Lung ischemia-reperfusion injury

肺缺血再灌注损伤
  • 文章类型: Journal Article
    原发性移植物功能障碍(PGD)是肺移植(LTx)中由肺缺血/再灌注损伤(I/R)引起的严重急性肺损伤,与移植后发病率和死亡率升高有关。通过NETosis释放过多的中性粒细胞胞外陷阱(NETs),在再灌注过程中浸润的中性粒细胞被确定为肺I/R损伤的关键贡献者。而肺泡巨噬细胞(AM)参与调节中性粒细胞趋化和浸润,它们在肺I/R期间NETosis中的作用仍未充分阐明。细胞外组蛋白构成了NETs的主要结构并能激活AMs。我们证实了肺I/R期间细胞外组蛋白诱导的AMsM1表型(M1-AMs)在驱动NETosis中的显着参与。使用分泌组分析,公共蛋白质数据库,以及AM和中性粒细胞的transwell共培养模型,我们确定源自AMs的组织蛋白酶C(CTSC)是NETosis的主要介质。进一步阐明分子机制,我们发现CTSC通过依赖于NADPH氧化酶介导的活性氧(ROS)产生的途径诱导NETosis。CTSC能显著激活p38MAPK,导致NADPH氧化酶亚基p47phox的磷酸化,从而促进细胞质亚基向细胞膜的运输并激活NADPH氧化酶。此外,CTSC上调并激活其底物膜蛋白酶3(mPR3),导致NETosis相关炎症因子的释放增加。抑制CTSC显示出减轻肺I/R期间NETosis相关损伤的巨大潜力。这些发现表明,来自AM的CTSC可能是介导肺I/R期间NETosis的关键因素,靶向CTSC抑制可能是LTx中PGD的一种新型干预措施。
    Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
    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:糖尿病(DM)可加重肺缺血再灌注(I/R)损伤,是肺移植后受体死亡的重要危险因素。二甲双胍可防止多种器官的I/R损伤。然而,二甲双胍对糖尿病肺I/R损伤的影响尚不清楚.因此,本研究旨在观察二甲双胍对2型糖尿病大鼠肺移植后肺缺血再灌注损伤的影响及机制。
    方法:SD大鼠随机分为6组:对照组+假手术组(CS组),控制+I/R组(CIR组),DM+假手术组(DS组),DM+I/R组(DIR组),DM+I/R+二甲双胍组(DIRM组)和DM+I/R+二甲双胍+化合物C组(DIRMC组)。对照组和糖尿病大鼠进行了假手术或左肺移植手术。肺功能,肺泡毛细血管通透性,炎症反应,氧化应激,再灌注24小时后测定细胞凋亡和p-AMPK/AMPK比值。
    结果:与CIR组相比,DIR组表现为肺功能下降,肺泡毛细血管通透性增加,炎症反应,氧化应激和坏死,但降低了p-AMPK/AMPK比值。二甲双胍改善移植肺功能,肺泡毛细血管通透性降低,炎症反应,氧化应激和坏死,并增加p-AMPK/AMPK比值。相比之下,复方C消除二甲双胍的保护作用。
    结论:二甲双胍通过AMPK途径减轻2型糖尿病肺移植受体大鼠的I/R损伤和坏死。
    BACKGROUND: Diabetes mellitus (DM) can aggravate lung ischemia-reperfusion (I/R) injury and is a significant risk factor for recipient mortality after lung transplantation. Metformin protects against I/R injury in a variety of organs. However, the effect of metformin on diabetic lung I/R injury remains unclear. Therefore, this study aimed to observe the effect and mechanism of metformin on lung I/R injury following lung transplantation in type 2 diabetic rats.
    METHODS: Sprague-Dawley rats were randomly divided into the following six groups: the control + sham group (CS group), the control + I/R group (CIR group), the DM + sham group (DS group), the DM + I/R group (DIR group), the DM + I/R + metformin group (DIRM group) and the DM + I/R + metformin + Compound C group (DIRMC group). Control and diabetic rats underwent the sham operation or left lung transplantation operation. Lung function, alveolar capillary permeability, inflammatory response, oxidative stress, necroptosis and the p-AMPK/AMPK ratio were determined after 24 h of reperfusion.
