Gene delivery

基因传递
  • 文章类型: Journal Article
    作为二维材料,金纳米三角形(GNTs)在基因传递领域很少研究。在这项研究中,开发了一种温度响应型GNTs作为基因传递的新型载体。将温敏共聚物PNIPAm-g-PEI接枝到GNTs表面,构建PNIPAm-g-PEI/GNTs复合可控释放平台。PNIPAm-g-PEI/GNTs的较低临界溶解温度(LCST)为42°C,在该温度下,PNIPAm-g-PEI/GNTs的粒径为150nm。凝胶电泳实验表明,PNIPAm-g-PEI/GNTs在20μg/mL时完全凝聚了DNA,PNIPAm-g-PEI/GNTs促进激光照射下DNA的释放。荧光素酶和绿色荧光蛋白报告基因检测表明,PNIPAm-g-PEI/GNTs的转染效率是PEI的1.5和7.2倍,分别。这些结果证明了温度响应性GNT作为有效和安全的基因递送载体的有希望的潜力。
    As a two-dimensional material, gold nanotriangles (GNTs) are rarely studied in the field of gene delivery. In this study, a temperature-responsive GNTs was developed as a novel carrier for gene delivery. The temperature-sensitive copolymer PNIPAm-g-PEI was grafted onto the surface of GNTs to construct a PNIPAm-g-PEI/GNTs composite controllable release platform. The lower critical solution temperature (LCST) of PNIPAm-g-PEI/GNTs was found to be 42 °C, and the particle size of PNIPAm-g-PEI/GNTs was 150 nm at this temperature. Gel electrophoresis experiments showed that PNIPAm-g-PEI/GNTs completely condensed DNA at 20 μg/mL, and PNIPAm-g-PEI/GNTs promoted the release of DNA under laser irradiation. Luciferase and green fluorescent protein reporter gene assays demonstrated that the transfection efficiency of PNIPAm-g-PEI/GNTs was 1.5 and 7.2 times that of PEI, respectively. These results demonstrated the promising potential of temperature-responsive GNTs as effective and safe gene delivery vectors.
    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
    基因治疗为治疗缺血性疾病提供了一个有希望的途径,然而,其临床疗效受到单一基因疗法的局限性和这种疾病的高氧化应激微环境特征的阻碍。脂质-聚合物杂合载体代表了一种通过利用脂质和聚合物的组合优势来增强基因治疗有效性的新方法。在这项研究中,我们设计了具有定制结构修饰的脂质-聚合物杂化纳米载体,以创建针对基因递送优化的多功能膜融合脂质-核靶向聚合物纳米递送系统(FLNP).我们的结果表明,FLNPs通过膜融合促进有效的细胞摄取和基因转染,溶酶体回避,和核瞄准机制。在封装肝细胞生长因子质粒(pHGF)和过氧化氢酶质粒(pCAT)后,制备HGF/CAT-FLNP,显著增强了C2C12细胞对H2O2诱导的体外损伤的抗性。体内研究进一步表明,HGF/CAT-FLNPs能有效缓解后肢缺血引起的坏疽,恢复电机功能,促进小鼠血液灌注恢复。代谢组学分析表明,FLNPs在基因转染过程中不会引起代谢紊乱。总之,FLNPs代表了多维辅助基因传递的通用平台,显着提高基因传递的效率,并有望使用pHGF和pCAT有效协同治疗下肢缺血。
    Gene therapy offers a promising avenue for treating ischemic diseases, yet its clinical efficacy is hindered by the limitations of single gene therapy and the high oxidative stress microenvironment characteristic of such conditions. Lipid-polymer hybrid vectors represent a novel approach to enhance the effectiveness of gene therapy by harnessing the combined advantages of lipids and polymers. In this study, we engineered lipid-polymer hybrid nanocarriers with tailored structural modifications to create a versatile membrane fusion lipid-nuclear targeted polymer nanodelivery system (FLNPs) optimized for gene delivery. Our results demonstrate that FLNPs facilitate efficient cellular uptake and gene transfection via membrane fusion, lysosome avoidance, and nuclear targeting mechanisms. Upon encapsulating Hepatocyte Growth Factor plasmid (pHGF) and Catalase plasmid (pCAT), HGF/CAT-FLNPs were prepared, which significantly enhanced the resistance of C2C12 cells to H2O2-induced injury in vitro. In vivo studies further revealed that HGF/CAT-FLNPs effectively alleviated hindlimb ischemia-induced gangrene, restored motor function, and promoted blood perfusion recovery in mice. Metabolomics analysis indicated that FLNPs didn\'t induce metabolic disturbances during gene transfection. In conclusion, FLNPs represent a versatile platform for multi-dimensional assisted gene delivery, significantly improving the efficiency of gene delivery and holding promise for effective synergistic treatment of lower limb ischemia using pHGF and pCAT.
