extracellular vesicles (EVs)

细胞外囊泡 ( EV )
  • 文章类型: Journal Article
    肝细胞癌(HCC)是原发性肝癌最常见的类型,其特点是早期诊断困难,容易发生远处转移,手术后复发率高。细胞外囊泡(EV)是一类细胞衍生的颗粒,包括外泌体,以磷脂双层为特征。它们是细胞间通讯货物的有效载体,包括蛋白质和核酸,并广泛参与肿瘤进展。它们作为潜在的肿瘤生物标志物和新的治疗途径正在被探索。我们简要概述了电动汽车的生物发生和特征,以更好地了解其分类标准。本文就EV相关蛋白在HCC领域的研究进展作一综述。EV相关蛋白参与HCC的肿瘤生长和调节,参与肿瘤微环境(TME)内的细胞间通讯,并且与肿瘤转移过程中的血管生成和上皮间质转化(EMT)事件有关。此外,EV相关蛋白对HCC显示出有希望的诊断功效。对于HCC的治疗,它们还显示出巨大的潜力,包括增强肿瘤疫苗的功效,瞄准货锚。面对当前的挑战,提出了该领域未来的研究方向。最重要的是,对EV相关蛋白的研究有可能增强我们对HCC的理解,并为开发新的治疗策略提供新的见解。
    Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, characterized by difficulties in early diagnosis, prone to distant metastasis, and high recurrence rates following surgery. Extracellular vesicles (EVs) are a class of cell-derived particles, including exosomes, characterized by a phospholipid bilayer. They serve as effective carriers for intercellular communication cargo, including proteins and nucleic acids, and are widely involved in tumor progression. They are being explored as potential tumor biomarkers and novel therapeutic avenues. We provide a brief overview of the biogenesis and characteristics of EVs to better understand their classification standards. The focus of this review is on the research progress of EV-associated proteins in the field of HCC. EV-associated proteins are involved in tumor growth and regulation in HCC, participate in intercellular communication within the tumor microenvironment (TME), and are implicated in events including angiogenesis and epithelial-mesenchymal transition (EMT) during tumor metastasis. In addition, EV-associated proteins show promising diagnostic efficacy for HCC. For the treatment of HCC, they also demonstrate significant potential including enhancing the efficacy of tumor vaccines, and as targeting cargo anchors. Facing current challenges, we propose the future directions of research in this field. Above all, research on EV-associated proteins offers the potential to enhance our comprehension of HCC and offer novel insights for developing new treatment strategies.
    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
    类风湿性关节炎(RA)仍然是一种具有挑战性的慢性自身免疫性疾病,其特征是持续的关节炎症和损伤。虽然现代再生策略,包括基于细胞/干细胞的疗法,基因治疗,和组织工程学,有先进的组织修复努力,RA的最终治愈仍然难以捉摸。因此,有越来越多的兴趣开发靶向治疗,直接解决潜在的机制驱动RA发病机制,例如细胞外囊泡(EV)。这些小膜结合颗粒可以调节受损软骨的炎症微环境内的免疫应答。为了发挥电动汽车的临床潜力,它们可以通过几种技术从各种细胞类型中分离出来。电动汽车可以携带各种生物活性分子和抗炎或促再生药物,将它们直接送到受影响的关节,影响受损细胞的行为,使它们成为RA患者靶向治疗和药物递送的令人信服的选择。然而,仍然有几个挑战和限制与基于EV的治疗,包括缺乏电动汽车隔离的标准化协议,表征,和交付。这篇综述全面概述了RA中EV的细胞来源,并深入研究了它们的治疗潜力和必须克服的障碍。
    Rheumatoid arthritis (RA) remains a challenging chronic autoimmune disorder characterized by persistent joint inflammation and damage. While modern regenerative strategies, encompassing cell/stem cell-based therapies, gene therapy, and tissue engineering, have advanced tissue repair efforts, a definitive cure for RA remains elusive. Consequently, there is growing interest in developing targeted therapies that directly address the underlying mechanisms driving RA pathogenesis, such as extracellular vesicles (EVs). These small membrane-bound particles can modulate immune responses within the inflammatory microenvironment of damaged cartilage. To launch the clinical potential of EVs, they can be isolated from various cell types through several techniques. EVs can carry various bioactive molecules and anti-inflammatory or pro-regenerative drugs, deliver them directly to the affected joints, and affect the behavior of injured cells, making them a compelling choice for targeted therapy and drug delivery in RA patients. However, there are still several challenges and limitations associated with EV-based therapy, including the absence of standardized protocols for EV isolation, characterization, and delivery. This review provides a comprehensive overview of the cellular sources of EVs in RA and delves into their therapeutic potential and the hurdles they must overcome.
