lung toxicity

肺毒性
  • 文章类型: Journal Article
    二氧化硅纳米颗粒(SiNP)对肺的毒性是已知的。我们之前证明了暴露于SiNPs会促进肺损伤,但确切的发病机制仍未阐明。Ferroptosis现在被认为是一种独特的氧化细胞死亡形式,但其是否参与SiNPs诱导的肺损伤尚不清楚.在这项工作中,我们通过气管内滴注建立了SiNPs亚慢性吸入暴露的大鼠模型,并进行了组织病理学检查,铁检测,以及与铁凋亡相关的脂质过氧化和蛋白质测定。此外,我们评估了SiNPs对上皮铁蛋白的影响,使用体外培养的人支气管上皮细胞(16HBE)细胞的可能机制,并评估了对成纤维细胞活化的影响。因此,大鼠肺发生纤维化病变,伴随着增强的脂质过氧化,铁过载,和铁中毒。始终如一,体外数据显示SiNPs引发氧化应激并导致脂质过氧化物的积累,导致铁中毒。重要的是,机制研究显示,miR-21-5p在SiNPs通过靶向GCLM清除GSH诱导的上皮铁生成过程中起关键作用.值得注意的是,通过SiNPs可以极大地抑制铁凋亡并减轻上皮损伤和随后的成纤维细胞活化。总之,我们的研究结果首次揭示了SiNPs通过miR-21-5p/GCLM信号传导触发上皮铁凋亡,从而促进纤维化病变的成纤维细胞活化,并强调了在SiNP暴露后抑制铁凋亡对抗肺损伤的治疗潜力。
    The toxicity of silica nanoparticles (SiNPs) to lung is known. We previously demonstrated that exposure to SiNPs promoted pulmonary impairments, but the precise pathogenesis remains elucidated. Ferroptosis has now been identified as a unique form of oxidative cell death, but whether it participated in SiNPs-induced lung injury remains unclear. In this work, we established a rat model with sub-chronic inhalation exposure of SiNPs via intratracheal instillation, and conducted histopathological examination, iron detection, and ferroptosis-related lipid peroxidation and protein assays. Moreover, we evaluated the effect of SiNPs on epithelial ferroptosis, possible mechanisms using in vitro-cultured human bronchial epithelial cells (16HBE), and also assessed the ensuing impact on fibroblast activation for fibrogenesis. Consequently, fibrotic lesions occurred in the rat lungs, concomitantly by enhanced lipid peroxidation, iron overload, and ferroptosis. Consistently, the in vitro data showed SiNPs triggered oxidative stress and caused the accumulation of lipid peroxides, resulting in ferroptosis. Importantly, the mechanistic investigation revealed miR-21-5p as a key player in the epithelial ferroptotic process induced by SiNPs via targeting GCLM for GSH depletion. Of note, ferrostatin-1 could greatly suppress ferroptosis and alleviate epithelial injury and ensuing fibroblast activation by SiNPs. In conclusion, our findings first revealed SiNPs triggered epithelial ferroptosis through miR-21-5p/GCLM signaling and thereby promoted fibroblast activation for fibrotic lesions, and highlighted the therapeutic potential of inhibiting ferroptosis against lung impairments upon SiNPs exposure.
