Airway epithelia

气道上皮
  • 文章类型: Journal Article
    据报道,污水处理工人和居住在污水处理厂附近的居民出现呼吸道症状。然而,关于城市污水呼吸危害的毒理学研究很少。本研究旨在深入了解城市废水中污染物混合物引起的综合呼吸危害。四个二级污水处理厂(SWTP)废水的综合呼吸危害,三级污水处理厂(TTP),使用正常人支气管上皮细胞(NHBE)生物测定法对人工湿地(CW)进行了评估,分析了各种处理技术的减毒效果。流出物引起细胞毒性,氧化损伤,NHBE中IL-6和CXCL8水平升高,屏障完整性受损,ZO-1和occludin表达降低。Further,流出物抑制了3D支气管球的发育,增加不规则表面和细胞碎片,并抑制管腔结构的形成。TTPE流出物显著增加MUC5AC在支气管球中的表达。在SWTP时,进水的综合生物标志物反应(IBR)被去除40.2%,TTP为18.2%,CW的36.6%,分别。SWTP最终流出物的IBR,TTP,和CW分别为7.2、7.7和7.7。硬脂醇与毒性生物标志物显著相关,邻甲酚,菲,丁基化羟基甲苯,和邻苯二甲酸二甲酯。本研究提供了有关与放电相关的不良呼吸影响的人类相关证据。深水处理的必要性,性能优化,并提出了改善水质和保护呼吸道健康的潜在手段。
    Respiratory symptoms have been reported in wastewater treatment workers and residents living close to sewage treatment plant. However, toxicological research about the respiratory hazards of municipal wastewater is scarce. The present study aims to gain insight into the comprehensive respiratory hazards induced by the contaminant mixtures in municipal wastewater. The integrated respiratory hazards of effluents from four secondary wastewater treatment plants (SWTPs), a tertiary wastewater treatment plant (TTP), and a constructed wetland (CW) were evaluated using normal human bronchial epithelial cells (NHBE) bioassay, and toxicity reduction efficiency of various treatment techniques was analyzed. Effluents caused cytotoxicity, oxidative damage, inflammation response with the increased levels of IL-6 and CXCL8, and impaired barrier integrity with decreased expressions of ZO-1 and occludin in NHBE. Further, the effluents inhibited the development of 3D bronchospheres, increased irregular surface and cell debris, and suppressed the formation of luminal structures. TTP E effluent significantly increased the expression of MUC5AC in bronchospheres. The integrated biomarker response (IBR) of the influent was removed by 40.2% at SWTPs, 18.2% at TTP, and 36.6% at CW, respectively. The IBR of the final effluents from SWTPs, TTP, and CW were 7.2, 7.7, and 7.7, respectively. Significant correlation with toxicity biomarkers was frequently observed for stearyl alcohol, o-cresol, phenanthrene, butylated hydroxytoluene, and dimethyl phthalate. The present study provided human relevant evidence concerning the adverse respiratory effects associated with discharge. The necessity for deep water treatment, performance optimization, and the potential means were suggested for improving water quality and protecting respiratory health.
    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
    遗传突变引起的囊性纤维化跨膜传导调节因子(CFTR)阴离子通道的功能障碍导致遗传性疾病囊性纤维化(CF)。涉及多种上皮功能紊乱的CF肺病可能是由于CFTR作为传导氯离子和碳酸氢根离子的阴离子通道的功能丧失及其作为调节膜和细胞溶胶蛋白活性的细胞调节剂的功能丧失所致。在没有CFTR活性的情况下,丰富的粘液积累,细菌感染和炎症表征CF气道,其中炎症相关的组织重塑和损伤逐渐破坏肺。破译CFTR功能障碍与CF气道细菌感染之间的联系可能揭示CF肺病的发病机制并指导新的治疗方法的发展。为实现这一目标而努力的研究,包括高盐,低音量,严格审查了气道表面液体酸中毒和异常粘液假设。
    Dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel by genetic mutations causes the inherited disease cystic fibrosis (CF). CF lung disease that involves multiple disorders of epithelial function likely results from loss of CFTR function as an anion channel conducting chloride and bicarbonate ions and its function as a cellular regulator modulating the activity of membrane and cytosol proteins. In the absence of CFTR activity, abundant mucus accumulation, bacterial infection and inflammation characterize CF airways, in which inflammation-associated tissue remodeling and damage gradually destroys the lung. Deciphering the link between CFTR dysfunction and bacterial infection in CF airways may reveal the pathogenesis of CF lung disease and guide the development of new treatments. Research efforts towards this goal, including high salt, low volume, airway surface liquid acidosis and abnormal mucus hypotheses are critically reviewed.
