acute lung injury and acute respiratory distress syndrome

  • 文章类型: Journal Article
    很少有药物干预措施能够提高急性肺损伤和急性呼吸窘迫综合征(ALI/ARDS)的死亡率。这项研究的目的是阐明内质网(ER)应激和c-Jun-N-末端激酶(JNK)-线粒体途径是否在ALI/ARDS中起重要作用,并确定关键成分Sab是否是潜在的治疗目标。当前的研究调查了ER应激的激活和JNK途径,通过蛋白质印迹分析,在ER应激和脂多糖(LPS)诱导的ALI/ARDS期间位于线粒体上的JNK含量。通过包括组织病理学评估在内的多种方法探索了位于线粒体上的JNK的选择性抑制剂Tat-SabKIM1的治疗效果。通过TUNEL测定法检测肺细胞凋亡,线粒体膜通透性和存活分析。结果证实,在LPS诱导的ALI/ARDS过程中,ER应激增强,并可诱导JNK途径和JNK线粒体定位以及线粒体功能障碍和细胞死亡的激活。Tat‑SabKIM1通过特异性抑制JNK定位于线粒体和mito‑JNK信号激活而不影响胞浆/核JNK激活,减轻了LPS注射诱导的肺损伤并提高了小鼠存活率。Tat-SabKIM1对ALI/ARDS的保护作用部分是由抑制线粒体介导的细胞凋亡和自噬的过度激活引起的。这些结果显示了Sab作为ALI/ARDS治疗靶点的重要作用以及Tat-SabKIM1的潜在治疗效果。总之,JNK线粒体途径的异常激活可以显著破坏肺细胞的正常生理功能,导致ALI/ARDS的发生和Tat-SabKIM1选择性抑制线粒体上的JNK对LPS诱导的ALI/ARDS小鼠内质网应激引起的线粒体功能障碍和细胞死亡具有保护作用。
    Few pharmacological interventions are able to improve the mortality rate of acute lung injury and acute respiratory distress syndrome (ALI/ARDS). The aim of this research was to elucidate whether endoplasmic reticulum (ER) stress and c‑Jun‑N‑terminal kinase (JNK)‑mitochondria pathways serve important roles in ALI/ARDS and to determine whether the key component Sab is a potential treatment target. The current study investigated the activation of ER stress and the JNK pathway, the content of JNK located on the mitochondria during ER stress and lipopolysaccharide (LPS)‑induced ALI/ARDS by western blot analysis. The treatment effects of Tat‑SabKIM1, a selective inhibitor of JNK located on mitochondria were explored by multiple methods including histopathological evaluation, lung cell apoptosis tested by TUNEL assay, mitochondrial membrane permeability and survival analysis. The results verified that ER stress was enhanced during LPS‑induced ALI/ARDS and could induce activation of the JNK pathway and JNK‑mitochondrial localization as well as mitochondrial dysfunction and cell death. Tat‑SabKIM1 alleviated LPS injection‑induced lung injury and improved mouse survival rates by specifically inhibiting JNK localization to mitochondria and mito‑JNK signal activation without affecting cytosolic/nuclear JNK activation. The protective effect of Tat‑SabKIM1 against ALI/ARDS was partly caused by inhibition of the excessive activation of mitochondria‑mediated apoptosis and autophagy. These results showed the important role of Sab as a treatment target of ALI/ARDS and the potential treatment effect of Tat‑SabKIM1. In conclusion, abnormal activation of the JNK‑mitochondrial pathway could significantly disrupt the normal physiological function of lung cells, resulting in the occurrence of ALI/ARDS and selective inhibit of JNK located on mitochondria by Tat‑SabKIM1 had a protective effect against the mitochondrial dysfunction and cell death caused by endoplasmic reticulum stress in mice with LPS‑induced ALI/ARDS.