    RESULTS: Compared with the CIR group, the DIR group exhibited decreased lung function, increased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, but decreased the p-AMPK/AMPK ratio. Metformin improved the function of lung grafts, decreased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, and increased the p-AMPK/AMPK ratio. In contrast, the protective effects of metformin were abrogated by Compound C.
    CONCLUSIONS: Metformin attenuates lung I/R injury and necroptosis through AMPK pathway in type 2 diabetic lung transplant recipient rats.
    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:本研究的目的是研究盐酸戊乙奎醚(PHC)如何影响肺缺血再灌注损伤大鼠模型中肺组织细胞焦亡的发生。
    方法:24只SD大鼠,重250克至270克,随机分为三个不同的组,如下所述:假手术组(S组),对照组(C组),和试验组(PHC组)。PHC组的大鼠接受3mg/kg剂量的PHC的初步静脉内注射。实验结束时,收集肺组织和血液样本并妥善储存用于后续分析.丙二醛的水平,超氧化物歧化酶,和肺组织中的髓过氧化物酶,以及血清中的IL-18和IL-1β,使用ELISA试剂盒进行评估。焦亡相关蛋白,包括Caspase1p20,GSDMD-N,和NLRP3,通过蛋白质印迹法检测。此外,还记录了肺组织的干湿比(D/W)和血气分析结果.
    结果:与对照组相比,PHC组显示氧合指标增强,减少氧化应激和炎症反应,肺损伤减少。此外,PHC组表现出降低的焦亡相关蛋白水平,包括gasderminD(GSDMD-N)的N端节段,caspase-1p20和核苷酸结合寡聚化结构域样受体蛋白3(NLRP3)。
    结论:预先给药PHC有可能通过抑制肺组织细胞的焦亡来减轻肺缺血再灌注损伤,减少炎症反应,增强肺功能。PHC的抗热作用背后的主要机制似乎涉及氧化应激的抑制。
    OBJECTIVE: The aim of this research was to examine how penehyclidine hydrochloride (PHC) impacts the occurrence of pyroptosis in lung tissue cells within a rat model of lung ischemia-reperfusion injury.
    METHODS: Twenty-four Sprague Dawley (SD) rats, weighing 250 g to 270 g, were randomly distributed into three distinct groups as outlined below: a sham operation group (S group), a control group (C group), and a test group (PHC group). Rats in the PHC group received a preliminary intravenous injection of PHC at a dose of 3 mg/kg. At the conclusion of the experiment, lung tissue and blood samples were collected and properly stored for subsequent analysis. The levels of malondialdehyde, superoxide dismutase, and myeloperoxidase in the lung tissue, as well as IL-18 and IL-1β in the blood serum, were assessed using an Elisa kit. Pyroptosis-related proteins, including Caspase1 p20, GSDMD-N, and NLRP3, were detected through the western blot method. Additionally, the dry-to-wet ratio (D/W) of the lung tissue and the findings from the blood gas analysis were also documented.
    RESULTS: In contrast to the control group, the PHC group showed enhancements in oxygenation metrics, reductions in oxidative stress and inflammatory reactions, and a decrease in lung injury. Additionally, the PHC group exhibited lowered levels of pyroptosis-associated proteins, including the N-terminal segment of gasdermin D (GSDMD-N), caspase-1p20, and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3).
    CONCLUSIONS: Pre-administration of PHC has the potential to mitigate lung ischemia-reperfusion injuries by suppressing the pyroptosis of lung tissue cells, diminishing inflammatory reactions, and enhancing lung function. The primary mechanism behind anti-pyroptotic effect of PHC appears to involve the inhibition of oxidative stress.
