Targeted gene delivery

靶向基因递送
  • 文章类型: Journal Article
    软骨肉瘤(CS)是一种恶性软骨形成性骨肿瘤,对化疗和放疗具有固有的抵抗力,将手术作为唯一的治疗选择。我们设计了一种肿瘤靶向噬菌体(噬菌体)衍生颗粒(PDP),用于将编码细胞因子的转基因靶向全身递送至实体瘤。噬菌体对哺乳动物细胞没有内在的向性;因此,它被设计为在衣壳上显示双环RGD4C配体以靶向肿瘤。诱导癌细胞死亡,我们构建了一个表达分泌型细胞因子肿瘤坏死因子相关凋亡诱导配体(sTRAIL)的转基因盒.我们检测到RGD4C配体的αvβ3和αvβ5整合素受体的高表达,和人CS细胞中的TRAIL受体2(SW1353),但不存在于原代正常软骨细胞中.RGD4C。携带荧光素酶报告基因的PDP-Luc颗粒,吕克,有效和选择性地介导基因传递到SW1353细胞,但不是原代软骨细胞.用RGD4C转导SW1353细胞。PDP-sTRAIL编码人sTRAIL,导致TRAIL的表达和随后的细胞死亡,而不损害正常软骨细胞。静脉内施用RGD4C。PDP-sTRAIL对具有建立的人CS的小鼠导致肿瘤大小和肿瘤活力的降低。总之,RGD4C。PDP-sTRAIL可用于靶向用sTRAIL全身治疗CS。
    Chondrosarcoma (CS) is a malignant cartilage-forming bone tumor that is inherently resistant to chemotherapy and radiotherapy, leaving surgery as the only treatment option. We have designed a tumor-targeted bacteriophage (phage)-derived particle (PDP), for targeted systemic delivery of cytokine-encoding transgenes to solid tumors. Phage has no intrinsic tropism for mammalian cells; therefore, it was engineered to display a double cyclic RGD4C ligand on the capsid to target tumors. To induce cancer cell death, we constructed a transgene cassette expressing a secreted form of the cytokine tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL). We detected high expression of αvβ3 and αvβ5 integrin receptors of the RGD4C ligand, and of the TRAIL receptor-2 in human CS cells (SW1353), but not in primary normal chondrocytes. The RGD4C.PDP-Luc particle carrying a luciferase reporter gene, Luc, effectively and selectively mediated gene delivery to SW1353 cells, but not primary chondrocytes. Transduction of SW1353 cells with RGD4C.PDP-sTRAIL encoding a human sTRAIL, resulted in the expression of TRAIL and subsequent cell death without harming the normal chondrocytes. Intravenous administration of RGD4C.PDP-sTRAIL to mice with established human CS resulted in a decrease in tumor size and tumor viability. Altogether, RGD4C.PDP-sTRAIL can be used to target systemic treatment of CS with the sTRAIL.
    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
    简介:具有严格控制活性的血脑屏障参与生物活性分子从血液到大脑的协调转移。在不同的交付方式中,基因传递被吹捧为治疗几种神经系统疾病的有希望的策略。外源遗传元件的转移受到缺乏合适载体的限制。作为相关的,设计用于基因传递的高效生物载体具有挑战性。本研究旨在使用CDX修饰的壳聚糖(CS)纳米颗粒(NP)将pEGFP-N1质粒递送到脑实质中。方法:这里,我们附上了CDX,16个氨基酸的肽,CS聚合物使用双官能聚乙二醇(PEG)与三磷酸钠(TPP)配制,采用离子凝胶法。使用DLS表征开发的NP及其与pEGFP-N1(CS-PEG-CDX/pEGFP)的纳米复合物,NMR,FTIR,和TEM分析。对于体外测定,大鼠C6神经胶质瘤细胞系用于细胞内化效率。使用体内成像和荧光显微镜在腹膜内注射后的小鼠模型中研究了纳米复合物的生物分布和脑定位。结果:我们的结果表明,胶质瘤细胞以剂量依赖的方式摄取CS-PEG-CDX/pEGFPNPs。体内成像显示成功进入脑实质,表明绿色荧光蛋白(GFP)作为报告蛋白的表达。然而,发育的NPs的生物分布在其他器官尤其是脾脏中也很明显,肝脏,心,还有肾脏.结论:根据我们的结果,CS-PEG-CDXNP可以为大脑基因递送到中枢神经系统(CNS)提供安全有效的纳米载体。
    Introduction: Blood-brain barrier with strictly controlled activity participates in a coordinated transfer of bioactive molecules from the blood to the brain. Among different delivery approaches, gene delivery is touted as a promising strategy for the treatment of several nervous system disorders. The transfer of exogenous genetic elements is limited by the paucity of suitable carriers. As a correlate, designing high-efficiency biocarriers for gene delivery is challenging. This study aimed to deliver pEGFP-N1 plasmid into the brain parenchyma using CDX-modified chitosan (CS) nanoparticles (NPs). Methods: Herein, we