首页 > 最新文献

American journal of physiology. Lung cellular and molecular physiology最新文献

英文 中文
Mucociliary clearance is impaired in small airways of cystic fibrosis pigs. 囊性纤维化猪小气道的黏膜纤毛清除能力受损
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-06 DOI: 10.1152/ajplung.00010.2024
Carley G Stewart, Brieanna M Hilkin, Nicholas D Gansemer, Ryan J Adam, David W Dick, John J Sunderland, David A Stoltz, Joseph Zabner, Mahmoud H Abou Alaiwa

Cystic fibrosis (CF) is a genetic disorder characterized by recurrent airway infections, inflammation, impaired mucociliary clearance, and progressive decline in lung function. The disease may start in the small airways; however, this is difficult to prove due to the limited accessibility of the small airways with the current single-photon mucociliary clearance assay. Here, we developed a dynamic positron emission tomography assay with high spatial and temporal resolution. We tested that mucociliary clearance is abnormal in the small airways of newborn cystic fibrosis pigs. Clearance of [68Ga]-tagged macroaggregated albumin from small airways started immediately after delivery and continued for the duration of the study. Initial clearance was fast but slowed down a few minutes after delivery. Cystic fibrosis pigs' small airways cleared significantly less than non-CF pigs' small airways (non-CF 25.1 ± 3.1% vs. CF 14.6 ± 0.1%). Stimulation of the cystic fibrosis airways with the purinergic secretagogue uridine-5'-triphosphate (UTP) further impaired clearance (non-CF with UTP 20.9 ± 0.3% vs. CF with UTP 13.0 ± 1.8%). None of the cystic fibrosis pigs treated with UTP (n = 6) cleared more than 20% of the delivered dose. These data indicate that mucociliary clearance in the small airways is fast and can easily be missed if the assay is not sensitive enough. The data also indicate that mucociliary clearance is impaired in the small airways of cystic fibrosis pigs. This defect is exacerbated by stimulation of mucus secretions with purinergic agonists.NEW & NOTEWORTHY We developed a novel positron emission tomography scan assay with unprecedented temporal and spatial resolution to measure mucociliary clearance in the small airways. We proved a long-standing but unproven assertion that mucociliary clearance is inherently abnormal in the small airways of newborn cystic fibrosis piglets that are otherwise free of infection or inflammation. This technique can be easily extended to other airway diseases such as asthma, idiopathic pulmonary fibrosis, or chronic obstructive pulmonary disease.

囊性纤维化是一种遗传性疾病,其特点是气道反复感染、炎症、粘液纤毛清除能力受损以及肺功能逐渐下降。这种疾病可能从小气道开始;然而,由于目前的单光子粘液纤毛清除测定对小气道的可及性有限,因此很难证明这一点。在这里,我们开发了一种具有高空间和时间分辨率的动态正电子发射断层扫描检测方法。我们测试了新生囊性纤维化猪小气道中的粘液纤毛清除率是否异常。小气道中[68Ga]标记的大聚集白蛋白的清除在分娩后立即开始,并持续整个研究过程。最初的清除速度很快,但在分娩几分钟后就减慢了。囊性纤维化猪小气道的清除率明显低于非囊性纤维化猪小气道(非囊性纤维化猪 25.1±3.1% vs. 囊性纤维化猪 14.6±0.1%)。用嘌呤能分泌物UTP刺激囊性纤维化气道会进一步降低清除率(使用UTP的非囊性纤维化猪为20.9±0.3%,使用UTP的囊性纤维化猪为13.0±1.8%)。使用UTP治疗的囊性纤维化猪(N = 6)的清除率均未超过给药剂量的20%。这些数据表明,小气道中的粘膜纤毛清除速度很快,如果检测灵敏度不够,很容易被漏掉。数据还表明,囊性纤维化猪小气道中的粘液纤毛清除能力受损。嘌呤能激动剂刺激粘液分泌会加剧这种缺陷。
{"title":"Mucociliary clearance is impaired in small airways of cystic fibrosis pigs.","authors":"Carley G Stewart, Brieanna M Hilkin, Nicholas D Gansemer, Ryan J Adam, David W Dick, John J Sunderland, David A Stoltz, Joseph Zabner, Mahmoud H Abou Alaiwa","doi":"10.1152/ajplung.00010.2024","DOIUrl":"10.1152/ajplung.00010.2024","url":null,"abstract":"<p><p>Cystic fibrosis (CF) is a genetic disorder characterized by recurrent airway infections, inflammation, impaired mucociliary clearance, and progressive decline in lung function. The disease may start in the small airways; however, this is difficult to prove due to the limited accessibility of the small airways with the current single-photon mucociliary clearance assay. Here, we developed a dynamic positron emission tomography assay with high spatial and temporal resolution. We tested that mucociliary clearance is abnormal in the small airways of newborn cystic fibrosis pigs. Clearance of [<sup>68</sup>Ga]-tagged macroaggregated albumin from small airways started immediately after delivery and continued for the duration of the study. Initial clearance was fast but slowed down a few minutes after delivery. Cystic fibrosis pigs' small airways cleared significantly less than non-CF pigs' small airways (non-CF 25.1 ± 3.1% vs. CF 14.6 ± 0.1%). Stimulation of the cystic fibrosis airways with the purinergic secretagogue uridine-5'-triphosphate (UTP) further impaired clearance (non-CF with UTP 20.9 ± 0.3% vs. CF with UTP 13.0 ± 1.8%). None of the cystic fibrosis pigs treated with UTP (<i>n</i> = 6) cleared more than 20% of the delivered dose. These data indicate that mucociliary clearance in the small airways is fast and can easily be missed if the assay is not sensitive enough. The data also indicate that mucociliary clearance is impaired in the small airways of cystic fibrosis pigs. This defect is exacerbated by stimulation of mucus secretions with purinergic agonists.<b>NEW & NOTEWORTHY</b> We developed a novel positron emission tomography scan assay with unprecedented temporal and spatial resolution to measure mucociliary clearance in the small airways. We proved a long-standing but unproven assertion that mucociliary clearance is inherently abnormal in the small airways of newborn cystic fibrosis piglets that are otherwise free of infection or inflammation. This technique can be easily extended to other airway diseases such as asthma, idiopathic pulmonary fibrosis, or chronic obstructive pulmonary disease.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892696","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The pontine Kölliker-Fuse nucleus is important for reduced postinspiratory airflow elicited by stimulation of the ventral respiratory parafacial region. 脑桥 Kölliker-Fuse 核对刺激腹侧呼吸面旁区引起的吸气后气流减少非常重要。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-06 DOI: 10.1152/ajplung.00155.2024
Karine C Flor, Octavio A C Maia, Ana C Takakura, Thiago S Moreira