    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
    本研究的目的是研究促红细胞生成素(EPO)对肺缺血再灌注损伤(LIRI)的影响。
    采用SpragueDawley大鼠和BEAS-2B细胞构建体内和体外缺血再灌注(I/R)诱导的模型,分别。之后,用不同浓度的EPO处理I/R大鼠和叔丁基过氧化氢(TBHP)诱导的细胞。此外,40名LIRI患者和健康对照者被纳入研究。
    观察到肺组织损伤,LIRI模型在体内和体外的细胞凋亡和BAX和caspase-3的表达均高于对照组,然而,Bcl-2、FGF23和FGFR4表达水平低于对照组。EPO给药可显着减少肺组织损伤和细胞凋亡,同时也上调FGF23和FGFR4的表达。挽救实验表明,EPO发挥了与FGF23/FGFR4/p-ERK1/2信号通路相关的保护作用。值得注意的是,血清EPO的表达,LIRI患者FGF23、FGFR4和Bcl-2降低,而caspase-3和BAX的表达较高。
    EPO可以有效提高LIRI,这可能与FGF23/FGFR4/p-ERK1/2信号通路的激活有关。
    UNASSIGNED: The purpose of the present study was to investigate the effect of erythropoietin (EPO) on lung ischemia-reperfusion injury (LIRI).
    UNASSIGNED: Sprague Dawley rats and BEAS-2B cells were employed to construct an ischemia-reperfusion (I/R)-induced model in vivo and in vitro, respectively. Afterward, I/R rats and tert-butyl hydroperoxide (TBHP)-induced cells were treated with different concentrations of EPO. Furthermore, 40 patients with LIRI and healthy controls were enrolled in the study.
    UNASSIGNED: It was observed that lung tissue damage, cell apoptosis and the expression of BAX and caspase-3 were higher in the LIRI model in vivo and in vitro than in the control group, nevertheless, the Bcl-2, FGF23 and FGFR4 expression level was lower than in the control group. EPO administration significantly reduced lung tissue damage and cell apoptosis while also up-regulating the expression of FGF23 and FGFR4. Rescue experiments indicated that EPO exerted a protective role associated with the FGF23/FGFR4/p-ERK1/2 signal pathway. Notably, the expression of serum EPO, FGF23, FGFR4 and Bcl-2 was decreased in patients with LIRI, while the expression of caspase-3 and BAX was higher.
    UNASSIGNED: EPO could effectively improve LIRI, which might be related to the activation of the FGF23/FGFR4/p-ERK1/2 signaling pathway.
    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
    肺缺血再灌注损伤(LIRI)是多种肺部疾病和外科手术中普遍存在的,包括肺切除和移植.LIRI可导致全身低氧血症和多器官衰竭。羟基柠檬酸(HCA),藤黄果皮中存在的主要酸,表现出抗炎,抗氧化剂,和抗癌特性。然而,HCA对LIRI的影响尚不清楚。为了研究HCA对小鼠LIRI的影响,将小鼠随机分为4组:对照组,I/R模型组,I/R+低剂量或高剂量HCA组。将人脐静脉内皮细胞(HUVEC)缺氧12小时,然后复氧6小时以模拟体外LIRI。结果表明,给予HCA可有效减轻肺损伤,炎症,缺血再灌注引起的水肿。此外,缺血再灌注损伤后,HCA治疗可显着降低丙二醛(MDA)和活性氧(ROS)水平,同时降低铁含量并增加超氧化物歧化酶(SOD)水平。机械上,HCA给药在体内和体外均显着抑制了Hif-1α和HO-1的上调。我们发现HCA还可以以浓度依赖的方式减轻H/R诱导的HUVECs中的内皮屏障损伤。此外,Hif-1α的过表达抵消了HCA介导的H/R诱导的内皮细胞铁凋亡的抑制。总之,这些结果表明,HCA通过Hif-1α途径抑制氧化应激和铁凋亡,减轻了LIRI。