    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
    二氧化硅纳米颗粒(SiNP)是二氧化硅的主要形式之一,并且由地球上最丰富的化合物组成。基于其优异的性能,SiNPs广泛用于食品生产,合成过程,医学诊断,药物输送,和其他领域。SiNP的大规模生产和广泛应用增加了人类暴露于SiNP的风险。在工作场所和环境中,SiNP主要通过呼吸道进入人体并到达肺部;因此,肺是SiNP的最重要和最具毒理学影响的靶器官。越来越多的研究表明,SiNP暴露会导致严重的肺毒性。然而,关于SiNPs在离体和体内环境中的毒性研究仍处于探索阶段。SiNP的肺毒性的分子机制是变化的并且尚未完全理解。因此,本文综述了SiNP诱导肺毒性的可能机制,如氧化应激,内质网应激,线粒体损伤,细胞死亡。此外,这项研究提供了由SiNPs引起的疾病进展的总结,从而为今后研究SiNP致肺毒性的机制奠定了理论基础。
    Silicon dioxide nanoparticles (SiNPs) are one of the major forms of silicon dioxide and are composed of the most-abundant compounds on earth. Based on their excellent properties, SiNPs are widely used in food production, synthetic processes, medical diagnostics, drug delivery, and other fields. The mass production and wide application of SiNPs increases the risk of human exposure to SiNPs. In the workplace and environment, SiNPs mainly enter the human body through the respiratory tract and reach the lungs; therefore, the lungs are the most important and most toxicologically affected target organ of SiNPs. An increasing number of studies have shown that SiNP exposure can cause severe lung toxicity. However, studies on the toxicity of SiNPs in ex vivo and in vivo settings are still in the exploratory phase. The molecular mechanisms underlying the lung toxicity of SiNPs are varied and not yet fully understood. As a result, this review summarizes the possible mechanisms of SiNP-induced lung toxicity, such as oxidative stress, endoplasmic reticulum stress, mitochondrial damage, and cell death. Moreover, this study provides a summary of the progression of diseases caused by SiNPs, thereby establishing a theoretical basis for future studies on the mechanisms of SiNP-induced lung toxicity.
    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: Comparative Study
    微/纳米塑料(MNP)在环境和多种生物体中无处不在。一些常见类型的MNP的毒性,例如,聚醚砜(PES)MNP,仍然知之甚少。在这项研究中使用了多组学方法来确定食源性和空气中的PESMNP对肝脏和肺的影响,分别。食源性MNP能够诱导肠道微生物菌群失调,肠道和血清代谢中断,和肝脏转录组失调,并影响血清抗氧化活性和肝功能,导致肝损伤。至于机载MNP,发现它们会引起鼻腔和肺部微生物菌群失调,血清和肺代谢紊乱,和肝脏转录组紊乱,并引起血清抗氧化活性破坏和肺损伤。发现食源性和空气传播的PESNP分别比MP引起更大的肝和肺毒性,这可能与NP和MP暴露之间的差异有关。相关结果表明,食源性PESMNP可以破坏“肠道微生物群-肠道-肝脏”轴并引起肝损伤,而空气中的PESMNP可能会影响“空气中的微生物群-肺”轴并引起肺损伤。这一发现有助于诊断食源性和空气中的PESMNP分别引起的肝和肺损伤,以及在人类生活环境中正确使用PES。
    Micro/nanoplastics (MNP) are ubiquitous in the environment and multiple living organisms. The toxicity of some common types of MNP, e.g., polyethersulfone (PES) MNP, remains poorly understood. Multi-omics approaches were used in this study to determine the effects of foodborne and airborne PES MNP on liver and lung, respectively. Foodborne MNP were capable of inducing gut microbial dysbiosis, gut and serum metabolic disruption, and liver transcriptomic dysregulation, and affecting serum antioxidant activity and liver function, resulting in liver injury. As for the airborne MNP, they were found to induce nasal and lung microbial dysbiosis, serum and lung metabolic disruption, and liver transcriptome disturbance, and cause disrupted serum antioxidant activity and lung injury. Foodborne and airborne PES NP were found to respectively induce greater liver and lung toxicity than MP, which could be associated with the differences between NP and MP exposures. The relevant results suggest that foodborne PES MNP could disrupt the \"gut microbiota-gut-liver\" axis and induce hepatic injury, while airborne PES MNP could affect the \"airborne microbiota-lung\" axis and cause lung injury. The findings could benefit the diagnoses of liver and lung injury respectively induced by foodborne and airborne PES MNP, as well as the proper use of PES in human living environment.