    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
    白细胞介素-12(IL-12)是一种具有显著抗癌特性的关键细胞因子,包括增强T细胞介导的癌细胞杀伤,抑制肿瘤血管生成。迄今为止,评估了许多方法来实现IL-12的原位过表达,从而使渗漏和随后的毒性最小化。这里,它专注于环状单链DNA(CssDNA),一种以其独特结构为特征的DNA,可以在哺乳动物中表达。发现CssDNA可以通过肌肉注射诱导持续半年的荧光素酶表达,并通过肿瘤内注射显示出有效的抗肿瘤效果。基于这些发现,现在开发了叶酸修饰的LNP系统,用于递送表达IL-12的CssDNA,用于治疗4T1三阴性乳腺癌(TNBC)。这种递送系统有效激活抗癌免疫反应,减缓肿瘤生长,显著延长动物模型的生存期,并防止肿瘤复发。经过6个月的长期观察,在治愈小鼠的淋巴结和血清中仍可检测到IL-12水平的升高。这项研究强调了CssDNA的长期持续表达能力及其抑制复发的能力,以及肿瘤靶向LNP在基于CssDNA的癌症治疗中的潜力,为基因过表达策略提供了新的见解。
    Interleukin-12 (IL-12) is a critical cytokine with notable anticancer properties, including enhancing T-cell-mediated cancer cell killing, and curbing tumor angiogenesis. To date, many approaches are evaluated to achieve in situ overexpression of IL-12, minimizing leakage and the ensuing toxicity. Here, it is focused on circular single-stranded DNA (Css DNA), a type of DNA characterized by its unique structure, which could be expressed in mammals. It is discovered that Css DNA can induce sustained luciferase expression for half a year by intramuscular injection and showed effective antitumor results by intratumoral injection. Motivated by these findings, a folate-modified LNP system is now developed for the delivery of Css DNA expressing IL-12 for the therapy of 4T1 triple-negative breast cancer (TNBC). This delivery system effectively activates anti-cancer immune responses, slows tumor growth, significantly prolongs survival in animal models, and prevents tumor recurrence. After 6 months of long-term observation, the elevated level of IL-12 is still detectable in the lymph nodes and serum of the cured mice. This study highlights the long-term sustained expression capacity of Css DNA and its ability to inhibit recurrence, and the potential of tumor-targeted LNPs for Css DNA-based cancer therapy, providing a new insight into gene overexpression strategy.
