primary human airway epithelial cells

  • 文章类型: Journal Article
    鼻病毒(RV)是哮喘急性发作的主要诱因,COPD,和其他呼吸道疾病。房车分为三种(RV-A,RV-B,和RV-C),包含超过160种血清型,很难开发出有效的疫苗。目前,目前尚无有效的RV感染治疗方法.肺表面活性物质是脂质和蛋白质的细胞外复合物,在调节肺的先天免疫中起重要作用。次要的肺表面活性物质脂质,棕榈酰-油酰基-磷脂酰甘油(POPG)和磷脂酰肌醇(PI),是炎症过程的有效调节剂,并对呼吸道合胞病毒(RSV)和甲型流感病毒(IAV)发挥抗病毒活性。在目前的研究中,我们检测了在气-液界面(ALI)分化的原代人气道上皮细胞(AECs)中POPG和PI对鼻病毒A16(RV-A16)的效力.在用RV-A16感染AECs后,PI使病毒RNA拷贝数减少70%,并下调(55-75%)抗病毒(MDA5、IRF7和IFN-lambda)和CXCL11趋化因子基因的表达。相比之下,POPG仅略微降低MDA5(24%)和IRF7(11%)基因表达,但不抑制AECs中IFN-λ基因表达或RV-A16复制。然而,POPG和PI均抑制(50-80%)IL6基因表达和蛋白质分泌以及CXCL11蛋白分泌。PI处理显著减弱了AECs中单独由RV-A16感染诱导的整体基因表达变化。观察到的抑制作用是间接的,主要是由于抑制了病毒复制。与PI处理相反的病毒调节基因的细胞类型富集分析显示,PI抑制的病毒诱导杯状细胞化生和病毒诱导的纤毛下调,俱乐部,和离子细胞类型。值得注意的是,PI处理还改变了RV-A16调节某些磷脂酰肌醇4激酶(PI4K)表达的能力;酰基辅酶A结合,包含域(ACBD);和低密度脂蛋白受体(LDLR)基因在宿主细胞中RV复制所需的复制细胞器(RO)的形成和功能中起关键作用。这些数据表明PI可以用作有效的,无毒,用于预防和治疗RV感染的抗病毒剂。
    Rhinoviruses (RVs) are major instigators of acute exacerbations of asthma, COPD, and other respiratory diseases. RVs are categorized into three species (RV-A, RV-B, and RV-C), which comprise more than 160 serotypes, making it difficult to develop an effective vaccine. Currently, no effective treatment for RV infection is available. Pulmonary surfactant is an extracellular complex of lipids and proteins that plays a central role in regulating innate immunity in the lung. The minor pulmonary surfactant lipids, palmitoyl-oleoyl-phosphatidylglycerol (POPG) and phosphatidylinositol (PI), are potent regulators of inflammatory processes and exert antiviral activity against respiratory syncytial virus (RSV) and influenza A viruses (IAV). In the current study, we examined the potencies of POPG and PI against rhinovirus A16 (RV-A16) in primary human airway epithelial cells (AECs) differentiated at an air-liquid interface (ALI). After AECs were infected with RV-A16, PI reduced the viral RNA copy number by 70% and downregulated (55-75%) the expression of antiviral (MDA5, IRF7, and IFN-lambda) and CXCL11 chemokine genes. In contrast, POPG only slightly decreased MDA5 (24%) and IRF7 (11%) gene expression but did not inhibit IFN-lambda gene expression or RV-A16 replication in AECs. However, both POPG and PI inhibited (50-80%) IL6 gene expression and protein secretion and CXCL11 protein secretion. PI treatment dramatically attenuated global gene expression changes induced by RV-A16 infection alone in AECs. The observed inhibitory effects were indirect and resulted mainly from the inhibition of virus replication. Cell-type enrichment analysis of viral-regulated genes opposed by PI treatment revealed the PI-inhibited viral induction of goblet cell metaplasia and the virus-induced downregulation of ciliated, club, and ionocyte cell types. Notably, the PI treatment also altered the ability of RV-A16 to regulate the expression of some phosphatidylinositol 4-kinase (PI4K); acyl-CoA-binding, domain-containing (ACBD); and low-density lipoprotein receptor (LDLR) genes that play critical roles in the formation and functioning of replication organelles (ROs) required for RV replication in host cells. These data suggest PI can be used as a potent, non-toxic, antiviral agent for RV infection prophylaxis and treatment.
