Hyperoxia

高氧
  • 文章类型: English Abstract
    OBJECTIVE: To investigate the effect of reactive oxygen species (ROS)/silent information regulator 1 (SIRT1) on hyperoxia-induced mitochondrial injury in BEAS-2B cells.
    METHODS: The experiment was divided into three parts. In the first part, cells were divided into H0, H6, H12, H24, and H48 groups. In the second part, cells were divided into control group, H48 group, H48 hyperoxia+SIRT1 inhibitor group (H48+EX 527 group), and H48 hyperoxia+SIRT1 agonist group (H48+SRT1720 group). In the third part, cells were divided into control group, 48-hour hyperoxia+N-acetylcysteine group (H48+NAC group), and H48 group. The ROS kit was used to measure the level of ROS. Western blot and immunofluorescent staining were used to measure the expression levels of SIRT1 and mitochondria-related proteins. Transmission electron microscopy was used to observe the morphology of mitochondria.
    RESULTS: Compared with the H0 group, the H6, H12, H24, and H48 groups had a significantly increased fluorescence intensity of ROS (P<0.05), the H48 group had significant reductions in the expression levels of SIRT1 protein and mitochondria-related proteins (P<0.05), and the H24 and H48 groups had a significant reduction in the fluorescence intensity of mitochondria-related proteins (P<0.05). Compared with the H48 group, the H48+SRT1720 group had significant increases in the expression levels of mitochondria-related proteins and the mitochondrial aspect ratio (P<0.05), and the H48+EX 527 group had a significant reduction in the mitochondrial area (P<0.05). Compared with the H48 group, the H48+NAC group had a significantly decreased fluorescence intensity of ROS (P<0.05) and significantly increased levels of SIRT1 protein, mitochondria-related proteins, mitochondrial area, and mitochondrial aspect ratio (P<0.05).
    CONCLUSIONS: The ROS/SIRT1 axis is involved in hyperoxia-induced mitochondrial injury in BEAS-2B cells.
    目的: 探讨活性氧簇(reactive oxygen species, ROS)/沉默信息调节因子1(silent information regulator 1, SIRT1)对高氧致BEAS-2B细胞线粒体损伤的影响。方法: 实验分为三部分:(1)细胞分为高氧0 h(H0)组、H6组、H12组、H24组、H48组。(2)细胞分为对照组、H48组、高氧48 h+SIRT1抑制剂(H48+EX 527)组和高氧48 h+SIRT1激动剂(H48+SRT1720)组。(3)细胞分为对照组、高氧48 h+乙酰半胱氨酸(H48+NAC)组和H48组。采用活性氧试剂盒检测ROS水平,Western blot法检测SIRT1和线粒体相关蛋白表达水平,免疫荧光染色法检测线粒体相关蛋白表达,透射电镜检测线粒体形态。结果: (1)与H0组相比,H6组、H12组、H24组和H48组ROS荧光强度增加(P<0.05),H48组SIRT1和线粒体相关蛋白表达水平降低(P<0.05),H24组和H48组线粒体相关蛋白荧光强度降低(P<0.05)。(2)与H48组相比,H48+SRT1720组线粒体相关蛋白表达水平升高,线粒体平均长宽比增加(P<0.05);H48+EX 527组线粒体平均面积减少(P<0.05)。(3)与H48组相比,H48+NAC组ROS荧光强度降低,SIRT1和线粒体相关蛋白表达水平升高,线粒体平均面积和平均长宽比增加(P<0.05)。结论: ROS/SIRT1轴参与了高氧诱导的BEAS-2B细胞线粒体损伤。.
    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
    MecROX是UK-ROX试验的一项机械性子研究,旨在评估重症监护中侵入性通气成人的保守氧疗方法的临床和成本效益。这是基于过量氧气有害的科学原理。肺泡表面活性剂缺乏引起的上皮细胞损伤是高氧急性肺损伤的特征。此外,高氧血症(过量的血氧水平)可能会加剧全身氧化应激,导致细胞死亡,自噬,线粒体功能障碍,生物能量衰竭和多器官衰竭导致不良的临床结果。然而,缺乏体内人体模型评估机械通气患者氧诱导器官损伤的机制.