    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
    靶向消除肿瘤内受损或过表达的蛋白质在调节细胞功能和抑制肿瘤细胞生长中起关键作用。研究人员一直在努力寻找更安全,更有效的蛋白质去除方法。这里,我们建议协同使用小分子降解剂(PROTAC)和siRNA来提高蛋白质清除效率和肿瘤治疗效果。制备共递送脂质体以促进PROTAC和siRNA的有效包封。具体来说,阳离子脂质体显著提高了不溶性PROTAC(DT2216)的溶解度。阳离子聚合物(F-PEI)实现了核酸药物的有效封装,从而促进内吞作用并增强药物的治疗效果。体内和体外实验都证明了靶蛋白的显著降解和共同递送系统对肿瘤细胞的抑制。总之,共递送脂质体提供了一个纳米递送系统,精通有效地包封亲水性和疏水性药物,从而提出了靶向联合治疗肿瘤的新策略。
    Targeted elimination of damaged or overexpressed proteins within the tumor serves a pivotal role in regulating cellular function and restraining tumor cell growth. Researchers have been striving to identify safer and more effective methods for protein removal. Here, we propose the synergistic employment of a small molecule degrading agent (PROTAC) and siRNA to attain enhanced protein clearance efficiency and tumor therapeutic effects. Co-delivery liposomes were prepared to facilitate the efficient encapsulation of PROTAC and siRNA. Specifically, the cationic liposome significantly improved the solubility of the insoluble PROTAC (DT2216). The cationic polymer (F-PEI) achieved efficient encapsulation of the nucleic acid drug, thereby promoting endocytosis and enhancing the therapeutic impact of the drug. Both in vivo and in vitro experiments demonstrated remarkable degradation of target proteins and inhibition of tumor cells by the co-delivery system. In conclusion, the co-delivery liposomes furnished a nano-delivery system proficient in effectively encapsulating both hydrophilic and hydrophobic drugs, thereby presenting a novel strategy for targeted combination therapy in treating 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
    缺氧可加重肿瘤的发生,发展,入侵,和转移,并极大地抑制光动力疗法(PDT)效应。在这里,开发了具有产氧功能的基于氮化碳(CN)的DNA和光敏剂共递送系统(BPSCN)来解决此问题。硒化物葡萄糖(Seglu)用作掺杂剂以制备红色/NIR活性CNs(SegluCNs)。肿瘤靶向单元Bio-PEG2000用于通过酯化反应构建BPSCNs纳米颗粒。此外,通过将P53基因与带正电荷的线粒体靶向近红外(NIR)光敏剂(MTTPY)混合来实现DNA疏水化,其被包封在非阳离子BPSCNs中以用于协同递送。BPSCNs@MTTPY-P53复合物中的酯键可以被肝脏中的脂肪酶破坏,以促进P53的释放,P53表达上调,并促进线粒体中HIF-1α的降解。此外,复合物产生的氧气改善了肝细胞癌(HCC)的缺氧微环境,线粒体中HIF-1α表达协同下调,促进线粒体衍生的铁死亡并增强MTTPY单位的PDT效应。体内和体外实验表明,转染的P53-DNA,通过HIF-1α/SLC7A11途径,这些复合物产生的O2和ROS协同导致肝癌细胞线粒体源性铁凋亡,完全避免缺氧引起的PDT抵抗,在HCC治疗中发挥重要的治疗作用。
    Hypoxia can aggravate tumor occurrence, development, invasion, and metastasis, and greatly inhibit the photodynamic therapy (PDT) effect. Herein, carbon nitride (CNs)-based DNA and photosensitizer co-delivery systems (BPSCNs) with oxygen-producing functions are developed to address this problem. Selenide glucose (Seglu) is used as the dopant to prepare red/NIR-active CNs (SegluCNs). The tumor-targeting unit Bio-PEG2000 is utilized to construct BPSCNs nanoparticles through esterification reactions. Furthermore, DNA hydrophobization is realized via mixing P53 gene with a positively charged mitochondrial-targeted near-infrared (NIR) emitting photosensitizer (MTTPY), which is encapsulated in non-cationic BPSCNs for synergistic delivery. Ester bonds in BPSCNs@MTTPY-P53 complexes can be disrupted by lipase in the liver to facilitate P53 release, upregulated P53 expression, and promoted HIF-1α degradation in mitochondria. In addition, the oxygen produced by the complexes improved the hypoxic microenvironment of hepatocellular carcinoma (HCC), synergistically downregulated HIF-1α expression in mitochondria, promoted mitochondrial-derived ferroptosis and enhanced the PDT effect of the MTTPY unit. Both in vivo and in vitro experiments indicated that the transfected P53-DNA, produced O2 and ROS by these complexes synergistically led to mitochondrial-derived ferroptosis in hepatoma cells through the HIF-1α/SLC7A11 pathway, and completely avoiding PDT resistance caused by hypoxia, exerting a significant therapeutic role in HCC 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
    基因递送疗法已成为治疗各种疾病的流行方法。然而,它仍然带来了在目标地点积累和减少脱靶效应的挑战。气雾剂基因递送用于治疗肺部疾病具有高肺积,特异性靶向和较少的全身副作用。然而,关键挑战是选择能够克服生理障碍的气溶胶基因递送的合适制剂。有许多现有的基因携带者正在研究中,包括病毒载体和非病毒载体。随着生物材料的发展,更多的生物相容性物质已经通过吸入应用基因递送。此外,许多类型的基因可以通过气溶胶吸入传递,比如DNA,mRNAsiRNA和CRISPR/Cas9。已证明不同类型基因的气溶胶递送在许多疾病如SARS-CoV-2、囊性纤维化和肺癌的治疗中是有效的。在本文中,我们对正在进行的气溶胶基因递送疗法的研究进行了全面的综述,包括基本的呼吸系统,不同类型的基因携带者,不同类型的携带基因和临床应用。