    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
    UNASSIGNED:分化的人气道上皮细胞的体外模型是研究严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)感染的有价值的工具。通过使用这些模型,已经表明,SARS-CoV-2进入基因在人气道上皮中的表达受多种因素的影响,例如年龄,性别,吸烟状况,和致病条件。在这项研究中,我们旨在确定细胞培养条件和供体人口统计学和临床特征对包括血管紧张素转换酶2(ACE2)在内的SARS-CoV-2进入基因表达的影响,跨膜丝氨酸蛋白酶2(TMPRSS2),组织蛋白酶L(CTSL),和酪氨酸蛋白激酶受体UFO(AXL)在原发性气道上皮细胞。
    UNASSIGNED:招募了11名患有或不患有慢性阻塞性肺疾病(COPD)或哮喘的肺癌患者。通过定量实时PCR分析从浸没或气液界面(ALI)培养物中分离的人支气管上皮细胞(HBEC)或小气道上皮细胞(SAEC)。我们还测试了与临床数据的相关性。
    未经批准:在ALI文化中,HBEC中AXL的表达明显高于SAEC。此外,与深层培养相比,在ALI条件下分化的HBEC和SAEC中ACE2,TMPRSS2和CTSL的表达均显着增加。在SAEC中SARS-CoV-2进入基因的表达与年龄之间发现了微不足道的关联,性别,吸烟状况,和COPD的并发症,细胞供体的哮喘或高血压。
    UNASSIGNED:这些结果表明,SARS-CoV-2进入基因在体外分化的原代气道上皮细胞中的表达受个体培养条件的影响比受个体供体的特定特征的影响大得多。
    UNASSIGNED: In-vitro models of differentiated primary human airway epithelial cells are a valuable tool to study severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Through the use of these models, it has been shown that the expression of SARS-CoV-2 entry genes in human airway epithelia is influenced by various factors such as age, sex, smoking status, and pathogenic conditions. In this study, we aimed to determine the effects of cell culture conditions and donor demographic and clinical characteristics on the expression of SARS-CoV-2 entry genes including angiotensin converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2), cathepsin L (CTSL), and tyrosine protein kinase receptor UFO (AXL) in primary airway epithelial cells.
    UNASSIGNED: Eleven lung cancer patients with or without chronic obstructive pulmonary disease (COPD) or asthma were recruited. Human bronchial epithelial cells (HBEC) or small airway epithelial cells (SAEC) isolated from submerged or air-liquid interface (ALI) cultures were analyzed by quantitative real-time PCR. We also tested for correlations with clinical data.
    UNASSIGNED: In ALI cultures, the expression of AXL was significantly higher in HBEC than in SAEC. In addition, the expression of ACE2, TMPRSS2, and CTSL was significantly increased in both HBEC and SAEC differentiated under ALI conditions compared with the submerged culture. Negligible association was found between the expression of SARS-CoV-2 entry genes in SAEC and the age, sex, smoking status, and complication of COPD, asthma or hypertension of the cell donors.
    UNASSIGNED: These results demonstrate that the expression of SARS-CoV-2 entry genes in differentiated primary airway epithelial cells in-vitro is much more influenced by individual culture conditions than by specific characteristics of individual donors.
    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
    通过B.1.1.7、B.1.351、B.1.617.2和B.1.1.29谱系(对应于α,beta,delta,和关注的omicron变体(VoC),分别)。然而,仍然缺乏生物学证据,这些VoC在多大程度上与祖先血统不同。通过利用人气道上皮细胞(HAEC)培养,非常类似于人类的气道结构和生理学,我们报告了不同呼吸组织中独特的SARS-CoV-2嗜性。总的来说,SARS-CoV-2VoC在HAEC中的主要感染和复制优于祖先USA-WA1分离株或BavPat1分离株,其中包含D614G突变,即使变体之间在其传染性方面几乎没有差异(即,病毒体每vRNA拷贝比)。我们还观察到在存在病毒的情况下,上呼吸道和下呼吸道组织之间的组织特异性先天免疫激活差异。我们的研究提供了对这种生理相关离体模型中不同VoC行为的更好理解。
    The emergence of new SARS-CoV-2 variants and the replacement of preceding isolates have been observed through B.1.1.7, B.1.351, B.1.617.2, and B.1.1.529 lineages (corresponding to alpha, beta, delta, and omicron variants of concern (VoC), respectively). However, there is still a lack of biological evidence to which extent those VoC differ from the ancestral lineages. By exploiting human airway epithelial cell (HAEC) cultures, which closely resemble the human airway architecture and physiology, we report distinctive SARS-CoV-2 tropism in different respiratory tissues. In general, SARS-CoV-2 VoC predominantly infect and replicate in HAEC better than the progenitor USA-WA1 isolate or the BavPat1 isolate, which contains the D614G mutation, even though there is little to no difference between variants regarding their infectivity (i.e., virion-per-vRNA copy ratio). We also observe differential tissue-specific innate immunity activation between the upper and lower respiratory tissues in the presence of the virus. Our study provides better comprehension of the behavior of the different VoC in this physiologically relevant ex vivo model.