attached CDX, a 16 amino acids peptide, to the CS polymer using bifunctional polyethylene glycol (PEG) formulated with sodium tripolyphosphate (TPP), by ionic gelation method. Developed NPs and their nanocomplexes with pEGFP-N1 (CS-PEG-CDX/pEGFP) were characterized using DLS, NMR, FTIR, and TEM analyses. For in vitro assays, a rat C6 glioma cell line was used for cell internalization efficiency. The biodistribution and brain localization of nanocomplexes were studied in a mouse model after intraperitoneal injection using in vivo imaging and fluorescent microscopy. Results: Our results showed that CS-PEG-CDX/pEGFP NPs were uptaken by glioma cells in a dose-dependent manner. In vivo imaging revealed successful entry into the brain parenchyma indicated with the expression of green fluorescent protein (GFP) as a reporter protein. However, the biodistribution of developed NPs was also evident in other organs especially the spleen, liver, heart, and kidneys. Conclusion: Based on our results, CS-PEG-CDX NPs can provide a safe and effective nanocarrier for brain gene delivery into the central nervous system (CNS).
    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
    暂无摘要。
    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
    骨关节炎(OA)是一种退行性关节疾病,其特征在于关节软骨的进行性恶化和丧失。目前没有逆转OA发作的治疗方法。因此,作为概念验证研究,我们开发了将基因转移至原代人软骨细胞的靶向递送策略.我们在基于M13丝状噬菌体(噬菌体)的颗粒的pIII小外壳蛋白上展示了软骨细胞亲和肽(CAP),该颗粒在巨细胞病毒CMV启动子和反向末端重复序列(ITR)顺式元件下携带哺乳动物转基因盒相关病毒血清型2(AAV-2)。原代人关节软骨细胞(HACs)被用作体外模型,并表征了CAP配体与HAC致病条件相关的选择性和结合特性。我们发现CAP配体对致病性HAC具有高度选择性。此外,稳定性,细胞毒性,和CAP展示噬菌体(CAP。噬菌体)进行了评估。我们发现噬菌体颗粒在很宽的温度和pH值范围内是稳定的,同时对HAC无细胞毒性。重要的是,CAP。噬菌体粒子,携带分泌型荧光素酶(Lucia)报告基因,有效和选择性地将转基因表达传递给HAC。总之,发现CAP配体优选与致病性软骨细胞结合,和CAP。噬菌体颗粒成功地靶向并将转基因递送至HAC。
    Osteoarthritis (OA) is a degenerative joint disease characterized by progressive deterioration and loss of articular cartilage. There is currently no treatment to reverse the onset of OA. Thus, we developed a targeted delivery strategy to transfer genes into primary human chondrocytes as a proof-of-concept study. We displayed a chondrocyte-affinity peptide (CAP) on the pIII minor coat protein of the M13 filamentous bacteriophage (phage)-based particle carrying a mammalian transgene cassette under cytomegalovirus CMV promoter and inverted terminal repeats (ITRs) cis elements of adeno-associated virus serotype 2 (AAV-2). Primary human articular chondrocytes (HACs) were used as an in vitro model, and the selectivity and binding properties of the CAP ligand in relation to the pathogenic conditions of HACs were characterized. We found that the CAP ligand is highly selective toward pathogenic HACs. Furthermore, the stability, cytotoxicity, and gene delivery efficacy of the CAP-displaying phage (CAP.Phage) were evaluated. We found that the phage particle is stable under a wide range of temperatures and pH values, while showing no cytotoxicity to HACs. Importantly, the CAP.Phage particle, carrying a secreted luciferase (Lucia) reporter gene, efficiently and selectively delivered transgene expression to HACs. In summary, it was found that the CAP ligand preferably binds to pathogenic chondrocytes, and the CAP.Phage particle successfully targets and delivers transgene to HACs.
