Radiation-Protective Agents

  • 文章类型: Journal Article
    线粒体氧化应激是细胞凋亡的重要因素。氧化铈纳米材料具有清除自由基和模拟超氧化物歧化酶(SOD)和过氧化氢酶(CAT)活性的巨大潜力。为解决氧化铈纳米材料靶向性差的问题,我们设计了白蛋白-氧化铈纳米簇(TPP-PCNLs),其目标是用磷酸三苯酯(TPP)修饰线粒体。TPP-PCNLs有望模拟超氧化物歧化酶的活性,不断去除活性氧,并在辐射防护中发挥持久作用。
    首先,二氧化铈纳米团簇(CNLs),聚乙二醇二氧化铈纳米团簇(PCNLs),TPP-PCNLs的形态和大小进行了表征,紫外光谱,分散稳定性和细胞摄取,和共同定位随后,TPP-PCNLs的抗辐射作用进行了体外和体内实验,包括细胞活力,凋亡,彗星化验,组织病理学,和剂量减少因子(DRF)。
    TPP-PCNLs表现出良好的稳定性和生物相容性。体外实验表明,TPP-PCNLs不仅可以很好地靶向线粒体,而且可以调节整个细胞中的活性氧(ROS)水平。更重要的是,TPP-PCNLs提高了L-02细胞线粒体的完整性和功能,从而间接消除线粒体氧化应激对细胞核DNA的持续损伤。TPP-PCNLs主要针对肝脏,脾,脾和其他髓外造血器官的辐射剂量降低因子为1.30。体内实验表明,TPP-PCNLs能有效提高小鼠的成活率,体重变化,受辐照动物的造血功能。Westernblot实验已证实TPP-PCNLs通过调节线粒体凋亡途径在辐射保护中发挥作用。
    TPP-PCNLs通过靶向髓外造血器官-肝细胞和线粒体以持续清除ROS而发挥放射学保护作用。
    UNASSIGNED: Mitochondrial oxidative stress is an important factor in cell apoptosis. Cerium oxide nanomaterials show great potential for scavenging free radicals and simulating superoxide dismutase (SOD) and catalase (CAT) activities. To solve the problem of poor targeting of cerium oxide nanomaterials, we designed albumin-cerium oxide nanoclusters (TPP-PCNLs) that target the modification of mitochondria with triphenyl phosphate (TPP). TPP-PCNLs are expected to simulate the activity of superoxide dismutase, continuously remove reactive oxygen species, and play a lasting role in radiation protection.
    UNASSIGNED: First, cerium dioxide nanoclusters (CNLs), polyethylene glycol cerium dioxide nanoclusters (PCNLs), and TPP-PCNLs were characterized in terms of their morphology and size, ultraviolet spectrum, dispersion stability and cellular uptake, and colocalization Subsequently, the anti-radiation effects of TPP-PCNLs were investigated using in vitro and in vivo experiments including cell viability, apoptosis, comet assays, histopathology, and dose reduction factor (DRF).
    UNASSIGNED: TPP-PCNLs exhibited good stability and biocompatibility. In vitro experiments indicated that TPP-PCNLs could not only target mitochondria excellently but also regulate reactive oxygen species (ROS)levels in whole cells. More importantly, TPP-PCNLs improved the integrity and functionality of mitochondria in irradiated L-02 cells, thereby indirectly eliminating the continuous damage to nuclear DNA caused by mitochondrial oxidative stress. TPP-PCNLs are mainly targeted to the liver, spleen, and other extramedullary hematopoietic organs with a radiation dose reduction factor of 1.30. In vivo experiments showed that TPP-PCNLs effectively improved the survival rate, weight change, hematopoietic function of irradiated animals. Western blot experiments have confirmed that TPP-PCNLs play a role in radiation protection by regulating the mitochondrial apoptotic pathway.
    UNASSIGNED: TPP-PCNLs play a radiologically protective role by targeting extramedullary hematopoietic organ-liver cells and mitochondria to continuously clear ROS.