    MecROX机制子研究的目的是评估肺表面活性物质组成和全身氧化还原状态,为UK-ROX试验结果提供机制和互补的科学依据。目标是量化体内表面活性剂组成,合成,以及具有氧化应激和全身氧化还原不平衡标记物的代谢(如“反应性物种相互作用组”的变化所证明的),以区分保守和通常的氧目标组。
    在随机进入UK-ROX试验后,将在两个试验地点招募100名成年参与者(保守组50名,常规护理组50名)。在输注3mg/kg甲基-D9-胆碱氯化物后0、48和72小时采集血液和气管内样品。这是非放射性的,胆碱(维生素)的稳定同位素,已被广泛用于研究人体表面活性剂磷脂动力学。这项研究将机械评估体内表面活性剂的合成和分解(通过水解和氧化),使用一系列分析平台从连续的血浆和支气管样品中获得氧化应激和氧化还原不平衡。我们将根据给药的氧气量比较保守和常规氧合组。试用注册:ISRCTNISRCTN61929838,27/03/2023https://doi.org/10.1186/ISRCTN61929838。
    UNASSIGNED: MecROX is a mechanistic sub-study of the UK-ROX trial which was designed to evaluate the clinical and cost-effectiveness of a conservative approach to oxygen therapy for invasively ventilated adults in intensive care. This is based on the scientific rationale that excess oxygen is harmful. Epithelial cell damage with alveolar surfactant deficiency is characteristic of hyperoxic acute lung injury. Additionally, hyperoxaemia (excess blood oxygen levels) may exacerbate whole-body oxidative stress leading to cell death, autophagy, mitochondrial dysfunction, bioenergetic failure and multi-organ failure resulting in poor clinical outcomes. However, there is a lack of in-vivo human models evaluating the mechanisms that underpin oxygen-induced organ damage in mechanically ventilated patients.
    UNASSIGNED: The aim of the MecROX mechanistic sub-study is to assess lung surfactant composition and global systemic redox status to provide a mechanistic and complementary scientific rationale to the UK-ROX trial findings. The objectives are to quantify in-vivo surfactant composition, synthesis, and metabolism with markers of oxidative stress and systemic redox disequilibrium (as evidenced by alterations in the \'reactive species interactome\') to differentiate between groups of conservative and usual oxygen targets.
    UNASSIGNED: After randomisation into the UK-ROX trial, 100 adult participants (50 in the conservative and 50 in usual care group) will be recruited at two trial sites. Blood and endotracheal samples will be taken at 0, 48 and 72 hours following an infusion of 3 mg/kg methyl-D 9-choline chloride. This is a non-radioactive, stable isotope of choline (vitamin), which has been extensively used to study surfactant phospholipid kinetics in humans. This study will mechanistically evaluate the in-vivo surfactant synthesis and breakdown (by hydrolysis and oxidation), oxidative stress and redox disequilibrium from sequential plasma and bronchial samples using an array of analytical platforms. We will compare conservative and usual oxygenation groups according to the amount of oxygen administered. Trial registration: ISRCTNISRCTN61929838, 27/03/2023 https://doi.org/10.1186/ISRCTN61929838.
    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
    支气管肺发育不良(BPD)是困扰新生儿的最重要并发症之一,可导致各种后遗症。HIF-1α/VEGF信号通路促进新生血管生成的能力在新生儿肺发育中具有重要作用。
    新生大鼠暴露于85%的氧气中。通过免疫荧光和WesternBlot分析评估高氧暴露对大鼠肺组织多形性腺瘤基因样2(PLAGL2)和HIF-1α/VEGF通路的影响。在细胞实验中,PLAGL2上调,高氧和PLAGL2对细胞活力的影响使用划痕试验进行评估,CCK-8测定,和EDU染色。通过WesternBlot和RT-PCR确定上调的PLAGL2在HIF-1α/VEGF通路中的作用。通过流式细胞术和活力测定确定凋亡和铁凋亡效应。
    与对照组相比,PLAGL2,HIF-1α的表达水平,VEGF,高氧暴露3、7和14天后肺组织中的SPC均降低。此外,高氧还抑制II型肺泡上皮细胞(AECII)的增殖和运动,并诱导AECII中的细胞凋亡。PLAGL2上调可恢复AECII的增殖和运动,抑制细胞凋亡和铁凋亡,同时HIF-1α/VEGF信号通路也得以恢复。
    我们证实了PLAGL2和HIF-1α/VEGF信号通路在高氧条件下促进BPD的积极作用,并提供了有希望的治疗靶点。
    UNASSIGNED: Bronchopulmonary dysplasia (BPD) is one of the most important complications plaguing neonates and can lead to a variety of sequelae. the ability of the HIF-1α/VEGF signaling pathway to promote angiogenesis has an important role in neonatal lung development.