    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
    A new pH-responsive cationic co-liposomal formulation was prepared in this study using the twin version of the amphiphile palmitoyl homocysteine, TPHC; natural zwitterionic lipid, DOPE; and cholesterol-based twin cationic lipid, C5C, at specified molar ratios. This co-liposome was further decorated with a newly designed fluorescently tagged, cholesterol-tethered EpCAM-targeting RNA aptamer for targeted gene delivery. This aptamer-guided nanoliposomal formulation, C5C/DOPE/TPHC at 8:24:1 molar ratio, could efficiently transport the genes in response to low pH of cellular endosomes selectively to the EpCAM overexpressing cancer stem cells. This particular observation was extended using siRNA against GFP to validate their transfection capabilities in response to EpCAM expression. Overall, the aptamer-guided nanoliposomal formulation was found to be an excellent transfectant for in vitro siRNA gene delivery.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    巨噬细胞是癌症和其他疾病的基因治疗的理想靶标。合成了聚乙二醇(PEG)和聚-L-赖氨酸(PLL)或聚{N-[N-(2-氨基乙基)-2-氨基乙基]天冬酰胺}(pAsp(DET))的阳离子二嵌段共聚物,并用于与用甘露糖部分修饰的质粒DNA(pDNA)形成复合物,作为巨噬细胞表面显示的C型凝集素受体的靶向配体。PEG-b-PLL共聚物因其细胞毒性而闻名,因此,使用可还原试剂二硫代双(琥珀酰亚胺基丙酸酯)(DSP)交联基于PEG-b-PLL的聚合复合物。交联复合物对小鼠胚胎成纤维细胞NIH/3T3细胞系和小鼠骨髓来源的巨噬细胞(BMMΦ)均显示低毒性。在巨噬细胞中,甘露糖修饰的复合物显示出约8倍的转染效率。聚合复合物的交联降低了毒性,但转染增强是适度的。PEG-b-pAsp(DET)共聚物对IC-21鼠巨噬细胞系显示低毒性,可用于生产非交联的pDNA聚合物复合物。获得的甘露糖修饰的复合物表现出大约。与无甘露糖复合物相比,IC-21巨噬细胞的转染活性高500倍。该结果大大超过了先前使用甘露糖受体靶向的非病毒基因递送系统描述的靶向基因转移效果。这些结果表明,Man-PEG-b-pAsp(DET)/pDNA复合物是基于免疫细胞的基因治疗的潜在载体。
    Macrophages are desirable targets for gene therapy of cancer and other diseases. Cationic diblock copolymers of polyethylene glycol (PEG) and poly-L-lysine (PLL) or poly{N-[N-(2-aminoethyl)-2-aminoethyl]aspartamide} (pAsp(DET)) are synthesized and used to form polyplexes with a plasmid DNA (pDNA) that are decorated with mannose moieties, serving as the targeting ligands for the C type lectin receptors displayed at the surface of macrophages. The PEG-b-PLL copolymers are known for its cytotoxicity, so PEG-b-PLL-based polyplexes are cross-linked using reducible reagent dithiobis(succinimidyl propionate) (DSP). The cross-linked polyplexes display low toxicity to both mouse embryonic fibroblasts NIH/3T3 cell line and mouse bone marrow-derived macrophages (BMMΦ). In macrophages mannose-decorated polyplexes demonstrate an ≈8 times higher transfection efficiency. The cross-linking of the polyplexes decrease the toxicity, but the transfection enhancement is moderate. The PEG-b-pAsp(DET) copolymers display low toxicity with respect to the IC-21 murine macrophage cell line and are used for the production of non-cross-linked pDNA-contained polyplexes. The obtained mannose modified polyplexes exhibit ca. 500-times greater transfection activity in IC-21 macrophages compared to the mannose-free polyplexes. This result greatly exceeds the targeting gene transfer effects previously described using mannose receptor targeted non-viral gene delivery systems. These results suggest that Man-PEG-b-pAsp(DET)/pDNA polyplex is a potential vector for immune cells-based gene 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
    腺相关病毒(AAV)是一种很有前途的基因治疗载体,但是如果传递的转基因在体内普遍表达时是有害的,那么它的广泛取向可能是有害的,即在非目标组织中。将目的转基因以足够高的水平递送至靶细胞以治疗有效,同时通过最小化向脱靶细胞的递送来维持安全性是基因治疗领域中的普遍挑战。我们已经开发了基于AAV9的蛋白酶可活化载体(前载体)平台,其可以被全身注射以通过对存在于疾病部位的细胞外蛋白酶做出响应而将治疗性转基因位点特异性地递送至患病细胞。前载体平台由插入病毒衣壳的肽组成,其破坏病毒结合细胞表面受体的能力。该肽含有阻断基序(天冬氨酸残基),其两侧两侧有被某些蛋白酶识别的切割序列。暴露于蛋白酶会将肽从衣壳上裂解下来,激活或“打开”前向量。为了响应激活,前载体恢复了结合和转导细胞的能力。这里,我们设计了一种由半胱氨酸天冬氨酸蛋白酶(caspases)激活的前载体,在炎症和细胞凋亡中起作用,因此在心力衰竭等疾病部位升高,神经退行性疾病,和缺血性中风。与AAV9相比,该前载体在OFF状态下的转导能力降低了200倍,从而降低了病毒转导脱靶健康组织的能力。在暴露于caspase-3并通过caspase-3进行蛋白水解后,与OFF状态相比,前载体的转导增加了95倍。发现可转换的转导行为是肽插入消除病毒与细胞结合能力的直接结果。进行体内研究以表征生物分布,血液循环时间,中和抗体形成,和caspase激活的前载体在心力衰竭模型中的靶向递送能力。