    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
    软骨损伤的恢复在骨科仍然是一个艰巨的挑战,主要归因于软骨中缺乏血管化和神经支配。脱细胞细胞外基质(dECM)来源于软骨,在通过去细胞化过程去除抗原之后,表现出显著的生物相容性和生物活性,使其成为软骨修复的可行候选者。此外,当与dECM结合时,从软骨产生的细胞外囊泡(EV)已经证明了协同作用,通过磷酸化4ebp可能减轻对蛋白质合成的抑制作用,从而促进软骨相关蛋白如胶原的合成。为了实现这个目标,我们创新了一种新型的生物墨水和修复支架,其特点是具有特殊的生物相容性,生物活性,和生物降解性,建立有利于软骨形成的组织特异性微环境。在大鼠骨软骨缺损中,生物活性支架成功促进透明软骨的形成,具有足够的机械强度,有利的弹性,和dECM沉积指示软骨。总之,这项研究有效地设计了一种为软骨修复量身定制的水凝胶生物墨水,并设计了一种生物活性软骨修复支架,该支架精通诱导细胞分化和促进软骨修复。
    The restoration of cartilage injuries remains a formidable challenge in orthopedics, chiefly attributed to the absence of vascularization and innervation in cartilage. Decellularized extracellular matrix (dECM) derived from cartilage, following antigenic removal through decellularization processes, has exhibited remarkable biocompatibility and bioactivity, rendering it a viable candidate for cartilage repair. Additionally, extracellular vesicles (EVs) generated from cartilage have demonstrated a synergistic effect when combined with dECM, potentially mitigating the inhibitory impact on protein synthesis by phosphorylating 4ebp, thereby promoting the synthesis of cartilage-related proteins such as collagen. In pursuit of this objective, we have innovated a novel bioink and repair scaffold characterized by exceptional biocompatibility, bioactivity, and biodegradability, establishing a tissue-specific microenvironment conducive to chondrogenesis. Within rat osteochondral defects, the biologically active scaffold successfully prompted the formation of transparent cartilage, featuring adequate mechanical strength, favorable elasticity, and dECM deposition indicative of cartilage. In summary, this study has effectively engineered a hydrogel bioink tailored for cartilage repair and devised a bioactive cartilage repair scaffold proficient in instigating cell differentiation and fostering cartilage 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
    这项研究提出并比较了两种方法,用于鉴定来自不同细胞系的细胞外囊泡(EV)的类型。通过SDS-PAGE分析,我们发现不同电动汽车中CD63与CD81的比例是一致和不同的,使其成为识别癌细胞分泌的电动汽车的可靠特征。然而,电泳和成像过程可能会在浓度值中引入误差,特别是在较低的浓度下,渲染这种方法可能不太有效。另一种方法涉及使用石英晶体微天平(QCM)和电分析叉指型电极(IDT)生物传感器进行EV类型识别和定量。电动汽车引起的QCM频移与其浓度成正比,虽然电分析依赖于测量I-V曲线的曲率作为一个显著特征,这也与EV浓度成正比。分别绘制各种EV类型的QCM频移和电分析曲率的线性回归线,使得能够在图形上估计未知EV类型的相应浓度。通过交叉两个生物传感器的结果,可以识别未知的EV类型。生物传感器分析方法被证明是分析来自不同细胞系的EV的类型和浓度的有效手段。
    This study presents and compares two methods for identifying the types of extracellular vesicles (EVs) from different cell lines. Through SDS-PAGE analysis, we discovered that the ratio of CD63 to CD81 in different EVs is consistent and distinct, making it a reliable characteristic for recognizing EVs secreted by cancer cells. However, the electrophoresis and imaging processes may introduce errors in the concentration