Nanovesicle

纳米囊泡
  • 文章类型: Editorial
    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
    囊泡,用超薄壁包围体积的微观单元,在生物材料中无处不在。然而,创造无机金属基人造产品仍然是一个巨大的挑战。这里,受到生物囊泡形成的启发,我们提出了一种新颖的仿生策略,将超薄纳米片卷曲成纳米囊泡,这是由界面应变驱动的。受到最初形成的衬底Rh层和随后形成的RhRu覆盖层之间的界面应变的影响,纳米片开始变形以释放一定量的应变。密度泛函理论(DFT)计算表明,Ru原子使纳米片的卷曲在热力学应用中更有利。由于独特的囊泡结构,RhRu纳米囊泡/C表现出优异的氢氧化反应(HOR)活性和稳定性,实验和DFT计算都证明了这一点。具体来说,RhRu纳米囊泡/C的HOR质量活性为7.52Amg(RhRu)-1,在旋转圆盘电极(RDE)水平的过电位为50mV;这是商业Pt/C(0.31mAmgPt-1)的24.19倍。此外,具有RhRu纳米囊泡/C的氢氧化物交换膜燃料电池(HEMFC)在H2-O2条件下显示出1.62Wcm-2的峰值功率密度,比商业Pt/C(1.18Wcm-2)好得多。这项工作创造了一种新的仿生策略来合成无机纳米材料,为设计催化反应器铺平了道路。
    Vesicle, a microscopic unit that encloses a volume with an ultrathin wall, is ubiquitous in biomaterials. However, it remains a huge challenge to create its inorganic metal-based artificial counterparts. Here, inspired by the formation of biological vesicles, we proposed a novel biomimetic strategy of curling the ultrathin nanosheets into nanovesicles, which was driven by the interfacial strain. Trapped by the interfacial strain between the initially formed substrate Rh layer and subsequently formed RhRu overlayer, the nanosheet begins to deform in order to release a certain amount of strain. Density functional theory (DFT) calculations reveal that the Ru atoms make the curling of nanosheets more favorable in thermodynamics applications. Owing to the unique vesicular structure, the RhRu nanovesicles/C displays excellent hydrogen oxidation reaction (HOR) activity and stability, which has been proven by both experiments and DFT calculations. Specifically, the HOR mass activity of RhRu nanovesicles/C are 7.52 A mg(Rh+Ru)-1 at an overpotential of 50 mV at the rotating disk electrode (RDE) level; this is 24.19 times that of commercial Pt/C (0.31 mA mgPt-1). Moreover, the hydroxide exchange membrane fuel cell (HEMFC) with RhRu nanovesicles/C displays a peak power density of 1.62 W cm-2 in the H2-O2 condition, much better than that of commercial Pt/C (1.18 W cm-2). This work creates a new biomimetic strategy to synthesize inorganic nanomaterials, paving a pathway for designing catalytic reactors.
