gene silence

基因沉默
  • 文章类型: Journal Article
    化疗和基因治疗的结合对于肿瘤的治疗和根除具有巨大的前景。然而,由于化学治疗剂和功能性核酸药物之间的物理化学性质的显着差异,直接整合到单个纳米剂中受到阻碍,阻碍了协同抗肿瘤治疗的有效共递送纳米平台的设计和构建。在这项研究中,我们通过2\'-氟取代的针对P-糖蛋白的反义DNA(2\'F-DNA)和化疗药物紫杉醇(PTX)的直接自组装,开发了一种mRNA反应型二合一纳米药物,用于有效的抗肿瘤治疗.DNA的2'-氟修饰可以显着增加治疗性核酸与化疗药物之间的相互作用,促进2个F-DNA/PTX纳米球(2个F-DNA/PTXNSs)的成功形成。由于一步自组装过程无需额外的载体材料,制备的2个F-DNA/PTXNSs表现出相当大的负载效率和PTX的生物利用度。在内源性P-糖蛋白mRNA的存在下,分解了2个F-DNA/PTXNS。释放的2'F-DNA可以下调P-糖蛋白的表达,降低了肿瘤细胞的多药耐药性,增强了PTX引起的化疗效果。这样,2'F-DNA/PTXNSs可协同诱导肿瘤细胞凋亡,实现联合抗肿瘤治疗。该策略可能为探索具有高生物利用度的功能性细胞内共递送纳米系统提供新的工具,并在肿瘤的准确诊断和治疗中显示出潜在的应用前景。
    The combination of chemotherapy and gene therapy holds great promise for the treatment and eradication of tumors. However, due to significant differences in physicochemical properties between chemotherapeutic agents and functional nucleic acid drugs, direct integration into a single nano-agent is hindered, impeding the design and construction of an effective co-delivery nano-platform for synergistic anti-tumor treatments. In this study, we have developed an mRNA-responsive two-in-one nano-drug for effective anti-tumor therapy by the direct self-assembly of 2\'-fluoro-substituted antisense DNA against P-glycoprotein (2\'F-DNA) and chemo drug paclitaxel (PTX). The 2\'-fluoro modification of DNA could significantly increase the interaction between the therapeutic nucleic acid and the chemotherapeutic drug, promoting the successful formation of 2\'F-DNA/PTX nanospheres (2\'F-DNA/PTX NSs). Due to the one-step self-assembly process without additional carrier materials, the prepared 2\'F-DNA/PTX NSs exhibited considerable loading efficiency and bioavailability of PTX. In the presence of endogenous P-glycoprotein mRNA, the 2\'F-DNA/PTX NSs were disassembled. The released 2\'F-DNA could down-regulate the expression of P-glycoprotein, which decreased the multidrug resistance of tumor cells and enhanced the chemotherapy effect caused by PTX. In this way, the 2\'F-DNA/PTX NSs could synergistically induce the apoptosis of tumor cells and realize the combined anti-tumor therapy. This strategy might provide a new tool to explore functional intracellular co-delivery nano-systems with high bioavailability and exhibit potential promising in the applications of accurate diagnosis and treatment of tumors.