    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
    目的:小肠是最容易受到电离辐射(IR)损伤的器官之一。然而,防止IR诱导的肠损伤的方法是有限的。CBLB502,一种来自沙门氏菌鞭毛蛋白的Toll样受体5(TLR5)激动剂,对各种组织和器官发挥辐射防护作用。然而,CBLB502对抗IR诱导的肠损伤的分子机制尚不清楚.因此,本研究旨在阐明IR诱导小鼠肠损伤的潜在机制以及CBLB502对这种情况的保护作用。
    方法:在不同时间点,在IR之前以不同剂量给予小鼠0.2mg/kgCBLB502,然后是存活率,体重,血象,并对小鼠进行组织病理学分析。
    结果:CBLB502减少IR诱导的肠损伤。RNA-seq分析显示不同剂量和持续时间的IR诱导不同的调控模式。CBLB502主要通过逆转IR诱导基因的表达并调节免疫过程和代谢途径来保护IR后的肠道损伤。
    结论:本研究初步阐述了CBLB502对IR诱导肠损伤的调控机制和潜在的分子保护机制,为鉴定介导IR诱导肠损伤保护的功能基因和分子机制提供了依据。
    OBJECTIVE: The small intestine is one of the organs most vulnerable to ionizing radiation (IR) damage. However, methods to protect against IR-induced intestinal injury are limited. CBLB502, a Toll-like receptor 5 (TLR5) agonist from Salmonella flagellin, exerts radioprotective effects on various tissues and organs. However, the molecular mechanisms by which CBLB502 protects against IR-induced intestinal injury remain unclear. Thus, this study aimed to elucidate the mechanisms underlying IR-induced intestinal injury and the protective effects of CBLB502 against this condition in mice.
    METHODS: Mice were administered 0.2 mg/kg CBLB502 before IR at different doses for different time points, and then the survival rate, body weight, hemogram, and histopathology of the mice were analyzed.
    RESULTS: CBLB502 reduced IR-induced intestinal injury. RNA-seq analysis revealed that different doses and durations of IR induced different regulatory patterns. CBLB502 protected against intestinal injury mainly after IR by reversing the expression of IR-induced genes and regulating immune processes and metabolic pathways.
    CONCLUSIONS: This study preliminarily describes the regulatory mechanism of IR-induced intestinal injury and the potential molecular protective mechanism of CBLB502, providing a basis for identifying the functional genes and molecular mechanisms that mediate protection against IR-induced injury.
    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:放射治疗是肿瘤的主要局部治疗方法,然而,它可能会导致并发症,如辐射诱发的心脏病(RIHD)。目前,没有预防RIHD的标准化方法。据报道,右美托咪定(Dex)具有心脏保护作用,而其在放射性心肌损伤中的作用尚不清楚。在目前的研究中,我们旨在评估右美托咪定对X线放射治疗小鼠的辐射防护作用.
    方法:18只雄性小鼠随机分为3组:对照组,16Gy,和16Gy+Dex。16Gy组接受单剂量16GyX射线辐射。16Gy+Dex组接受右美托咪定预处理(30µg/kg,腹膜内注射)X射线照射前30分钟。对照组用生理盐水治疗,不接受X线照射。X线照射16周后收集心肌组织。苏木精-伊红染色用于组织病理学检查。进行末端脱氧核苷酸转移酶dUTP缺口末端标记染色以评估凋亡细胞的状态。免疫组化染色检测CD34分子和血管性血友病因子的表达。此外,采用westernblot检测凋亡相关蛋白(BCL2凋亡调节因子和BCL2相关X)以及自噬相关蛋白(微管相关蛋白1轻链3、beclin1和螯合体1)。
    结果:研究结果表明,16GyX射线辐射导致心肌组织发生明显变化,心肌细胞凋亡增加,并激活自噬。右美托咪定预处理通过抑制细胞凋亡和自噬对16GyX射线辐射诱导的小鼠心肌损伤具有保护作用。
    结论:总之,我们的研究证实了右美托咪定减轻16GyX射线辐射诱导的心肌细胞凋亡和自噬的辐射防护作用。
    BACKGROUND: Radiotherapy is a primary local treatment for tumors, yet it may lead to complications such as radiation-induced heart disease (RIHD). Currently, there is no standardized approach for preventing RIHD. Dexmedetomidine (Dex) is reported to have cardio-protection effects, while its role in radiation-induced myocardial injury is unknown. In the current study, we aimed to evaluate the radioprotective effect of dexmedetomidine in X-ray radiation-treated mice.