    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
    有,除了Remdesivir,没有批准用于治疗SARS-CoV-2感染的抗病毒药物。为了帮助寻找对抗这种病毒的抗病毒药物,我们探索了在气液界面(ALI)生长的人气管气道上皮细胞(HtAEC)和人小气道上皮细胞(HsAEC)的使用。这些培养物在顶端用两种不同的SARS-CoV-2分离株之一感染。每个病毒都被证明可以长时间(至少8天)复制高滴度,特别是在刺突(S)蛋白中含有D614G的分离物在35℃比37℃更有效。探索了在系统的基底外侧添加到培养基中的一组选定的参考药物的效果。Remdesivir,GS-441524(remdesivir的母体核苷),EIDD-1931(莫那普拉韦的母体核苷)和IFN(β1和λ1)均导致病毒RNA的剂量依赖性抑制,并在顶端收集感染性病毒滴度。然而,AT-511(目前在临床试验中的AT-527的游离碱基形式)不能抑制这些体外原代细胞模型中的病毒复制。一起,这些结果为进一步选择SARS-CoV-2抑制剂进行临床前和临床开发提供了参考。
    There are, besides remdesivir, no approved antivirals for the treatment of SARS-CoV-2 infections. To aid in the search for antivirals against this virus, we explored the use of human tracheal airway epithelial cells (HtAEC) and human small airway epithelial cells (HsAEC) grown at the air-liquid interface (ALI). These cultures were infected at the apical side with one of two different SARS-CoV-2 isolates. Each virus was shown to replicate to high titers for extended periods of time (at least 8 days) and, in particular an isolate with the D614G in the spike (S) protein did so more efficiently at 35 °C than 37 °C. The effect of a selected panel of reference drugs that were added to the culture medium at the basolateral side of the system was explored. Remdesivir, GS-441524 (the parent nucleoside of remdesivir), EIDD-1931 (the parent nucleoside of molnupiravir) and IFN (β1 and λ1) all resulted in dose-dependent inhibition of viral RNA and infectious virus titers collected at the apical side. However, AT-511 (the free base form of AT-527 currently in clinical testing) failed to inhibit viral replication in these in vitro primary cell models. Together, these results provide a reference for further studies aimed at selecting SARS-CoV-2 inhibitors for further preclinical and clinical development.
    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
    由迄今未描述的机制引发的过度炎症是严重急性呼吸综合征冠状病毒2(SARS-CoV-2)感染的标志,并且与增强的致病性和死亡率相关。
    补体过度激活促进肺损伤,并在中东呼吸综合征相关冠状病毒患者中观察到,SARS-CoV-1和SARS-CoV-2感染。因此,我们从补体成分3(C3)介导的作用方面研究了人气道上皮细胞在暴露于SARS-CoV-2时的首次相互作用。
    为此,我们使用了感染SARS-CoV-2患者分离株的高分化原代人3维组织模型.关于感染,病毒载量,病毒感染性,细胞内补体激活,炎症机制,和组织破坏通过实时RT-PCR分析,高含量筛选,斑块检测,luminex分析,和跨上皮电阻测量。
    这里,我们表明,原发性正常人支气管和小气道上皮细胞通过膨胀的局部C3动员对SARS-CoV-2感染作出反应。SARS-CoV-2感染导致细胞内补体激活过度,并破坏了单层培养的人气道细胞和高度分化的上皮完整性,伪分层,粘液产生,纤毛呼吸组织模型。SARS-CoV-2感染的3维培养物分泌的C3a和促炎细胞因子IL-6,单核细胞趋化蛋白1,IL-1α的水平显着升高,和RANTES。
    最重要的是,我们在此首次说明,靶向非免疫呼吸细胞中的过敏毒素受体C3a受体和C5a受体可以预防内在肺部炎症和组织损伤.这为COVID-19的治疗开辟了令人兴奋的可能性。
    Excessive inflammation triggered by a hitherto undescribed mechanism is a hallmark of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and is associated with enhanced pathogenicity and mortality.