    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
    CXCL14是趋化因子家族中进化上最保守的成员之一,在多器官中表达。这表明它参与了系统的稳态维持。CXCL14在结肠上皮细胞中高表达,在临床结肠癌样本中表现出明显的基因沉默,表明其沉默与癌细胞的免疫逃逸有关。在本文中,我们分析了多个人类临床结肠癌数据集和小鼠结肠癌模型的表达谱,以揭示结肠炎期间CXCL14表达的变化趋势,结肠息肉,原发性结肠癌,和肝转移。通过结直肠癌甲基化数据库揭示CXCL14基因沉默与启动子高甲基化之间的关系。结果提示CXCL14是结直肠癌的抑癌基因,在肿瘤的发生和恶化过程中先被激活后被沉默。启动子甲基化是CXCL14沉默的主要原因。CXCL14的甲基化水平与肿瘤发生的解剖部位有关,与患者年龄呈正相关,并与预后相关。逆转CXCL14的过度甲基化可能是结肠癌的表观遗传疗法。
    CXCL14 is one of the most evolutionarily conserved members of the chemokine family and is constitutionally expressed in multiple organs, suggesting that it is involved in the homeostasis maintenance of the system. CXCL14 is highly expressed in colon epithelial cells and shows obvious gene silencing in clinical colon cancer samples, suggesting that its silencing is related to the immune escape of cancer cells. In this paper, we analyzed the expression profiles of multiple human clinical colon cancer datasets and mouse colon cancer models to reveal the variation trend of CXCL14 expression during colitis, colon polyps, primary colon cancer, and liver metastases. The relationship between CXCL14 gene silencing and promoter hypermethylation was revealed through the colorectal carcinoma methylation database. The results suggest that CXCL14 is a tumor suppressor gene in colorectal carcinoma which is activated first and then silenced during the process of tumor occurrence and deterioration. Promoter hypermethylation is the main cause of CXCL14 silencing. The methylation level of CXCL14 is correlated with the anatomic site of tumor occurrence, positively correlated with patient age, and associated with prognosis. Reversing the hypermethylation of CXCL14 may be an epigenetic therapy for colon 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
    基因编辑技术(GET)可以诱导基因敲低或基因敲除,用于生物医学应用。通过RNAi疗法的基因沉默的临床成功关注其他GET作为治疗方法。这篇评论旨在强调GET,类别,机制,挑战,当前使用,和未来的应用。不同的学术搜索引擎,电子数据库,和选定文章的参考书目被用于本综述的编写,重点是基本考虑因素。目前的结果表明,在GET中,与其他GET相比,CRISPR/Cas9具有更高的编辑效率和靶向特异性,删除,修改,或在宿主基因组中的特定位置替换基因。因此,CRISPR/Cas9在分子生产方面很有才华,组织,cell,和器官疗法。因此,GET可用于发现遗传疾病的创新疗法,大流行,癌症,无望的疾病,器官衰竭。具体来说,GET已被用于生产基因修饰的动物,以避免人体器官衰竭。转基因猪在临床试验中用作心脏来源,肝脏,肾脏,和用于人类异种移植(XT)的肺。病毒,非病毒,和混合载体已被用于递送GET,但有一些限制。因此,细胞外囊泡(EV)被认为是临床应用中GET递送的智能和未来货物。这项研究得出结论,GET有望用于生产分子,细胞,和器官疗法。GET作为XT的使用仍处于早期阶段,它们存在道德和生物安全问题。
    Gene editing technologies (GETs) could induce gene knockdown or gene knockout for biomedical applications. The clinical success of gene silence by RNAi therapies pays attention to other GETs as therapeutic approaches. This review aims to highlight GETs, categories, mechanisms, challenges, current use, and prospective applications. The different academic search engines, electronic databases, and bibliographies of selected articles were used in the preparation of this review with a focus on the fundamental considerations. The present results revealed that, among GETs, CRISPR/Cas9 has higher editing efficiency and targeting specificity compared to other GETs to insert, delete, modify, or replace the gene at a specific location in the host genome. Therefore, CRISPR/Cas9 is talented in the production of molecular, tissue, cell, and organ therapies. Consequently, GETs could be used in the discovery of innovative therapeutics for genetic diseases, pandemics, cancer, hopeless diseases, and organ failure. Specifically, GETs have been used to produce gene-modified animals to spare human organ failure. Genetically modified pigs are used in clinical trials as a source of heart, liver, kidneys, and lungs for xenotransplantation (XT) in humans. Viral, non-viral, and hybrid vectors have been utilized for the delivery of GETs with some limitations. Therefore, extracellular vesicles (EVs) are proposed as intelligent and future cargoes for GETs delivery in clinical applications. This study concluded that GETs are promising for the production of molecular, cellular, and organ therapies. The use of GETs as XT is still in the early stage as well and they have ethical and biosafety issues.