Considering that the retrotrapezoid nucleus/respiratory parafacial region (RTN/pFRG) would be an important center in the central nervous system involved in the maintenance and modulation of respiratory activity, we hypothesized that neurons in this nucleus would also be involved in the postinspiratory (post-I) phase of the respiratory cycle through a connection with the pontine Kölliker-Fuse (KF) region. Here, we performed pharmacogenetic manipulation (AAV-hM3D(Gq)-mCherry or AAV-hM4D(Gi)-mCherry) in VGlut2-cre, Ai6 conscious mice to evaluate breathing parameters through whole body plethysmography under baseline conditions (normoxia: [Formula: see text] = 0.21) or under hypercapnia or hypoxia challenges ([Formula: see text] = 0.07 or [Formula: see text] = 0.08). Under normoxia, selective stimulation of RTN/pFRG resulted in a smaller increase in V̇e (1,272 ± 102.5, vs. RTN/pFRG stimulation: 1,878 ± 122.1 mL/kg/min), due to a smaller increase in VT (5.4 ± 0.35, vs. RTN/pFRG stimulation: 7.77 ± 0.21 mL/kg) without changing fR in a condition of KF inhibition. However, inhibition of the VGlut2 neurons in the KF did affect the TE1 produced by selective activation of RTN/pFRG (119.9 ± 2.53, vs. RTN/pFRG stimulation: 104 ± 2.46 ms). Both the hypercapnia and hypoxia ventilatory response were reduced after inhibition of VGlut2-expressing KF neurons. Therefore, consistent with anatomical projections RTN/pFRG neurons regulate lung ventilation by controlling all aspects of breathing, i.e., breathing frequency, inspiration, postinspiration, and active expiration. All the modulation seems to be dependent on the integrity of the glutamatergic neurons in the KF region.NEW & NOTEWORTHY Our research reveals specific roles and interactions between the retrotrapezoid nucleus/respiratory parafacial region (RTN/pFRG) and the pontine Kölliker-Fuse (KF) region in controlling respiratory phases. RTN/pFRG neurons are key in regulating all aspects of breathing, including frequency, inspiration, postinspiration, and active expiration. This regulation depends on the functional integrity of glutamatergic neurons in the KF region, aligning with anatomical projections.

考虑到蛛网膜后核/呼吸面旁区(RTN/pFRG)是中枢神经系统中参与维持和调节呼吸活动的一个重要中枢,我们假设该核中的神经元也会通过与桥脑柯利克-傅斯(KF)区的连接参与呼吸周期的后吸气阶段。在此,我们对 VGlut2-cre、Ai6 意识清醒的小鼠进行了药物遗传学操作(AAV-hM3D(Gq)-mCherry 或 AAV-hM4D(Gi)-mCherry),在基线条件(常氧:FiO2 = 0.21)或高碳酸血症或低氧挑战(FiCO2 = 0.07 或 FiO2 = 0.08)下通过全身胸透评估呼吸参数。在常氧状态下,选择性刺激 RTN/pFRG 导致 VE 增加较少(1,272 ± 102.5,与 RTN/pFRG 刺激相比:1,878 ± 122.1 毫升/千克/分钟),这是因为在 KF 抑制条件下 VT 增加较少(5.4 ± 0.35,与 RTN/pFRG 刺激相比:7.77 ± 0.21 毫升/千克),而 fR 不变。然而,抑制 KF 中的 VGlut2 神经元确实会影响 RTN/pFRG 选择性激活产生的 TE1(119.9 ± 2.53,对比:RTN/pFRG 刺激:104 ± 2.46 ms)。抑制 VGlut2 表达的 KF 神经元后,高碳酸血症和低氧通气反应均减弱。因此,与解剖投射一致,RTN/pFRG 神经元通过控制呼吸的各个方面(即呼吸频率、吸气、吸气后和主动呼气)来调节肺通气。所有这些调节似乎都取决于 KF 区域谷氨酸能神经元的完整性。
{"title":"The pontine Kölliker-Fuse nucleus is important for reduced postinspiratory airflow elicited by stimulation of the ventral respiratory parafacial region.","authors":"Karine C Flor, Octavio A C Maia, Ana C Takakura, Thiago S Moreira","doi":"10.1152/ajplung.00155.2024","DOIUrl":"10.1152/ajplung.00155.2024","url":null,"abstract":"<p><p>Considering that the retrotrapezoid nucleus/respiratory parafacial region (RTN/pFRG) would be an important center in the central nervous system involved in the maintenance and modulation of respiratory activity, we hypothesized that neurons in this nucleus would also be involved in the postinspiratory (post-I) phase of the respiratory cycle through a connection with the pontine Kölliker-Fuse (KF) region. Here, we performed pharmacogenetic manipulation (AAV-hM3D(Gq)-mCherry or AAV-hM4D(Gi)-mCherry) in VGlut2-cre, Ai6 conscious mice to evaluate breathing parameters through whole body plethysmography under baseline conditions (normoxia: [Formula: see text] = 0.21) or under hypercapnia or hypoxia challenges ([Formula: see text] = 0.07 or [Formula: see text] = 0.08). Under normoxia, selective stimulation of RTN/pFRG resulted in a smaller increase in V̇e (1,272 ± 102.5, vs. RTN/pFRG stimulation: 1,878 ± 122.1 mL/kg/min), due to a smaller increase in V<sub>T</sub> (5.4 ± 0.35, vs. RTN/pFRG stimulation: 7.77 ± 0.21 mL/kg) without changing <i>f</i><sub>R</sub> in a condition of KF inhibition. However, inhibition of the VGlut2 neurons in the KF did affect the <i>T</i><sub>E1</sub> produced by selective activation of RTN/pFRG (119.9 ± 2.53, vs. RTN/pFRG stimulation: 104 ± 2.46 ms). Both the hypercapnia and hypoxia ventilatory response were reduced after inhibition of VGlut2-expressing KF neurons. Therefore, consistent with anatomical projections RTN/pFRG neurons regulate lung ventilation by controlling all aspects of breathing, i.e., breathing frequency, inspiration, postinspiration, and active expiration. All the modulation seems to be dependent on the integrity of the glutamatergic neurons in the KF region.<b>NEW & NOTEWORTHY</b> Our research reveals specific roles and interactions between the retrotrapezoid nucleus/respiratory parafacial region (RTN/pFRG) and the pontine Kölliker-Fuse (KF) region in controlling respiratory phases. RTN/pFRG neurons are key in regulating all aspects of breathing, including frequency, inspiration, postinspiration, and active expiration. This regulation depends on the functional integrity of glutamatergic neurons in the KF region, aligning with anatomical projections.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892698","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effect of physical activity in lymphocytes senescence burden in patients with COPD. 体育锻炼对慢性阻塞性肺病患者淋巴细胞衰老负担的影响
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-06 DOI: 10.1152/ajplung.00151.2024
Enrique Alfaro, Elena Díaz-García, Sara García-Tovar, Raúl Galera, Raquel Casitas, Elisabet Martínez-Cerón, María Torres-Vargas, José M Padilla, Cristina López-Fernández, Paula Pérez-Moreno, Francisco García-Río, Carolina Cubillos-Zapata