values, especially at lower concentrations, rendering this method potentially less effective. An alternative approach involves the use of quartz crystal microbalance (QCM) and electroanalytical interdigitated electrode (IDT) biosensors for EV type identification and quantification. The QCM frequency shift caused by EVs is directly proportional to their concentration, while electroanalysis relies on measuring the curvature of the I-V curve as a distinguishing feature, which is also proportional to EV concentration. Linear regression lines for the QCM frequency shift and the electroanalysis curvature of various EV types are plotted separately, enabling the estimation of the corresponding concentration for an unknown EV type on the graphs. By intersecting the results from both biosensors, the unknown EV type can be identified. The biosensor analysis method proves to be an effective means of analyzing both the type and concentration of EVs from different cell lines.
    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
    脑肿瘤,and,特别是,胶质母细胞瘤(GBM),是最具侵略性的癌症之一。尽管现有的治疗方法取得了进展,诊断和治疗仍无法确保GBM患者的无病理生存期超过12-15个月.在应对脑肿瘤的能力仍然很差的基础上,我们可以考虑:(i)肿瘤内异质性;(ii)当我们比较不同患者时,肿瘤特性的异质性;(iii)血脑屏障(BBB),这使得分离肿瘤特异性生物标志物和向大脑输送治疗药物变得困难。最近,越来越清楚的是,癌细胞释放大量运输代谢物的细胞外囊泡(EV),蛋白质,不同种类的RNA,DNA,和脂质。这些结构参与病理过程并表征任何特定形式的癌症。此外,EV能够在两个方向上穿过BBB。从这些观察开始,研究人员现在正在评估使用从有机液体中纯化的电动汽车的可能性(首先,血液和唾液),为了获得,通过非侵入性方法(液体活检),肿瘤生物标志物,and,也许,还用于获得用于靶向递送药物的纳米载体。
    Brain tumors, and, in particular, glioblastoma (GBM), are among the most aggressive forms of cancer. In spite of the advancement in the available therapies, both diagnosis and treatments are still unable to ensure pathology-free survival of the GBM patients for more than 12-15 months. At the basis of the still poor ability to cope with brain tumors, we can consider: (i) intra-tumor heterogeneity; (ii) heterogeneity of the tumor properties when we compare different patients; (iii) the blood-brain barrier (BBB), which makes difficult both isolation of tumor-specific biomarkers and delivering of therapeutic drugs to the brain. Recently, it is becoming increasingly clear that cancer cells release large amounts of extracellular vesicles (EVs) that transport metabolites, proteins, different classes of RNAs, DNA, and lipids. These structures are involved in the pathological process and characterize any particular form of cancer. Moreover, EVs are able to cross the BBB in both directions. Starting from these observations, researchers are now evaluating the possibility to use EVs purified from organic fluids (first of all, blood and saliva), in order to obtain, through non-invasive methods (liquid biopsy), tumor biomarkers, and, perhaps, also for obtaining nanocarriers for the targeted delivering of drugs.