    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
    对于气道上皮功能障碍和缺陷的再生,尚无根治性治疗。在临床研究中,将聚丙烯和胶原海绵制成的人工气管用于切除后重建气管。对于人工气管腔表面的早期上皮化,建立了一个模型,也就是说,将覆盖有人类诱导的多能干细胞来源的气道上皮细胞(hiPSC-AECs)的人工气管移植到免疫缺陷大鼠的气管缺损中.与目前用于临床研究的hiPSC衍生细胞的细胞类型不同,AECs通过使基底细胞增殖和分化为不断构成AECs的各种细胞类型来维持组织。因此,移植后,每种细胞类型的比例,如纤毛和杯状细胞,可能会改变;然而,没有研究证实这种可能性。在这项研究中,使用我们的hiPSC-AEC移植的大鼠模型,我们调查了hiPSC-AECs移植前和移植后各细胞类型比例的变化.因此,移植后每种细胞类型的比例发生了变化.纤毛的比例,基底,俱乐部细胞增加了,移植后杯状细胞比例下降。此外,移植的hiPSC-AECs中每种细胞类型的比例与正常近端气道组织中每种细胞类型的比例比移植前每种细胞类型的比例更相似。这项研究的结果对于开发使用hiPSC-AEC移植的治疗技术很有用。
    No radical treatment is available for the regeneration of dysfunction and defects in airway epithelia. Artificial tracheae made of polypropylene and collagen sponge were used in clinical studies to reconstitute tracheae after resection. For early epithelialization of the luminal surface of the artificial trachea, a model was established, that is, an artificial trachea covered with human-induced pluripotent stem cell-derived airway epithelial cells (hiPSC-AECs) was transplanted into a tracheal defect in an immunodeficient rat. Unlike the cell types of hiPSC-derived cells that are currently used in clinical studies, AECs maintain tissues by proliferation and differentiation of basal cells into various cell types that constitute AECs constantly. Therefore, post-transplantation, the proportion of each cell type, such as ciliated and goblet cells, may change; however, no studies have examined this possibility. In this study, using our hiPSC-AEC-transplanted rat model, we investigated changes in the proportion of each cell type in hiPSC-AECs pre-transplantation and post-transplantation. As a result, the proportion of each cell type changed post-transplantation. The proportion of ciliated, basal, and club cells increased, and the proportion of goblet cells decreased post-transplantation. In addition, the proportion of each cell type in engrafted hiPSC-AECs is more similar to the proportion of each cell type in normal proximal airway tissue than the proportion of each cell type pre-transplantation. The results of this study are useful for the development of therapeutic techniques using hiPSC-AEC 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
    已经做出了相当大的努力来开发用于囊性纤维化(CF)的基因治疗的基于腺相关病毒(AAV)的载体。作为定向进化和衣壳混洗技术的结果,AAV衣壳对气道上皮细胞具有广泛的向性。例如,AAV2.5T和AAV6.2是两个进化的衣壳,其具有优于其亲本血清型的改进的气道上皮细胞转导特性。然而,有限的研究集中在确定它们的特定细胞嗜性上。恢复囊性纤维化跨膜传导调节因子(CFTR)在表面柱状上皮细胞中的表达对于纠正CF气道表型是必要的。基础细胞是负责补充表面上皮细胞(包括分泌细胞和离子细胞)的传导气道的祖细胞群。纠正这种细胞群对于长期的基因治疗策略至关重要。在这项研究中,我们研究了AAV衣壳对高分化的人气道上皮(HAE)细胞和原代人气道基底细胞的原代培养物中三种细胞类型的嗜性。我们观察到AAV2.5T转导表面上皮细胞优于AAV6.2,而AAV6.2转导气道基底细胞优于AAV2.5T。我们还调查了一个最近开发的衣壳,AAV6.2FF,其中有两个表面酪氨酸转化为苯丙氨酸。接下来,我们整合了相互突变,以创建具有进一步改善的表面和基底细胞转导特征的AAV衣壳.最后,我们成功地采用了使用AAV的分裂-内含素方法递送腺嘌呤碱基编辑器(ABE)来修复CFTRR553X突变.我们的结果表明,合理掺入AAV衣壳突变可改善气道表面和祖细胞的AAV转导,并最终可能导致CF患者的肺功能改善。