Chronic obstructive pulmonary disease (COPD) is regarded as an accelerated-age disease in which chronic inflammation, maladaptive immune responses, and senescence cell burden coexist. Accordingly, cellular senescence has emerged as a potential mechanism involved in COPD pathophysiology. In this study, 25 stable patients with COPD underwent a daily physical activity promotion program for 6 mo. We reported that increase of physical activity was related to a reduction of the senescent cell burden in circulating lymphocytes of patients with COPD. Senescent T-lymphocyte population, characterized by absence of surface expression of CD28, was reduced after physical activity intervention, and the reduction was associated to the increase of physical activity level. In addition, the mRNA expression of cyclin-dependent kinase inhibitors, a hallmark of cell senescence, was reduced and, in accordance, the proliferative capacity of lymphocytes was improved postintervention. Moreover, we observed an increase in functionality in T cells from patients after intervention, including improved markers of activation, enhanced cytotoxicity, and altered cytokine secretions in response to viral challenge. Lastly, physical activity intervention reduced the potential of lymphocytes' secretome to induce senescence in human primary fibroblasts. In conclusion, our study provides, for the first time, evidence of the potential of physical activity intervention in patients with COPD to reduce the senescent burden in circulating immune cells.NEW & NOTEWORTHY For the first time, we identified in patients with COPD a relation between physical activity intervention with respiratory function improvement and cellular senescence burden in lymphocytes that improved the T cell functionality and proliferative capacity of patients. In addition, our experiments highlight the possible impact of T-cell senescence in other cell types which could be related to some of the clinical lung complications observed in COPD.

慢性阻塞性肺病(COPD)被认为是一种加速衰老的疾病,其中慢性炎症、不适应性免疫反应和衰老细胞负担并存。因此,细胞衰老已成为慢性阻塞性肺病病理生理学的一个潜在机制。在这项研究中,25 名病情稳定的慢性阻塞性肺病患者接受了为期 6 个月的日常体育锻炼促进计划。我们发现,体力活动的增加与慢性阻塞性肺病患者循环淋巴细胞中衰老细胞负担的减少有关。体力活动干预后,以表面不表达 CD28 为特征的衰老 T 淋巴细胞数量减少了,而且这种减少与体力活动水平的增加有关。此外,作为细胞衰老标志的细胞周期蛋白依赖性激酶抑制剂的 mRNA 表达量也有所减少,因此,干预后淋巴细胞的增殖能力也有所提高。此外,我们还观察到患者的 T 细胞在干预后功能增强,包括活化标志物改善、细胞毒性增强以及细胞因子分泌对病毒挑战的反应发生改变。最后,体育锻炼干预降低了淋巴细胞分泌组诱导人类原代成纤维细胞衰老的潜力。总之,我们的研究首次提供了证据,证明对慢性阻塞性肺病患者进行体育锻炼干预有可能减少循环免疫细胞的衰老负担。
{"title":"Effect of physical activity in lymphocytes senescence burden in patients with COPD.","authors":"Enrique Alfaro, Elena Díaz-García, Sara García-Tovar, Raúl Galera, Raquel Casitas, Elisabet Martínez-Cerón, María Torres-Vargas, José M Padilla, Cristina López-Fernández, Paula Pérez-Moreno, Francisco García-Río, Carolina Cubillos-Zapata","doi":"10.1152/ajplung.00151.2024","DOIUrl":"10.1152/ajplung.00151.2024","url":null,"abstract":"<p><p>Chronic obstructive pulmonary disease (COPD) is regarded as an accelerated-age disease in which chronic inflammation, maladaptive immune responses, and senescence cell burden coexist. Accordingly, cellular senescence has emerged as a potential mechanism involved in COPD pathophysiology. In this study, 25 stable patients with COPD underwent a daily physical activity promotion program for 6 mo. We reported that increase of physical activity was related to a reduction of the senescent cell burden in circulating lymphocytes of patients with COPD. Senescent T-lymphocyte population, characterized by absence of surface expression of CD28, was reduced after physical activity intervention, and the reduction was associated to the increase of physical activity level. In addition, the mRNA expression of cyclin-dependent kinase inhibitors, a hallmark of cell senescence, was reduced and, in accordance, the proliferative capacity of lymphocytes was improved postintervention. Moreover, we observed an increase in functionality in T cells from patients after intervention, including improved markers of activation, enhanced cytotoxicity, and altered cytokine secretions in response to viral challenge. Lastly, physical activity intervention reduced the potential of lymphocytes' secretome to induce senescence in human primary fibroblasts. In conclusion, our study provides, for the first time, evidence of the potential of physical activity intervention in patients with COPD to reduce the senescent burden in circulating immune cells.<b>NEW & NOTEWORTHY</b> For the first time, we identified in patients with COPD a relation between physical activity intervention with respiratory function improvement and cellular senescence burden in lymphocytes that improved the T cell functionality and proliferative capacity of patients. In addition, our experiments highlight the possible impact of T-cell senescence in other cell types which could be related to some of the clinical lung complications observed in COPD.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chronic hyperglycemia aggravates lung function in a Scnn1b-Tg murine model. 慢性高血糖会加重 Scnn1b-Tg 小鼠模型的肺功能。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-07-16 DOI: 10.1152/ajplung.00279.2023
Guiying Cui, Dina A Moustafa, Shilin Zhao, Analia Vazquez Cegla, James T Lyles, Joanna B Goldberg, Joshua D Chandler, Nael A McCarty