    Complement hyperactivation promotes lung injury and was observed in patients suffering from Middle East respiratory syndrome-related coronavirus, SARS-CoV-1, and SARS-CoV-2 infections. Therefore, we investigated the very first interactions of primary human airway epithelial cells on exposure to SARS-CoV-2 in terms of complement component 3 (C3)-mediated effects.
    For this, we used highly differentiated primary human 3-dimensional tissue models infected with SARS-CoV-2 patient isolates. On infection, viral load, viral infectivity, intracellular complement activation, inflammatory mechanisms, and tissue destruction were analyzed by real-time RT-PCR, high content screening, plaque assays, luminex analyses, and transepithelial electrical resistance measurements.
    Here, we show that primary normal human bronchial and small airway epithelial cells respond to SARS-CoV-2 infection by an inflated local C3 mobilization. SARS-CoV-2 infection resulted in exaggerated intracellular complement activation and destruction of the epithelial integrity in monolayer cultures of primary human airway cells and highly differentiated, pseudostratified, mucus-producing, ciliated respiratory tissue models. SARS-CoV-2-infected 3-dimensional cultures secreted significantly higher levels of C3a and the proinflammatory cytokines IL-6, monocyte chemoattractant protein 1, IL-1α, and RANTES.
    Crucially, we illustrate here for the first time that targeting the anaphylotoxin receptors C3a receptor and C5a receptor in nonimmune respiratory cells can prevent intrinsic lung inflammation and tissue damage. This opens up the exciting possibility in the treatment of COVID-19.
    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
    Significant progress has been achieved in developing precision therapies for cystic fibrosis; however, highly effective treatments that target the ion channel, CFTR, are not yet available for many patients. As numerous CFTR therapeutics are currently in the clinical pipeline, reliable screening tools capable of predicting drug efficacy to support individualized treatment plans and translational research are essential. The utilization of bronchial, nasal, and rectal tissues from individual cystic fibrosis patients for drug testing using in vitro assays such as electrophysiological measurements of CFTR activity and evaluation of fluid movement in spheroid cultures, has advanced the prediction of patient-specific responses. However, for precise prediction of drug effects, in vitro models of CFTR rescue should incorporate the inflamed cystic fibrosis airway environment and mimic the complex tissue structures of airway epithelia. Furthermore, novel assays that monitor other aspects of successful CFTR rescue such as restoration of mucus characteristics, which is important for predicting mucociliary clearance, will allow for better prognoses of successful therapies in vivo. Additional cystic fibrosis treatment strategies are being intensively explored, such as development of drugs that target other ion channels, and novel technologies including pluripotent stem cells, gene therapy, and gene editing. The multiple therapeutic approaches available to treat the basic defect in cystic fibrosis combined with relevant precision medicine models provide a framework for identifying optimal and sustained treatments that will benefit all cystic fibrosis patients.
    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
    The choice of model used to study human respiratory syncytial virus (RSV) infection is extremely important. RSV is a human pathogen that is exquisitely adapted to infection of human hosts. Rodent models, such as mice and cotton rats, are semi-permissive to RSV infection and do not faithfully reproduce hallmarks of RSV disease in humans. Furthermore, immortalized airway-derived cell lines, such as HEp-2, BEAS-2B, and A549 cells, are poorly representative of the complexity of the respiratory epithelium. The development of a well-differentiated primary pediatric airway epithelial cell models (WD-PAECs) allows us to simulate several hallmarks of RSV infection of infant airways. They therefore represent important additions to RSV pathogenesis modeling in human-relevant tissues. The following protocols describe how to culture and differentiate both bronchial and nasal primary pediatric airway epithelial cells and how to use these cultures to study RSV cytopathogenesis.
    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