    Considerable effort has been devoted to developing adeno-associated virus (AAV)-based vectors for gene therapy in cystic fibrosis (CF). As a result of directed evolution and capsid shuffling technology, AAV capsids are available with widespread tropism for airway epithelial cells. For example, AAV2.5T and AAV6.2 are two evolved capsids with improved airway epithelial cell transduction properties over their parental serotypes. However, limited research has been focused on identifying their specific cellular tropism. Restoring cystic fibrosis transmembrane conductance regulator (CFTR) expression in surface columnar epithelial cells is necessary for the correction of the CF airway phenotype. Basal cells are a progenitor population of the conducting airways responsible for replenishing surface epithelial cells (including secretory cells and ionocytes), making correction of this cell population vital for a long-lived gene therapy strategy. In this study, we investigate the tropism of AAV capsids for three cell types in primary cultures of well-differentiated human airway epithelial (HAE) cells and primary human airway basal cells. We observed that AAV2.5T transduced surface epithelial cells better than AAV6.2, while AAV6.2 transduced airway basal cells better than AAV2.5T. We also investigated a recently developed capsid, AAV6.2FF, which has two surface tyrosines converted to phenylalanines. Next, we incorporated reciprocal mutations to create AAV capsids with further improved surface and basal cell transduction characteristics. Lastly, we successfully employed a split-intein approach using AAV to deliver an adenine base editor (ABE) to repair the CFTR R553X mutation. Our results suggest that rational incorporation of AAV capsid mutations improves AAV transduction of the airway surface and progenitor cells and may ultimately lead to improved pulmonary function in people with CF.
    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
    CC16(俱乐部细胞分泌蛋白)是由肺内的俱乐部细胞和其他非纤毛上皮细胞产生的蛋白质。CC16已被证明可以预防阻塞性肺疾病的发展并减轻肺部病原体负担。尽管最近在理解CC16对循环的影响方面取得了进展,CC16在肺上皮反应中的生物学机制尚未阐明.
    我们试图确定CC16缺乏是否会损害上皮驱动的宿主反应,并鉴定在肺上皮内表达的新型受体,CC16通过这些受体赋予活性。
    我们利用质谱和定量蛋白质组学研究CC16缺乏如何影响顶端分泌的肺上皮蛋白。小鼠气管上皮细胞(MTECS),在原始条件下和Mp攻击后研究了人鼻上皮细胞(HNEC)和小鼠。
    我们鉴定出8种抗菌蛋白被CC16-/-MTECS显著降低,其中6例通过在重度哮喘研究计划(SARP)队列中的mRNA表达进行了验证。短腭肺和鼻上皮克隆1(SPLUNC1)是差异表达最多的蛋白质(66倍),是本研究的重点。使用MTEC和HNEC的组合,我们发现CC16通过受体复合物极晚期抗原-2(VLA-2)的信号传导增强肺上皮驱动的SPLUNC1表达,并且给予小鼠rCC16可增强肺SPLUNC1的产生并降低肺炎支原体(Mp)负担.同样,rSPLUNC1导致缺乏CC16小鼠的Mp负荷降低。rCC16内的VLA-2整联蛋白结合位点对于SPLUNC1的诱导和Mp负荷的减少是必需的。
    我们的发现证明了CC16通过上调抗微生物剂在上皮驱动的宿主防御中的新作用,并定义了CC16的新型上皮受体VLA-2,通过该受体信号传导是增强SPLUNC1产生所必需的。
    CC16 (Club Cell Secretory Protein) is a protein produced by club cells and other non-ciliated epithelial cells within the lungs. CC16 has been shown to protect against the development of obstructive lung diseases and attenuate pulmonary pathogen burden. Despite recent advances in understanding CC16 effects in circulation, the biological mechanisms of CC16 in pulmonary epithelial responses have not been elucidated.