    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
    对胰腺导管腺癌(PDAC)的临床治疗仍然存在难以察觉的检查和治疗效果不佳的问题。目前,尽管5-氟尿嘧啶(5-FU),作为临床一线FOLFIRINOX化疗药物,取得了显著的治疗效果。然而,这些不可避免的因素,如低溶解度,缺乏生物特异性,容易诱导免疫抑制环境的形成,严重限制了他们在PDAC的治疗。作为许多肿瘤细胞的重要能量来源,色氨酸(Trp),容易被吲哚胺2,3-双加氧酶1(IDO1)降解为犬尿氨酸(Kyn),激活Kyn-AHR轴以形成促进肿瘤生长和转移的特殊抑制性免疫微环境。然而,我们的研究发现,5-FU可以诱导有效的免疫原性细胞死亡(ICD),通过激活免疫系统进一步治疗肿瘤,而干扰素-γ(IFN-γ)的分泌重新诱导Kyn-AHR轴的激活,导致治疗效率低下。因此,金属基质蛋白酶-2(MMP-2)和内源性GSH双响应脂质体基纳米囊泡,与5-FU(抗癌药物)和NLG919(IDO1抑制剂)共负载,已建成(命名为ENP919@5-FU)。多功能ENP919@5-FU可有效重塑肿瘤免疫抑制微环境,增强化疗疗效,从而有效抑制癌症生长。机械上,高表达MMP-2的PDAC将通过在纳米囊泡表面上脱落PEG推动制备的纳米囊泡停留在肿瘤区域,有效增强肿瘤的摄取。随后,通过高内源性GSH切割含有S-S键的纳米囊泡,导致5-FU和NLG919的持续释放,从而使循环化学免疫疗法能够有效地引起肿瘤消融。此外,ENP919@5-FU联合PD-L1抗体(αPD-L1)对腹腔转移的PDAC模型具有协同抗肿瘤作用。总的来说,ENP919@5-FU纳米囊泡,作为PDAC治疗策略,通过重塑肿瘤微环境循环肿瘤化学免疫疗法扩增显示出优异的抗肿瘤疗效,在精准医学方法中具有很好的潜力。
    Imperceptible examination and unideal treatment effect are still intractable difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). At present, despite 5-fluorouracil (5-FU), as a clinical first-line FOLFIRINOX chemo-drug, has achieved significant therapeutic effects. Nevertheless, these unavoidable factors such as low solubility, lack of biological specificity and easy to induce immunosuppressive surroundings formation, severely limit their treatment in PDAC. As an important source of energy for many tumor cells, tryptophan (Trp), is easily degraded to kynurenine (Kyn) by indolamine 2,3- dioxygenase 1 (IDO1), which activates the axis of Kyn-AHR to form special suppressive immune microenvironment that promotes tumor growth and metastasis. However, our research findings that 5-FU can induce effectively immunogenic cell death (ICD) to further treat tumor by activating immune systems, while the secretion of interferon-γ (IFN-γ) re-induce the Kyn-AHR axis activation, leading to poor treatment efficiency. Therefore, a metal matrix protease-2 (MMP-2) and endogenous GSH dual-responsive liposomal-based nanovesicle, co-loading with 5-FU (anti-cancer drug) and NLG919 (IDO1 inhibitor), was constructed (named as ENP919@5-FU). The multifunctional ENP919@5-FU can effectively reshape the tumor immunosuppression microenvironment to enhance the effect of chemoimmunotherapy, thereby effectively inhibiting cancer growth. Mechanistically, PDAC with high expression of MMP-2 will propel the as-prepared nanovesicle to dwell in tumor region via shedding PEG on the nanovesicle surface, effectively enhancing tumor uptake. Subsequently, the S-S bond containing nanovesicle was cut via high endogenous GSH, leading to the continued release of 5-FU and NLG919, thereby enabling circulating chemoimmunotherapy to effectively cause tumor ablation. Moreover, the combination of ENP919@5-FU and PD-L1 antibody (αPD-L1) showed a synergistic anti-tumor effect on the PDAC model with abdominal cavity metastasis. Collectively, ENP919@5-FU nanovesicle, as a PDAC treatment strategy, showed excellent antitumor efficacy by remodeling tumor microenvironment to circulate tumor chemoimmunotherapy amplification, which has promising potential in a precision medicine approach.