    METHODS: 18 male mice were randomized into 3 groups: control, 16 Gy, and 16 Gy + Dex. The 16 Gy group received a single dose of 16 Gy X-ray radiation. The 16 Gy + Dex group was pretreated with dexmedetomidine (30 µg/kg, intraperitoneal injection) 30 min before X-ray radiation. The control group was treated with saline and did not receive X-ray radiation. Myocardial tissues were collected 16 weeks after X-ray radiation. Hematoxylin-eosin staining was performed for histopathological examination. Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was performed to assess the state of apoptotic cells. Immunohistochemistry staining was performed to examine the expression of CD34 molecule and von Willebrand factor. Besides, western blot assay was employed for the detection of apoptosis-related proteins (BCL2 apoptosis regulator and BCL2-associated X) as well as autophagy-related proteins (microtubule-associated protein 1 light chain 3, beclin 1, and sequestosome 1).
    RESULTS: The findings demonstrated that 16 Gy X-ray radiation resulted in significant changes in myocardial tissues, increased myocardial apoptosis, and activated autophagy. Pretreatment with dexmedetomidine significantly protects mice against 16 Gy X-ray radiation-induced myocardial injury by inhibiting apoptosis and autophagy.
    CONCLUSIONS: In summary, our study confirmed the radioprotective effect of dexmedetomidine in mitigating cardiomyocyte apoptosis and autophagy induced by 16 Gy X-ray radiation.
    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
    迄今为止,很少有FDA批准的医学对策可用于解决造血急性放射综合征(H-ARS).在这项研究中,我们介绍了我们的最新研究结果,重点是评估一种新型的辐射缓解剂,称为缓解氨基酸混合物(MAAM)。MAAM由五种氨基酸组成,作为最近报道的用于缓解胃肠道(GI)-ARS的基于氨基酸的口服补液溶液。CD2F1雄性和雌性小鼠暴露于9.0或9.5Gy的60Co-γ全身照射(TBI)。辐照后,小鼠口服MAAM或生理盐水载体对照,每天一次,持续14天,在TBI后24小时开始。照射后30天监测小鼠存活和体重变化。全血细胞计数(CBC),骨髓(BM)干细胞和祖细胞存活(克隆形成),使用来自第30天存活小鼠的样品分析血清细胞因子抗体阵列。我们的数据显示,MAAM治疗显着提高了受照射的雄性CD2F1小鼠的存活率,在9.0GyTBI后,存活率从载体对照组的25%增加到MAAM治疗组的60%(p<0.05)。在9.0GyTBI后,BM集落的数目从41.8±6.4/104细胞(在媒介物组中)显著增加至78.5±17.0/104细胞(在MAAM组中)。此外,MAAM治疗导致6种细胞因子/蛋白质[分化簇40(CD40),白细胞介素(IL)-17A,C-X-C基序趋化因子10(CXCL10/CRG-2),皮肤T细胞吸引趋化因子(CTACK),巨噬细胞炎性蛋白(MIP)-3β,和IL-1β]以及其他五种细胞因子/蛋白质[IL-3Rβ,IL-5,瘦素,9.0GyTBI后,小鼠血清中的IL-6和干细胞因子(SCF)]与媒介物组相比。然而,在受辐照的CD2F1雌性小鼠中未观察到MAAM的类似缓解作用。与经照射的雄性小鼠相比,经照射的雌性小鼠中的血清细胞因子谱是不同的。总之,我们的数据表明,辐射暴露后缓解性氨基酸联合治疗的有益效果可能取决于性别.