    UNASSIGNED: Newborn rats were exposed to 85% oxygen. The effects of hyperoxia exposure on Pleomorphic Adenoma Gene like-2 (PLAGL2) and the HIF-1α/VEGF pathway in rats lung tissue were assessed through immunofluorescence and Western Blot analysis. In cell experiments, PLAGL2 was upregulated, and the effects of hyperoxia and PLAGL2 on cell viability were evaluated using scratch assays, CCK-8 assays, and EDU staining. The role of upregulated PLAGL2 in the HIF-1α/VEGF pathway was determined by Western Blot and RT-PCR. Apoptosis and ferroptosis effects were determined through flow cytometry and viability assays.
    UNASSIGNED: Compared with the control group, the expression levels of PLAGL2, HIF-1α, VEGF, and SPC in lung tissues after 3, 7, and 14 days of hyperoxia exposure were all decreased. Furthermore, hyperoxia also inhibited the proliferation and motility of type II alveolar epithelial cells (AECII) and induced apoptosis in AECII. Upregulation of PLAGL2 restored the proliferation and motility of AECII and suppressed cell apoptosis and ferroptosis, while the HIF-1α/VEGF signaling pathway was also revived.
    UNASSIGNED: We confirmed the positive role of PLAGL2 and HIF-1α/VEGF signaling pathway in promoting BPD in hyperoxia conditions, and provided a promising therapeutic targets.
    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
    肺巨噬细胞在健康和疾病中起着至关重要的作用。性二态性显着影响组织驻留巨噬细胞的表型和功能。导致支气管肺发育不良(BPD)性二态结局的主要机制尚未确定。我们检验了生物学性别在肺泡巨噬细胞的转录状态中起关键作用的假设。使用新生儿小鼠高氧诱导的肺损伤作为人BPD的相关模型。测量了新生儿高氧暴露(95%FiO2,PND1-5:囊状阶段)在损伤后和正常氧恢复期间对肺骨髓细胞的影响。对雄性和雌性新生鼠肺暴露的室内空气和高氧的肺泡巨噬细胞(AM)进行大量RNA测序。损伤后暴露于高氧的肺中的AMs明显耗尽,随后两性都恢复了。肺泡巨噬细胞的转录组受新生儿高氧暴露和性别作为生物学变量的影响。与DNA损伤和干扰素信号相关的通路在雌性AMs中呈正富集;与葡萄糖和碳水化合物代谢相关的代谢通路在雄性AMs中呈正富集。而氧化磷酸化是负富集的。这些途径与来自插管的男性和女性人类早产新生儿的单核细胞和气道巨噬细胞共享。
    The lung macrophages play a crucial role in health and disease. Sexual dimorphism significantly impacts the phenotype and function of tissue-resident macrophages. The primary mechanisms responsible for sexually dimorphic outcomes in bronchopulmonary dysplasia (BPD) remain unidentified. We tested the hypothesis that biological sex plays a crucial role in the transcriptional state of alveolar macrophages, using neonatal murine hyperoxia-induced lung injury as a relevant model for human BPD. The effects of neonatal hyperoxia exposure (95 % FiO2, PND1-5: saccular stage) on the lung myeloid cells acutely after injury and during normoxic recovery were measured. Alveolar macrophages (AM) from room air- and hyperoxia exposed from male and female neonatal murine lungs were subjected to bulk-RNA Sequencing. AMs are significantly depleted in the hyperoxia-exposed lung acutely after injury, with subsequent recovery in both sexes. The transcriptome of the alveolar macrophages is impacted by neonatal hyperoxia exposure and by sex as a biological variable. Pathways related to DNA damage and interferon-signaling were positively enriched in female AMs. Metabolic pathways related to glucose and carbohydrate metabolism were positively enriched in the male AMs, while oxidative phosphorylation was negatively enriched. These pathways were shared with monocytes and airway macrophages from intubated male and female human premature neonates.