    Adeno-associated virus (AAV) is a promising vector for gene therapy, but its broad tropism can be detrimental if the transgene being delivered is harmful when expressed ubiquitously in the body, i.e. in non-target tissues. Delivering the transgene of interest to target cells at levels high enough to be therapeutically effective while maintaining safety by minimizing delivery to off-target cells is a prevalent challenge in the field of gene therapy. We have developed a protease activatable vector (provector) platform based on AAV9 that can be injected systemically to deliver therapeutic transgenes site-specifically to diseased cells by responding to extracellular proteases present at the disease site. The provector platform consists of a peptide insertion into the virus capsid which disrupts the virus\' ability to bind to cell surface receptors. This peptide contains a blocking motif (aspartic acid residues) flanked on either side by cleavage sequences that are recognized by certain proteases. Exposure to proteases cleaves the peptides off the capsid, activating or \"switching ON\" the provector. In response to the activation, the provectors regain their ability to bind and transduce cells. Here, we have designed a provector that is activated by cysteine aspartic proteases (caspases), which have roles in inflammation and apoptosis and thus are elevated at sites of diseases such as heart failure, neurodegenerative diseases, and ischemic stroke. This provector demonstrates a 200-fold reduction in transduction ability in the OFF state compared to AAV9, reducing the virus\' ability to transduce off-target healthy tissue. Following exposure to and proteolysis by caspase-3, the provector shows a 95-fold increase in transduction compared to the OFF state. The switchable transduction behavior was found to be a direct result of the peptide insertion ablating the ability of the virus to bind to cells. In vivo studies were conducted to characterize the biodistribution, blood circulation time, neutralizing antibody formation, and targeted delivery ability of the caspase-activatable provector in a model of heart failure.
    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
    Merging targeted systemic gene delivery and systemic chemotherapy against cancer, chemovirotherapy, has the potential to improve chemotherapy and gene therapy treatments and overcome cancer resistance. We introduced a bacteriophage (phage) vector, named human adeno-associated virus (AAV)/phage or AAVP, for the systemic targeting of therapeutic genes to cancer. The vector was designed as a hybrid between a recombinant adeno-associated virus genome (rAAV) and a filamentous phage capsid. To achieve tumor targeting, we displayed on the phage capsid the double-cyclic CDCRGDCFC (RGD4C) ligand that binds the alpha-V/beta-3 (αvβ3) integrin receptor. Here, we investigated a combination of doxorubicin chemotherapeutic drug and targeted gene delivery by the RGD4C/AAVP vector. Firstly, we showed that doxorubicin boosts transgene expression from the RGD4C/AAVP in two-dimensional (2D) cell cultures and three-dimensional (3D) tumor spheres established from human and murine cancer cells, while preserving selective gene delivery by RGD4C/AAVP. Next, we confirmed that doxorubicin does not increase vector attachment to cancer cells nor vector cell entry. In contrast, doxorubicin may alter the intracellular trafficking of the vector by facilitating nuclear accumulation of the RGD4C/AAVP genome through destabilization of the nuclear membrane. Finally, a combination of doxorubicin and RGD4C/AAVP-targeted suicide gene therapy exerts a synergistic effect to destroy human and murine tumor cells in 2D and 3D tumor sphere settings.