    To date, few FDA-approved medical countermeasures are available for addressing hematopoietic acute radiation syndrome (H-ARS). In this study, we present our latest research findings focusing on the evaluation of a novel radiation mitigator known as the mitigating amino acid mixture (MAAM). MAAM is composed of five amino acids as the recently reported amino acid-based oral rehydration solution for mitigating gastrointestinal (GI)-ARS. CD2F1 male and female mice were exposed to 60Co-γ total body irradiation (TBI) at 9.0 or 9.5 Gy. Following irradiation, mice were orally administered with MAAM or a saline vehicle control once daily for a duration of 14 days, commencing 24 h after TBI. Mouse survival and body weight change were monitored for 30 days after irradiation. Complete blood counts (CBCs), bone marrow (BM) stem and progenitor cell survival (clonogenicity), and a serum cytokine antibody array were analyzed using samples from day 30 surviving mice. Our data revealed that MAAM treatment significantly enhanced survival rates in irradiated male CD2F1 mice, and the survival rate increased from 25% in the vehicle control group to 60% in the MAAM-treated group (p < 0.05) after 9.0 Gy TBI. The number of BM colonies significantly increased from 41.8 ± 6.4 /104 cells (in the vehicle group) to 78.5 ± 17.0 /104 cells (in the MAAM group) following 9.0 Gy TBI. Furthermore, MAAM treatment led to a decrease in the levels of six cytokines/proteins [cluster of differentiation 40 (CD40), interleukin (IL)-17A, C-X-C motif chemokine 10 (CXCL10/CRG-2), cutaneous T cell-attracting chemokine (CTACK), macrophage inflammatory protein (MIP)-3β, and IL-1β] and an increase in the levels of five other cytokines/proteins [IL-3Rβ, IL-5, leptin, IL-6, and stem cell factor (SCF)] in mouse serum compared to the vehicle group after 9.0 Gy TBI. However, similar alleviating effects of MAAM were not observed in the irradiated CD2F1 female mice. The serum cytokine profile in the irradiated female mice was different compared to the irradiated male mice. In summary, our data suggest that the beneficial effects of the mitigative amino acid combination treatment after radiation exposure may depend on sex.
    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
    在高水平辐射暴露的患者中,胃肠道损伤是导致死亡的主要原因。尽管胃肠道损伤严重,没有具体的治疗选择。牛磺熊去氧胆酸(TUDCA)是熊去氧胆酸的缀合形式,其抑制内质网(ER)应激并调节各种细胞信号传导途径。我们研究了TUDCA前用药在减轻肠道损伤和提高C57BL/6小鼠的存活中的作用,这些小鼠施用了致死剂量(15Gy)的局灶性腹部照射。在辐射暴露前1小时对小鼠施用TUDCA,照射后12小时空肠隐窝的凋亡减少。在稍后的时间点(3.5天),辐照小鼠表现出肠道形态学变化,通过组织学检查检测到。辐射暴露后,TUDCA降低了炎性细胞因子水平,并减弱了血清瓜氨酸水平的降低。虽然辐射引起的内质网应激,TUDCA预处理降低了辐照肠细胞中的ER应激。TUDCA的作用表明肿瘤细胞中癌症的放射治疗的可能性。TUDCA不影响肠上皮细胞的增殖和凋亡。TUDCA降低了CT26转移性结肠癌细胞系的侵袭能力。TUDCA治疗后侵袭性降低与基质金属蛋白酶(MMP)-7和MMP-13表达降低有关,在侵袭和转移中起重要作用。这项研究显示了TUDCA在防止辐射诱导的肠道损伤和抑制肿瘤细胞迁移方面的潜在作用,而没有任何辐射和放射治疗作用。
    In patients with high-level radiation exposure, gastrointestinal injury is the main cause of death. Despite the severity of damage to the gastrointestinal tract, no specific therapeutic option is available. Tauroursodeoxycholic acid (TUDCA) is a conjugated form of ursodeoxycholic acid that suppresses endoplasmic reticulum (ER) stress and regulates various cell-signaling pathways. We investigated the effect of TUDCA premedication in alleviating intestinal damage and enhancing the survival of C57BL/6 mice administered a lethal dose (15Gy) of focal abdominal irradiation. TUDCA was administered to mice 1 h before radiation exposure, and reduced apoptosis of the jejunal crypts 12 h after irradiation. At later timepoint (3.5 days), irradiated mice manifested intestinal morphological changes that were detected via histological examination. TUDCA decreased the inflammatory cytokine levels and attenuated the decrease in serum citrulline levels after radiation exposure. Although radiation induced ER stress, TUDCA pretreatment decreased ER stress in the irradiated intestinal cells. The effect of TUDCA indicates the possibility of radiation therapy for cancer in tumor cells. TUDCA did not affect cell proliferation and apoptosis in the intestinal epithelium. TUDCA decreased the invasive ability of the CT26 metastatic colon cancer cell line. Reduced invasion after TUDCA treatment was associated with decreased matrix metalloproteinase (MMP)-7 and MMP-13 expression, which play important roles in invasion and metastasis. This study shows a potential role of TUDCA in protecting against radiation-induced intestinal damage and inhibiting tumor cell migration without any radiation and radiation therapy effect.