Cystic fibrosis-related diabetes (CFRD), the most common comorbidity in cystic fibrosis (CF), leads to increased mortality by accelerating the decline in lung function. Scnn1b-Tg transgenic mice overexpressing the epithelial sodium channel β subunit exhibit spontaneous CF-like lung disease, including airway mucus obstruction and chronic inflammation. Here, we established a chronic CFRD-like model using Scnn1b-Tg mice made diabetic by injection of streptozotocin (STZ). In Ussing chamber recordings of the trachea, Scnn1b-Tg mice exhibited larger amiloride-sensitive currents and forskolin-activated currents, without a difference in adenosine triphosphate (ATP)-activated currents compared with wild-type (WT) littermates. Both diabetic WT (WT-D) and diabetic Scnn1b-Tg (Scnn1b-Tg-D) mice on the same genetic background exhibited substantially elevated blood glucose at 8 wk; glucose levels also were elevated in bronchoalveolar lavage fluid (BALF). Bulk lung RNA-seq data showed significant differences between WT-D and Scnn1b-Tg-D mice. Neutrophil counts in BALF were substantially increased in Scnn1b-Tg-D lungs compared with controls (Scnn1b-Tg-con) and compared with WT-D lungs. Lung histology data showed enhanced parenchymal destruction, alveolar wall thickening, and neutrophilic infiltration in Scnn1b-Tg-D mice compared with WT-D mice, consistent with the development of a spontaneous lung infection. We intranasally administered Pseudomonas aeruginosa to induce lung infection in these mice for 24 h, which led to severe lung leukocytic infiltration and an increase in pro-inflammatory cytokine levels in the BALF. In summary, we established a chronic CFRD-like lung mouse model using the Scnn1b-Tg mice. The model can be used for future studies toward understanding the mechanisms underlying the lung pathophysiology associated with CFRD and developing novel therapeutics.NEW & NOTEWORTHY We established a chronic CFRD-like mouse model using the Scnn1b-Tg transgenic mice overexpressing the epithelial sodium channel β subunit made diabetic by injection of streptozotocin. The results underscore the urgent need to develop novel therapeutics for CF lung disease.

囊性纤维化相关糖尿病(CFRD)是囊性纤维化(CF)中最常见的并发症,它通过加速肺功能衰退而导致死亡率上升。过表达上皮钠通道β亚基的Scnn1b-Tg转基因小鼠表现出自发性CFR样肺病,包括气道粘液阻塞和慢性炎症。在这里,我们利用通过注射链脲佐菌素使小鼠患上糖尿病的 Scnn1b-Tg 小鼠建立了慢性 CFRD 样模型。在气管乌星室记录中,Scnn1b-Tg 小鼠表现出更大的阿米洛利敏感电流和福斯可林激活电流,与野生型(WT)小鼠相比,ATP 激活电流没有差异。具有相同遗传背景的糖尿病 WT(WT-D)小鼠和糖尿病 Scnn1b-Tg (Scnn1b-Tg-D)小鼠在 8 周时都表现出血糖大幅升高;支气管肺泡灌洗液(BALF)中的葡萄糖水平也升高 Bulk 肺 RNA-seq 数据显示 WT-D 和 Scnn1b-Tg-D 小鼠之间存在显著差异。与对照组(Scnn1b-Tg-con)和 WT-D 小鼠肺部相比,Scnn1b-Tg-D 肺部 BALF 中的中性粒细胞数量大幅增加。肺组织学数据显示,与 WT-D 小鼠相比,Scnn1b-Tg-D 小鼠的肺实质破坏、肺泡壁增厚和中性粒细胞浸润均有所增强,这与自发性肺部感染的发生是一致的。我们通过鼻内注射铜绿假单胞菌诱导这些小鼠肺部感染 24 小时,这导致了严重的肺部白细胞浸润和 BALF 中促炎细胞因子水平的升高。总之,我们利用 Scnn1b-Tg 小鼠建立了慢性 CFRD 样肺小鼠模型。该模型可用于未来的研究,以了解与 CFRD 相关的肺部病理生理学机制并开发新型疗法。
{"title":"Chronic hyperglycemia aggravates lung function in a <i>Scnn1b</i>-Tg murine model.","authors":"Guiying Cui, Dina A Moustafa, Shilin Zhao, Analia Vazquez Cegla, James T Lyles, Joanna B Goldberg, Joshua D Chandler, Nael A McCarty","doi":"10.1152/ajplung.00279.2023","DOIUrl":"10.1152/ajplung.00279.2023","url":null,"abstract":"<p><p>Cystic fibrosis-related diabetes (CFRD), the most common comorbidity in cystic fibrosis (CF), leads to increased mortality by accelerating the decline in lung function. <i>Scnn1b-</i>Tg transgenic mice overexpressing the epithelial sodium channel β subunit exhibit spontaneous CF-like lung disease, including airway mucus obstruction and chronic inflammation. Here, we established a chronic CFRD-like model using <i>Scnn1b-</i>Tg mice made diabetic by injection of streptozotocin (STZ). In Ussing chamber recordings of the trachea, <i>Scnn1b-</i>Tg mice exhibited larger amiloride-sensitive currents and forskolin-activated currents, without a difference in adenosine triphosphate (ATP)-activated currents compared with wild-type (WT) littermates. Both diabetic WT (WT-D) and diabetic <i>Scnn1b-</i>Tg (<i>Scnn1b</i>-Tg-D) mice on the same genetic background exhibited substantially elevated blood glucose at 8 wk; glucose levels also were elevated in bronchoalveolar lavage fluid (BALF). Bulk lung RNA-seq data showed significant differences between WT-D and <i>Scnn1b</i>-Tg-D mice. Neutrophil counts in BALF were substantially increased in <i>Scnn1b-</i>Tg-D lungs compared with controls (<i>Scnn1b</i>-Tg-con) and compared with WT-D lungs. Lung histology data showed enhanced parenchymal destruction, alveolar wall thickening, and neutrophilic infiltration in <i>Scnn1b-</i>Tg-D mice compared with WT-D mice, consistent with the development of a spontaneous lung infection. We intranasally administered <i>Pseudomonas aeruginosa</i> to induce lung infection in these mice for 24 h, which led to severe lung leukocytic infiltration and an increase in pro-inflammatory cytokine levels in the BALF. In summary, we established a chronic CFRD-like lung mouse model using the <i>Scnn1b</i>-Tg mice. The model can be used for future studies toward understanding the mechanisms underlying the lung pathophysiology associated with CFRD and developing novel therapeutics.<b>NEW & NOTEWORTHY</b> We established a chronic CFRD-like mouse model using the <i>Scnn1b</i>-Tg transgenic mice overexpressing the epithelial sodium channel β subunit made diabetic by injection of streptozotocin. The results underscore the urgent need to develop novel therapeutics for CF lung disease.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619054","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
New Faces: Introducing the newest Editorial Board Fellows of the American Journal of Physiology-Lung Cellular and Molecular Physiology. 新面孔:介绍《美国生理学杂志-肺细胞与分子生理学》的最新编委成员。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 DOI: 10.1152/ajplung.00304.2024
Larissa A Shimoda, Cristina M Alvira, Julie A Bastarache, Rodney D Britt, Wolfgang M Kuebler, Thiago S Moreira, Eric P Schmidt
{"title":"New Faces: Introducing the newest Editorial Board Fellows of the <i>American Journal of Physiology-Lung Cellular and Molecular Physiology</i>.","authors":"Larissa A Shimoda, Cristina M Alvira, Julie A Bastarache, Rodney D Britt, Wolfgang M Kuebler, Thiago S Moreira, Eric P Schmidt","doi":"10.1152/ajplung.00304.2024","DOIUrl":"https://doi.org/10.1152/ajplung.00304.2024","url":null,"abstract":"","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142339337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Citrullination, a novel posttranslational modification of elastin, is involved in COPD pathogenesis. 弹性蛋白的一种新型翻译后修饰--瓜氨酸化参与了慢性阻塞性肺病的发病机制。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-13 DOI: 10.1152/ajplung.00185.2024
Mark P Murphy, Marina Zieger, Michael Henry, Paula Meleady, Christian Mueller, Noel G McElvaney, Emer P Reeves