    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
    常规药物递送技术面临与靶向和不良反应相关的挑战。近年来,基于纳米颗粒的药物载体取得了重大进展。然而,对它们的安全性和代谢不足的担忧仍然存在。使用细胞及其衍生物,如细胞膜和细胞外囊泡(EV),作为药物载体有效地解决了与纳米颗粒载体相关的挑战。然而,有效地将药物装载到这些载体中仍然是一个重要的障碍。随着微流控技术的进步及其在微观和纳米尺度上的精确操纵优势,以及最小的样本损失,它在使用细胞及其衍生物装载药物方面得到了广泛的应用,从而促进了药物装载技术的发展。本文概述了利用细胞及其衍生物作为药物载体的特点和益处,特别是那些根植于微流体技术。微流控技术通过采用细胞及其衍生物的药物递送系统在靶向疾病治疗中的巨大潜力,是预见的。
    Conventional drug delivery techniques face challenges related to targeting and adverse reactions. Recent years have witnessed significant advancements in nanoparticle-based drug carriers. Nevertheless, concerns persist regarding their safety and insufficient metabolism. Employing cells and their derivatives, such as cell membranes and extracellular vesicles (EVs), as drug carriers effectively addresses the challenges associated with nanoparticle carriers. However, an essential hurdle remains in efficiently loading drugs into these carriers. With the advancement of microfluidic technology and its advantages in precise manipulation at the micro- and nanoscales, as well as minimal sample loss, it has found extensive application in the loading of drugs using cells and their derivatives, thereby fostering the development of drug-loading techniques. This paper outlines the characteristics and benefits of utilizing cells and their derivatives as drug carriers and provides an overview of current drug-loading techniques, particularly those rooted in microfluidic technology. The significant potential for microfluidic technology in targeted disease therapy through drug delivery systems employing cells and their derivatives, is foreseen.
    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
    长冠状病毒病2019(COVID-19)与急性后遗症影响肺和肺外器官系统的风险增加有关。高达20%的COVID-19患者可能会发展为更严重的形式,比如重症肺炎,急性呼吸窘迫综合征(ARDS),或肺纤维化。尽管如此,大多数患者可能只有轻度,自我限制的疾病。特别值得关注的是长期COVID-19患者的实质纤维化和肺功能障碍的可能性。此外,据观察,多达43%的COVID-19住院患者也有急性肾损伤(AKI).照顾肾脏,大脑,肺,心血管,肝脏,眼,和组织损伤应包括在急性COVID-19治疗后。作为再生医学中强大的免疫调节工具,牙科干细胞(DSC)引起了极大的兴趣。大量的免疫细胞和细胞因子参与过度的炎症反应,这对组织再生也有显著的影响。一种用于治疗急性肺损伤(ALI)的独特的干细胞(SCs)库,肝损伤,神经系统疾病,心血管问题,肾脏损伤可能在牙齿组织中发现,根据许多研究。此外,越来越多的体内研究正在连接DSC衍生的细胞外囊泡(DSC-EV),它们是必不可少的旁分泌效应物,对DSC的有益作用。DSC-EV,含有生物活性成分和某些疾病的治疗潜力,已被证明是治疗COVID-19后组织损伤的潜在有效疗法。因此,在这项工作中,我们探索了DSC的特性。接下来,我们将看看SARS-CoV-2如何影响组织损伤。最后,我们研究了DSC和DSC-EV在管理COVID-19和慢性组织损伤中的应用,比如对心脏的伤害,大脑,肺,和其他组织。
    Long coronavirus disease 2019 (COVID-19) is linked to an increased risk of post-acute sequelae affecting the pulmonary and extrapulmonary organ systems. Up to 20% of COVID-19 patients may proceed to a more serious form, such as severe pneumonia, acute respiratory distress syndrome (ARDS), or pulmonary fibrosis. Still, the majority of patients may only have mild, self-limiting sickness. Of particular concern is the possibility of parenchymal fibrosis and lung dysfunction in long-term COVID-19 patients. Furthermore, it has been observed that up to 43% of individuals hospitalized with COVID-19 also had acute renal injury (AKI). Care for kidney, brain, lung, cardiovascular, liver, ocular, and tissue injuries should be included in post-acute COVID-19 treatment. As a powerful immunomodulatory tool in regenerative medicine, dental stem cells (DSCs) have drawn much interest. Numerous immune cells and cytokines are involved in the excessive inflammatory response, which also has a significant effect on tissue regeneration. A unique reservoir of stem cells (SCs) for treating acute lung injury (ALI), liver damage, neurological diseases, cardiovascular issues, and renal damage may be found in tooth tissue, according to much research. Moreover, a growing corpus of in vivo research is connecting DSC-derived extracellular vesicles (DSC-EVs), which are essential paracrine effectors, to the beneficial effects of DSCs. DSC-EVs, which contain bioactive components and therapeutic potential in certain disorders, have been shown as potentially effective therapies for tissue damage after COVID-19. Consequently, we explore the properties of DSCs in this work. Next, we\'ll look at how SARS-CoV-2 affects tissue damage. Lastly, we have looked at the use of DSCs and DSC-EVs in managing COVID-19 and chronic tissue damage, such as injury to the heart, brain, lung, and other tissues.