    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
    小细胞外囊泡(sEV)有望在心肌梗死后进行基于细胞的心脏修复。这些电动汽车封装了有力的货物,包括microRNAs(miRs),在双层膜内,当施用于细胞时,有助于sEV摄取。然而,尽管它们的功效,sEV疗法受限于从亲本细胞释放的sEV的不一致性和货物包封的可变性。具有人工双层膜的合成sEV模拟物允许货物控制,但在体内施用时具有差的稳定性和快速清除。这里,我们使用电穿孔技术开发了一种类似电动汽车的车辆(ELV),在我们之前发表的作品的基础上,并研究了在体外和体内将带有促血管生成miR-126的电穿孔ELV递送至缺血再灌注大鼠模型的效力。我们表明,电穿孔的miR-126+ELV在给予心脏内皮细胞的2D培养物时改善了管形成参数,并且在缺血再灌注损伤后递送至大鼠左心室时改善了超声心动图和组织学参数。这项工作强调了使用电穿孔的ELV作为递送用于心脏修复的选择miR货物的载体的价值。
    Small extracellular vesicles (sEVs) are promising for cell-based cardiac repair after myocardial infarction. These sEVs encapsulate potent cargo, including microRNAs (miRs), within a bilayer membrane that aids sEV uptake when administered to cells. However, despite their efficacy, sEV therapies are limited by inconsistencies in the sEV release from parent cells and variability in cargo encapsulation. Synthetic sEV mimics with artificial bilayer membranes allow for cargo control but suffer poor stability and rapid clearance when administered in vivo. Here, we developed an sEV-like vehicle (ELV) using an electroporation technique, building upon our previously published work, and investigated the potency of delivering electroporated ELVs with pro-angiogenic miR-126 both in vitro and in vivo to a rat model of ischemia-reperfusion. We show that electroporated miR-126+ ELVs improve tube formation parameters when administered to 2D cultures of cardiac endothelial cells and improve both echocardiographic and histological parameters when delivered to a rat left ventricle after ischemia reperfusion injury. This work emphasizes the value of using electroporated ELVs as vehicles for delivery of select miR cargo for cardiac repair.
    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
    已经研究了治疗性寡核苷酸的脑靶向递送作为各种脑疾病的新治疗方式。比如脑肿瘤。然而,由于血脑屏障,进入大脑的递送效率受到限制。在这项研究中,脑靶向外泌体模拟细胞膜纳米囊泡(CMNV)旨在增强治疗性寡核苷酸向脑内的递送.首先,用分离的C6细胞膜片段通过挤出产生CMNV。然后,CMNV用胆固醇连接的T7肽作为疏水相互作用的靶向配体进行修饰,生产T7-CMNV。T7-CMNV在水溶液中保持其纳米颗粒尺寸超过21天。在原位胶质母细胞瘤动物模型中评估T7-CMNVs的靶向和递送效果。将2'-O-甲基和胆固醇-TEG修饰的抗microRNA-21寡核苷酸(AMO21c)加载到T7-CMNVs中,生物分布实验表明,T7-CMNVs比CMNVs更有效地将AMO21c传递到大脑中,加扰的T7-CMNVs,lipofectamine,和全身给药后的裸AMO21c。此外,在T7-CMNVs组中,AMO21c最有效地下调成胶质细胞瘤组织中的miRNA-21(miR-21)水平。这种增强的miR-21抑制导致PDCD4和PTEN的上调。最终,T7-CMNVs组比其他对照组更有效地缩小了脑肿瘤大小.有了稳定性,低毒性,和目标效率,T7-CMNVs可用于开发针对脑肿瘤的寡核苷酸疗法。
    The brain-targeted delivery of therapeutic oligonucleotides has been investigated as a new treatment modality for various brain diseases, such as brain tumors. However, delivery efficiency into the brain has been limited due to the blood-brain barrier. In this research, brain-targeted exosome-mimetic cell membrane nanovesicles (CMNVs) were designed to enhance the delivery of therapeutic oligonucleotides into the brain. First, CMNVs were produced by extrusion with isolated C6 cell membrane fragments. Then, CMNVs were decorated with cholesterol-linked T7 peptides as a targeting ligand by hydrophobic interaction, producing T7-CMNV. T7-CMNV was in aqueous solution maintained its nanoparticle size for over 21 days. The targeting and delivery effects of T7-CMNVs were evaluated in an orthotopic glioblastoma animal model. 2\'-O-metyl and cholesterol-TEG modified anti-microRNA-21 oligonucleotides (AMO21c) were loaded into T7-CMNVs, and biodistribution experiments indicated that T7-CMNVs delivered AMO21c more efficiently into the brain than CMNVs, scrambled T7-CMNVs, lipofectamine, and naked AMO21c after systemic administration. In addition, AMO21c down-regulated miRNA-21 (miR-21) levels in glioblastoma tissue most efficiently in the T7-CMNVs group. This enhanced suppression of miR-21 resulted in the up-regulation of PDCD4 and PTEN. Eventually, brain tumor size was reduced in the T7-CMNVs group more efficiently than in the other control groups. With stability, low toxicity, and targeting efficiency, T7-CMNVs may be useful to the development of oligonucleotide therapy for brain tumors.