    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
    铅基钙钛矿纳米颗粒(Pb-PNP)已在各个领域得到广泛应用。然而,由于稳定性差和相对较强的水溶性,Pb-PNP在制造过程中释放到环境中的潜在毒性,用法,和处置引起了极大的关注。吸入是人类暴露于Pb-PNP的主要途径。在这里,研究了Pb-PNPs在呼吸系统中的毒性作用和潜在的分子机制。使用多种生物测定和电子显微镜研究了CsPbBr3纳米颗粒在BEAS-2B细胞中的体外细胞毒性。不同浓度的CsPbBr3纳米颗粒诱导过度氧化应激和细胞凋亡。此外,CsPbBr3纳米颗粒特异性募集TGF-β1,随后诱导上皮-间质转化。此外,鼻内给药后,研究ICR小鼠中代表性CsPbBr3纳米颗粒的生物分布和肺毒性。这些发现表明CsPbBr3纳米颗粒在小鼠模型中显著诱导肺部炎症和上皮-间质转化,甚至可以导致肺纤维化。以上发现揭示了Pb-PNP在肺中的不良反应和分子机制,拓宽了Pb-PNP的安全数据。
    Lead-based perovskite nanoparticles (Pb-PNPs) have found extensive applications across diverse fields. However, because of poor stability and relatively strong water solubility, the potential toxicity of Pb-PNPs released into the environment during their manufacture, usage, and disposal has attracted significant attention. Inhalation is a primary route through which human exposure to Pb-PNPs occurs. Herein, the toxic effects and underlying molecular mechanisms of Pb-PNPs in the respiratory system are investigated. The in vitro cytotoxicity of CsPbBr3 nanoparticles in BEAS-2B cells is studied using multiple bioassays and electron microscopy. CsPbBr3 nanoparticles of different concentrations induce excessive oxidative stress and cell apoptosis. Furthermore, CsPbBr3 nanoparticles specifically recruit the TGF-β1, which subsequently induces epithelial-mesenchymal transition. In addition, the biodistribution and lung toxicity of representative CsPbBr3 nanoparticles in ICR mice are investigated following intranasal administration. These findings indicate that CsPbBr3 nanoparticles significantly induce pulmonary inflammation and epithelial-mesenchymal transition and can even lead to pulmonary fibrosis in mouse models. Above findings expose the adverse effects and molecular mechanisms of Pb-PNPs in the lung, which broadens the safety data of Pb-PNPs.
    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
    细颗粒物(PM2.5)通过促进炎症细胞因子的释放而对健康造成不良影响。Rosavidin(Ro),具有多种生物活性的苯丙素类化合物,是从大花红景天中提取的,一种药食同源植物。然而,Ro在PM2.5引起的肺毒性中的保护作用和机制尚未被研究。本研究旨在探讨Ro对PM2.5肺毒性的保护作用及其机制。采用不同剂量Ro预处理(50mg/kg和100mg/kg)后气管滴注PM2.5混悬液建立肺毒性大鼠模型,评价Ro对PM2.5肺毒性的影响。结果表明,Ro减轻了病理变化,水肿,和大鼠的炎症反应。PI3K/AKT信号通路可能与Ro对肺毒性的保护作用有关。随后,我们验证了PI3K/AKT在PM2.5暴露肺组织中的作用。此外,p-PI3K和p-AKT的表达水平较低,还有NLRP3,ASC,裂解的caspase-1,裂解的IL-1β,PM2.5组的GSDMD-N高于对照组。而Ro的预施用逆转了这些蛋白质在肺组织中的表达趋势。值得注意的是,在使用Ro与尼加霉素或LY294002的组合进行预处理后,未观察到Ro的这些保护作用.这些结果表明,Ro通过激活PI3K/AKT信号通路抑制NLRP3炎性体介导的焦亡,从而减轻PM2.5引起的肺毒性。
    Fine particulate matter (PM2.5) contributes to adverse health effects through the promotion of inflammatory cytokine release. Rosavidin (Ro), a phenylpropanoid compound having multiple biological activities, is extracted from Rhodiola crenulata, a medicine and food homology plant. However, the protective role and mechanism of Ro in PM2.5-induced lung toxicity have not been previously studied. This study aimed to investigate the potential protective effect and mechanism of Ro in PM2.5-induced lung toxicity. A lung toxicity rat model was established through trachea drip of PM2.5 suspension after the different dose pretreatment of Ro (50 mg/kg and 100 mg/kg) to evaluate the effect of Ro on PM2.5 caused lung toxicity. The results showed that Ro attenuated the pathological changes, edema, and inflammation response in rats. The PI3K/AKT signaling pathway may be associated with the protective effect of Ro against pulmonary toxicity. Subsequently, we verified the role of PI3K/AKT in the PM2.5 exposure lung tissue. Moreover, expression levels of p-PI3K and p-AKT were lower, and those of NLRP3, ASC, cleaved caspase-1, cleaved IL-1β, and GSDMD-N were higher in PM2.5 group compared to those in control group. Whereas pre-administration of Ro reversed the expression trends of these proteins in lung tissue. Notably, those protective effects of Ro were not observed after pretreatment with a combination of Ro with nigericin or LY294002. These results indicate that Ro mitigates PM2.5-caused lung toxicity by inhibiting NLRP3 inflammasome-mediated pyroptosis through activation of the PI3K/AKT signaling pathway.