    Gene delivery therapy has emerged as a popular approach for the treatment of various diseases. However, it still poses the challenges of accumulation in target sites and reducing off-target effects. Aerosol gene delivery for the treatment of pulmonary diseases has the advantages of high lung accumulation, specific targeting and fewer systemic side effects. However, the key challenge is selecting the appropriate formulation for aerosol gene delivery that can overcome physiological barriers. There are numerous existing gene carriers under study, including viral vectors and non-viral vectors. With the development of biomaterials, more biocompatible substances have applied gene delivery via inhalation. Furthermore, many types of genes can be delivered through aerosol inhalation, such as DNA, mRNA, siRNA and CRISPR/Cas9. Aerosol delivery of different types of genes has proven to be efficient in the treatment of many diseases such as SARS-CoV-2, cystic fibrosis and lung cancer. In this paper, we provide a comprehensive review of the ongoing research on aerosol gene delivery therapy, including the basic respiratory system, different types of gene carriers, different types of carried genes and clinical applications.
    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
    肝细胞核中共价闭合环状DNA(cccDNA)的持续存在对实现乙型肝炎病毒(HBV)的全面治疗构成了重大障碍。CRISPR/Cas9用于靶向和消除cccDNA的当前应用已经局限于体外研究,这是由于在动物模型中稳定的cccDNA表达的挑战和递送系统的有限的非免疫原性。本研究通过引入利用双子表面活性剂(GS)的新型非病毒基因递送系统来解决这些限制。开发的系统通过用红细胞膜(RBCM)或肝细胞膜(HCM)修饰,产生具有带负电荷表面的稳定和靶向的CRISPR/Cas9纳米药物,产生GS-pDNA@Cas9-CMs复合物。这些GS-pDNA复合物显示以4:lw/w比率完全形成。GS-pDNA-HCM的体外转染效率达到54.61%,显示同型靶向和优异的安全性。此外,该研究从GS-pDNA@Cas9-HCM递送的6个序列中鉴定出最有效的单向导RNA(sgRNA).使用GS-pDNA@Cas9-HCM,观察到在体外HBVcccDNA和体内HBVcccDNA减少52.34%的显着减少96.47%,随着其他HBV相关标志物的显着下降。AML-12细胞在不同时间和温度条件下对GS复合物摄取的研究表明,网格蛋白介导的GS-pDNA内吞作用(CME)和谷蛋白介导的GS-pDNA-HCM和GS-pDNA-RBCM的内吞作用(CVME)。总之,这项研究提出了基于GS(GS-pDNA@Cas9-CMs)的仿生基因编辑纳米载体,并探索了使用CRISPR/Cas9对cccDNA的精确和靶向清除,在体外和体内都证明了良好的生物相容性。这种创新的方法为推进HBV的治愈提供了一个有前途的治疗策略。
    The persistent presence of covalently closed circular DNA (cccDNA) in hepatocyte nuclei poses a significant obstacle to achieving a comprehensive cure for hepatitis B virus (HBV). Current applications of CRISPR/Cas9 for targeting and eliminating cccDNA have been confined to in vitro studies due to challenges in stable cccDNA expression in animal models and the limited non-immunogenicity of delivery systems. This study addresses these limitations by introducing a novel non-viral gene delivery system utilizing Gemini Surfactant (GS). The developed system creates stable and targeted CRISPR/Cas9 nanodrugs with a negatively charged surface through modification with red blood cell membranes (RBCM) or hepatocyte membranes (HCM), resulting in GS-pDNA@Cas9-CMs complexes. These GS-pDNA complexes demonstrated complete formation at a 4:1 w/w ratio. The in vitro transfection efficiency of GS-pDNA-HCM reached 54.61%, showing homotypic targeting and excellent safety. Additionally, the study identified the most effective single-guide RNA (sgRNA) from six sequences delivered by GS-pDNA@Cas9-HCM. Using GS-pDNA@Cas9-HCM, a significant reduction of 96.47% in in vitro HBV cccDNA and a 52.34% reduction in in vivo HBV cccDNA were observed, along with a notable decrease in other HBV-related markers. The investigation of GS complex uptake by AML-12 cells under varied time and temperature conditions revealed clathrin-mediated endocytosis (CME) for GS-pDNA and caveolin-mediated endocytosis (CVME) for GS-pDNA-HCM and GS-pDNA-RBCM. In summary, this research presents biomimetic gene-editing nanovectors based on GS (GS-pDNA@Cas9-CMs) and explores their precise and targeted clearance of cccDNA using CRISPR/Cas9, demonstrating good biocompatibility both in vitro and in vivo. This innovative approach provides a promising therapeutic strategy for advancing the cure of HBV.