    Lung ischemia-reperfusion injury (LIRI) is a prevalent occurrence in various pulmonary diseases and surgical procedures, including lung resections and transplantation. LIRI can result in systemic hypoxemia and multi-organ failure. Hydroxycitric acid (HCA), the primary acid present in the peel of Garcinia cambogia, exhibits anti-inflammatory, antioxidant, and anticancer properties. However, the effects of HCA on LIRI remain unknown. To investigate the impact of HCA on LIRI in mice, the mice were randomly divided into four groups: the control group, the I/R model group, and the I/R + low- or high-dose HCA groups. Human umbilical vein endothelial cells (HUVECs) were subjected to hypoxia for 12 h followed by reoxygenation for 6 h to simulate in vitro LIRI. The results demonstrated that administration of HCA effectively attenuated lung injury, inflammation, and edema induced by ischemia reperfusion. Moreover, HCA treatment significantly reduced malondialdehyde (MDA) and reactive oxygen species (ROS) levels while decreasing iron content and increasing superoxide dismutase (SOD) levels after ischemia-reperfusion insult. Mechanistically, HCA administration significantly inhibited Hif-1α and HO-1 upregulation both in vivo and in vitro. We found that HCA could also alleviate endothelial barrier damage in H/R-induced HUVECs in a concentration-dependent manner. In addition, overexpression of Hif-1α counteracted HCA-mediated inhibition of H/R-induced endothelial cell ferroptosis. In summary, these results indicate that HCA alleviated LIRI by inhibiting oxidative stress and ferroptosis through the Hif-1α pathway.
    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:肺缺血再灌注(I/R)损伤是一个严重的临床问题,没有有效的治疗方法。增强支链氨基酸(BCAA)代谢可以防止心脏I/R损伤,这可能与BCAA代谢物产生的生物活性分子有关。L-β-氨基异丁酸(L-BAIBA),BCAA的代谢产物,具有多器官保护作用,但它是否能预防肺I/R损伤尚不清楚。
    方法:为了评估L-BAIBA对肺I/R损伤的保护作用,通过夹住左肺门产生动物模型,然后在C57BL/6小鼠中释放夹钳。将具有肺I/R损伤的小鼠用L-BAIBA(150mg/kg/天)预处理或后处理,通过管饲法或腹膜内注射给予。通过测量肺水肿和分析血气来评估肺损伤。通过测量支气管肺泡灌洗液(BALF)中的促炎细胞因子来评估炎症,通过髓过氧化物酶活性测量肺的中性粒细胞浸润。分子生物学方法,包括蛋白质印迹和免疫荧光,用于检测A549和BEAS-2B细胞中的潜在信号传导机制。
    结果:我们发现L-BAIBA可以通过抑制铁凋亡来保护肺免受I/R损伤,这取决于C57BL/6小鼠中GPX4和SLC7A11表达的上调。此外,我们证明Nrf-2信号通路是L-BAIBA抑制A549和BEAS-2B细胞铁凋亡的关键。L-BAIBA可诱导Nrf-2的核转位。干扰Nrf-2的表达消除了L-BAIBA对铁凋亡的保护作用。筛选潜在的信号通路显示L-BAIBA可以增加AMPK的磷酸化,化合物C可以阻断L-BAIBA诱导的Nrf-2核易位。化合物C的存在还阻断了L-BAIBA对C57BL/6小鼠肺I/R损伤的保护作用。
    结论:我们的研究表明,L-BAIBA通过AMPK/Nrf-2信号通路保护肺I/R损伤,这可能是一个治疗目标。
    L-BAIBA通过激活AMPK/Nrf-2/GPX4/SLC7A11信号通路上调GPX4和SLC7A11的表达,从而防止I/R诱导的肺中ROS和铁凋亡的增加。
    BACKGROUND: Lung ischemia-reperfusion (I/R) injury is a serious clinical problem without effective treatment. Enhancing branched-chain amino acids (BCAA) metabolism can protect against cardiac I/R injury, which may be related to bioactive molecules generated by BCAA metabolites. L-β-aminoisobutyric acid (L-BAIBA), a metabolite of BCAA, has multi-organ protective effects, but whether it protects against lung I/R injury is unclear.
