gold nanocages

金纳米笼
  • 文章类型: Journal Article
    作为肝功能的重要指标,胆红素对临床诊断具有重要意义。基于未标记的金纳米笼(GNC)催化的胆红素氧化,已经建立了一种非酶传感器,用于灵敏的胆红素检测。通过一锅法制备具有双局部表面等离子体共振(LSPR)峰的GNC。500nm附近的一个峰归因于金纳米粒子(AuNP),另一个位于近红外区域的是GNC的典型峰。GNC对胆红素的催化氧化伴随着笼状结构的破坏,从纳米笼子中释放出自由的AuNPs.这种转变以相反的趋势改变了双峰强度,并使以比率方式实现胆红素的比色传感成为可能。吸光度比值在0.20~3.60μmol/L范围内与胆红素浓度呈良好的线性关系,检出限为39.35nM(3σ,n=3)。该传感器对胆红素的选择性优于其他共存物质。检测到真实人血清样品中的胆红素,回收率为94.5%至102.6%。胆红素测定方法简便,敏感,没有复杂的生物标签。
    As an essential indicator of liver function, bilirubin is of great significance for clinical diagnosis. A non-enzymatic sensor has been established for sensitive bilirubin detection based on the bilirubin oxidation catalyzed by unlabeled gold nanocages (GNCs). GNCs with dual-localized surface plasmon resonance (LSPR) peaks were prepared by a one-pot method. One peak around 500 nm was ascribed to gold nanoparticles (AuNPs), and the other located in the near-infrared region was the typical peak of GNCs. The catalytic oxidation of bilirubin by GNCs was accompanied by the disruption of cage structure, releasing free AuNPs from the nanocage. This transformation changed the dual peak intensities in opposite trend, and made it possible to realize the colorimetric sensing of bilirubin in a ratiometric mode. The absorbance ratios showed good linearity to bilirubin concentrations in the range of 0.20~3.60 μmol/L with a detection limit of 39.35 nM (3σ, n = 3). The sensor exhibited excellent selectivity for bilirubin over other coexisting substances. Bilirubin in real human serum samples was detected with recoveries ranging from 94.5 to 102.6%. The method for bilirubin assay is simple, sensitive and without complex biolabeling.
    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
    胰腺导管腺癌(PDAC)由于其化疗效果差和多药耐药性,仍然是最高的恶性肿瘤之一。化疗失败的主要原因是由于过度表达的细胞外基质(ECM)基质,药物在PDAC肿瘤组织中的积累不良。这构成了限制化疗药物深层组织渗透的主要障碍。在这里,我们报道了一种肿瘤微环境(TME)反应性纳米药物,基于PDAC细胞膜包覆的金纳米笼(AuNC),共同递送化疗剂(GEM)和氮氧化物(NO)供体(L-Arg)以增强药物积累并降低化学抗性。高谷胱甘肽(GSH)水平可以触发纳米药物上的二硫键的裂解以释放GEM。此外,升高的ROS水平可以激活L-Arg产生NO,其通过PDAC肿瘤组织中的血管舒张和血管正常化协同地促进GEM渗透到深部组织中。此外,AuNC不仅用作化疗的光热剂,但也产生光声信号来监测药物的积累和分布。不出所料,该策略在治疗不同的异种移植小鼠模型中表现出显著的效果,特别是在原位和患者来源的异种移植(PDX)模型中。当前的研究定义了用于治疗PDAC肿瘤的有用的治疗工具。
    Pancreatic ductal adenocarcinoma (PDAC) remains to be one of the highest malignant tumors due to its poor chemotherapeutic efficacy and multidrug resistance. A major reason for the failure in chemotherapy is poor drug accumulation into PDAC tumor tissues due to the overexpressed extracellular matrix (ECM) stroma, which forms a major obstacle limiting the deep tissue penetration of chemotherapeutics. Herein, we report a tumor microenvironment (TME)-responsive nanodrug, based