    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
    磷酸化(p)-JNK到线粒体的定位可导致功能性线粒体疾病,导致能量供应和膜电位下降,以及活性氧产生和细胞凋亡的增加。JNK参与急性肺损伤(ALI)的发生,JNK通路的激活是导致损伤的关键因素之一。本研究的目的是调查JNK-线粒体(mitoJNK)位置是否参与了ALI和急性呼吸窘迫综合征(ALI/ARDS)的发生。本研究检查了JNK途径的激活,位于线粒体上的JNK的含量以及细胞渗透性肽Tat‑SabKIM1的治疗效果,该肽可选择性地抑制JNK在线粒体上的位置。通过蛋白质印迹分析检测蛋白质的表达水平。通过组织学检查评估肺损伤,湿干重量比,以及肺组织中的H2O2和丙二醛浓度。使用TUNEL测定评价肺细胞凋亡。结果表明,JNK在海水吸入诱导的ALI/ARDS过程中被磷酸化和激活,不仅在常规JNK途径中,而且在mitoJNK途径中。还发现Tat‑SabKIM1可以特异性抑制JNK定位于线粒体和mitoJNK信号的激活。此外,Tat‑SabKIM1可抑制Bcl‑2调节的自噬和线粒体介导的细胞凋亡。总之,mitoJNK定位破坏了ALI/ARDS期间线粒体的正常生理功能,选择性抑制JNK和线粒体SH3BP5(也称为Sab)与Tat‑SabKIM1的结合可以阻断ALI/ARDS的恶化。
    Localization of phosphorylated (p)‑JNK to the mitochondria can lead to functional mitochondrial disorder, resulting in a decrease in energy supply and membrane potential, as well as an increase in reactive oxygen species production and apoptosis. JNK is involved in the occurrence of acute lung injury (ALI), and activation of the JNK pathway is one of the crucial factors resulting in injury. The aim of the present study was to investigate whether the JNK‑mitochondria (mitoJNK) location participated in the occurrence of ALI and acute respiratory distress syndrome (ALI/ARDS). The present study examined the activation of the JNK pathway, the content of JNK located on the mitochondria and the treatment effects of a cell‑permeable peptide Tat‑SabKIM1, which can selectively inhibit the location of JNK on mitochondria. The expression levels of proteins were detected by western blot analysis. Lung injuries were evaluated by histological examination, wet‑to‑dry weight ratios, and H2O2 and malondialdehyde concentrations in the lung tissues. Lung cells apoptosis was evaluated using TUNEL assay. The results demonstrated that JNK was phosphorylated and activated during seawater inhalation‑induced ALI/ARDS, not only in the routine JNK pathway but also in the mitoJNK pathway. It was also found that Tat‑SabKIM1 could specifically inhibit JNK localization to mitochondria and the activation of mitoJNK signaling. Furthermore, Tat‑SabKIM1 could inhibit Bcl‑2‑regulated autophagy and mitochondria‑mediated apoptosis. In conclusion, mitoJNK localization disrupted the normal physiological functions of the mitochondria during ALI/ARDS, and selective inhibition of JNK and mitochondrial SH3BP5 (also known as Sab) binding with Tat‑SabKIM1 can block deterioration from ALI/ARDS.
    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
    肾素-血管紧张素系统(RAS)是电解质稳态和血压最重要的调节系统,并通过血管紧张素转换酶(ACE)/血管紧张素II(AngII)/AngII1型(AT1)受体轴和血管紧张素转换酶2(ACE2)/血管紧张素(1-7)/MAS受体轴发挥作用。RAS功能障碍与急性肺损伤(ALI)和急性呼吸窘迫综合征(ARDS)的发生、发展有关,导致严重的预后甚至死亡。ALI/ARDS可以通过各种方式诱发,其中之一是病毒感染,比如SARS-CoV,SARS-CoV-2、H5N1、H7N9和EV71。本文综述了RAS功能障碍如何影响病毒感染引起的ALI/ARDs的具体机制。SARS-CoV和SARS-CoV-2通过与ACE2结合进入宿主细胞。H5N1和H7N9禽流感病毒降低体内ACE2水平,EV71增加AngII浓度。血管紧张素转换酶抑制剂和血管紧张素AT1受体阻滞剂治疗可缓解ALI/ARDS症状。本综述为病毒感染引起的肺损伤的治疗提供建议。
    The renin-angiotensin system (RAS) is the most important regulatory system of electrolyte homeostasis and blood pressure and acts through angiotensin-converting enzyme (ACE)/angiotensin II (Ang II)/Ang II type 1 (AT1) receptor axis and angiotensin-converting enzyme 2 (ACE2)/angiotensin (1-7)/MAS receptor axis. RAS dysfunction is related to the occurrence and development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) and causes a serious prognosis and even death. ALI/ARDS can be induced by various ways, one of which is viral infections, such as SARS-CoV, SARS-CoV-2, H5N1, H7N9, and EV71. This article reviews the specific mechanism on how RAS dysfunction affects ALI/ARDs caused by viral infections. SARS-CoV and SARS-CoV-2 enter the host cells by binding with ACE2. H5N1 and H7N9 avian influenza viruses reduce the ACE2 level in the body, and EV71 increases Ang II concentration. Treatment with angiotensin-converting enzyme inhibitor and angiotensin AT1 receptor blocker can alleviate ALI/ARDS symptoms. This review provides suggestions for the treatment of lung injury caused by viral infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Inhalation or systemic administration of lipopolysaccharide (LPS) can induce acute pulmonary inflammation and lung injury. The pulmonary vasculature is composed of pulmonary microvascular endothelial cells (PMVECs), which form a semiselective membrane for gas exchange. The miRNA miR-642a-5p has previously been reported to be up-regulated in patients with acute respiratory distress syndrome; thus, here, we examined whether this miRNA is involved in the effects of LPS on PMVECs. The levels of miR-642a-5p and mRNA encoding eukaryotic elongation factor 2 (eEF2) were detected by quantitative RT-PCR. Moesin and eEF2 protein levels were tested by western blot assay. Dual-luciferase reporter assay was used to examine the relationship between miR-642a-5p and eEF2. Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and cell permeability was analyzed using the transendothelial electrical resistance assay. We report that miR-642a-5p levels are significantly up-regulated in LPS-stimulated PMVECs, and miR-642a-5p contributes to LPS-induced hyperpermeability and apoptosis of PMVECs. LPS treatment results in down-regulation of eEF2 in PMVECs. Overexpression of eEF2, a direct target of miR-642a-5p, inhibited the effect of LPS on PMVECs. miR-642a-5p promoted LPS-induced hyperpermeability and apoptosis by targeting eEF2. Thus, miR-642a-5p and eEF2 may serve as potential targets for acute lung injury/acute respiratory distress syndrome diagnosis or treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号