    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
    骨肉瘤(OS)是一种罕见的影响软组织的恶性肿瘤,具有很高的肺转移率和死亡率。OS的主要治疗包括术前化疗,手术切除病灶,和术后化疗。然而,OS化疗提出了与治疗毒性和多药耐药性相关的严峻挑战。为了应对这些挑战,纳米技术已经开发出可以直接向OS细胞释放药物的纳米系统,降低药物的毒性。细胞外囊泡(EV)是纳米尺寸的脂质双层结合的囊泡,其充当几种癌症的细胞衍生载体和药物递送系统。本研究旨在通过共同递送Hdac1siRNA和唑来膦酸(zol)来利用EV进行OS管理。电动汽车表面用叶酸(FA)修饰,并将其靶向能力与天然电动汽车进行比较。结果表明,电动汽车的靶向能力取决于亲本细胞来源,FA共轭进一步增强了它。此外,EV用作共负载药物(zol)和小RNA(Hdac-1)的载体。这种使用表面工程电动汽车作为货物装载和交付载体的方法可能是骨肉瘤管理的有希望的策略。
    Osteosarcoma (OS) is a rare malignant tumor that affects soft tissue and has high rates of lung metastasis and mortality. The primary treatments for OS include preoperative chemotherapy, surgical resection of the lesion, and postoperative chemotherapy. However, OS chemotherapy presents critical challenges related to treatment toxicity and multiple drug resistance. To address these challenges, nanotechnology has developed nanosystems that release drugs directly to OS cells, reducing the drug\'s toxicity. Extracellular vesicles (EVs) are nanosized lipid-bilayer bound vesicles that act as cell-derived vehicles and drug delivery systems for several cancers. This study aims to utilize EVs for OS management by co-delivering Hdac1 siRNA and zoledronic acid (zol). The EVs\' surface is modified with folic acid (FA) and their targeting ability is compared to that of native EVs. The results showed that the EVs\' targeting ability depends on the parent cell source, and FA conjugation further enhanced it. Furthermore, EVs were used as the carrier for co-loading drug (zol) and small RNA (Hdac-1). This approach of using surface engineered EVs as carriers for cargo loading and delivery can be a promising strategy for osteosarcoma management.
    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
    间充质干细胞衍生的细胞外囊泡(MSC-EV)已被越来越多地研究用于癌症治疗和药物递送。他们提供了先进的无细胞治疗选择。然而,它们的整体效果和功效取决于各种因素,包括MSC来源和货物内容。在这项研究中,我们从人未成熟牙髓干细胞(hIDPSC-EVs)的条件培养基中分离出EVs,并研究了其对两种甲状腺乳头状癌(PTC)细胞系(BCPAP和TPC1)的影响.我们观察到两种PTC细胞系对hIDPSC-EV的有效摄取,对基因调控有显著影响,特别是在BCPAP细胞的Wnt信号通路中。然而,未观察到对细胞增殖的显著影响。相反,处理120小时后,hIDPSC-EV显着降低了两种PTC细胞系的侵袭能力。这些体外发现表明hIDPSC-EV在癌症管理中的治疗潜力,并强调需要进一步研究以开发新的有效治疗策略。此外,PTC细胞系成功内化hIDPSC-EV强调了它们作为纳米载体用于抗癌药物的潜在用途.