    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: Meta-Analysis
    目的:间变性淋巴瘤激酶酪氨酸激酶抑制剂(ALKTKIs)在非小细胞肺癌(NSCLC)患者中显示出显著的临床活性。然而,肺炎是NSCLC患者ALKTKIs的严重副作用。在这个荟萃分析中,我们旨在确定ALK-TKI相关性肺炎的发生率.
    方法:我们搜索了电子数据库,以确定直到2022年8月发表的相关研究。当没有观察到实质性异质性时,使用固定效应模型计算肺炎的发生率。否则,使用随机效应模型。对不同治疗组进行亚组分析。使用STATA17.0进行统计分析。
    结果:涉及4752名患者的26项临床试验符合分析条件。所有级别肺炎发生率为2.92%(95%置信区间[CI]:1.79%-4.27%),高级别(3-4级)肺炎发生率为1.42%(95%CI:0.84%-2.12%),5级肺炎发生率为0.09%(95%CI:0.00%-0.28%).亚组分析显示,布格替尼与全级别和高级别肺炎的发生率最高(7.09%和3.06%,分别)。与一线ALKTKI治疗相比,化疗后ALKTKI治疗的全级别和高级别肺炎发生率更高(7.73%vs.2.26%和3.64%与1.26%,分别)。来自日本试验的队列具有较高的全级别和高级别肺炎发生率。
    结论:我们的研究提供了接受ALKTKIs治疗的患者肺炎发生率的精确数据。总的来说,ALKTKIs具有可耐受的肺毒性。需要早期肺炎的识别和治疗,以防止接受布格替尼治疗的患者和先前接受化疗的患者进一步恶化。特别是在日本人口中。
    Anaplastic lymphoma kinase tyrosine kinase inhibitors (ALK TKIs) have shown remarkable clinical activity in patients with non-small-cell lung cancer (NSCLC). However, pneumonitis is a serious side effect of ALK TKIs in NSCLC patients. In this meta-analysis, we aimed to determine the incidence of ALK-TKI-associated pneumonitis.
    We searched electronic databases to identify relevant studies published until August 2022. The incidence of pneumonitis was calculated using a fixed-effects model when no substantial heterogeneity was observed. Otherwise, a random-effects model was used. Subgroup analyses of different treatment groups were performed. Statistical analyses were conducted using STATA 17.0.
    Twenty-six clinical trials involving 4752 patients were eligible for analysis. All-grade pneumonitis incidence was 2.92% (95% confidence interval [CI]: 1.79%-4.27%), high-grade (Grade 3-4) pneumonitis incidence was 1.42% (95% CI: 0.84%-2.12%) and Grade 5 pneumonitis incidence was 0.09% (95% CI: 0.00%-0.28%). The subgroup analysis showed that brigatinib was associated with the highest incidence of both all-grade and high-grade pneumonitis (7.09% and 3.06%, respectively). ALK TKI treatment after chemotherapy was associated with a higher incidence of all-grade and high-grade pneumonitis than first-line ALK TKI treatment (7.73% vs. 2.26% and 3.64% vs. 1.26%, respectively). Cohorts from Japanese trials had a higher incidence of all-grade and high-grade pneumonitis.