    METHODS: To assess the protective effect of L-BAIBA against lung I/R injury, an animal model was generated by clamping the hilum of the left lung, followed by releasing the clamp in C57BL/6 mice. Mice with lung I/R injury were pre-treated or post-treated with L-BAIBA (150 mg/kg/day), given by gavage or intraperitoneal injection. Lung injury was assessed by measuring lung edema and analyzing blood gases. Inflammation was assessed by measuring proinflammatory cytokines in bronchoalveolar lavage fluid (BALF), and neutrophil infiltration of the lung was measured by myeloperoxidase activity. Molecular biological methods, including western blot and immunofluorescence, were used to detect potential signaling mechanisms in A549 and BEAS-2B cells.
    RESULTS: We found that L-BAIBA can protect the lung from I/R injury by inhibiting ferroptosis, which depends on the up-regulation of the expressions of GPX4 and SLC7A11 in C57BL/6 mice. Additionally, we demonstrated that the Nrf-2 signaling pathway is key to the inhibitory effect of L-BAIBA on ferroptosis in A549 and BEAS-2B cells. L-BAIBA can induce the nuclear translocation of Nrf-2. Interfering with the expression of Nrf-2 eliminated the protective effect of L-BAIBA on ferroptosis. A screening of potential signaling pathways revealed that L-BAIBA can increase the phosphorylation of AMPK, and compound C can block the Nrf-2 nuclear translocation induced by L-BAIBA. The presence of compound C also blocked the protective effects of L-BAIBA on lung I/R injury in C57BL/6 mice.
    CONCLUSIONS: Our study showed that L-BAIBA protects against lung I/R injury via the AMPK/Nrf-2 signaling pathway, which could be a therapeutic target.
    L-BAIBA upregulates the expression of GPX4 and SLC7A11 by activating the AMPK/Nrf-2/GPX4/SLC7A11 signaling pathway, thereby protecting against I/R-induced increase in ROS and ferroptosis in the lung.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    肺缺血再灌注损伤(IRI),以炎症为特征,血管通透性,和肺水肿,是肺移植后原发性移植物功能障碍的主要原因。我们最近报道,内皮细胞(EC)TRPV4通道在IR后肺水肿和功能障碍中起重要作用。然而,肺IR诱导的内皮TRPV4通道激活的细胞机制尚不清楚.在小鼠IRI的左肺门结扎模型中,我们发现肺IR增加了细胞外ATP(eATP)通过EC膜上的pannexin1(Panx1)通道的流出。eATP通过嘌呤能P2Y2受体(P2Y2R)信号通过内皮TRPV4通道激活基本Ca2内流信号。在离体和体外肺IR替代模型中,在人和小鼠肺微血管内皮中也观察到了TRPV4通道的P2Y2R依赖性激活。P2Y2R的内皮特异性缺失,小鼠中的TRPV4和Panx1对肺IR诱导的内皮TRPV4通道激活具有实质性的保护作用,肺水肿,炎症,和功能障碍。这些结果确定内皮P2Y2R是肺水肿的新介质,炎症,和IR后的功能障碍,并显示内皮细胞Panx1-P2Y2R-TRPV4信号通路的破坏可能代表一种有希望的预防移植后肺IRI的治疗策略。
    Lung ischemia-reperfusion injury (IRI), characterized by inflammation, vascular permeability, and lung edema, is the major cause of primary graft dysfunction after lung transplantation. We recently reported that endothelial cell (EC) TRPV4 channels play a central role in lung edema and dysfunction after IR. However, the cellular mechanisms for lung IR-induced activation of endothelial TRPV4 channels are unknown. In a left-lung hilar ligation model of IRI in mice, we found that lung IR increases the efflux of extracellular ATP (eATP) through pannexin 1 (Panx1) channels at the EC membrane. Elevated eATP activated elementary Ca2+ influx signals through endothelial TRPV4 channels through purinergic P2Y2 receptor (P2Y2R) signaling. P2Y2R-dependent activation of TRPV4 channels was also observed in human and mouse pulmonary microvascular endothelium in ex vivo and in vitro surrogate models of lung IR. Endothelium-specific deletion of P2Y2R, TRPV4, and Panx1 in mice had substantial protective effects against lung IR-induced activation of endothelial TRPV4 channels, lung edema, inflammation, and dysfunction. These results identify endothelial P2Y2R as a novel mediator of lung edema, inflammation, and dysfunction after IR, and show that disruption of endothelial Panx1-P2Y2R-TRPV4 signaling pathway could represent a promising therapeutic strategy for preventing lung IRI after transplantation.