    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
    早产新生儿气道上皮不断暴露于环境应激源。患有肺病的新生儿中的这些应激源之一包括高于周围大气的氧气(O2)张力-称为高氧(>21%O2)。高氧对气道的影响取决于各种因素,包括气道的发育阶段,高氧的程度,以及暴露的持续时间,可变的暴露可能导致独特的表型。虽然对高氧对新生儿肺泡化和气道高反应性的影响进行了广泛的研究,关于高氧对人新生儿气道上皮细胞的短期和长期潜在影响知之甚少。其主要原因是缺乏有效的体外模型来研究人新生儿气道上皮发育和功能。这里,我们描述了一种利用人新生儿气管抽吸物分离和扩增人新生儿气管气道上皮细胞(nTAEC)并在气液界面(ALI)培养物中培养这些细胞的方法。我们证明nTAEC在ALI培养物中形成成熟的极化细胞单层,并经历粘膜纤毛分化。我们还提出了一种使用专门的培养箱在ALI培养物中适度高氧暴露细胞单层的方法。此外,我们描述了一种使用荧光定量测量ALI培养物中高氧暴露后细胞氧化应激的测定法,这证实了中度高氧暴露会诱导细胞氧化应激,但不会引起明显的细胞膜损伤或细胞凋亡。该模型可用于模拟新生儿重症监护病房(NICU)中新生儿气道遇到的临床相关高氧暴露,并用于研究O2对新生儿气道上皮编程的短期和长期影响。使用该模型的研究可用于探索减轻早期气道氧化损伤的方法。这与前早产儿长期气道疾病的发展有关。
    The preterm neonatal airway epithelium is constantly exposed to environmental stressors. One of these stressors in neonates with lung disease includes oxygen (O2) tension higher than the ambient atmosphere - termed hyperoxia (>21% O2). The effect of hyperoxia on the airway depends on various factors, including the developmental stage of the airway, the degree of hyperoxia, and the duration of exposure, with variable exposures potentially leading to unique phenotypes. While there has been extensive research on the effect of hyperoxia on neonatal lung alveolarization and airway hyperreactivity, little is known about the short and long-term underlying effect of hyperoxia on human neonatal airway epithelial cells. A major reason for this is the scarcity of an effective in vitro model to study human neonatal airway epithelial development and function. Here, we describe a method for isolating and expanding human neonatal tracheal airway epithelial cells (nTAECs) utilizing human neonatal tracheal aspirates and culturing these cells in air-liquid interface (ALI) culture. We demonstrate that nTAECs form a mature polarized cell-monolayer in ALI culture and undergo mucociliary differentiation. We also present a method for moderate hyperoxia exposure of the cell monolayer in ALI culture using a specialized incubator. Additionally, we describe an assay to measure cellular oxidative stress following hyperoxia exposure in ALI culture using fluorescent quantification, which confirms that moderate hyperoxia exposure induces cellular oxidative stress but does not cause significant cell membrane damage or apoptosis. This model can potentially be used to simulate clinically relevant hyperoxia exposure encountered by neonatal airways in the Neonatal Intensive Care Unit (NICU) and used to study the short and long-lasting effects of O2 on neonatal airway epithelial programming. Studies using this model could be utilized to explore ways to mitigate early-life oxidative injury to developing airways, which is implicated in the development of long-term airway diseases in former premature infants.
    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
    补充氧气(高氧)可改善低氧运动期间的身体表现。基于人体运动员血液样本中代谢组和铁稳态的分析,我们表明,在恢复期高氧会干扰低氧运动后的代谢改变。这可能会损害对运动和/或缺氧的有益适应性,并突出缺氧中补充氧气的风险。
    Supplemental oxygen (hyperoxia) improves physical performance during hypoxic exercise. Based on the analysis of metabolome and iron homeostasis from human athlete blood samples, we show that hyperoxia during recovery periods interferes with metabolic alterations following hypoxic exercise. This may impair beneficial adaptations to exercise and/or hypoxia and highlights risks of oxygen supplementation in hypoxia.