on PDAC cell membrane-coated gold nanocages (AuNCs), to co-deliver the chemotherapeutics (GEM) and nitrogen oxide (NO) donor (L-Arg) to enhance drug accumulation and reduce chemoresistance. The high glutathione (GSH) level can trigger the cleavage of the disulfide bond on nanodrug to release GEM. Moreover, the elevated ROS level could activate L-Arg to generate NO, which synergistically facilitate GEM to penetrate into deep tissues by means of vasodilation and normalization of blood vessels in the PDAC tumor tissue. In addition, AuNCs not only serve as a photothermal agent for chemotherapy, but also generate photoacoustic signals to monitor drug accumulation and distribution. As expected, the strategy demonstrates to be remarkable in treating different xenograft mice models, especially in orthotopic and patient-derived xenograft (PDX) models. The current study defines a useful therapeutic tool for treating PDAC 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
    癌症免疫疗法代表了医学上的突破,但仍有许多患者由于反应率低而无法从中受益。免疫抑制性肿瘤微环境(TME)是免疫治疗的主要障碍。减轻肿瘤内免疫抑制对于提高免疫治疗效果至关重要。这项工作通过使用基于金纳米笼(AuNC)的光热效应与佐剂和PD-L1抑制剂的组合开发了原位疫苗接种策略。具体而言,这种治疗策略包括三个组成部分:AuNCs作为通过光热消融产生肿瘤抗原的诱导剂,CpG寡脱氧核苷酸作为增强免疫反应的佐剂,和JQ1作为PD-L1抑制因子来抑制免疫检查点。结果显示,原位疫苗接种有效地激活树突状细胞并引发T细胞,并且在携带黑色素瘤的小鼠中表现出高治疗功效。这种治疗策略可以增加细胞毒性T淋巴细胞的浸润,抑制PD-L1在肿瘤中的表达,并将肿瘤相关巨噬细胞从肿瘤前M2重新极化为抗肿瘤M1表型,从而通过调节先天免疫和适应性免疫应答来重塑TME。
    Cancer immunotherapy represents a medical breakthrough, but there are still many patients unable to benefit from it because of the low response rate. The immunosuppressive tumor microenvironment (TME) is the main barrier to immunotherapy. Alleviating intratumoral immunosuppression is critical for improving the immune therapeutic efficacy. This work developed an in situ vaccination strategy by using gold nanocage (AuNC)-based photothermal effect in combination with an adjuvant and PD-L1 suppressor. In specific, this therapeutic strategy included three components: AuNCs as an inducer for tumor antigen production via photothermal ablation, CpG oligodeoxynucleotides as an adjuvant to amplify immune responses, and JQ1 as a PD-L1 suppressor to inhibit an immune checkpoint. The results showed that the in situ vaccination efficiently activated dendritic cells and primed T cells and exhibited a high therapeutic efficacy in the melanoma-bearing mice. This therapeutic strategy can increase the infiltration of cytotoxic T lymphocytes, suppress the PD-L1 expression in the tumor, and repolarize tumor-associated macrophages from pro-tumor M2 to the anti-tumor M1 phenotype, thereby remodeling the TME via regulating the innate immune and adaptive immune responses.