    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
    简介:地塞米松(DEX),作为一种重要的持久效应糖皮质激素(GC),由于其免疫调节特性,在肺缺血再灌注损伤(LIRI)综合治疗领域具有广阔的前景,如诱导细胞凋亡和细胞周期分布。然而,由于多种内部生理障碍,其有效的抗炎应用仍然受到限制。方法:这里,我们开发了涂覆有光敏剂/封端剂/荧光探针修饰的介孔二氧化硅(UCNPs@mSiO2[DEX]-Py/β-CD/FITC,USDPFs)用于精确的DEX释放协同LIRI综合治疗。UCNP是通过在YOF:Yb上覆盖惰性YOF:Yb外壳来设计的,Tm核心在近红外(NIR)激光照射下实现高强度蓝色和红色上转换发射。结果:在适宜的配伍条件下,光敏剂的分子结构会随着封端剂的脱落而受损,这赋予了USDPF进行DEX释放控制和荧光指示剂靶向的杰出能力。此外,DEX的混合封装显着提高了纳米药物的利用率,提高水溶性和生物利用度,这有利于开发USDPFs在复杂临床环境中的抗炎性能。讨论:DEX在肺内微环境中的反应控制释放可减少正常细胞损伤,能有效避免纳米药物在抗炎应用中的副作用。同时,多波长UCNPs赋予纳米药物在肺内微环境中的荧光发射成像能力,为LIRI提供精确的指导。
    Introduction: Dexamethasone (DEX), as an important enduring-effect glucocorticoid (GC), holds great promise in the field of lung ischemia-reperfusion injury (LIRI) comprehensive therapy owing to its immunomodulatory properties, such as inducing apoptosis and cell cycle distribution. However, its potent anti-inflammatory application is still restricted because of multiple internal physiologic barriers. Methods: Herein, we developed upconversion nanoparticles (UCNPs) coated with photosensitizer/capping agent/fluorescent probe-modified mesoporous silica (UCNPs@mSiO2[DEX]-Py/β-CD/FITC, USDPFs) for precise DEX release synergistic LIRI comprehensive therapy. The UCNPs were designed by covering an inert YOF:Yb shell on the YOF:Yb, Tm core to achieve high-intensity blue and red upconversion emission upon Near-Infrared (NIR) laser irradiation. Results: Under suitable compatibility conditions, the molecular structure of photosensitizer can be damaged along with capping agent shedding, which endowed USDPFs with an outstanding capability to carry out DEX release controlling and fluorescent indicator targeting. Furthermore, the hybrid encapsulating of DEX significantly increased utilization of nano-drugs, improving the water solubility and bioavailability, which was conducive to developing the anti-inflammatory performance of USDPFs in the complex clinical environment. Discussion: The response-controlled release of DEX in the intrapulmonary microenvironment can reduce normal cell damage, which can effectively avoid the side effects of nano-drugs in anti-inflammatory application. Meanwhile, the multi-wavelength of UCNPs endowed nano-drugs with the fluorescence emission imaging capacity in an intrapulmonary microenvironment, providing precise guidance for LIRI.