    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
    器官发生在低氧水平(4%)下的子宫中。早产使未成熟的新生儿暴露在高氧环境中,可以诱导大量产生活性氧并可能影响器官发育,导致坏死性小肠结肠炎等疾病。β3-肾上腺素受体(β3-AR)具有氧依赖性调节机制,它的活化发挥了抗氧化作用。为了检验β3-AR可以保护出生后回肠发育免受高氧水平的负面影响的假设,Sprague-Dawley幼鼠出生后的前2周在常氧(21%)或高氧(85%)下饲养,并使用或不使用选择性β3-AR激动剂BRL37344治疗,1、3或6mg/kg。高氧改变回肠粘膜形态,导致细胞脂质氧化副产物增加,β3-AR阳性驻留细胞的存在减少,连接蛋白表达降低,破坏了刷子边界,粘蛋白过度生产,血管化受损.用3mg/kg的BRL37344治疗可以防止这些改变,虽然不完全,而较低的1毫克/千克剂量是无效的,较高的6mg/kg剂量是有毒的。我们的发现表明β3-AR激动作为一种新的治疗方法来抵消高氧诱导的回肠改变的潜力,更一般地说,与超生理氧暴露相关的早产儿疾病。
    Organogenesis occurs in the uterus under low oxygen levels (4%). Preterm birth exposes immature newborns to a hyperoxic environment, which can induce a massive production of reactive oxygen species and potentially affect organ development, leading to diseases such as necrotizing enterocolitis. The β3-adrenoreceptor (β3-AR) has an oxygen-dependent regulatory mechanism, and its activation exerts an antioxidant effect. To test the hypothesis that β3-AR could protect postnatal ileal development from the negative impact of high oxygen levels, Sprague-Dawley rat pups were raised under normoxia (21%) or hyperoxia (85%) for the first 2 weeks after birth and treated or not with BRL37344, a selective β3-AR agonist, at 1, 3, or 6 mg/kg. Hyperoxia alters ileal mucosal morphology, leading to increased cell lipid oxidation byproducts, reduced presence of β3-AR-positive resident cells, decreased junctional protein expression, disrupted brush border, mucin over-production, and impaired vascularization. Treatment with 3 mg/kg of BRL37344 prevented these alterations, although not completely, while the lower 1 mg/kg dose was ineffective, and the higher 6 mg/kg dose was toxic. Our findings indicate the potential of β3-AR agonism as a new therapeutic approach to counteract the hyperoxia-induced ileal alterations and, more generally, the disorders of prematurity related to supra-physiologic oxygen exposure.
    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:支气管肺发育不良(BPD)是一种主要影响早产儿的慢性肺部疾病。氧疗,BPD的常用治疗方法,通常会导致高氧引起的肺损伤,特别是靶向肺泡上皮细胞(AECs)。至关重要的是,破坏的肺上皮-成纤维细胞相互作用显著有助于BPD的发病机制。先前关于白细胞介素-11(IL-11)在肺部疾病中的研究产生了矛盾的结果。最近的研究,然而,强调IL-11是纤维化的关键调节因子,基质炎症,和上皮功能障碍。尽管如此,IL-11在BPD中的具体作用仍未得到充分研究。我们对正常和高氧暴露的鼠肺组织的转录组分析显示IL-11RNA的表达增加。本研究旨在探讨IL-11在调节BPD中AECs与成纤维细胞之间的相互作用中的作用。
    方法:通过将C57BL/6J新生小鼠暴露于高氧下,在体内对BPD进行建模。用苏木精-伊红染色评估肺组织的组织病理学变化,而肺纤维化使用Masson染色和免疫组织化学(IHC)进行评估。探讨IL-11在导致BPD的肺损伤中的作用,通过在暴露于高氧的小鼠中腹膜内施用IL-11RαFc来降低IL-11水平。此外,收集经受95%氧的MLE-12细胞并与小鼠肺成纤维细胞(MPF)共培养以测量α-SMA和胶原I表达水平。使用酶联免疫吸附测定(ELISA)定量上清液中的IL-11水平。
    结果:IHC和Masson染色均显示抑制IL-11的表达减轻了高氧诱导的新生小鼠肺纤维化,同时降低肺组织中纤维化标志物α-SMA和I型胶原的表达。体外分析显示用高氧处理的MLE-12细胞的上清液中IL-11水平显著增加。在MLE-12细胞中沉默IL-11表达降低了与高氧处理的MLE-12细胞的上清液共培养的MPF中的α-SMA和胶原蛋白I浓度。此外,ERK抑制剂降低了与高氧处理的MLE-12细胞的上清液共培养的MPF中的α-SMA和胶原蛋白I水平。临床研究发现,BPD婴儿的气管抽吸物(TA)中IL-11水平升高。
    结论:这项研究表明,高氧诱导肺上皮分泌IL-11。此外,来自肺上皮的IL-11通过ERK信号通路作为肌成纤维细胞分化的关键介质,强调其在BPD治疗中的潜在治疗价值。
    BACKGROUND: Bronchopulmonary dysplasia (BPD) is a chronic lung disorder predominantly affecting preterm infants. Oxygen therapy, a common treatment for BPD, often leads to hyperoxia-induced pulmonary damage, particularly targeting alveolar epithelial cells (AECs). Crucially, disrupted lung epithelium-fibroblast interactions significantly contribute to BPD\'s pathogenesis. Previous studies on interleukin-11 (IL-11) in lung diseases have yielded conflicting results. Recent research, however, highlights IL-11 as a key regulator of fibrosis, stromal inflammation, and epithelial dysfunction. Despite this, the specific role of IL-11 in BPD remains underexplored. Our transcriptome analysis of normal and hyperoxia-exposed murine lung tissues revealed an increased expression of IL-11 RNA. This study aimed to investigate IL-11\'s role in modulating the disrupted interactions between AECs and fibroblasts in BPD.