    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
    本研究遵循PRISMA报告指南来呈现结果。对PubMed等电子数据库进行了全面搜索,Scopus,WebofSciences,科学直接。最初,检索到413篇文章。删除重复项并应用特定的纳入和排除标准后,本系统综述最终纳入10篇文章。
    综述的研究表明,硒纳米颗粒具有抗炎和抗氧化特性。它们有效地保护了肾脏,肝脏,和睾丸受损。此外,有证据表明所检查的器官具有有效的放射防护作用,没有明显的副作用。
    本系统综述强调了使用硒纳米颗粒防止电离辐射的负面影响的潜在优势。重要的是,这些保护作用是在没有引起明显副作用的情况下实现的。这些发现表明硒纳米颗粒作为辐射防护剂的潜在作用,提供可能的治疗应用,以减少医学影像和放射治疗程序中与电离辐射暴露相关的风险。
    UNASSIGNED: This study followed the PRISMA reporting guidelines to present the results. A comprehensive search was performed on electronic databases such as PubMed, Scopus, Web of Sciences, and Science Direct. Initially, 413 articles were retrieved. After removing duplicates and applying specific inclusion and exclusion criteria, 10 articles were finally included in this systematic review.
    UNASSIGNED: The reviewed studies showed that selenium nanoparticles had anti-inflammatory and antioxidant properties. They effectively protected the kidneys, liver, and testicles from damage. Furthermore, there was evidence of efficient radioprotection for the organs examined without significant side effects.
    UNASSIGNED: This systematic review emphasizes the potential advantages of using selenium nanoparticles to prevent the negative effects of ionizing radiation. Importantly, these protective effects were achieved without causing noticeable side effects. These findings suggest the potential role of selenium nanoparticles as radioprotective agents, offering possible therapeutic applications to reduce the risks related to ionizing radiation exposure in medical imaging and radiotherapy procedures.
    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
    暴露于高,轻微致死剂量或更高的电离辐射,有意或意外,导致各种器官受伤。目前,只有有限数量的安全和有效的辐射对策由美国食品和药物管理局批准的此类伤害。这些批准的药物仅对急性放射综合征的造血成分有效,并且必须仅在暴露事件后才能施用:目前,没有FDA批准的药物可以预防性使用。营养食品,γ-生育三烯酚(GT3)已被发现是这种暴露相关伤害的有前途的辐射防护剂,特别是那些造血性质的,当在啮齿动物或非人类灵长类动物中测试时。我们调查了由非致死水平(4.0Gy)引起的特定器官系统/组织中GT3的损伤性质和可能的保护作用,以及潜在的致命水平(5.8Gy)的电离辐射,作为全身或部分身体暴露递送。结果表明,最严重的,剂量依赖性损伤发生在具有强大自我更新能力的器官系统内(例如,淋巴造血和胃肠道系统),而在其他组织中(例如,肝脏,肾,肺)具有较少的自我更新能力,注意到的病理倾向于不那么明显,并且不太依赖于暴露剂量水平或所应用的暴露方案。预防性使用测试营养品,GT3,似乎限制了血液形成组织内辐射相关病理的程度,在某种程度上,在胃肠道的小肠内。没有明显的区别,全球身体保护模式与代理人的使用有关,尽管暗示了可能的辐射防护益处,不仅是通过减少特定器官系统内的明显损伤,但也通过注意到在选择GT3治疗的动物中缺乏早期的垂死性。
    Exposure to high, marginally lethal doses or higher of ionizing radiation, either intentional or accidental, results in injury to various organs. Currently, there is only a limited number of safe and effective radiation countermeasures approved by US Food and Drug Administration for such injuries. These approved agents are effective for only the hematopoietic component of the acute radiation syndrome and must be administered only after the exposure event: currently, there is no FDA-approved agent that can be used prophylactically. The nutraceutical, gamma-tocotrienol (GT3) has been found to be a promising radioprotector of such exposure-related injuries, especially those of a hematopoietic