    Mesenchymal stem-cell-derived extracellular vesicles (MSC-EVs) have been increasingly investigated for cancer therapy and drug delivery, and they offer an advanced cell-free therapeutic option. However, their overall effects and efficacy depend on various factors, including the MSC source and cargo content. In this study, we isolated EVs from the conditioned medium of human immature dental pulp stem cells (hIDPSC-EVs) and investigated their effects on two papillary thyroid cancer (PTC) cell lines (BCPAP and TPC1). We observed efficient uptake of hIDPSC-EVs by both PTC cell lines, with a notable impact on gene regulation, particularly in the Wnt signaling pathway in BCPAP cells. However, no significant effects on cell proliferation were observed. Conversely, hIDPSC-EVs significantly reduced the invasive capacity of both PTC cell lines after 120 h of treatment. These in vitro findings suggest the therapeutic potential of hIDPSC-EVs in cancer management and emphasize the need for further research to develop novel and effective treatment strategies. Furthermore, the successful internalization of hIDPSC-EVs by PTC cell lines underscores their potential use as nanocarriers for anti-cancer agents.
    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
    COVID-19大流行在全球范围内造成了显著的发病率和死亡率。血浆来源的细胞外囊泡(EV)在小儿COVID-19患者中的作用尚不清楚。
    我们使用连续超速离心技术从健康对照(n=13)和儿科COVID-19患者(n=104)中分离出严重程度不同的EV。EV对健康PBMC的影响,幼稚CD4+T细胞,和单核细胞通过体外试验进行评估,流式细胞术,和ELISA。
    我们的研究结果表明,COVID-19的严重程度与多种免疫反应相关。严重急性病例显示细胞因子水平升高,IFNγ水平降低,降低CD4+T细胞和单核细胞计数,提示免疫抑制。重症急性患者的EV刺激健康细胞表达较高的PDL1,增加Th2和Treg细胞,减少IFNγ分泌,并改变了Th1/Th17比率。患者来源的EV显着降低单核细胞产生的促炎细胞因子(轻度,p<.001,严重病例的p=.0025)和刺激的健康PBMC中CD4T细胞(p=.043)和单核细胞(p=.033)群体减少。
    这项研究揭示了免疫反应与EV介导的作用之间的复杂关系,强调COVID-19严重程度的影响。我们强调了血浆来源的电动汽车在重症COVID-19患者早期免疫抑制中的潜在作用。
    UNASSIGNED: The COVID-19 pandemic has caused significant morbidity and mortality globally. The role of plasma-derived extracellular vesicles (EVs) in pediatric COVID-19 patients remains unclear.
    UNASSIGNED: We isolated EVs from healthy controls (n = 13) and pediatric COVID-19 patients (n = 104) with varying severity during acute and convalescent phases using serial ultracentrifugation. EV effects on healthy PBMCs, naïve CD4+ T cells, and monocytes were assessed through in vitro assays, flow cytometry, and ELISA.
    UNASSIGNED: Our findings indicate that COVID-19 severity correlates with diverse immune responses. Severe acute cases exhibited increased cytokine levels, decreased IFNγ levels, and lower CD4+ T cell and monocyte counts, suggesting immunosuppression. EVs from severe acute patients stimulated healthy cells to express higher PDL1, increased Th2 and Treg cells, reduced IFNγ secretion, and altered Th1/Th17 ratios. Patient-derived EVs significantly reduced proinflammatory cytokine production by monocytes (p < .001 for mild, p = .0025 for severe cases) and decreased CD4+ T cell (p = .043) and monocyte (p = .033) populations in stimulated healthy PBMCs.
    UNASSIGNED: This study reveals the complex relationship between immunological responses and EV-mediated effects, emphasizing the impact of COVID-19 severity. We highlight the potential role of plasma-derived EVs in early-stage immunosuppression in severe COVID-19 patients.
    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