    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
    随着纳米技术的进步,各种新型药物递送系统(DDS)已经出现,并在癌症的诊断和治疗中发挥了关键作用。在过去的二十年里,金纳米笼(AuNC)因其优异的性能而备受关注。这篇综述总结了当前内生、外源性,和双/多刺激响应性AuNC在药物递送中的应用。这篇评论的重点是属性,临床翻译潜力,以及用于癌症诊断和治疗的刺激响应性AuNC的局限性。
    With advances in nanotechnology, various new drug delivery systems (DDSs) have emerged and played a key role in the diagnosis and treatment of cancers. Over the last two decades, gold nanocages (AuNCs) have been attracting considerable attention because of their outstanding properties. This review summarizes current advancements in endogenous, exogenous, and dual/multi-stimuli responsive AuNCs in drug delivery. This review focuses on the properties, clinical translation potential, and limitations of stimuli-responsive AuNCs for cancer diagnosis and 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
    肝细胞癌(HCC)是一种具有有限治疗选择的癌症。表没食子儿茶素没食子酸酯(EGCG)是茶叶中主要的生物活性成分之一。大量研究表明EGCG对多种肿瘤具有预防和治疗作用。此外,近红外(NIR)响应型纳米平台的开发一直吸引着癌症治疗。在这项工作中,我们设计并合成了金纳米笼(AuNCs)作为控制EGCG释放的有效载体用于抗肿瘤,以实现NIR反应的协同作用并抑制肿瘤细胞增殖。AuNC的直径为约50nm并且具有中空多孔(8nm)结构。热成像图形研究证明,获得的AuNC-EGCG在近红外光下对激光辐照具有光热响应,并且在多次激光辐照后仍保持光稳定性。结果AuNCs-EGCG在48h时将HepG2细胞的增殖率降低至50%。Westernblot分析表明,NIR响应的AuNCs-EGCG可以促进HepG2细胞凋亡相关蛋白HSP70,细胞色素C,Caspase-9、Caspase-3和Bax,而Bcl-2的表达受到抑制。细胞共聚焦显微镜分析证明,NIR照射的AuNCs-EGCG显著上调Caspase-3近2倍,下调Bcl-2近0.33倍,有利于促进HepG2细胞凋亡。这项研究为NIR响应性AuNCs-EGCG作为HCC的新型纳米药物提供了有用的信息。
    Hepatocellular carcinoma (HCC) is a type of cancer that has a restricted therapy option. Epigallocatechin gallate (EGCG) is one of the main biologically active ingredients in tea. A large number of studies have shown that EGCG has preventive and therapeutic effects on various tumors. In addition, the development of near-infrared (NIR)-responsive nano-platforms has been attracting cancer treatment. In this work, we designed and synthesized a strategy of gold nanocages (AuNCs) as an efficient carrier for controlling release of EGCG for anti-tumor to achieve the synergistic functions of NIR-response and inhibited tumor cell proliferation. The diameter of AuNCs is about 50 nm and has a hollow porous (8 nm) structure. Thermal imaging-graphic studies proved that the AuNCs-EGCG obtained have photothermal response to laser irradiation under near-infrared light and still maintain light stability after multiple cycles of laser irradiation. The resulted AuNCs-EGCG reduced the proliferation rate of HepG2 cells to 50% at 48 h. Western blot analysis showed that NIR-responsive AuNCs-EGCG can promote the expression of HepG2 cell apoptosis-related proteins HSP70, Cytochrome C, Caspase-9, Caspase-3, and Bax, while the expression of Bcl-2 is inhibited. Cell confocal microscopy analysis proved that AuNCs-EGCG irradiated by NIR significantly upregulates Caspase-3 by nearly 2-fold and downregulates Bcl-2 by nearly 0.33-fold, which is beneficial to promote HepG2 cell apoptosis. This study provides useful information for the NIR-responsive AuNCs-EGCG as a new type of nanomedicine for HCC.
    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
    未经证实:由于缺乏可以选择性地将治疗性基因传递给癌细胞的有效纳米载体,因此阻碍了基因治疗治疗前列腺癌的使用。为了克服这一点,我们假设缀合乳铁蛋白,肿瘤靶向配体,和二氨基丁酸聚丙烯亚胺树枝状聚合物进入金纳米笼,然后与质粒DNA复合,在不使用外部刺激的情况下,将增强前列腺癌细胞中的基因表达和抗增殖活性。
    UNASSIGNED:合成并表征了具有乳铁蛋白并与二氨基丁酸聚丙烯亚胺树枝状聚合物(AuNC-DAB-Lf)缀合的新型金纳米笼。与质粒DNA复合后,他们的基因表达,在PC-3前列腺癌细胞上评估细胞摄取和抗增殖功效.