    Our study provides precise data on the incidence of pneumonitis in patients receiving treatment with ALK TKIs. Overall, ALK TKIs have tolerable pulmonary toxicity. Early pneumonitis identification and treatment are required to prevent further deterioration in patients receiving treatment with brigatinib and in those who received prior chemotherapy, particularly in the Japanese population.
    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
    暴露于细颗粒物(PM2.5),一种环境污染物,与呼吸系统疾病相关的发病率和死亡风险显著增加。Sipeimine(Sip)是贝母中的甾体生物碱,具有抗氧化和抗炎作用。然而,Sip对肺毒性的保护作用及其机制至今仍知之甚少。在本研究中,通过建立大鼠经口气管滴注PM2.5(7.5mg/kg)混悬液的肺毒性模型,研究Sip的肺保护作用。Sprague-Dawley大鼠每天腹腔注射Sip(15mg/kg或30mg/kg)或赋形剂,连续3天,然后滴注PM2.5悬浮液以建立肺毒性模型。结果发现Sip明显改善肺组织的病理损伤,减轻炎症反应,并抑制肺组织焦亡。我们还发现PM2.5激活了NLRP3炎性体,如NLRP3,裂解的caspase-1和ASC蛋白的上调水平所证明的。重要的是,PM2.5可以通过增加与焦亡相关的蛋白质水平来触发焦亡,包括IL-1β,裂解的IL-1β,和GSDMD-N,膜孔形成,和线粒体肿胀.不出所料,所有这些有害的改变都被Sip预处理逆转。Sip的这些作用被NLRP3激活剂尼德霉素阻断。此外,网络药理学分析显示Sip可能通过PI3K/AKT信号通路发挥作用,这表明Sip通过抑制PI3K和AKT的磷酸化来抑制NLRP3炎性体介导的焦亡。我们的发现表明,Sip通过激活PM2.5诱导的肺毒性中的PI3K/AKT通路来抑制NLRP3介导的细胞焦亡,具有很好的应用价值和发展前景。
    Exposure to fine particulate matter (PM2.5), an environmental pollutant, significantly contributes to the incidence of and risk of mortality associated with respiratory diseases. Sipeimine (Sip) is a steroidal alkaloid in fritillaries that exerts antioxidative and anti-inflammatory effects. However, protective effect of Sip for lung toxicity and its mechanism to date remains poorly understood. In the present study, we investigated the lung-protective effect of Sip via establishing the lung toxicity model of rats with orotracheal instillation of PM2.5 (7.5 mg/kg) suspension. Sprague-Dawley rats were intraperitoneally administered with Sip (15 mg/kg or 30 mg/kg) or vehicle daily for 3 days before instillation of PM2.5 suspension to establish the model of lung toxicity. The results found that Sip significantly improved pathological damage of lung tissue, mitigated inflammatory response, and inhibited lung tissue pyroptosis. We also found that PM2.5 activated the NLRP3 inflammasome as evidenced by the upregulation levels of NLRP3, cleaved-caspase-1, and ASC proteins. Importantly, PM2.5 could trigger pyroptosis by increased levels of pyroptosis-related proteins, including IL-1β, cleaved IL-1β, and GSDMD-N, membrane pore formation, and mitochondrial swelling. As expected, all these deleterious alterations were reversed by Sip pretreatment. These effects of Sip were blocked by the NLRP3 activator nigericin. Moreover, network pharmacology analysis showed that Sip may function via the PI3K/AKT signaling pathway and animal experiment validate the results, which revealed that Sip inhibited NLRP3 inflammasome-mediated pyroptosis by suppressing the phosphorylation of PI3K and AKT. Our findings demonstrated that Sip inhibited NLRP3-mediated cell pyroptosis through activation of the PI3K/AKT pathway in PM2.5-induced lung toxicity, which has a promising application value and development prospect against lung injury in the future.