    We sought to determine if CC16 deficiency impairs epithelial-driven host responses and identify novel receptors expressed within the pulmonary epithelium through which CC16 imparts activity.
    We utilized mass spectrometry and quantitative proteomics to investigate how CC16 deficiency impacts apically secreted pulmonary epithelial proteins. Mouse tracheal epithelial cells (MTECS), human nasal epithelial cells (HNECs) and mice were studied in naïve conditions and after Mp challenge.
    We identified 8 antimicrobial proteins significantly decreased by CC16-/- MTECS, 6 of which were validated by mRNA expression in Severe Asthma Research Program (SARP) cohorts. Short Palate Lung and Nasal Epithelial Clone 1 (SPLUNC1) was the most differentially expressed protein (66-fold) and was the focus of this study. Using a combination of MTECs and HNECs, we found that CC16 enhances pulmonary epithelial-driven SPLUNC1 expression via signaling through the receptor complex Very Late Antigen-2 (VLA-2) and that rCC16 given to mice enhances pulmonary SPLUNC1 production and decreases Mycoplasma pneumoniae (Mp) burden. Likewise, rSPLUNC1 results in decreased Mp burden in mice lacking CC16 mice. The VLA-2 integrin binding site within rCC16 is necessary for induction of SPLUNC1 and the reduction in Mp burden.
    Our findings demonstrate a novel role for CC16 in epithelial-driven host defense by up-regulating antimicrobials and define a novel epithelial receptor for CC16, VLA-2, through which signaling is necessary for enhanced SPLUNC1 production.
    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
    患有囊性纤维化(pwCF)的人患有慢性和复发性细菌肺部感染,这种感染在生命的早期就开始并导致进行性肺衰竭。CF是由CF跨膜传导调节因子(CFTR)基因突变引起的,其编码对于维持肺表面的适当水合作用重要的离子通道。当CFTR功能消融或受损时,气道增厚,粘附粘液,导致感染和炎症的恶性循环。pwCF的治疗学,称为CFTR调制器,直接针对CFTR缺陷,恢复气道表面水化和粘膜纤毛清除。然而,即使使用CFTR调节剂治疗,细菌感染持续存在。建立病变气道上皮的相关模型,我们建立了持续铜绿假单胞菌感染的人气道上皮细胞培养系统。我们使用这个模型来检查CFTR调节剂对CFTR成熟的影响,CFTR函数,和细菌的持久性。我们发现铜绿假单胞菌的存在增加了CFTRmRNA,蛋白质,和功能。我们还发现CFTR调节剂引起铜绿假单胞菌负荷的降低。这些结果表明,包括活细菌对CF肺准确建模的重要性,了解感染对CFTR调节剂挽救CFTR的影响对于评估和优化所有pwCF的药物治疗至关重要。
    People with cystic fibrosis (pwCF) suffer from chronic and recurring bacterial lung infections that begin very early in life and contribute to progressive lung failure. CF is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, which encodes an ion channel important for maintaining the proper hydration of pulmonary surfaces. When CFTR function is ablated or impaired, airways develop thickened, adherent mucus that contributes to a vicious cycle of infection and inflammation. Therapeutics for pwCF, called CFTR modulators, target the CFTR defect directly, restoring airway surface hydration and mucociliary clearance. However, even with CFTR modulator therapy, bacterial infections persist. To develop a relevant model of diseased airway epithelium, we established a primary human airway epithelium culture system with persistent Pseudomonas aeruginosa infection. We used this model to examine the effects of CFTR modulators on CFTR maturation, CFTR function, and bacterial persistence. We found that the presence of P. aeruginosa increased CFTR mRNA, protein, and function. We also found that CFTR modulators caused a decrease in P. aeruginosa burden. These results demonstrate the importance of including live bacteria to accurately model the CF lung, and that understanding the effects of infection on CFTR rescue by CFTR modulators is critical to evaluating and optimizing drug therapies for all pwCF.