    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
    甲烷(CH4)对肺缺血再灌注(I/R)损伤具有保护作用,但机制尚不清楚,尤其是肺表面活性物质的作用。因此,本研究旨在探讨吸入CH4对肺表面活性物质在大鼠肺I/R损伤中的作用及其机制。大鼠随机分为三组(n=6):假手术组,I/R控制,和I/RCH4组。在假小组中,只对大鼠进行了开胸手术。在I/R控制和I/RCH4组中,大鼠左肺门闭塞90分钟,然后再灌注180分钟,分别用O2或2.5%CH4通气。与假手术组相比,肺表面活性物质中大型表面活性物质聚集体(LA)的水平,肺顺应性,氧合减少,小表面活性剂聚集体(SAs),炎症反应,氧化应激损伤,对照组细胞凋亡增加(P<0.05)。与对照治疗相比,CH4增加LA(0.42±0.06vs.0.31±0.09mg/kg),氧合(201±11vs.151±14mmHg),和肺顺应性(16.8±1.0vs.11.5±1.3ml/kg),以及总抗氧化能力和Nrf2蛋白表达降低炎症反应和凋亡细胞数(P<0.05)。总之,CH4吸入减少氧化应激损伤,炎症反应,和细胞凋亡,并通过Nrf2介导的肺表面活性物质调节大鼠肺I/R损伤改善肺功能。
    Methane (CH4) exerted protective effects against lung ischemia-reperfusion (I/R) injury, but the mechanism remains unclear, especially the role of pulmonary surfactant. Therefore, this study aimed to explore the effects of CH4 inhalation on pulmonary surfactant in rat lung I/R injury and to elucidate the mechanism. Rats were randomly divided into three groups (n = 6): the sham, I/R control, and I/R CH4 groups. In the sham group, only thoracotomy was performed on the rats. In the I/R control and I/R CH4 groups, the rats underwent left hilum occlusion for 90 min, followed by reperfusion for 180 min and ventilation with O2 or 2.5% CH4, respectively. Compared with those of the sham group, the levels of large surfactant aggregates (LAs) in pulmonary surfactant, lung compliance, oxygenation decreased, the small surfactant aggregates (SAs), inflammatory response, oxidative stress injury, and cell apoptosis increased in the control group (P < 0.05). Compared to the control treatment, CH4 increased LA (0.42 ± 0.06 vs. 0.31 ± 0.09 mg/kg), oxygenation (201 ± 11 vs. 151 ± 14 mmHg), and lung compliance (16.8 ± 1.0 vs. 11.5 ± 1.3 ml/kg), as well as total antioxidant capacity and Nrf2 protein expression and decreased the inflammatory response and number of apoptotic cells (P < 0.05). In conclusion, CH4 inhalation decreased oxidative stress injury, inflammatory response, and cell apoptosis, and improved lung function through Nrf2-mediated pulmonary surfactant regulation in rat lung I/R 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
    In previous study, we reported that kaempferol ameliorates significantly lung ischemia-reperfusion injury (LIRI), and may be achieved by targeting the SIRT 1 pathway. This study further explored the anti-LIRI mechanism of kaempferol. In vitro, the rat alveolar epithelial cells L2 was cultured and subjected to anoxia/reoxygenation (A/R) insult. In vivo, SD rats were operated to establish LIRI model. The related indicators of oxidative stress and apoptosis in L2 cells and rats lung tissues were detected. Results showed that kaempferol pre-treatment significantly increased the cell viability, improved mitochondrial membrane potential, inhibited the opening of mitochondrial permeability transition pores, reduced the levels of oxidative stress and apoptosis, increased the expressions of Bcl-2 and mitochondrial cytochrome c, and decreased the expressions of Bax and cytoplasmic cytochrome c in L2 cells after A/R insult. In vivo, kaempferol improved the pathological injury, inhibited the levels of oxidative stress and apoptosis, increased the expressions of Bcl-2 and mitochondrial cytochrome c, and decreased the expressions of Bax and cytoplasmic cytochrome c in rats lung tissues after I/R. However, the aforementioned effects of kaempferol were significantly attenuated by the SIRT 1 inhibitor EX527 or the PGC-1α inhibitor SR-18292. What\'s more, SR-18292 has not reversed the effect of kaempferol on increasing the protein activity of SIRT 1. Above results suggest that kaempferol ameliorates LIRI by improving mitochondrial function, reducing oxidative stress and inhibiting cell apoptosis. Its molecular mechanism of action includes the SIRT 1/PGC-1α/mitochondria signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号