    METHODS: BPD was modeled in vivo by exposing C57BL/6J neonatal mice to hyperoxia. Histopathological changes in lung tissue were evaluated with hematoxylin-eosin staining, while lung fibrosis was assessed using Masson staining and immunohistochemistry (IHC). To investigate IL-11\'s role in pulmonary injury contributing to BPD, IL-11 levels were reduced through intraperitoneal administration of IL-11RαFc in hyperoxia-exposed mice. Additionally, MLE-12 cells subjected to 95% oxygen were collected and co-cultured with mouse pulmonary fibroblasts (MPFs) to measure α-SMA and Collagen I expression levels. IL-11 levels in the supernatants were quantified using an enzyme-linked immunosorbent assay (ELISA).
    RESULTS: Both IHC and Masson staining revealed that inhibiting IL-11 expression alleviated pulmonary fibrosis in neonatal mice induced by hyperoxia, along with reducing the expression of fibrosis markers α-SMA and collagen I in lung tissue. In vitro analysis showed a significant increase in IL-11 levels in the supernatant of MLE-12 cells treated with hyperoxia. Silencing IL-11 expression in MLE-12 cells reduced α-SMA and collagen I concentrations in MPFs co-cultured with the supernatant of hyperoxia-treated MLE-12 cells. Additionally, ERK inhibitors decreased α-SMA and collagen I levels in MPFs co-cultured with the supernatant of hyperoxia-treated MLE-12 cells. Clinical studies found increased IL-11 levels in tracheal aspirates (TA) of infants with BPD.
    CONCLUSIONS: This research reveals that hyperoxia induces IL-11 secretion in lung epithelium. Additionally, IL-11 derived from lung epithelium emerged as a crucial mediator in myofibroblast differentiation via the ERK signaling pathway, highlighting its potential therapeutic value in BPD 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
    支气管肺发育不良(BPD)是早产儿常见的严重并发症。没有有效的手段控制它。高氧毁伤是BPD的重要机制之一。证实了BPD中存在焦亡。右美托咪定是一种新的,高特异性α2受体激动剂。先前的研究基础发现右美托咪定对BPD具有保护作用。探讨右美托咪定如何通过调节焦亡改善新生小鼠高氧肺损伤。将新生大鼠随机分为4组:正常对照组,高氧损伤组,空气加右美托咪定组,高氧加右美托咪定组。七天后抽取各组大鼠的肺部,并测量肺的湿干重量比。采用苏木精-伊红染色观察大鼠肺损伤情况。此外,核苷酸结合寡聚化结构域样受体热蛋白结构域相关蛋白3(NLRP3)的表达和定位,凋亡相关斑点样蛋白(ASC),使用免疫荧光染色在大鼠的肺中检查了gasderminD(GSDMD)蛋白。NLRP3、ASC、使用实时PCR测定大鼠肺中的caspase-1和白介素18(IL-18)。此外,NLRP3,ASC,caspase-1/裂解的caspase-1,白细胞介素1β(IL-1β),使用Westernblot检测大鼠肺中的IL-18和膜坏死因子α(TNF-α)。透射电镜观察各组肺组织线粒体损伤程度。与高氧损伤组相比,高氧+右美托咪定组新生大鼠肺组织损伤明显改善。此外,焦亡相关蛋白如NLRP3、ASC、cleaved-caspase-1和GSDMD显著降低,肺组织中炎症因子的表达。通过抑制NLRP3/caspase-1/GSDMD途径,右美托咪定可减少新生小鼠高氧肺损伤时炎症因子的激活和释放,并具有保护作用。
    Bronchopulmonary dysplasia (BPD) is a common serious complication of premature babies. No effective means control it. Hyperoxia damage is one of the important mechanisms of BPD. The reaserach confirmed pyroptosis existed in BPD. Dexmedetomidine is a new, high-specific α2 receptor agonist. Previous research foundation found that dexmedetomidine has a protective effect on BPD. To investigate how dexmedetomidine improves hyperoxic lung injury in neonatal mice by regulating pyroptosis. Neonatal rats were randomly divided into four groups: normal control group, hyperoxic injury group, air plus dexmedetomidine group, and hyperoxia plus dexmedetomidine group. After seven days the lungs of rats in each group were extracted, and the wet-to-dry weight ratio of the lung