    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
    化学动力学疗法(CDT)已成为一种有希望的癌症治疗策略。但是CDT效率受到细胞内过氧化氢(H2O2)水平不足的限制。在这里,我们提出了一种通过siRNA沉默过氧化氢酶(CAT)基因以增强CDT在肿瘤细胞中积累H2O2的方法。Cu-siRNA纳米复合材料是通过Cu2和CATsiRNA的自组装制备的,然后用透明质酸(HA)修饰以进行主动肿瘤靶向。肿瘤细胞摄取后,释放的Cu2+被高表达的谷胱甘肽(GSH)还原为Cu+,然后催化H2O2产生有毒的羟基自由基(•OH)杀死肿瘤细胞。CATsiRNA可以有效地沉默CATmRNA以抑制H2O2的消耗,从而导致H2O2的积累。Cu2+介导的GSH消除和siRNA诱导的内源性H2O2富集都增强了CDT。Cu-siRNA@HA表现出良好的生物相容性和治疗效率。因此,这项工作为CDT中的H2O2提供了一种新的途径,并且可能具有临床应用的潜力。
    Chemodynamic therapy (CDT) has been emerging as a promising strategy for cancer treatment. But the CDT efficiency is restricted by the insufficient intracellular hydrogen peroxide (H2O2) level. Herein, we present a method for H2O2 accumulation in tumor cells by silencing the catalase (CAT) gene with siRNA to achieve enhanced CDT. Cu-siRNA nanocomposites are fabricated by self-assembly of Cu2+ and CAT siRNA and then modified with hyaluronic acid (HA) for active tumor targeting. After tumor cell uptake, the released Cu2+ is reduced by highly expressed glutathione (GSH) to Cu+, which then catalyzes H2O2 to produce toxic hydroxyl radicals (•OH) to kill tumor cells. CAT siRNA can efficiently silence the CAT mRNA to inhibit the consumption of H2O2, resulting in H2O2 accumulation. The Cu2+-mediated GSH elimination and siRNA-induced endogenous H2O2 enrichment both potentiate CDT. Cu-siRNA@HA exhibits good biocompatibility and therapeutic efficiency. This work thus paves a new way to supply H2O2 in CDT and may hold potential for clinical application.
    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
    脊髓损伤(SCI)后的神经性疼痛仍然是一个难题,影响超过80%的SCI患者。越来越多的证据表明,神经炎症反应在SCI后的神经性疼痛中起关键作用。短发夹RNA(shRNA)干扰是SCI后敲低疾病相关特异性基因表达的有效工具,但没有足够的数据可用于建立指南。在这项研究中,我们构建了瞬时受体电位锚蛋白1(TRPA1)shRNA编码慢病毒载体(LV-shTRPA1)和P38MAPKshRNA编码慢病毒载体(LV-shP38),以研究shRNA的沉默效应及其重新编程的能力神经炎症反应,从而增强SCI后的体感恢复。我们使用HEK293-FT和活化的巨噬细胞的体外数据表明,递送的LV-shRNA显示出高转导功效而没有细胞毒性。此外,发现LV-shP38和LV-shTRPA1的组合在抑制靶基因方面最有效,切断脊髓背角和背根神经节中促炎症和促伤害性因子的表达,从而有助于减轻SCI后的神经元超敏反应。总的来说,我们的数据表明,LV-shP38/shTRPA1组合在免疫调节方面产生了协同作用,并降低了神经性疼痛,且具有良好的风险-获益比.总的来说,我们的LV介导的shRNA递送将为基因沉默治疗方法提供有效的工具,以治疗各种无法治愈的疾病.