nature, when tested in either rodents or nonhuman primates. We investigated the nature of injuries and the possible protective effects of GT3 within select organ systems/tissues caused by both non-lethal level (4.0 Gy), as well as potentially lethal level (5.8 Gy) of ionizing radiation, delivered as total-body or partial-body exposure. Results indicated that the most severe, dose-dependent injuries occurred within those organ systems with strong self-renewing capacities (e.g., the lymphohematopoietic and gastrointestinal systems), while in other tissues (e.g., liver, kidney, lung) endowed with less self-renewal, the pathologies noted tended to be less pronounced and less dependent on the level of exposure dose or on the applied exposure regimen. The prophylactic use of the test nutraceutical, GT3, appeared to limit the extent of irradiation-associated pathology within blood forming tissues and, to some extent, within the small intestine of the gastrointestinal tract. No distinct, global pattern of bodily protection was noted with the agent\'s use, although a hint of a possible radioprotective benefit was suggested not only by a lessening of apparent injury within select organ systems, but also by way of noting the lack of early onset of moribundity within select GT3-treated animals.
    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
    辐射对正常组织造成损害,导致氧化应激增加,炎症,和纤维化,强调需要对健康组织进行选择性辐射防护,而不会阻碍癌症的放射治疗效果。这项研究表明,脂联素,脂肪细胞分泌的脂肪因子,保护正常组织免受体外和体内的辐射损伤。具体来说,脂联素(APN)可降低辐照小鼠的慢性氧化应激和纤维化。重要的是,APN也没有保护前列腺癌细胞免受辐射。脂肪组织是循环内源性脂联素的主要来源。然而,这项研究表明,脂肪组织对辐射暴露敏感,表现出形态变化和持续的氧化损伤。此外,辐射导致暴露于盆腔照射的小鼠和人类前列腺癌患者的脂肪组织中血液APN水平显着慢性降低。在前列腺癌患者中,APN水平与肠毒性和与放疗相关的总体毒性呈负相关。因此,保护,或调节APN信号可以改善接受放疗的前列腺癌患者的预后.
    Radiation causes damage to normal tissues that leads to increased oxidative stress, inflammation, and fibrosis, highlighting the need for the selective radioprotection of healthy tissues without hindering radiotherapy effectiveness in cancer. This study shows that adiponectin, an adipokine secreted by adipocytes, protects normal tissues from radiation damage invitro and invivo. Specifically, adiponectin (APN) reduces chronic oxidative stress and fibrosis in irradiated mice. Importantly, APN also conferred no protection from radiation to prostate cancer cells. Adipose tissue is the primary source of circulating endogenous adiponectin. However, this study shows that adipose tissue is sensitive to radiation exposure exhibiting morphological changes and persistent oxidative damage. In addition, radiation results in a significant and chronic reduction in blood APN levels from adipose tissue in mice and human prostate cancer patients exposed to pelvic irradiation. APN levels negatively correlated with bowel toxicity and overall toxicities associated with radiotherapy in prostate cancer patients. Thus, protecting, or modulating APN signaling may improve outcomes for prostate cancer patients undergoing radiotherapy.