    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
    酒精使用障碍(AUD)是一个重要的公共卫生问题,患有AUD的人更有可能因呼吸道感染而发展为严重的急性呼吸窘迫综合征(ARDS)。为了检查AUD是否是SARS-CoV-2感染更严重结局的危险因素,我们使用培养的分化支气管上皮细胞检查了对感染的早期反应,这些细胞来自AUD患者或无AUD患者的刷洗。未感染细胞的RNA-seq分析确定,与非AUD细胞相比,针对表皮基因的表达富集了AUD细胞。AUD患者支气管上皮细胞在感染后72小时显示屏障功能显著下降,由跨上皮电阻确定。相比之下,SARS-CoV-2感染后72小时,非AUD细胞的屏障功能增强。AUD细胞显示claudin-7未与小带闭塞1共定位,表明紧密连接混乱。然而,SARS-CoV-2感染后,AUD和非AUD细胞均显示β-catenin表达降低。为了确定AUD对SARS-CoV-2感染的炎症反应的影响,通过多重分析测量细胞因子分泌。SARS-CoV-2感染的AUD支气管细胞分泌多种促炎细胞因子,包括TNFα,IL-1β,和IFNγ,而不是非AUD细胞。相比之下,非AUD支气管细胞的屏障保护性细胞因子EGF和GM-CSF的分泌增强。一起来看,这些数据支持以下假设:AUD是COVID-19的一个危险因素,在COVID-19中,酒精引发气道上皮细胞增加的炎症和屏障功能障碍,以响应SARS-CoV-2的感染.
    Alcohol use disorder (AUD) is a significant public health concern and people with AUD are more likely to develop severe acute respiratory distress syndrome (ARDS) in response to respiratory infections. To examine whether AUD was a risk factor for more severe outcome in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we examined early responses to infection using cultured differentiated bronchial epithelial cells derived from brushings obtained from people with AUD or without AUD. RNA-seq analysis of uninfected cells determined that AUD cells were enriched for expression of epidermal genes as compared with non-AUD cells. Bronchial epithelial cells from patients with AUD showed a significant decrease in barrier function 72 h postinfection, as determined by transepithelial electrical resistance. In contrast, barrier function of non-AUD cells was enhanced 72 h after SARS-CoV-2 infection. AUD cells showed claudin-7 that did not colocalize with zonula occludens-1 (ZO-1), indicative of disorganized tight junctions. However, both AUD and non-AUD cells showed decreased β-catenin expression following SARS-CoV-2 infection. To determine the impact of AUD on the inflammatory response to SARS-CoV-2 infection, cytokine secretion was measured by multiplex analysis. SARS-CoV-2-infected AUD bronchial cells had enhanced secretion of multiple proinflammatory cytokines including TNFα, IL-1β, and IFNγ as opposed to non-AUD cells. In contrast, secretion of the barrier-protective cytokines epidermal growth factor (EGF) and granulocyte macrophage-colony stimulating factor (GM-CSF) was enhanced for non-AUD bronchial cells. Taken together, these data support the hypothesis that AUD is a risk factor for COVID-19, where alcohol primes airway epithelial cells for increased inflammation and increased barrier dysfunction and increased inflammation in response to infection by SARS-CoV-2.NEW & NOTEWORTHY Alcohol use disorder (AUD) is a significant risk factor for severe acute respiratory distress syndrome. We found that AUD causes a phenotypic shift in gene expression in human bronchial epithelial cells, enhancing expression of epidermal genes. AUD cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had higher levels of proinflammatory cytokine secretion and barrier dysfunction not present in infected non-AUD cells, consistent with increased early COVID-19 severity due to AUD.