    Neuropathic pain following spinal cord injury (SCI) remains a difficult problem that affects more than 80% of SCI patients. Growing evidence indicates that neuroinflammatory responses play a key role in neuropathic pain after SCI. Short hairpin RNA (shRNA) interference is an efficient tool for the knockdown of disease-related specific gene expression after SCI, yet insufficient data is available to establish guidelines. In this study, we have constructed the transient receptor potential ankyrin 1 (TRPA1) shRNA encoded-lentiviral vector (LV-shTRPA1) and P38 MAPK shRNA encoded-lentiviral vector (LV-shP38) to investigate the silencing effects of shRNAs and their ability to reprogram the neuroinflammatory responses, thereby enhancing somatosensory recovery after SCI. Our in vitro data employing HEK293-FT and activated macrophages demonstrated that delivered LV-shRNAs showed high transduction efficacy with no cytotoxicity. Furthermore, a combination of LV-shP38 and LV-shTRPA1 was found to be most effective at suppressing target genes, cutting the expression of pro-inflammatory and pro-nociceptive factors in the dorsal horn of the spinal cord and dorsal root ganglia, thus contributing to the alleviation of neuronal hypersensitivities after SCI. Overall, our data demonstrated that the combination LV-shP38/shTRPA1 produced a synergistic effect for immunomodulation and reduced neuropathic pain with a favorable risk-to-benefit ratio. Collectively, our LV-mediated shRNA delivery will provide an efficient tool for gene silencing therapeutic approaches to treat various incurable disorders.
    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
    胰岛素样生长因子1受体(IGF1R),具有酪氨酸激酶(TK)活性的细胞表面受体,配体在急性白血病中异常表达,多发性骨髓瘤,乳房,前列腺,子宫颈,非小细胞肺癌,尤因肉瘤,和其他恶性肿瘤。IGF1R介导恶性增殖,入侵,肿瘤细胞通过多种信号转导途径转移,它还参与肿瘤血管生成和肿瘤细胞抗凋亡。在这项研究中,我们实验室的中性cytidinyl脂质DNCA和胱氨酸骨架阳离子脂质CLD可以被优化以封装反义寡核苷酸(ASO)CT102以形成稳定和均匀的Mix/CT102纳米颗粒(NP),通过静脉给药可以特异性靶向体内高表达IGF1R的肿瘤细胞。与裸CT102相比,脂质复合物能促进HepG2和Huh-7细胞的摄取和晚期凋亡,有效抑制细胞增殖。我们还发现Mix/CT102可以在2小时左右进入细胞核,有效下调IGF1R的mRNA水平。体内功效实验表明,在接受最佳剂量的Mix/CT102的组中,与裸剂量组相比,肿瘤体积减少了8倍。同时,体内分布研究表明,纳米颗粒在肝组织中具有主要的积累能力。这些结果表明,临床医生可以预期Mix/CT102纳米复合材料在减少临床施用CT102的剂量和频率方面非常有效,从而减少ASO的副作用。
    Insulin-like growth factor 1 receptor (IGF1R), a cell surface receptor with tyrosine kinase (TK) activity, has ligands abnormally expressed in acute leukemia, multiple myeloma, breast, prostate, cervical, and nonsmall cell lung cancers, Ewing\'s sarcoma, and other malignant tumors. IGF1R mediates the malignant proliferation, invasion, and metastasis of tumor cells through a variety of signal transduction pathways, and it is also involved in tumor angiogenesis and tumor cell antiapoptosis. In this study, the neutral cytidinyl lipid DNCA and cystine skeleton cationic lipid CLD from our laboratory could be optimized to encapsulate antisense oligonucleotide (ASO) CT102 to form stable and uniform Mix/CT102 nanoparticles (NPs), which could specifically target tumor cells that highly expressed IGF1R in vivo by intravenous administration. Compared with naked CT102, the lipid complex could promote the uptake and late apoptosis levels of HepG2 and Huh-7 cells, inhibiting cell proliferation efficiently. We also found that Mix/CT102 could enter nucleus in about 2 h, effectively downregulating the mRNA level of IGF1R. The in vivo efficacy experiment demonstrated that in the group that received the optimal dose of Mix/CT102, tumor volume was reduced 8-fold compared with the naked dose group. Meanwhile, in vivo distribution studies showed that the nanoparticles had a predominant accumulation capacity in liver tissue. These results indicated that clinicians can expect the Mix/CT102 nanocomposite to be very effective in reducing the dose and frequency of clinically administered CT102, thereby reducing the side effects of ASOs.