Elastin is an extracellular matrix protein (ECM) that supports elasticity of the lung, and in patients with chronic obstructive pulmonary disease (COPD) and emphysema, the structural changes that reduce the amount of elastic recoil, lead to loss of pulmonary function. We recently demonstrated that elastin is a target of peptidyl arginine deiminase (PAD) enzyme-induced citrullination, thereby leading to enhanced susceptibility of this ECM protein to proteolysis. This study aimed to investigate the impact of PAD activity in vivo and furthermore assessed whether pharmacological inhibition of PAD activity protects against pulmonary emphysema. Using a Serpina1a-e knockout mouse model, previously shown to develop inflammation-mediated emphysema, we validated the involvement of PADs in airway disease. In line with emphysema development, intratracheal administration of lipopolysaccharide in combination with PADs provoked significant airspace enlargement (P < 0.001) and diminished lung function, including loss of lung tissue elastance (P = 0.0217) and increases in lung volumes (P = 0.0463). Intraperitoneal treatment of mice with the PAD inhibitor, BB-Cl-amidine, prevented PAD/LPS-mediated lung function decline and emphysema and reduced levels of citrullinated airway elastin (P = 0.0199). These results provide evidence for the impact of PADs on lung function decline, indicating promising potential for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.NEW & NOTEWORTHY This study provides evidence for the impact of peptidyl arginine deiminase (PAD) enzymes on lung function decline, indicating promising potential for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.

弹性蛋白是一种细胞外基质蛋白(ECM),它支撑着肺部的弹性,在慢性阻塞性肺病(COPD)和肺气肿患者中,结构性变化会减少弹性反冲力,导致肺功能丧失。我们最近证实,弹性蛋白是肽酰精氨酸脱氨酶(PAD)诱导的瓜氨酸化作用的靶标,从而导致这种 ECM 蛋白更易被蛋白水解。本研究旨在调查 PAD 活性在体内的影响,并进一步评估药物抑制 PAD 活性是否能预防肺气肿。我们利用先前被证明会发生炎症介导的肺气肿的 Serpina1a-e 基因敲除小鼠模型,验证了 PAD 在气道疾病中的参与作用。与肺气肿的发展相一致,气管内给予脂多糖和 PADs 会导致气道显著扩大(P < 0.001)和肺功能减退,包括肺组织弹性损失(P = 0.0217)和肺体积增大(P = 0.0463)。用 PAD 抑制剂 BB-Cl-amidine 对小鼠进行腹腔治疗可防止 PAD/LPS 介导的肺功能下降和肺气肿,并降低瓜氨酸化气道弹性蛋白的水平(P = 0.0199)。这些结果为 PAD 对肺功能下降的影响提供了证据,为未来开发基于 PAD 的治疗方法以保护慢性阻塞性肺病患者的肺功能提供了广阔的前景。
{"title":"Citrullination, a novel posttranslational modification of elastin, is involved in COPD pathogenesis.","authors":"Mark P Murphy, Marina Zieger, Michael Henry, Paula Meleady, Christian Mueller, Noel G McElvaney, Emer P Reeves","doi":"10.1152/ajplung.00185.2024","DOIUrl":"10.1152/ajplung.00185.2024","url":null,"abstract":"<p><p>Elastin is an extracellular matrix protein (ECM) that supports elasticity of the lung, and in patients with chronic obstructive pulmonary disease (COPD) and emphysema, the structural changes that reduce the amount of elastic recoil, lead to loss of pulmonary function. We recently demonstrated that elastin is a target of peptidyl arginine deiminase (PAD) enzyme-induced citrullination, thereby leading to enhanced susceptibility of this ECM protein to proteolysis. This study aimed to investigate the impact of PAD activity in vivo and furthermore assessed whether pharmacological inhibition of PAD activity protects against pulmonary emphysema. Using a <i>Serpina1a-e</i> knockout mouse model, previously shown to develop inflammation-mediated emphysema, we validated the involvement of PADs in airway disease. In line with emphysema development, intratracheal administration of lipopolysaccharide in combination with PADs provoked significant airspace enlargement (<i>P</i> < 0.001) and diminished lung function, including loss of lung tissue elastance (<i>P</i> = 0.0217) and increases in lung volumes (<i>P</i> = 0.0463). Intraperitoneal treatment of mice with the PAD inhibitor, BB-Cl-amidine, prevented PAD/LPS-mediated lung function decline and emphysema and reduced levels of citrullinated airway elastin (<i>P</i> = 0.0199). These results provide evidence for the impact of PADs on lung function decline, indicating promising potential for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.<b>NEW & NOTEWORTHY</b> This study provides evidence for the impact of peptidyl arginine deiminase (PAD) enzymes on lung function decline, indicating promising potential for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141974845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Developing a mouse model of human coronavirus NL63 infection: comparison with rhinovirus-A1B and effects of prior rhinovirus infection. 开发人类冠状病毒 NL63 感染的小鼠模型:与鼻病毒-A1B 的比较以及之前鼻病毒感染的影响。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-27 DOI: 10.1152/ajplung.00149.2023
J Kelley Bentley, Jordan E Kreger, Haley A Breckenridge, Shilpi Singh, Jing Lei, Yiran Li, Susan C Baker, Carey N Lumeng, Marc B Hershenson