was measured. The lung injury in rats was observed using hematoxylin-eosin staining. Additionally, the expression and localization of nucleotide-binding oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), and gasdermin D (GSDMD) proteins were examined in the lungs of rats using immunofluorescence staining. The mRNA levels of NLRP3, ASC, caspase-1, and interleukin 18 (IL-18) in the lungs of rats were determined using real-time PCR. Moreover, the protein levels of NLRP3, ASC, caspase-1/cleaved caspase-1, interleukin 1beta (IL-1β), IL-18, and tunor necrosis factor alpha (TNF-α) were detected in lungs of rats using Western blot. The extent of mitochondrial damage in lung tissues of each group was observed by transmission electron microscopy. The lung tissue injury of the neonatal rats was significantly improved in the hyperoxia plus dexmedetomidine group compared to the hyperoxic injury group. Furthermore, the expressions of pyroptosis-related proteins such as NLRP3, ASC, cleaved-caspase-1, and GSDMD were significantly decreased, along with the expressions of inflammatory factors in lung tissues. By inhibiting the NLRP3/caspase-1/GSDMD pyroptosis pathway, dexmedetomidine reduces the activation and release of inflammatory factors and provides a protective effect against hyperoxic lung injury in neonatal mice.
    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
    早产儿经常暴露于高氧。然而,关于新生儿高氧暴露及其对早产成人心肾功能障碍的影响的机制基础的数据有限.我们的目的是确定新生儿高氧是否会在成年期引起全身血管僵硬和心肾功能障碍。从出生后的第1天到第14天,新生大鼠被随机分配到室内空气(RA)或高氧(85%O2),然后在RA中恢复,直到生命的1年。动脉僵硬度,心-肾组织形态计量学,和主动脉纤维化,心,并对肾脏进行了评估。还进行了主动脉和肾脏的RNA测序(RNA-seq)。暴露于新生儿高氧的成年大鼠的主动脉和肠系膜动脉硬度增加,如线和压力肌电图所示。他们还有心肌细胞肥大,肾小球肿大,和管状损伤。高氧暴露改变了与主动脉和肾脏纤维化和基质重塑相关的转录组概况。主动脉中TGF-β1水平和纤维化也增加,左心室,还有肾.总之,在1岁大鼠中,新生儿高氧暴露与全身血管和心-肾改变相关.指出了进一步的研究,以确定在新生儿高氧暴露后靶向治疗如何重新编程心肾损伤。
    Premature infants are often exposed to hyperoxia. However, there is limited data regarding the mechanistic underpinnings linking neonatal hyperoxia exposure and its contribution to cardio-renal dysfunction in adults born preterm. Our objective was to determine whether neonatal hyperoxia induces systemic vascular stiffness and cardio-renal dysfunction in adulthood. Newborn rats were randomly assigned to room air (RA) or hyperoxia (85% O2) from postnatal day 1 to 14, then recovered in RA until 1 year of life. Arterial stiffness, cardio-renal histomorphometry, and fibrosis in the aorta, heart, and kidney were assessed. RNA-sequencing (RNA-seq) of the aorta and kidney was also done. Adult rats exposed to neonatal hyperoxia had increased aortic and mesenteric artery stiffness as demonstrated by wire and pressure myography. They also had cardiomyocyte hypertrophy, glomerulomegaly, and tubular injury. Hyperoxia exposure altered the transcriptome profile associated with fibrosis and matrix remodeling in the aorta and kidney. There was also increased TGF-β1 levels and fibrosis in the aorta, left ventricle, and kidney. In conclusion, neonatal hyperoxia exposure was associated with systemic vascular and cardio-renal alterations in 1-year-old rats. Further studies to determine how targeted therapies could reprogram cardio-renal injury after neonatal hyperoxia exposure are indicated.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号