    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
    细颗粒物(PM2.5)污染已成为公共健康的主要威胁,并与肺毒性有关。河马信号系统的关键监管机构之一,Yes相关蛋白1(YAP1),推测在铁凋亡发育中起作用。这里,我们重点研究了YAP1在焦凋亡和铁凋亡中的功能,旨在探索其在PM2.5引起的肺毒性中的治疗潜力。PM2.5诱导的肺毒性在野生型WT和条件YAP1敲除小鼠中诱导,肺上皮细胞受到PM2.5的体外刺激。我们用了蛋白质印迹,透射电子显微镜,和荧光显微镜来研究焦化瘤和铁还原相关特性。我们发现PM2.5通过涉及焦化凋亡和铁凋亡的机制导致肺毒性。YAP1敲低阻碍了焦亡,铁性凋亡,和PM2.5引起的肺损伤,如组织病理学增加所示,更高水平的促炎细胞因子,GSDMD蛋白,脂质过氧化,和铁的积累,以及增加NLRP3炎性体激活和降低SLC7A11表达。YAP1沉默持续促进NLRP3炎性体激活并降低SLC7A11水平,加重PM2.5引起的细胞损伤。相比之下,过表达YAP1的细胞抑制NLRP3炎性体激活并增加SLC7A11水平,防止焦亡和铁凋亡。总的来说,我们的数据提示YAP1通过抑制NLRP3介导的焦凋亡和SL7A11依赖性的铁凋亡,改善PM2.5诱导的肺损伤.
    Pollution from fine particulate matter (PM2.5) has become a major threat to public health and has been related to lung toxicity. One of the key regulators of the Hippo signaling system, Yes-associated protein 1 (YAP1), is speculated to play a role in ferroptosis development. Here, we focused on investigating the function of YAP1 in pyroptosis and ferroptosis, aiming to explore its therapeutic potential in PM2.5-induced lung toxicity. PM2.5-induced lung toxicity was induced in Wild-type WT and conditional YAP1-knockout mice, and lung epithelial cells were stimulatd by PM2.5 in vitro. We used western blot, transmission electron microscopy, and fluorescence microscopy to investigate pyroptosis- and ferroptosis-related characteristics. We found that PM2.5 leads to lung toxicity using mechanisms involving pyroptosis and ferroptosis. YAP1 knockdown impeded pyroptosis, ferroptosis, and PM2.5-induced lung damage, as shown by increased histopathology, higher levels of proinflammatory cytokines, GSDMD protein, lipid peroxidation, and iron accumulation, as well as increased NLRP3 inflammasome activation and decreased SLC7A11 expression. YAP1 silencing consistently promoted NLRP3 inflammasome activation and reduced SLC7A11 levels, aggravating PM2.5-induced cellular damage. In contrast, YAP1-overexpressing cells inhibited NLRP3 inflammasome activation and increased SLC7A11 levels, preventing pyroptosis and ferroptosis. Overall, our data suggest that YAP1 ameliorates PM2.5-induced lung injury by inhibiting NLRP3-mediated pyroptosis and SL7A11-dependent ferroptosis.
    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
    目的:研究臭氧诱导肺上皮细胞线粒体自噬与细胞凋亡的调控关系。
    结果:首先,用不同剂量的臭氧处理I型原代肺泡上皮细胞和雄性Wistar大鼠。Ⅰ型原代肺泡上皮细胞ATP含量和线粒体膜电位显著下降。线粒体自噬相关标志物和PINK1/Parkin通路相关蛋白,以及LC3,Parkin,I型肺泡上皮细胞中的线粒体表明臭氧暴露会触发线粒体自噬。另一方面,活性氧(ROS)抑制剂NAC可以明显减轻上皮细胞的线粒体自噬。用线粒体自噬抑制剂MDIVI-1治疗后,NLRP3炎性体的水平,细胞中裂解的caspase-1和N-gasderminD(N-GSDMD)显着降低。总之,这些结果表明,臭氧暴露可以触发线粒体自噬,并随后诱导由NLRP3炎性体介导的细胞死亡。最后,NLRP3的过表达和敲低证实了这一结论。
    结论:臭氧暴露诱导氧化损伤,导致线粒体结构和功能损伤。臭氧诱导的ROS通过激活PINK1/Parkin信号通路触发线粒体自噬,然后通过激活NLRP3炎性体而导致焦亡。
    OBJECTIVE: To study the regulatory relationship between ozone-induced mitophagy and pyroptosis in lung epithelial cells.