Human coronavirus (HCoV)-NL63 causes respiratory tract infections in humans and uses angiotensin-converting enzyme 2 (ACE2) as a receptor. We sought to establish a mouse model of HCoV-NL63 and determine whether prior rhinovirus (RV)-A1B infection affected HCoV-NL63 replication. HCoV-NL63 was propagated in LLC-MK2 cells expressing human ACE2. RV-A1B was grown in HeLa-H1 cells. C57BL6/J or transgenic mice expressing human ACE2 were infected intranasally with sham LLC-MK2 cell supernatant or 1 × 105 tissue culture infectious dose (TCID50) units HCoV-NL63. Wild-type mice were infected with 1 × 106 plaque-forming units (PFU) RV-A1B. Lungs were assessed for vRNA, bronchoalveolar lavage (BAL) cells, histology, HCoV-NL63 nonstructural protein 3 (nsp3), and host gene expression by next-generation sequencing and qPCR. To evaluate sequential infections, mice were infected with RV-A1B followed by HCoV-NL63 infection 4 days later. We report that hACE2 mice infected with HCoV-NL63 showed evidence of replicative infection with increased levels of vRNA, BAL neutrophils and lymphocytes, peribronchial and perivascular infiltrates, and expression of nsp3. Viral replication peaked 3 days after infection and inflammation persisted 6 days after infection. HCoV-NL63-infected hACE2 mice showed increased mRNA expression of IFNs, IFN-stimulated proteins, and proinflammatory cytokines. Infection with RV-A1B 4 days before HCoV-NL63 significantly decreased both HCoV-NL63 vRNA levels and airway inflammation. Mice infected with RV-A1B prior to HCoV-NL63 showed increased expression of antiviral proteins compared with sham-treated mice. In conclusion, we established a mouse model of HCoV-NL63 replicative infection characterized by relatively persistent viral replication and inflammation. Prior infection with RV-A1B reduced HCoV-NL63 replication and airway inflammation, indicative of viral interference.NEW & NOTEWORTHY We describe a mouse model of human coronavirus (HCoV) infection. Infection of transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with HCoV-NL63 produced a replicative infection with peribronchial inflammation and nonstructural protein 3 expression. Mice infected with RV-A1B 4 days before HCoV-NL63 showed decreased HCoV-NL63 replication and airway inflammation and increased expression of antiviral proteins compared with sham-treated mice. This research may shed light on human coronavirus infections, viral interference, and viral-induced asthma exacerbations.

人类冠状病毒(HCoV)-NL63 会导致人类呼吸道感染,并利用血管紧张素转换酶 2(ACE2)作为受体。我们试图建立一个 HCoV-NL63 小鼠模型,并确定之前的 RV-A1B 感染是否会影响 HCoV-NL63 的复制。HCoV-NL63 在表达人类 ACE2 的 LLC-MK2 细胞中繁殖。RV-A1B 在 HeLa-H1 细胞中生长。用假的 LLC-MK2 细胞上清或 1 x 105 TCID50 单位的 HCoV-NL63 经鼻感染 C57BL6/J 或表达人类 ACE2 的转基因小鼠。野生型小鼠用 1 x 106 PFU RV-A1B 感染。通过新一代测序和 qPCR 评估肺部的 vRNA、支气管肺泡灌洗液 (BAL) 细胞、组织学、HCoV-NL63 非结构蛋白 3 (nsp3) 和宿主基因表达。为了评估连续感染,小鼠先感染 RV-A1B,四天后再感染 HCoV-NL63。我们报告说,感染 HCoV-NL63 的 hACE2 小鼠显示出复制感染的证据,其 vRNA、BAL 中性粒细胞和淋巴细胞、支气管周围和血管周围浸润以及 nsp3 表达水平均有所增加。病毒复制在感染三天后达到高峰,炎症在感染六天后持续存在。感染了 HCoV-NL63 的 hACE2 小鼠显示 IFNs、IFN 刺激蛋白和促炎细胞因子的 mRNA 表达增加。在感染 HCoV-NL63 前四天感染 RV-A1B 可显著降低 HCoV-NL63 vRNA 水平和气道炎症。与假治疗小鼠相比,在感染 HCoV-NL63 前感染 RV-A1B 的小鼠抗病毒蛋白表达量增加。总之,我们建立了一种以相对持久的病毒复制和炎症为特征的 HCoV-NL63 复制感染小鼠模型。事先感染 RV-A1B 可减少 HCoV-NL63 的复制和气道炎症,这表明病毒干扰。
{"title":"Developing a mouse model of human coronavirus NL63 infection: comparison with rhinovirus-A1B and effects of prior rhinovirus infection.","authors":"J Kelley Bentley, Jordan E Kreger, Haley A Breckenridge, Shilpi Singh, Jing Lei, Yiran Li, Susan C Baker, Carey N Lumeng, Marc B Hershenson","doi":"10.1152/ajplung.00149.2023","DOIUrl":"10.1152/ajplung.00149.2023","url":null,"abstract":"<p><p>Human coronavirus (HCoV)-NL63 causes respiratory tract infections in humans and uses angiotensin-converting enzyme 2 (ACE2) as a receptor. We sought to establish a mouse model of HCoV-NL63 and determine whether prior rhinovirus (RV)-A1B infection affected HCoV-NL63 replication. HCoV-NL63 was propagated in LLC-MK2 cells expressing human ACE2. RV-A1B was grown in HeLa-H1 cells. C57BL6/J or transgenic mice expressing human ACE2 were infected intranasally with sham LLC-MK2 cell supernatant or 1 × 10<sup>5</sup> tissue culture infectious dose (TCID<sub>50</sub>) units HCoV-NL63. Wild-type mice were infected with 1 × 10<sup>6</sup> plaque-forming units (PFU) RV-A1B. Lungs were assessed for vRNA, bronchoalveolar lavage (BAL) cells, histology, HCoV-NL63 nonstructural protein 3 (nsp3), and host gene expression by next-generation sequencing and qPCR. To evaluate sequential infections, mice were infected with RV-A1B followed by HCoV-NL63 infection 4 days later. We report that hACE2 mice infected with HCoV-NL63 showed evidence of replicative infection with increased levels of vRNA, BAL neutrophils and lymphocytes, peribronchial and perivascular infiltrates, and expression of nsp3. Viral replication peaked 3 days after infection and inflammation persisted 6 days after infection. HCoV-NL63-infected hACE2 mice showed increased mRNA expression of IFNs, IFN-stimulated proteins, and proinflammatory cytokines. Infection with RV-A1B 4 days before HCoV-NL63 significantly decreased both HCoV-NL63 vRNA levels and airway inflammation. Mice infected with RV-A1B prior to HCoV-NL63 showed increased expression of antiviral proteins compared with sham-treated mice. In conclusion, we established a mouse model of HCoV-NL63 replicative infection characterized by relatively persistent viral replication and inflammation. Prior infection with RV-A1B reduced HCoV-NL63 replication and airway inflammation, indicative of viral interference.<b>NEW & NOTEWORTHY</b> We describe a mouse model of human coronavirus (HCoV) infection. Infection of transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with HCoV-NL63 produced a replicative infection with peribronchial inflammation and nonstructural protein 3 expression. Mice infected with RV-A1B 4 days before HCoV-NL63 showed decreased HCoV-NL63 replication and airway inflammation and increased expression of antiviral proteins compared with sham-treated mice. This research may shed light on human coronavirus infections, viral interference, and viral-induced asthma exacerbations.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142071779","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of FGFR4 as a regulator of myofibroblast differentiation in Pulmonary Fibrosis. 确定 FGFR4 是肺纤维化肌成纤维细胞分化的调节因子
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 DOI: 10.1152/ajplung.00184.2023
Mada Ghanem, Aurélien Justet, Madeleine Jaillet, Eirini Vasarmidi, Tiara Boghanim, Mouna Hachem, Aurelie Vadel, Audrey Joannes, Pierre Mordant, Agshin Balayev, Taylor Adams, Hervé Mal, Aurélie Cazes, Nicolas Poté, Arnaud Mailleux, Bruno Crestani

