可获得的DC-Chol进行了比较。这揭示了氢键,盐桥,可能涉及静电相互作用。在存在不同辅助脂质和聚乙二醇化脂质的情况下,通过动态激光散射对这些脂质的自组装行为进行了深入研究和评估。它们的质粒结合能力,转染效率,溶血毒性,和细胞毒性进行了充分的研究。IZ-Chol-LNP被证明具有与DNA有效复合的高度潜力,通过pH敏感的荧光探针BCFL验证了质子海绵效应介导的内体逃逸机制。
    ABSTACTThe medicinal properties of genetic drugs are highly dependent on the design of delivery systems. Ionizable cationic lipids are considered core materials in delivery systems. However, there has not yet been a widespread consensus on the relationship between the wide diversity of lipid structure design and gene delivery efficiency. The aims of the research work were to synthesize ionizable cholesterol derivatives (iChol-lipids) and to evaluate their potential applications as gene delivery vector. A series of iChol-lipids with different head groups were synthesized with carbamate bond spacer. The chemical structures were characterized by 1H NMR, MS, melting range, and pKa. The interactions between iChol-lipids and MALAT1-siRNA were studied by molecular dynamics simulations and compared with market available DC-Chol, which revealed that hydrogen bonds, salt-bridge, and electrostatic interaction were probably involved. The self-assemble behaviors of these lipids were intensively investigated and evaluated by dynamic laser scattering in the presence of different helper lipids and PEGylated lipids. Their plasmid binding ability, transfection efficiency, hemolytic toxicity, and cytotoxicity were fully studied. IZ-Chol-LNPs was proved to be highly potential to effectively complex with DNA, and endosome escape mechanisms mediated by proton sponge effect was verified by pH-sensitive fluorescence probe BCFL.
    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
    细胞屏障是生物活性化合物进入细胞以完成其生物学功能的主要瓶颈。这限制了它们的生物医学应用。纳米载体已经证明了通过克服细胞内屏障的级联来包封生物活性化合物并有效地将其递送到靶细胞中以实现期望的治疗和诊断效果的高潜力和益处。在这次审查中,我们在药物输送和纳米载体之前引入了细胞屏障,以及总结纳米载体增加细胞内化的最新进展和策略,促进细胞内贩运,克服耐药性,靶向亚细胞位置和控制药物释放。最后,讨论了纳米载体用于细胞内药物递送的未来前景,主要关注潜在挑战和未来方向。我们的综述概述了纳米载体的细胞内药物递送,这可能会鼓励纳米载体的未来发展,以有效和精确地将药物递送到广泛的细胞和亚细胞靶标中。
    The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
    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
    RNA疗法可以操纵基因表达或蛋白质产生,使它们适合治疗各种疾病。理论上,任何具有明确生物学靶标的疾病都可能从基于RNA的疗法中找到可行的治疗方法。许多使用RNA疗法对抗癌症的临床试验,传染病或遗传性疾病已被报道,并取得了理想的治疗效果。到目前为止,来自各种动物实验研究的令人鼓舞的发现也证实了基于RNA的疗法在治疗风湿性关节炎(RA)方面的巨大潜力.然而,体内多种生理屏障仍然严重损害RNA药物的治疗效果。因此,RNA治疗的安全和有效的递送策略对于它们在RA治疗中的进一步和广泛应用是非常必要的.在这次审查中,我们将讨论使用基于RNA的疗法取得的最新进展,并重点讨论可以克服RA治疗中体内递送障碍的递送策略。此外,本文还讨论了目前用于RA治疗的RNA递送系统中存在的问题.
    RNA therapeutics can manipulate gene expression or protein production, making them suitable for treating a wide range of diseases. Theoretically, any disease that has a definite biological target would probably find feasible therapeutic approach from RNA-based therapeutics. Numerous clinical trials using RNA therapeutics fighting against cancer, infectious diseases or inherited diseases have been reported and achieved desirable therapeutic efficacy. So far, encouraging findings from various animal experimental studies have also confirmed the great potential of RNA-based therapies in the treatment of rheumatic arthritis (RA). However, the in vivo multiple physiological barriers still seriously compromise the therapeutic efficacy of RNA drugs. Thus, safe and effective delivery strategies for RNA therapeutics are quite essential for their further and wide application in RA therapy. In this review, we will discuss the recent progress achieved using RNA-based therapeutics and focus on delivery strategies that can overcome the in vivo delivery barriers in RA treatment. Furthermore, discussion about the existing problems in current RNA delivery systems for RA therapy has been also included here.