    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
    饮食来源的外泌体样纳米囊泡是一类与哺乳动物外泌体具有相似结构和功能的天然活性物质。泌阳花蘑菇及其活性提取物已被发现具有辐射防护作用,并深入探索其新型活性物质,本研究研究了泌阳花蘑菇来源的外泌体样纳米囊泡(BFMELNs)的辐射防护作用。结果表明,这些带表面负电荷的囊泡具有理想的尺寸,并且对温度和胃肠道消化等环境变化具有良好的稳定性。此外,BFMELN可以通过网格蛋白和动力蛋白介导的细胞吞噬作用被HL-7702细胞和Caco-2细胞有效吸收。强调,具有外泌体样形态的BFMELN含有RNA,蛋白质,脂质,多酚和黄酮在体外发挥良好的抗氧化和辐射防护作用。同时,BFMELNs还通过恢复外周血指标表现出良好的辐射防护作用,减轻对器官的损害,调节小鼠的氧化还原状态。总的来说,BFMELNs有望作为新型和天然的辐射防护纳米剂,用于预防IR诱导的氧化应激损伤。
    Diet-derived exosome-like nanovesicles are a class of natural active substances that have similar structures and functions to mammalian exosomes. Biyang floral mushrooms and their active extracts have been found to possess radioprotective effects and to deeply explore their novel active substances, the radioprotective effects of Biyang floral mushroom-derived exosome-like nanovesicles (BFMELNs) were investigated in this study. Results showed that these surface-negatively charged vesicles possessed an ideal size and good stability against environmental changes such as temperature and gastrointestinal digestion. Furthermore, BFMELNs could effectively be taken up by HL-7702 cells and Caco-2 cells through cellular phagocytosis mediated by clathrin and dynein. Emphatically, BFMELNs with an exosome-like morphology contained RNA, proteins, lipids, polyphenols and flavonoids to exert good antioxidant and radioprotective effects in vitro. Meanwhile, BFMELNs also exhibited good radioprotective effects by restoring peripheral blood indexes, mitigating damage to organs, and regulating the redox state in mice. Collectively, BFMELNs showed promise as novel and natural radioprotective nano-agents for preventing IR-induced oxidative stress damage.
    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
    放射性肠损伤是腹部或盆腔实体瘤放疗过程中最常见的副作用,严重影响患者的生活质量,甚至导致预后不良。直到现在,口服用于肠道辐射防护的常规制剂仍然具有挑战性,没有可用于减轻小肠辐射毒性的优选方法.我们先前的研究表明,源自益生菌孢子涂层的纳米材料具有优异的抗炎作用,甚至可以预防癌症的进展。这项工作的目的是确定孢子涂层(表示为孢子鬼,SGs)来自三种临床批准的益生菌(B.凝块,枯草芽孢杆菌和地衣芽孢杆菌)。三种SGs均表现出优异的活性氧(ROS)清除能力和优异的抗炎作用。此外,这些SGs可以通过抑制有害细菌和增加乳酸菌的丰度来逆转肠道菌群的平衡。因此,SGs的给药通过缓解腹泻显著减少放射性肠损伤,预防X射线诱导的小肠上皮细胞凋亡和促进屏障完整性恢复的预防性研究。值得注意的是,SGs显着改善接受全腹部X射线辐射的小鼠的体重增加和存活。这项工作可能提供有前途的辐射防护剂,以有效减轻辐射诱发的胃肠道综合征,并促进新的肠道好感的发展。
    Radiation-induced intestinal injury is the most common side effect during radiotherapy of abdominal or pelvic solid tumors, significantly impacting patients\' quality of life and even resulting in poor prognosis. Until now, oral application of conventional formulations for intestinal radioprotection remains challenging with no preferred method available to mitigate radiation toxicity in small intestine. Our previous study revealed that nanomaterials derived from spore coat of probiotics exhibit superior anti-inflammatory effect and even prevent the progression of cancer. The aim of this work is to determine the radioprotective effect of spore coat (denoted as spore ghosts, SGs) from three clinically approved probiotics (B.coagulans, B.subtilis and B.licheniformis). All the three SGs exhibit outstanding reactive oxygen species (ROS) scavenging ability and excellent anti-inflammatory effect. Moreover, these SGs can reverse the balance of intestinal flora by inhibiting harmful bacteria and increasing the abundance of Lactobacillus. Consequently, administration of SGs significantly reduce radiation-induced intestinal injury by alleviating diarrhea, preventing X-ray induced apoptosis of small intestinal epithelial cells and promoting restoration of barrier integrity in a prophylactic study. Notably, SGs markedly improve weight gain and survival of mice received total abdominal X-ray radiation. This work may provide promising radioprotectants for efficiently attenuating radiation-induced gastrointestinal syndrome and promote the development of new intestinal predilection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号