Introduction IPF is a devastating lung disease with limited therapeutic options. FGFR4 is a known receptor for several paracrine Fibroblast growth factors (FGFs). FGFR4 is also the main receptor for FGF19, an endocrine FGF that was demonstrated by our group to have anti-fibrotic properties in the lung. We aimed to determine whether FGFR4 could modulate pulmonary fibrogenesis. Methods We assessed FGFR4 mRNA and protein levels in IPF and control lungs. In vitro, we determined the effect of TGF-b, Endothelin-1 and PDGF on FGFR4 expression in human lung fibroblasts. We determined the effect FGFR4 inhibition, using a specific pharmacological inhibitor (FGF401), or genetic deletion in murine embryonic fibroblasts (MEFs) on TGF-b-induced myofibroblastic differentiation. In vivo, we evaluated the development of bleomycin-induced lung fibrosis in Fgfr4-deficient (Fgfr4-/-) mice compared to Wild Type littermates (WT), and after FGF401 treatment in WT mice compared to a control group receiving the solvent only. Results FGFR4 was decreased in IPF lungs as compared to control lungs, at mRNA and protein levels. In vitro, FGFR4 was downregulated after treatment by TGF- β, Endothelin-1 and PDGF. In vitro, FGFR4 inhibition by FGF401 prevented TGF-b1-induced collagen and ACTA2 increase in lung fibroblasts. Similar results were observed in Fgfr4-/- MEFs. In vivo, FGFR4 genetic deficiency or FGFR4 pharmacological inhibition did not modulate bleomycin-induced pulmonary fibrosis. Conclusion Our data suggest that FGFR4 exerts pro-fibrotic properties by enhancing TGF- β signaling in vitro. However, the inhibition of FGFR4 is not sufficient to prevent the development of pulmonary fibrosis in vivo.

导言 IPF 是一种破坏性肺病,治疗方法有限。FGFR4是已知的几种旁分泌型成纤维细胞生长因子(FGF)的受体。FGFR4 也是 FGF19 的主要受体,FGF19 是一种内分泌性 FGF,我们的研究小组已证实其在肺部具有抗纤维化特性。我们旨在确定 FGFR4 是否能调节肺纤维化。方法 我们评估了 IPF 和对照肺的 FGFR4 mRNA 和蛋白水平。在体外,我们测定了 TGF-b、内皮素-1 和 PDGF 对人肺成纤维细胞中 FGFR4 表达的影响。我们使用特异性药理抑制剂(FGF401)或在小鼠胚胎成纤维细胞(MEFs)中进行基因缺失来确定 FGFR4 抑制对 TGF-b 诱导的肌成纤维细胞分化的影响。在体内,我们评估了Fgfr4缺陷(Fgfr4-/-)小鼠与野生型小鼠(WT)相比,以及WT小鼠经FGF401处理后与仅接受溶剂的对照组相比,博莱霉素诱导的肺纤维化的发展情况。结果 与对照组相比,IPF 肺中的 FGFR4 在 mRNA 和蛋白质水平上都有所下降。在体外,经 TGF-β、内皮素-1 和 PDGF 处理后,FGFR4 下调。在体外,用 FGF401 抑制 FGFR4 可防止 TGF-b1 诱导的肺成纤维细胞胶原和 ACTA2 的增加。在 Fgfr4-/- MEFs 中也观察到了类似的结果。在体内,FGFR4 基因缺失或 FGFR4 药物抑制并不能调节博莱霉素诱导的肺纤维化。结论 我们的数据表明,FGFR4 在体外通过增强 TGF- β 信号传导发挥促纤维化特性。然而,抑制 FGFR4 并不足以阻止肺纤维化在体内的发展。
{"title":"Identification of FGFR4 as a regulator of myofibroblast differentiation in Pulmonary Fibrosis.","authors":"Mada Ghanem, Aurélien Justet, Madeleine Jaillet, Eirini Vasarmidi, Tiara Boghanim, Mouna Hachem, Aurelie Vadel, Audrey Joannes, Pierre Mordant, Agshin Balayev, Taylor Adams, Hervé Mal, Aurélie Cazes, Nicolas Poté, Arnaud Mailleux, Bruno Crestani","doi":"10.1152/ajplung.00184.2023","DOIUrl":"https://doi.org/10.1152/ajplung.00184.2023","url":null,"abstract":"<p><p>Introduction IPF is a devastating lung disease with limited therapeutic options. FGFR4 is a known receptor for several paracrine Fibroblast growth factors (FGFs). FGFR4 is also the main receptor for FGF19, an endocrine FGF that was demonstrated by our group to have anti-fibrotic properties in the lung. We aimed to determine whether FGFR4 could modulate pulmonary fibrogenesis. Methods We assessed FGFR4 mRNA and protein levels in IPF and control lungs. In vitro, we determined the effect of TGF-b, Endothelin-1 and PDGF on FGFR4 expression in human lung fibroblasts. We determined the effect FGFR4 inhibition, using a specific pharmacological inhibitor (FGF401), or genetic deletion in murine embryonic fibroblasts (MEFs) on TGF-b-induced myofibroblastic differentiation. In vivo, we evaluated the development of bleomycin-induced lung fibrosis in <i>Fgfr4</i>-deficient (<i>Fgfr4</i>-/-) mice compared to Wild Type littermates (WT), and after FGF401 treatment in WT mice compared to a control group receiving the solvent only. Results FGFR4 was decreased in IPF lungs as compared to control lungs, at mRNA and protein levels. In vitro, FGFR4 was downregulated after treatment by TGF- β, Endothelin-1 and PDGF. In vitro, FGFR4 inhibition by FGF401 prevented TGF-b1-induced collagen and ACTA2 increase in lung fibroblasts. Similar results were observed in <i>Fgfr4</i>-/- MEFs. In vivo, FGFR4 genetic deficiency or FGFR4 pharmacological inhibition did not modulate bleomycin-induced pulmonary fibrosis. Conclusion Our data suggest that FGFR4 exerts pro-fibrotic properties by enhancing TGF- β signaling in vitro. However, the inhibition of FGFR4 is not sufficient to prevent the development of pulmonary fibrosis in vivo.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142339331","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Piezo channels modulate human lung fibroblast function. 压电通道调节人类肺成纤维细胞的功能
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-27 DOI: 10.1152/ajplung.00356.2023
Mengning Zheng, Yang Yao, Niyati A Borkar, Michael A Thompson, Emily Zhang, Li Y Drake, Xianwei Ye, Elizabeth R Vogel, Christina M Pabelick, Y S Prakash