    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
    耐药性是导致临床化疗结果不足的原因。一氧化氮(NO)征服耐药性的新作用已被认为是一种潜在的策略。然而,它仍然是一个巨大的挑战,以实现有针对性的递送以及准确的释放NO在所需的位置。在这里,我们开发了一种聚乙二醇化吲哚菁绿(mPEG-ICG)集成纳米囊泡系统(PIDA),以同时加载盐酸多柔比星(DOX·HCl)和NO供体L-精氨酸(L-Arg),可以通过NIR光照射触发产生NO,并进行多模式治疗以敏化耐药癌症。在808nm照射下,从PIDA释放的NO导致线粒体膜电位降低,K562/ADR细胞中ROS的增加和ATP的显著耗竭,从而抑制细胞生长,解决耐药问题。因此,对携带K562/ADR的裸鼠的体内实验表明,PIDA纳米囊泡具有显着的抗癌功效,肿瘤抑制率为80.8%。最重要的是,PIDA纳米囊泡为设计用于耐药性癌症治疗的纳米平台提供指导。
    Drug resistance is accountable for the inadequate outcome of chemotherapy in clinics. The newly emerging role of nitric oxide (NO) to conquer drug resistance has been recognized as a potential strategy. However, it remains a great challenge to realize targeted delivery as well as accurate release of NO at desired sites. Herein, we developed a PEGylated indocyanine green (mPEG-ICG) integrated nanovesicle system (PIDA) to simultaneously load doxorubicin hydrochloride (DOX⋅HCl) and the NO donor L-arginine (L-Arg), which can produce NO triggered by NIR light irradiation and exert multimodal therapy to sensitize drug-resistant cancers. Upon 808 nm irradiation, the NO released from PIDA led to a decrease in mitochondrial membrane potential, an increase in ROS and significant ATP depletion in K562/ADR cells, thus inhibiting cell growth and resolving the problem of drug resistance. Consequently, the in vivo experiment on K562/ADR-bearing nude mice indicated that PIDA nanovesicles achieved significant anticancer efficacy with a tumor inhibition rate of 80.8%. Above all, PIDA nanovesicles offer guidance for designing nanoplatforms for drug-resistant cancer treatment.
    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
    外泌体是细胞间传递信息并构成新的细胞间通讯模式的纳米级生物胞外囊泡。外泌体作为一种理想的纳米载体具有许多优点,包括良好的生物相容性,渗透性,低毒性,低免疫原性。最近,外泌体已被用于递送化疗剂,天然药物,核酸药物,和其他抗肿瘤药物来治疗许多类型的肿瘤。由于外泌体的产量有限,合成的外泌体模拟物已经被开发为药物递送的理想平台。本文综述了近年来外泌体和外泌体模拟药物在癌症治疗中的应用进展。
    Exosomes are nano-sized biological extracellular vesicles transmitting information between cells and constituting a new intercellular communication mode. Exosomes have many advantages as an ideal drug delivery nanocarrier, including good biocompatibility, permeability, low toxicity, and low immunogenicity. Recently, exosomes have been used to deliver chemotherapeutic agents, natural drugs, nucleic acid drugs, and other antitumor drugs to treat many types of tumors. Due to the limited production of exosomes, synthetic exosome-mimics have been developed as an ideal platform for drug delivery. This review summarizes recent advances in the application of exosomes and exosome-mimics delivering therapeutic drugs in treating cancers.