    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
    使用小干扰RNA(siRNA)进行特定基因沉默已成为治疗无法治愈的疾病如恶性肿瘤的新型治疗方法。然而,siRNA能否安全有效地递送到靶细胞中仍然是一个挑战。因此,我们在siRNA递送系统中合成了荧光碳点(CD)作为基因载体,该系统在体外诱导有效的基因敲低,同时结合适体AS1411解决了细胞靶向困难。我们发现具有足够生物相容性的CD可以提高siRNA的细胞摄取效率。CLSM和FCM结果表明,CD主要定位于细胞质中,并发出明亮的绿色荧光。此外,由适配体AS1411介导的CD/siRNA递送系统有效沉默了脆性X智力低下蛋白(FMRP)的表达,并成功抑制了肝细胞癌(HCC)细胞的迁移和侵袭倾向。总之,我们已经合成了一种有价值的siRNA递送载体,不仅能够进行生物成像,而且能够有效下调基因表达,这表明基因传递和治疗的有效潜力。
    Using small interfering RNA (siRNA) for the specific gene-silencing has been a novel therapeutic method for the treatment of incurable diseases such as malignancies. However, it remains a challenge whether siRNA can be safely and effectively delivered into target cells. Therefore, we synthesized fluorescent carbon dots (CDs) as a gene vector at the siRNA delivery system that induced efficient gene knockdown in vitro while binding aptamer AS1411 to resolve the difficulty in cell targeting. We found that CDs with adequate biocompatibility can improve the efficiency of cellular uptake of siRNA. CLSM and FCM results showed that CDs were mainly localized in the cytoplasm and emitted bright green fluorescence. In addition, the CD/siRNA delivery system mediated by the aptamer AS1411 effectively silenced the expression of Fragile X mental retardation protein (FMRP) and successfully inhibited the migration and invasive propensity of hepatocellular carcinoma (HCC) cells. In summary, we have synthesized a valuable siRNA delivery vector enabling not only bioimaging but also effective downregulation of gene expression, which is indicative of an efficient potential for gene delivery and therapy.
    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
    过量的淀粉样β肽(Aβ)处理导致的小胶质细胞的炎症功能障碍是阿尔茨海默病(AD)中被忽视但致病的事件。这里,我们利用了天然高密度脂蛋白(HDL)启发的纳米电容器(pHDL/Cur-siBACE1),结合了三位一体的磷脂酸功能化的HDL(pHDL),姜黄素(Cur),和β位点APP切割酶1靶向siRNA(siBACE1)以调节小胶质细胞功能障碍。通过模仿天然的脂蛋白运输路线,pHDL可以穿透血脑屏障并依次靶向Aβ斑块,其中Aβ分解代谢加速而没有小胶质细胞功能障碍。受益结果来自三管齐下的调制策略,包括以抗体样Aβ结合亲和力促进Aβ清除,通过阻断NF-κB途径使小胶质细胞功能障碍正常化,通过基因沉默减少了Aβ的产生(44%)。治疗后,APPswe/PSEN1dE9小鼠的记忆缺陷和神经炎症被逆转。总的来说,这项研究强调了小胶质细胞的双刃剑作用,为AD治疗中调节小胶质细胞功能障碍提供了一种有希望的策略.