    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
    鼻腔被呼吸道上皮覆盖,包括纤毛细胞,消除粘液中的外来物质。在遗传性疾病,如原发性纤毛运动障碍和囊性纤维化,呼吸道上皮功能不可逆地受损;然而,尚未建立根治性治疗。因此,我们认为,将正常气道上皮移植到鼻腔上皮是未来可能导致根治性治疗的策略之一。在我们之前的研究中,将玻璃胶膜上的人诱导多能干细胞来源的气道上皮(hiPSC-AE)移植到裸大鼠鼻中隔粘膜的刮削区域。虽然人类来源的纤毛细胞,俱乐部细胞,观察到基底细胞,它们位于粘膜下肉芽组织的囊肿中,但不位于鼻粘膜上皮中,移植的细胞可能对这种情况下的鼻粘膜功能没有贡献。因此,为了实现更多的功能移植,在这项研究中,我们通过将hiPSC-AE中的胶原海绵包裹在玻璃化胶膜上来制备不同的移植物。因此,我们发现移植的细胞在鼻粘膜上皮中存活。这些结果表明,移植到鼻腔中的hiPSC-AE可以在鼻粘膜中存活。此外,我们的方法导致建立鼻粘膜人源化大鼠,用于开发药物和治疗鼻呼吸道上皮遗传性疾病的方法。
    The nasal cavity is covered with respiratory epithelia, including ciliated cells that eliminate foreign substances trapped in the mucus. In hereditary diseases such as primary ciliary dyskinesia and cystic fibrosis, respiratory epithelial functions are irreversibly impaired; however, no radical treatment has been established yet. Thus, we considered that the transplantation of normal airway epithelia (AE) into the nasal epithelia is one of the strategies that could lead to radical treatment in the future. In our previous study, human induced pluripotent stem cell-derived AE (hiPSC-AE) on the vitrigel membrane were transplanted into the scraped area of the nasal septal mucosa of nude rats. Although human-derived ciliated cells, club cells, and basal cells were observed, they were located in the cysts within the submucosal granulation tissue but not in the nasal mucosal epithelia and the transplanted cells may not contribute to the function of the nasal mucosa with this condition. Therefore, to achieve more functional transplantation, we prepared the graft differently in this study by wrapping the collagen sponge in hiPSC-AE on the vitrigel membrane. As a result, we found the transplanted cells surviving in the nasal mucosal epithelia. These results suggest that hiPSC-AE transplanted into the nasal cavity could be viable in the nasal mucosa. In addition, our method leads to the establishment of nasal mucosa-humanized rats that are used for the development of the drugs and therapeutic methods for hereditary diseases of nasal respiratory epithelia.
    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
    背景:慢性肺部疾病如哮喘和COPD会增加中东呼吸综合征冠状病毒(MERS-CoV)感染期间的发病率和死亡率。我们假设有这种合并症的个体由于其受体表达增加而更容易感染MERS-CoV,二肽基肽酶4(DPP4)。
    方法:我们通过用Th2细胞因子IL-13治疗原发性人气道上皮来模拟慢性气道疾病,检查其如何影响DPP4蛋白水平以及MERS-CoV进入和复制。
    结果:IL-13暴露3天导致DPP4蛋白丰度增加,而21天的治疗会增加DPP4水平并导致杯状细胞化生。令人惊讶的是,尽管受体可用性增加,IL-13治疗对MERS-CoV进入和复制没有显著影响。
    结论:我们的结果表明,在Th2炎症的情况下,DPP4丰度增加可能不是导致MERS严重程度增加的主要机制。转录谱分析强调了IL-13诱导的气道上皮变化的复杂性,包括先天免疫相关基因表达的改变,抗病毒反应,和维持细胞外粘液屏障。这些数据表明,其他因素可能与DPP4丰度相互作用,以确定MERS-CoV感染结果。
    BACKGROUND: Chronic pulmonary conditions such as asthma and chronic obstructive pulmonary disease increase the risk of morbidity and mortality during infection with the Middle East respiratory syndrome coronavirus (MERS-CoV). We hypothesized that individuals with such comorbidities are more susceptible to MERS-CoV infection due to increased expression of its receptor, dipeptidyl peptidase 4 (DPP4).
    METHODS: We modeled chronic airway disease by treating primary human airway epithelia with the Th2 cytokine interleukin 13 (IL-13), examining how this affected DPP4 protein levels with MERS-CoV entry and replication.
    RESULTS: IL-13 exposure for 3 days led to greater DPP4 protein abundance, while a 21-day treatment raised DPP4 levels and caused goblet cell metaplasia. Surprisingly, despite this increase in receptor availability, MERS-CoV entry and replication were not significantly affected by IL-13 treatment.
    CONCLUSIONS: Our results suggest that greater DPP4 abundance is likely not the primary mechanism leading to increased MERS severity in the setting of Th2 inflammation. Transcriptional profiling analysis highlighted the complexity of IL-13-induced changes in airway epithelia, including altered expression of genes involved in innate immunity, antiviral responses, and maintenance of the extracellular mucus barrier. These data suggest that additional factors likely interact with DPP4 abundance to determine MERS-CoV infection outcomes.