    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
    信使RNA(mRNA)是蛋白质生物合成的模板,正在成为对抗各种疾病的重要活性分子,包括病毒感染和癌症.尤其是,基于mRNA的疫苗,作为一种新型疫苗,在抗击当前新冠肺炎全球大流行方面发挥了主导作用。然而,固有的缺点,包括大尺寸,负电荷,和不稳定性,阻碍其作为治疗剂的使用。脂质载体是mRNA递送的可区分和有前途的载体,拥有封装并将带负电荷的药物递送到目标组织并在所需时间释放货物的能力。这里,首先总结了不同脂质载体的结构和性质,如脂质体,脂质体状纳米颗粒,固体脂质纳米粒,脂质-聚合物混合纳米颗粒,纳米乳液,外泌体和脂蛋白颗粒,以及它们在传递mRNA中的应用。然后,讨论并强调了基于脂质的制剂作为疫苗递送系统的发展.强调了COVID-19mRNA疫苗的最新进展。最后,我们描述了我们在这一领域的未来愿景和观点。
    Messenger RNA (mRNA) is the template for protein biosynthesis and is emerging as an essential active molecule to combat various diseases, including viral infection and cancer. Especially, mRNA-based vaccines, as a new type of vaccine, have played a leading role in fighting against the current global pandemic of COVID-19. However, the inherent drawbacks, including large size, negative charge, and instability, hinder its use as a therapeutic agent. Lipid carriers are distinguishable and promising vehicles for mRNA delivery, owning the capacity to encapsulate and deliver negatively charged drugs to the targeted tissues and release cargoes at the desired time. Here, we first summarized the structure and properties of different lipid carriers, such as liposomes, liposome-like nanoparticles, solid lipid nanoparticles, lipid-polymer hybrid nanoparticles, nanoemulsions, exosomes and lipoprotein particles, and their applications in delivering mRNA. Then, the development of lipid-based formulations as vaccine delivery systems was discussed and highlighted. Recent advancements in the mRNA vaccine of COVID-19 were emphasized. Finally, we described our future vision and perspectives in this field.
    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
    我们先前证明了基于聚(dA)和裂孔多糖形成的复合物的反义寡核苷酸(AS-ODN)递送系统,一种β-1,3-葡聚糖。该复合物使得AS-ODN的有效细胞内递送成为可能。在这份通讯中,我们研究了复合物本身的细胞质易位及其对mRNA的作用机制。因此,我们发现复合体在保持其结构的同时移入细胞质,AS-ODN与靶mRNA杂交。该结果促进了复合物的药物应用。
    We previously demonstrated antisense oligonucleotides (AS-ODNs) delivery system based on the complex formed with poly (dA) and schizophyllan, a type of β-1,3-glucan. This complex enables efficient intracellular delivery of AS-ODNs. In this communication, we investigated the cytoplasmic translocation of the complex itself and its mechanism of action on mRNA. As a result, we found that the complex moved into the cytoplasm while keeping its structure, and AS-ODN hybridized with the target mRNA. This result encourages pharmaceutical applications of the complex.
    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
    在肝窦内皮细胞上表达的非DNA结合稳定剂-2/HARE受体特异性结合并内化几类硫代磷酸酯反义寡核苷酸(PS-ASO)。稳定蛋白介导的摄取后,PS-ASO在内体(>97%-99%)内贩运,最终导致溶酶体的破坏。内体中的ASO截留降低了治疗功效,从而增加患者的总剂量。这里,我们使用共聚焦显微镜来表征稳定蛋白受体介导的稳定重组稳定蛋白-1和-2细胞系摄取后PS-ASO的细胞内途径。我们发现PS-ASO以及Stabilin-2受体横贯经典路径:网格蛋白包被的囊泡-早期内体-晚期内体-溶酶体。与未处理的细胞相比,氯喹暴露促进了PS-ASO的内体逃逸,导致目标敲低50%以上。导致PS-ASO功效增加。我们还将细胞溶质半乳糖凝集素描述为PS-ASO逃逸的新贡献者。Galectin敲低可通过调节EEA1、Rab5C、和Rab7AmRNA表达,导致内体囊泡成熟过程延迟。总的来说,我们的结果为通过增强内体逃逸来增加PS-ASO功效提供了额外的见解,其可以进一步用于其他基于核酸的模态。
    Non-DNA-binding Stabilin-2/HARE receptors expressed on liver sinusoidal endothelial cells specifically bind to and internalize several classes of phosphorothioate antisense oligonucleotides (PS-ASOs). After Stabilin-mediated uptake, PS-ASOs are trafficked within endosomes (>97%-99%), ultimately resulting in destruction in the lysosome. The ASO entrapment in endosomes lowers therapeutic efficacy, thereby increasing the overall dose for patients. Here, we use confocal microscopy to characterize the intracellular route transverse by PS-ASOs after Stabilin receptor-mediated uptake in stable recombinant Stabilin-1 and -2 cell lines. We found that PS-ASOs as well as the Stabilin-2 receptor transverse the classic path: clathrin-coated vesicle-early endosome-late endosome-lysosome. Chloroquine exposure facilitated endosomal escape of PS-ASOs leading to target knockdown by more than 50% as compared to untreated cells, resulting in increased PS-ASO efficacy. We also characterize cytosolic galectins as novel contributor for PS-ASO escape. Galectins knockdown enhances ASO efficacy by more than 60% by modulating EEA1, Rab5C, and Rab7A mRNA expression, leading to a delay in the endosomal vesicle maturation process. Collectively, our results provide additional insight for increasing PS-ASO efficacy by enhancing endosomal escape, which can further be utilized for other nucleic acid-based modalities.