    The inflammatory dysfunction of microglia from excess amyloid-β peptide (Aβ) disposal is an overlooked but pathogenic event in Alzheimer\'s disease (AD). Here, we exploit a native high-density lipoprotein (HDL)-inspired nanoscavenger (pHDL/Cur-siBACE1) that combines the trinity of phosphatidic acid-functionalized HDL (pHDL), curcumin (Cur), and β-site APP cleavage enzyme 1 targeted siRNA (siBACE1) to modulate microglial dysfunction. By mimicking the natural lipoprotein transport route, pHDL can penetrate the blood-brain barrier and sequentially target Aβ plaque, where Aβ catabolism is accelerated without microglial dysfunction. The benefit results are from a three-pronged modulation strategy, including promoted Aβ clearance with an antibody-like Aβ binding affinity, normalized microglial dysfunction by blocking the NF-κB pathway, and reduced Aβ production by gene silence (44%). After treatment, the memory deficit and neuroinflammation of APPswe/PSEN 1dE9 mice are reversed. Collectively, this study highlights the double-edged sword role of microglia and provides a promising tactic for modulating microglial dysfunction in AD 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
    DNA生物大分子的功能已被广泛挖掘,作为治疗药物,承运人,和官能化改性衍生物。在这项研究中,我们开发了一系列的DNA四面体纳米笼(Td),通过在四面体DNA纳米笼的四个顶点上同步缀合不同数量的i-(X)和治疗性siRNA(aX-Td@bsiRNA,a+b=4)。该i基序缀合的Td在A549肿瘤细胞中表现出良好的内体逃逸行为,逃逸效率受i-motif数量的影响。此外,在A549细胞中验证了由该siRNA嵌入的Td引起的表皮生长因子受体(EGFR)的mRNA和蛋白表达水平下调。2X-Td@2siRNA治疗组在荷瘤小鼠中的肿瘤生长抑制效率显著高于非i-基序缀合的Td@2siRNA(3.14倍)和游离siRNA(3.63倍)。这些结果证明了赋予DNA纳米结构内体逃逸行为以实现有效的体内基因递送和治疗的一般策略。
    The functionality of DNA biomacromolecules has been widely excavated, as therapeutic drugs, carriers, and functionalized modification derivatives. In this study, we developed a series of DNA tetrahedron nanocages (Td), via synchronous conjugating different numbers of i-(X) and therapeutic siRNA on four vertexes of tetrahedral DNA nanocage (aX-Td@bsiRNA, a+b = 4). This i-motif-conjugated Td exhibited good endosomal escape behaviours in A549 tumor cells, and the escape efficiency was affected by the number of i-motif. Furthermore, the downregulating mRNA and protein expression level of epidermal growth factor receptor (EGFR) caused by this siRNA embedded Td were verified in A549 cells. The tumor growth inhibition efficiency of the 2X-Td@2siRNA treated group in tumor-bearing mice was significantly higher than that of non-i-motif-conjugated Td@2siRNA (3.14-fold) and free siRNA (3.63-fold). These results demonstrate a general strategy for endowing DNA nanostructures with endosomal escape behaviours to achieve effective in vivo gene delivery and therapy.
    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: Journal Article
    Pancreatic carcinoma (PC) is greatly induced by the KRAS gene mutation, but effective targeted delivery for gene therapy has not existed. Small interfering ribonucleic acid (siRNA) serves as an advanced therapeutic modality and holds great promise for cancer treatment. However, the development of a non-toxic and high-efficiency carrier system to accurately deliver siRNA into cells for siRNA-targeted gene silencing is still a prodigious challenge. Herein, polyethylenimine (PEI)-modified hydroxyapatite (HAp) nanoparticles (HAp-PEI) were fabricated. The siRNA of the KRAS gene (siKras) was loaded onto the surface of HAp-PEI via electrostatic interaction between siRNA and PEI to design the functionalized HAp-PEI nanoparticle (HAp-PEI/siKras). The HAp-PEI/siKras was internalized into the human PC cells PANC-1 to achieve the maximum transfection efficiency for active tumor targeting. HAp-PEI/siKras effectively knocked down the expression of the KRAS gene and downregulated the expression of the Kras protein in vitro. Furthermore, the treatment with HAp-PEI/siKras resulted in greater anti-PC cells\' (PANC-1, BXPC-3, and CFPAC-1) efficacy in vitro. Additionally, the HAp-PEI exhibited no obvious in vitro cytotoxicity in normal pancreatic HPDE6-C7 cells. These findings provided a promising alternative for the therapeutic route of siRNA-targeted gene engineering for anti-pancreatic cancer therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号