    未经鉴定:AuNC-DAB-Lf能够以5:1以上的缀合物:DNA重量比复合DNA。基因表达在用AuNC-DAB-Lf以10:1的重量比处理后达到最高,这是由于在PC-3细胞中以该比例由缀合物介导的DNA摄取显著增加。因此,与编码TNFα的DAB树枝状复合物相比,编码TNFα的AuNC-DAB-Lf-DNA的抗增殖活性显着提高了9倍。
    UNASSIGNED:带有乳铁蛋白的树枝状聚合物缀合的金纳米笼是用于治疗前列腺癌的非常有前途的基因递送系统。
    UNASSIGNED: The use of gene therapy to treat prostate cancer is hampered by the lack of effective nanocarriers that can selectively deliver therapeutic genes to cancer cells. To overcome this, we hypothesize that conjugating lactoferrin, a tumor-targeting ligand, and the diaminobutyric polypropylenimine dendrimer into gold nanocages, followed by complexation with a plasmid DNA, would enhance gene expression and anti-proliferation activity in prostate cancer cells without the use of external stimuli.
    UNASSIGNED: Novel gold nanocages bearing lactoferrin and conjugated to diaminobutyric polypropylenimine dendrimer (AuNCs-DAB-Lf) were synthesized and characterized. Following complexation with a plasmid DNA, their gene expression, cellular uptake and anti-proliferative efficacies were evaluated on PC-3 prostate cancer cells.
    UNASSIGNED: AuNCs-DAB-Lf was able to complex DNA at conjugate: DNA weight ratios 5:1 onwards. Gene expression was at its highest after treatment with AuNCs-DAB-Lf at a weight ratio of 10:1, as a result of a significant increase in DNA uptake mediated by the conjugate at that ratio in PC-3 cells. Consequently, the anti-proliferative activity of AuNCs-DAB-Lf-DNA encoding TNFα was significantly improved by up to 9-fold compared with DAB dendriplex encoding TNFα.
    UNASSIGNED: Lactoferrin-bearing dendrimer-conjugated gold nanocages are highly promising gene delivery systems for the treatment of prostate 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
    使用理想的生物材料来治疗受伤的骨骼可以加速愈合过程并同时表现出抗菌特性;从而保护患者免受细菌感染。因此,这项工作的目的是合成含有硅酸盐基生物活性玻璃和不同类型贵金属结构的复合材料(即,AgI金字塔,AgIAu复合材料,Au纳米笼,添加AgI的Au纳米笼)。生物活性玻璃被用作骨传导性骨替代品,Ag被用于其抗菌特性,同时包含Au以加速新骨的形成。为了研究这些复合材料中的协同效应,以两种方式进行两种合成:一步添加AgIAu复合材料或分别添加AgI金字塔和Au纳米笼。所有复合材料均显示出良好的体外生物活性。生物活性玻璃中的AgI转化为Ag纳米颗粒和其他银物种导致良好的抗菌行为。观察到Ag纳米颗粒保留在Au纳米笼中,这在抗菌性能方面也是有益的。Au纳米颗粒的存在有助于复合材料实现高细胞活力。通过在生物活性玻璃中连续添加贵金属获得了最杰出的结果,导致高抗菌效果和良好的细胞活力。
    Using an ideal biomaterial to treat injured bones can accelerate the healing process and simultaneously exhibit antibacterial properties; thus protecting the patient from bacterial infections. Therefore, the aim of this work was to synthesize composites containing silicate-based bioactive glasses and different types of noble metal structures (i.e., AgI pyramids, AgIAu composites, Au nanocages, Au nanocages with added AgI). Bioactive glass was used as an osteoconductive bone substitute and Ag was used for its antibacterial character, while Au was included to accelerate the formation of new bone. To investigate the synergistic effects in these composites, two syntheses were carried out in two ways: AgIAu composites were added in either one step or AgI pyramids and Au nanocages were added separately. All composites showed good in vitro bioactivity. Transformation of AgI in bioactive glasses into Ag nanoparticles and other silver species resulted in good antibacterial behavior. It was observed that the Ag nanoparticles remained in the Au nanocages, which was also beneficial in terms of antibacterial properties. The presence of Au nanoparticles contributed to the composites achieving high cell viability. The most outstanding result was obtained by the consecutive addition of noble metals into the bioactive glasses, resulting in both a high antibacterial effect and good cell viability.