    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
    最近的癌症治疗研究发现,基于壳聚糖(Ch)的纳米颗粒显示出靶向基因递送的巨大潜力。壳聚糖,一种生物相容性和生物可降解的聚合物,有特殊的属性,使其成为治疗基因的理想载体。这些纳米颗粒可以响应特定的刺激,如pH值,温度,和酶,实现基因的精确传递和调控释放。在癌症治疗中,这些纳米粒子已经被证明可以有效地将基因传递给肿瘤细胞,减缓肿瘤生长。调整纳米粒子的表面,包封保护剂,和使用靶向配体也提高了基因递送效率。基于壳聚糖的智能纳米颗粒已显示出通过选择性释放基因以响应肿瘤条件来改善结果的希望。加强有针对性的交付,减少脱靶效应。此外,纳米粒子表面上的靶向配体增加了吸收和有效性。尽管需要进一步的研究来优化这些纳米粒子的结构和组成,并评估其长期安全性,这些进步为创新的基因聚焦癌症疗法铺平了道路。
    Recent cancer therapy research has found that chitosan (Ch)-based nanoparticles show great potential for targeted gene delivery. Chitosan, a biocompatible and biodegradable polymer, has exceptional properties, making it an ideal carrier for therapeutic genes. These nanoparticles can respond to specific stimuli like pH, temperature, and enzymes, enabling precise delivery and regulated release of genes. In cancer therapy, these nanoparticles have proven effective in delivering genes to tumor cells, slowing tumor growth. Adjusting the nanoparticle\'s surface, encapsulating protective agents, and using targeting ligands have also improved gene delivery efficiency. Smart nanoparticles based on chitosan have shown promise in improving outcomes by selectively releasing genes in response to tumor conditions, enhancing targeted delivery, and reducing off-target effects. Additionally, targeting ligands on the nanoparticles\' surface increases uptake and effectiveness. Although further investigation is needed to optimize the structure and composition of these nanoparticles and assess their long-term safety, these advancements pave the way for innovative gene-focused cancer therapies.