    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
    核蛋白,像组蛋白H2A,是有前途的基因传递的非病毒载体,因为它们是生物相容的,可生物降解,承受固有的核定位信号,并且易于修改。向组蛋白H2A添加表面蛋白结合配体可以提高其DNA递送效率。肿瘤微环境(TME)是基因治疗的有希望的靶标,因为其表面蛋白库比癌细胞更稳定。癌相关成纤维细胞(CAFs)是TME的重要组成部分,它们的表面标志物之一是β型血小板衍生生长因子受体(PDGFRβ)。在这项研究中,我们将组蛋白H2A与PDGFRβ结合肽融合,YG2,以创建一种新型的非病毒成纤维细胞靶向DNA载体,H2A-YG2。具有编码双顺反子报告子的pDNA的组蛋白复合物的转染效率(增强的绿色荧光蛋白,EGFP,和萤火虫荧光素酶)通过荧光素酶测定和流式细胞术估计PDGFRβ阳性和PDGFRβ阴性细胞。荧光素酶活性,转染细胞的百分比,由于仅在PDGFRβ阳性细胞中用YG2进行组蛋白修饰,因此整体EGFP荧光增加。我们还使用四甲基-罗丹明标记的pDNA估算了DNA-载体复合物的内化效率。配体融合仅在PDGFRβ阳性细胞中增加DNA内化。总之,我们证明H2A-YG2载体靶向基因递送至PDGFRβ阳性肿瘤基质细胞。
    Nuclear proteins, like histone H2A, are promising non-viral carriers for gene delivery since they are biocompatible, biodegradable, bear intrinsic nuclear localization signal, and are easy to modify. The addition of surface-protein-binding ligand to histone H2A may increase its DNA delivery efficiency. Tumor microenvironment (TME) is a promising target for gene therapy since its surface protein repertoire is more stable than that of cancer cells. Cancer-associated fibroblasts (CAFs) are important components of TME, and one of their surface markers is beta-type platelet-derived growth factor receptor (PDGFRβ). In this study, we fused histone H2A with PDGFRβ-binding peptide, YG2, to create a novel non-viral fibroblast-targeting DNA carrier, H2A-YG2. The transfection efficiency of histone complexes with pDNA encoding a bicistronic reporter (enhanced green fluorescent protein, EGFP, and firefly luciferase) in PDGFRβ-positive and PDGFRβ-negative cells was estimated by luciferase assay and flow cytometry. The luciferase activity, percentage of transfected cells, and overall EGFP fluorescence were increased due to histone modification with YG2 only in PDGFRβ-positive cells. We also estimated the internalization efficiency of DNA-carrier complexes using tetramethyl-rhodamine-labeled pDNA. The ligand fusion increased DNA internalization only in the PDGFRβ-positive cells. In conclusion, we demonstrated that the H2A-YG2 carrier targeted gene delivery to PDGFRβ-positive tumor stromal cells.
    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
    基因治疗的进展已经预示了其治疗涉及遗传功能障碍的广泛疾病的潜力。然而,它的治疗效率和成功的结果高度依赖于理想的基因传递系统的发展。在这个问题上,基于二氧化硅的载体由于其增加的安全性而转移了病毒和其他类型的非病毒载体的一些注意力,易于修改的结构和表面,高稳定性,和成本效益。硅烷化学的多功能性以及二氧化硅与其他材料的结合,如聚合物,脂质,或无机颗粒,导致了具有强大装载能力的载体的发展,有效保护和结合遗传物质的能力,有针对性的交付,和货物的刺激响应释放。使用这些纳米系统作为不同潜在治疗领域的多功能平台,在体外和体内都获得了有希望的结果。比如癌症或脑部治疗,有时结合成像功能。在这里,综述了用于基因治疗的硅基系统的最新进展,包括它们的主要属性,制造方法,表面改性,和潜在的治疗应用。
    Advances in gene therapy have been foreshadowing its potential for the treatment of a vast range of diseases involving genetic malfunctioning. However, its therapeutic efficiency and successful outcome are highly dependent on the development of the ideal gene delivery system. On that matter, silica-based vectors have diverted some attention from viral and other types of non-viral vectors due to their increased safety, easily modifiable structure and surface, high stability, and cost-effectiveness. The versatility of silane chemistry and the combination of silica with other materials, such as polymers, lipids, or inorganic particles, has resulted in the development of carriers with great loading capacities, ability to effectively protect and bind genetic material, targeted delivery, and stimuli-responsive release of cargos. Promising results have been obtained both in vitro and in vivo using these nanosystems as multifunctional platforms in different potential therapeutic areas, such as cancer or brain therapies, sometimes combined with imaging functions. Herein, the current advances in silica-based systems designed for gene therapy are reviewed, including their main properties, fabrication methods, surface modifications, and potential therapeutic applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号