    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
    Bacterial infection and its severe oxidative stress reaction will cause damage to skin cell mitochondria, resulting in long-lasting wound healing and great pain to patients. Thus, delayed wound healing in diabetic patients with Staphylococcus aureus infection is a principal challenge worldwide. Therefore, novel biomaterials with multifunction of bacterial membrane destruction and skin cell mitochondrial protection are urgently needed to be developed to address this challenge. In this work, novel gold cage (AuNCs) modified with epigallocatechin gallate (EGCG) were prepared to treat delayed diabetic wounds. The results showed that Au-EGCG had a high and stable photothermal conversion efficiency under near-infrared irradiation, and the scavenging rate of Au-EGCG for S. aureus could reach 95%. The production of large amounts of reactive oxygen species (ROS) leads to the disruption of bacterial membranes, inducing bacterial lysis and apoptosis. Meanwhile, Au-EGCG fused into hydrogel (Au-EGCG@H) promoted the migration and proliferation of human umbilical cord endothelial cells, reduced cellular mitochondrial damage and oxidative stress in the presence of infection, and significantly increased the basic fibroblast growth factor expression and vascular endothelial growth factor. In addition, animal studies showed that wound closure was 97.2% after 12 days of treatment, and the healing of chronic diabetic wounds was significantly accelerated. Au-EGCG nanoplatforms were successfully prepared to promote cell migration and angiogenesis in diabetic rats while removing S. aureus, reducing oxidative stress in cells, and restoring impaired mitochondrial function. Au-EGCG provides an effective, biocompatible, and multifunctional therapeutic strategy for chronic diabetic wounds.
    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
    Continuous high doses of radiation can cause irreversible side effects and radiation resistance; thus, advanced radiosensitizers are urgently needed. To overcome this problem, we developed a nano platelet radiosensitization system (PCA) by coating the chemotherapeutic drug cisplatin (CDDP) loaded gold nanocages (AuNs) within the platelet membrane. The developed PCA system may enable AuNs to have immune escape and targeting capabilities. After administration, PCA will actively target tumor cells and avoid being cleared by the immune system. Subsequently, CDDP, which destroys tumor cell DNA, can not only kill tumor cells directly but also combine with AuNs, which deposit radiation energy into tumor tissues, reducing RT resistance. In vivo and in vitro studies revealed that the combination of PCA with RT (2Gy) efficiently inhibits tumor proliferation without causing side effects such as inflammation. To conclude, this is the first attempt to use platelet membranes to correctly transport AuNs while also accomplishing low-dose RT, which could help AuNs-based tumor RT become more effective.
    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
    微泡是临床上用于诊断超声检查的血管内造影剂,超声心动图,和放射学成像应用。然而,到目前为止,创建具有多种功能的微泡的想法(例如,多模态成像,光动力疗法)仍然是一个挑战。一种可能的解决方案是通过引入负责此类功能的特定化合物来修饰气泡壳。在目前的工作中,空气核心微泡,外壳由牛血清白蛋白组成,白蛋白包被的金纳米笼,使用超声方法制备了锌酞菁。各种物理化学参数,例如随时间的稳定性,尺寸,和浓度进行了研究,以证明这些微泡作为造影剂的潜在用途。这项工作表明,混合微泡具有多模态成像的所有必要特性(超声,光栅扫描显微镜,和荧光断层扫描),这对潜在的治疗和相关的生物医学应用表现出优异的特性。
    Microbubbles are intravascular contrast agents clinically used in diagnostic sonography, echocardiography, and radiology imaging applications. However, up to date, the idea of creating microbubbles with multiple functionalities (e.g., multimodal imaging, photodynamic therapy) remained a challenge. One possible solution is the modification of bubble shells by introducing specific compounds responsible for such functions. In the present work, air-core microbubbles with the shell consisting of bovine serum albumin, albumin-coated gold nanocages, and zinc phthalocyanine were prepared using the sonication method. Various physicochemical parameters such as stability over time, size, and concentration were investigated to prove the potential use of these microbubbles as contrast agents. This work shows that hybrid microbubbles have all the necessary properties for multimodal imaging (ultrasound, raster-scanning microscopy, and fluorescence tomography), which demonstrate superior characteristics for potential theranostic and related biomedical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号