    RESULTS: First, type I primary alveolar epithelial cells and male Wistar rats were treated with ozone at different dosages. The ATP content and mitochondrial membrane potential significantly decreased in type I primary alveolar epithelial cells. The mitophagy-related markers and PINK1/Parkin pathway-related proteins, and the co-localization of LC3, Parkin, and mitochondria in type I alveolar epithelial cells indicated that ozone exposure triggered mitophagy. On the other hand, the reactive oxygen species (ROS) inhibitor NAC could significantly alleviate mitophagy in epithelial cells. After treatment with the mitophagy inhibitor MDIVI-1, the levels of the NLRP3 inflammasome, cleaved caspase-1, and N-gasdermin D (N-GSDMD) significantly decreased in the cells. Altogether, these results indicated that mitophagy can be triggered by ozone exposure, and subsequently induces cell death mediated by the NLRP3 inflammasome. Finally, the overexpression and knockdown of NLRP3 confirmed this conclusion.
    CONCLUSIONS: Ozone exposure induced oxidative damage, leading to mitochondrial structural and functional damage. Ozone-induced ROS triggered mitophagy through the activation of the PINK1/Parkin signaling pathway, then pyroptosis through activation of the NLRP3 inflammasome.
    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
    暴露于空气污染中的颗粒物(PM)2.5是全球范围内的严重健康问题。目前,PM2.5引起的肺毒性缺乏有效的预防措施和治疗方式。本研究阐明了黄芪甲苷(Ast)的保护作用,黄芪膜质Bunge的天然产品,对PM2.5引起的肺毒性及其可能的分子机制。通过气管内滴注PM2.5粉尘悬浮液建立小鼠肺毒性模型。调查是用Ast或与尼格林联合进行的,它是NOD样受体蛋白3(NLRP3)激活剂。结果表明,PM2.5通过上调促炎细胞因子并引起与NLRP3炎性体介导的焦亡途径相关的氧化应激,导致显著的肺部炎症并促进细胞死亡的焦亡模式。通过抑制NLRP3/caspase-1轴抑制NLRP3炎性体介导的焦亡,防止PM2.5产生肺毒性,从而保护肺免受PM2.5引起的肺部炎症和氧化损伤,最终导致小鼠存活时间延长。Nigericin部分逆转了Ast的保护作用。本研究为Ast的治疗潜力提供了新的见解,证明它可能是预防PM2.5引起的呼吸系统疾病的候选药物。靶向NLRP3炎性体可能是PM2.5引起的呼吸系统疾病的一种新的治疗策略。
    Exposure to particulate matter (PM)2.5 in air pollution is a serious health issue worldwide. At present, effective prevention measures and modalities of treatment for PM2.5-caused lung toxicity are lacking. This study elucidated the protective effect of astragaloside IV (Ast), a natural product from Astragalus membranaceous Bunge, against PM2.5-caused lung toxicity and its possible molecular mechanisms. The mice model of lung toxicity was performed by intratracheal instillation of PM2.5 dust suspension. The investigation was performed with Ast or in combination with nigericin, which is a NOD-like receptor protein 3 (NLRP3) activator. The results revealed that PM2.5 lead significant lung inflammation and promoted the pyroptosis pattern of cell death by upregulating pro-inflammatory cytokines and causing oxidative stress related to the NLRP3 inflammasome-mediated pyroptosis pathway. Ast protected against PM2.5 resulted lung toxicity via suppressing NLRP3 inflammasome-mediated pyroptosis via NLRP3/caspase-1 axis inhibition, thereby protecting the lung against PM2.5-induced lung inflammation and oxidative damage, eventually resulting in prolonged survival in mice. Nigericin partially reversed the protective effects of Ast. The present research provides new insights into the therapeutic potential of Ast, demonstrating that it might be a possible candidate for the prevention of PM2.5-caused respiratory diseases. Targeting the NLRP3 inflammasome might be a novel therapeutic tactic for PM2.5-caused respiratory diseases.
    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