    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
    血小板在中性粒细胞的募集中起着至关重要的作用,由P-选择素介导,CCL5和ICAM-2。在这项研究中,我们用活化的血小板制备了血小板膜纳米囊泡。活化的血小板膜纳米囊泡能否募集中性粒细胞尚未见报道,在抗肿瘤免疫中也没有作用。SDS-PAGE结果表明,血小板膜纳米囊泡几乎保留了血小板的所有蛋白质。Western印迹显示活化的血小板和血小板膜纳米囊泡均表达P-选择素,ICAM-2和CCL5。乳腺癌移植瘤小鼠模型的体内结果显示肿瘤体积显著减小,Ki-67阳性肿瘤细胞减少,用活化的血小板膜纳米囊泡(aPNs)治疗后,肿瘤中TUNEL阳性肿瘤细胞增加。用aPN治疗后,不仅仅是中性粒细胞的数量,CD8+,CD4+T细胞,B细胞增加,还有IL-12,TNF-α,肿瘤组织中IFN-γ水平显著升高。
    Platelets play a crucial role in the recruitment of neutrophils, mediated by P-selectin, CCL5, and ICAM-2. In this study, we prepared platelet membrane nanovesicles from activated platelets. Whether activated platelet membrane nanovesicles can recruit neutrophils has not been reported, nor has their role in antitumor immunity. The results of SDS-PAGE showed that the platelet membrane nanovesicles retained almost all the proteins of platelets. Western blotting showed that both the activated platelets and the platelet membrane nanovesicles expressed P-selectin, ICAM-2, and CCL5. In vivo results of a mouse model of breast cancer-transplanted tumor showed that tumor volume reduced significantly, Ki-67-positive tumor cells decreased, and TUNEL-positive tumor cells increased in tumors after treatment with activated platelet membrane nanovesicles (aPNs). After treatment with aPNs, not only the number of neutrophils, CD8+, CD4+ T cells, and B cells increased, but also IL-12, TNF-α, and IFN-γ levels elevated significantly in tumor tissues.
    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
    背景:作为一种经典的自噬抑制剂,CQ被认为可以增加肿瘤对化疗药物的敏感性。然而,实验室研究和临床应用之间存在相当大的差距,这在很大程度上与CQ的独特药代动力学行为有关。方法:基于两亲性共聚物PPAP,pH响应药物诱导的自组装纳米囊泡,命名为DC-DIV/C,构造为加载DOX·HCl和CQ。表征了DC-DIV/C的理化性质。为了验证DOX·HCl和CQ的协同作用和递送同步性,细胞毒性,凋亡,在DOX·HCl耐药癌细胞中研究了细胞摄取和自噬测定。评价DC-DIV/C在大鼠和荷瘤裸鼠中的药代动力学特征和抗肿瘤作用。分别。结果:DC-DIV/C可以同时包封DOX·HCl和CQ,最佳比例为1:2。体外和体内测试证实,DC-DIV/C是在血液循环过程中同步递送DOX·HCl和CQ的优异载体,细胞摄取和细胞内释放。此外,CQ完成自噬抑制以降低DOX·HCl抗性癌细胞的IC50。因此,DC-DIV/C对K562/ADR肿瘤的抗肿瘤作用有84.52%的TIR显著改善。结论:本研究为精确联合治疗提供了一种有希望的有效策略,可以提高治疗效果。
    Background: As a classical autophagy inhibitor, CQ has been supposed to increase the sensitivity of tumors to chemotherapeutics. However, there exists a quite huge gap between laboratory research and clinical application, which is related to the distinct pharmacokinetic behavior of CQ to a great extent. Methods: Based on amphiphilic copolymer PPAP, a pH-responsive drug-induced self-assembled nanovesicle, named DC-DIV/C, was constructed to load DOX⋅HCl and CQ. The physicochemical properties of DC-DIV/C were characterized. To validate the cooperative action and delivery synchronism of DOX⋅HCl and CQ, cytotoxicity, apoptosis, cellular uptake and autophagy assay were investigated in DOX⋅HCl resistant cancer cells. The pharmacokinetic character and antitumor effect of DC-DIV/C were evaluated on rats