    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
    基因编辑策略对于治疗遗传性肺部疾病如囊性纤维化(CF)具有吸引力。然而,挑战包括开发用于气道上皮基因编辑的安全有效的载体系统和报告其效率和耐久性的模型系统。家雪貂(Mustelaputoriusfuro)在人类的肺细胞解剖学中具有高度的保守性,并为许多类型的肺部疾病提供了极好的模型,包括CF。在这项研究中,我们使用SpCas9和AsCas12a(Cpf1)核糖核蛋白(RNP)评估了两亲性穿梭肽S10用于蛋白质递送和基因编辑的效率.这些方法在增殖雪貂气道基底细胞中进行了评估,体外极化气道上皮,和体内的肺,通过使用报告雪貂访问编辑效率并测量雪貂CFTR基因座处的indel。我们的结果表明,穿梭肽可以有效地将报告蛋白/肽和基因编辑SpCas9或Cpf1RNP复合物在体外和体内递送到雪貂气道上皮细胞。我们在体外测量了绿色荧光蛋白(GFP)-核定位信号(NLS)蛋白或SpCas9RNP进入雪貂气道基底细胞和完全分化的纤毛和非纤毛上皮细胞的S10递送效率。使用转基因原代细胞和雪貂,通过Cas/LoxP-gRNARNP介导的ROSA-TGCre重组酶报告基因转化来确定体外和体内基因编辑效率。S10/Cas9RNP更有效,相对于S10/Cpf1RNP在ROSA-TG基因座的基因编辑。与GFP-NLS蛋白或D-Retro-Inverso(DRI)-NLS肽结合的S10穿梭的气管内肺部递送显示蛋白质递送的效率约为3倍或14倍,分别,使用S10/Cas9/LoxP-gRNA在ROSA-TG基因座处进行基因编辑的效率。Cpf1RNP在LoxP基因座的基因编辑方面不如SpCas9有效。这些数据证明了将CasRNP穿梭肽递送到雪貂气道的可行性,以及开发用于遗传性肺病(例如CF)的基于离体干细胞和体内基因编辑疗法的潜在效用。
    Gene editing strategies are attractive for treating genetic pulmonary diseases such as cystic fibrosis (CF). However, challenges have included the development of safe and effective vector systems for gene editing of airway epithelia and model systems to report their efficiency and durability. The domestic ferret (Mustela putorius furo) has a high degree of conservation in lung cellular anatomy with humans, and has served as an excellent model for many types of lung diseases, including CF. In this study, we evaluated the efficiency of amphiphilic shuttle peptide S10 for protein delivery and gene editing using SpCas9, and AsCas12a (Cpf1) ribonucleoproteins (RNPs). These approaches were evaluated in proliferating ferret airway basal cells, polarized airway epithelia in vitro, and lungs in vivo, by accessing the editing efficiency using reporter ferrets and measuring indels at the ferret CFTR locus. Our results demonstrate that shuttle peptides efficiently enable delivery of reporter proteins/peptides and gene editing SpCas9 or Cpf1 RNP complexes to ferret airway epithelial cells in vitro and in vivo. We measured S10 delivery efficiency of green fluorescent protein (GFP)-nuclear localization signal (NLS) protein or SpCas9 RNP into ferret airway basal cells and fully differentiated ciliated and nonciliated epithelial cells in vitro. In vitro and in vivo gene editing efficiencies were determined by Cas/LoxP-gRNA RNP-mediated conversion of a ROSA-TG Cre recombinase reporter using transgenic primary cells and ferrets. S10/Cas9 RNP was more effective, relative to S10/Cpf1 RNP at gene editing of the ROSA-TG locus. Intratracheal lung delivery of the S10 shuttle combined with GFP-NLS protein or D-Retro-Inverso (DRI)-NLS peptide demonstrated efficiencies of protein delivery that were ∼3-fold or 14-fold greater, respectively, than the efficiency of gene editing at the ROSA-TG locus using S10/Cas9/LoxP-gRNA. Cpf1 RNPs was less effective than SpCas9 at gene editing of LoxP locus. These data demonstrate the feasibility of shuttle peptide delivery of Cas RNPs to the ferret airways and the potential utility for developing ex vivo stem cell-based and in vivo gene editing therapies for genetic pulmonary diseases such as CF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号