    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
    腹主动脉瘤(AAA)是一种危及生命的血管疾病,而目前缺乏阻止AAA进展的药物干预措施。为了解决AAA的多方面病理问题,这项工作开发了一种新型的多功能基因递送系统,以同时递送靶向MMP-2和MMP-9的两种siRNA。系统(TPNs-siRNA),通过表没食子儿茶素没食子酸酯(EGCG)的氧化聚合和自组装形成,在自组装过程中有效地封装siRNA。TPNs-siRNA保护siRNA免受生物降解,促进细胞内siRNA转染,促进溶酶体逃逸,并释放siRNA以沉默MMP-2和MMP-9。此外,TPNs,作为一种多种生物活性材料,减轻氧化应激和炎症,促进巨噬细胞的M1-M2复极化,抑制细胞钙化和凋亡。在AAA小鼠的实验中,TPNs-siRNA在静脉内递送后在动脉瘤组织中积累并持续存在,证明TPNs-siRNA可以显著分布在与AAA发病机制相关的巨噬细胞和VSMC中。利用载体固有的多生物活性特性,通过TPN的靶向siRNA递送对于增强的AAA治疗具有协同作用.此外,TPNs-siRNA逐渐代谢并从体内排出,导致优异的生物相容性。因此,TPNs作为一种有前途的多生物活性纳米疗法和用于有效AAA疗法的靶向递送纳米载体出现。
    Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease, while there is a lack of pharmaceutical interventions to halt AAA progression presently. To address the multifaceted pathology of AAA, this work develops a novel multifunctional gene delivery system to simultaneously deliver two siRNAs targeting MMP-2 and MMP-9. The system (TPNs-siRNA), formed through the oxidative polymerization and self-assembly of epigallocatechin gallate (EGCG), efficiently encapsulates siRNAs during self-assembly. TPNs-siRNA safeguards siRNAs from biological degradation, facilitates intracellular siRNA transfection, promotes lysosomal escape, and releases siRNAs to silence MMP-2 and MMP-9. Additionally, TPNs, serving as a multi-bioactive material, mitigates oxidative stress and inflammation, fosters M1-to-M2 repolarization of macrophages, and inhibits cell calcification and apoptosis. In experiments with AAA mice, TPNs-siRNA accumulated and persisted in aneurysmal tissue after intravenous delivery, demonstrating that TPNs-siRNA can be significantly distributed in macrophages and VSMCs relevant to AAA pathogenesis. Leveraging the carrier\'s intrinsic multi-bioactive properties, the targeted siRNA delivery by TPNs exhibits a synergistic effect for enhanced AAA therapy. Furthermore, TPNs-siRNA is gradually metabolized and excreted from the body, resulting in excellent biocompatibility. Consequently, TPNs emerges as a promising multi-bioactive nanotherapy and a targeted delivery nanocarrier for effective AAA 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
    金属有机骨架(MOF)由于其优异的孔隙率和高表面积而具有有效运载货物的潜力。然而,传统的MOFs及其衍生物在转运核酸和其他小分子时表现出低效率,以及具有差的胶体稳定性。在这项研究中,制备了负载氧化铁纳米粒子和Au纳米棒的ZIF-90,然后用聚乙烯亚胺(PEI)进行表面官能化,以产生具有pH值的多功能纳米复合材料(AFZP25k),光热,和磁响应。AFZP25k可以浓缩质粒DNA,形成AFZP25k/DNA复合物,最大结合效率为92.85%。DNA释放试验显示出显著的光和pH响应性,孵育6小时后累积释放超过80%。当施加外部磁场时,HeLa细胞的细胞摄取效率达到81.51%,具有低细胞毒性和特异性分布。体外转染实验证明HeLa细胞的基因转染效率为44.77%。在近红外照射之后,AFZP25k在HeLa细胞中的摄取效率和转染效率分别提高了21.3%和13.59%。这些发现表明AFZP25k作为癌症基因治疗中有效和靶向的基因递送载体的潜力。
    Metal-organic frameworks (MOFs) have the potential to efficiently carry cargo due to their excellent porosity and high surface area. Nevertheless, conventional MOFs and their derivatives exhibit low efficiency in transporting nucleic acids and other small molecules, as well as having poor colloidal stability. In this study, a ZIF-90 loaded with iron oxide nanoparticles and Au nanorods was prepared, and then surface-functionalized with polyethyleneimine (PEI) to create a multifunctional nanocomposite (AFZP25k) with pH, photothermal, and magnetic responsiveness. AFZP25k can condense plasmid DNA to form AFZP25k/DNA complexes, with a maximum binding efficiency of 92.85 %. DNA release assay showed significant light and pH responsiveness, with over 80 % cumulative release after 6 h of incubation. When an external magnetic field is applied, the cellular uptake efficiency in HeLa cells reached 81.51 %, with low cytotoxicity and specific distribution. In vitro transfection experiments demonstrated a gene transfection efficiency of 44.77 % in HeLa cells. Following near-infrared irradiation, the uptake efficiency and transfection efficiency of AFZP25k in HeLa cells increased by 21.3 % and 13.59 % respectively. The findings indicate the potential of AFZP25k as an efficient and targeted gene delivery vector in cancer gene therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号