    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
    基于细胞外囊泡(EV)的低密度脂蛋白受体(Ldlr)mRNA递送在治疗家族性高胆固醇血症(FH)方面显示出出色的治疗效果。然而,基于EV的mRNA递送的装载低效率提出了重大挑战。最近,RNA结合蛋白(RBP)已经与EV膜蛋白融合,用于选择性地包封靶向RNA以提高加载效率。然而,治疗性RNA和RBP之间的强相互作用阻止RNA从内体释放到受体细胞中的胞质溶胶。在这项研究中,开发了一种改进的策略,用于将LdlrmRNA有效封装到供体细胞的EV中,并在受体细胞中可控释放.方法:MS2噬菌体外壳蛋白(CD9-MCP)融合蛋白,LdlrmRNA,并且在供体细胞中表达与LdlrmRNA匹配的定制的包含RNA适体碱基对的MS2。接受上述治疗性EV的细胞同时用含有“Ldlr释放剂”的EV处理,其序列与LdlrmRNA中的识别位点相似。在通过尾静脉接受EV治疗的Ldlr-/-小鼠中分析治疗效果。结果:体外实验表明,通过MS2-MCP相互作用,LdlrmRNA在EV中的加载效率提高。“Ldlr释放剂”的处理与MS2适体竞争性相互作用,具有更高的亲和力,并从CD9-MCP释放LdlrmRNA以进行有效翻译。当组合EV被递送到受体肝细胞时,稳健的LDLR表达在Ldlr-/-小鼠中提供了治疗益处。结论:我们提出了一种基于EV的mRNA递送策略,用于增强治疗性mRNA在EV中的包封,并将RNA释放到细胞质中,以在具有基因治疗潜力的受体细胞中进行翻译。
    Extracellular vesicle (EV)-based low-density lipoprotein receptor (Ldlr) mRNA delivery showed excellent therapeutic effects in treating familial hypercholesterolemia (FH). Nevertheless, the loading inefficiency of EV-based mRNA delivery presents a significant challenge. Recently, RNA-binding proteins (RBPs) have been fused to EV membrane proteins for selectively encapsulating targeted RNAs to promote loading efficiency. However, the strong interaction between therapeutic RNAs and RBPs prevents RNA release from endosomes to the cytosol in the recipient cells. In this study, an improved strategy was developed for efficient encapsulation of Ldlr mRNA into EVs in donor cells and controllable release in recipient cells. Methods: The MS2 bacteriophage coat protein (CD9-MCP) fusion protein, Ldlr mRNA, and a customized MS2 containing RNA aptamer base-pair matched with Ldlr mRNA were expressed in donor cells. Cells receiving the above therapeutic EVs were simultaneously treated with EVs containing \"Ldlr releaser\" with a sequence similar to the recognition sites in Ldlr mRNA. Therapeutic effects were analyzed in Ldlr-/- mice receiving EV treatments via the tail vein. Results: In vitro experiments demonstrated improved loading efficiency of Ldlr mRNA in EVs via MS2-MCP interaction. Treatment of \"Ldlr releaser\" competitively interacted with MS2 aptamer with higher affinity and released Ldlr mRNA from CD9-MCP for efficient translation. When the combinatory EVs were delivered into recipient hepatocytes, the robust LDLR expression afforded therapeutic benefits in Ldlr-/- mice. Conclusion: We proposed an EV-based mRNA delivery strategy for enhanced encapsulation of therapeutic mRNAs in EVs and RNA release into the cytosol for translation in recipient cells with great potential for gene therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号