and nude mice bearing xenograft drug-resistant K562/ADR tumors, respectively. Results: DC-DIV/C could simultaneously encapsulate DOX·HCl and CQ at the optimal ratio of 1:2. In vitro and in vivo tests confirmed that DC-DIV/C acted as an excellent vehicle for the synchronous delivery of DOX⋅HCl and CQ during the process of blood circulation, cellular uptake and intracellular release. Furthermore, CQ accomplished autophagy inhibition to reduce the IC50 of DOX⋅HCl resistant cancer cells. Consequently, DC-DIV/C exhibited the extremely improved anti-tumor effect with 84.52% TIR on K562/ADR tumor. Conclusion: This study provides a promising and powerful strategy to achieve enhanced treatment outcomes for the precise combination 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
    具有抗氧化能力的伤口敷料,抗炎,和有效的血管生成诱导有望有效促进伤口愈合。在这里,提出了一种新型的核壳透明质酸(HA)微针(MN)贴片,该贴片具有铁-间充质干细胞衍生的人工纳米囊泡(Fe-MSC-NVs)和包裹在针尖中的聚多巴胺纳米颗粒(PDANP),用于伤口愈合。含有多功能治疗性细胞因子的Fe-MSC-NVs被包封在MN尖端的内部HA核心中,用于加速血管生成。PDANP被封装在MN尖端的外部甲基丙烯酸酯化透明质酸(HAMA)壳中以克服来自活性氧(ROS)衍生的氧化应激的不利影响。随着HAMA贴剂在皮肤中的逐渐降解,PDANP在病变处持续释放,以抑制ROS诱导的炎症反应,而Fe-MSC-NVs显着增加了迁移,扩散,和人脐静脉内皮细胞(HUVEC)的管形成。更吸引人的是,PDANP和Fe-MSC-NVs的组合进一步促进M2巨噬细胞极化,从而抑制伤口炎症。通过体内实验,Fe-MSC-NVs/PDAMN贴剂对糖尿病伤口愈合显示出优异的效果。这些抗氧化的特点,抗炎,和促血管生成表明所提出的复合核壳MN贴片对于临床伤口愈合应用是有价值的。
    Wound dressing with the capacities of antioxidation, antiinflammation, and efficient angiogenesis induction is expected for effectively promoting wound healing. Herein, a novel core-shell hyaluronic acid (HA) microneedle (MN) patch with ferrum-mesenchymal stem cell-derived artificial nanovesicles (Fe-MSC-NVs) and polydopamine nanoparticles (PDA NPs) encapsulated in the needle tips is presented for wound healing. Fe-MSC-NVs containing multifunctional therapeutic cytokines are encapsulated in the inner HA core of the MN tips for accelerating angiogenesis. The PDA NPs are encapsulated in the outer methacrylated hyaluronic acid (HAMA) shell of the MN tips to overcome the adverse impacts from reactive oxygen species (ROS)-derived oxidative stress. With the gradual degradation of HAMA patch tips in the skin, the PDA NPs are sustainably released at the lesion to suppress the ROS-induced inflammation reaction, while the Fe-MSC-NVs significantly increase the migration, proliferation, and tube formation of human umbilical vein endothelial cells (HUVEC). More attractively, the combination of PDA NPs and Fe-MSC-NVs further promotes M2 macrophage polarization, thereby suppressing wound inflammation. Through in vivo experiment, the Fe-MSC-NVs/PDA MN patch shows an excellent effect for diabetic wound healing. These features of antioxidation, antiinflammation, and pro-angiogenesis indicate the proposed composite core-shell MN patch is valuable for clinical wound healing applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号