Bronchial airways and lung parenchyma undergo both static and dynamic stretch in response to normal breathing as well as in the context of insults such as mechanical ventilation (MV) or in diseases such as asthma and chronic obstructive pulmonary disease (COPD) which lead to airway remodeling involving increased extracellular matrix (ECM) production. Here, the role of fibroblasts is critical, but the relationship between stretch- and fibroblast-induced ECM remodeling under these conditions is not well-explored. Piezo (PZ) channels play a role in mechanotransduction in many cell and organ systems, but their role in mechanical stretch-induced airway remodeling is not known. To explore this, we exposed human lung fibroblasts to 10% static stretch on a background of 5% oscillations for 48 h, with no static stretch considered controls. Collagen I, fibronectin, alpha-smooth muscle actin (α-SMA), and Piezo 1 (PZ1) expression was determined in the presence or absence of Yoda1 (PZ1 agonist) or GsMTx4 (PZ1 inhibitor). Collagen I, fibronectin, and α-SMA expression was increased by stretch and Yoda1, whereas pretreatment with GsMTx4 or knockdown of PZ1 by siRNA blunted this effect. Acute stretch in the presence and absence of Yoda1 demonstrated activation of the ERK pathway but not Smad. Measurement of [Ca2+]i responses to histamine showed significantly greater responses following stretch, effects that were blunted by knockdown of PZ1. Our findings identify an essential role for PZ1 in mechanical stretch-induced production of ECM mediated by ERK phosphorylation and Ca2+ influx in lung fibroblasts. Targeting PZ channels in fibroblasts may constitute a novel approach to ameliorate airway remodeling by decreasing ECM deposition.NEW & NOTEWORTHY The lung is an inherently mechanosensitive organ that can respond to mechanical forces in adaptive or maladaptive ways, including via remodeling resulting in increased fibrosis. We explored the mechanisms that link mechanical forces to remodeling using human lung fibroblasts. We found that mechanosensitive Piezo channels increase with stretch and mediate extracellular matrix formation and the fibroblast-to-myofibroblast transition that occurs with stretch. Our data highlight the importance of Piezo channels in lung mechanotransduction toward remodeling.

支气管气道和肺实质在正常呼吸时会发生静态和动态拉伸,但在机械通气(MV)或哮喘和慢性阻塞性肺病等疾病的情况下也会发生拉伸,从而导致细胞外基质(ECM)生成增加的气道重塑。在这种情况下,成纤维细胞的作用至关重要,但在这些条件下,拉伸与成纤维细胞诱导的 ECM 重塑之间的关系还没有得到很好的探讨。压电(PZ)通道在许多细胞和器官系统的机械传导中发挥作用,但它们在机械拉伸诱导的气道重塑中的作用尚不清楚。为了探讨这个问题,我们将人肺成纤维细胞暴露在 5%振荡背景下的 10%静态拉伸环境中 48 小时,并将无静态拉伸作为对照。在 Yoda1(PZ1 激动剂)或 GsMTx4(PZ1 抑制剂)存在或不存在的情况下,测定胶原 I、纤连蛋白、α-SMA 和 Piezo 1(PZ1)的表达。拉伸和 Yoda1 可增加胶原 I、纤连蛋白和 α-SMA 的表达,而 GsMTx4 的预处理或通过 siRNA 敲除 PZ1 可减弱这种效应。在有 Yoda1 和没有 Yoda1 的情况下,急性拉伸显示了 ERK 通路的激活,但没有 Smad 的激活。对组胺的[Ca2+] i 反应的测量显示,拉伸后的反应明显增大:PZ1 的敲除会减弱这种效应。我们的研究结果确定了 PZ1 在机械拉伸诱导的由 ERK 磷酸化和 Ca2+ 流入介导的肺成纤维细胞 ECM 生成中的重要作用。靶向成纤维细胞中的 PZ 通道可能是通过减少 ECM 沉积来改善气道重塑的一种新方法。
{"title":"Piezo channels modulate human lung fibroblast function.","authors":"Mengning Zheng, Yang Yao, Niyati A Borkar, Michael A Thompson, Emily Zhang, Li Y Drake, Xianwei Ye, Elizabeth R Vogel, Christina M Pabelick, Y S Prakash","doi":"10.1152/ajplung.00356.2023","DOIUrl":"10.1152/ajplung.00356.2023","url":null,"abstract":"<p><p>Bronchial airways and lung parenchyma undergo both static and dynamic stretch in response to normal breathing as well as in the context of insults such as mechanical ventilation (MV) or in diseases such as asthma and chronic obstructive pulmonary disease (COPD) which lead to airway remodeling involving increased extracellular matrix (ECM) production. Here, the role of fibroblasts is critical, but the relationship between stretch- and fibroblast-induced ECM remodeling under these conditions is not well-explored. Piezo (PZ) channels play a role in mechanotransduction in many cell and organ systems, but their role in mechanical stretch-induced airway remodeling is not known. To explore this, we exposed human lung fibroblasts to 10% static stretch on a background of 5% oscillations for 48 h, with no static stretch considered controls. Collagen I, fibronectin, alpha-smooth muscle actin (α-SMA), and Piezo 1 (PZ1) expression was determined in the presence or absence of Yoda1 (PZ1 agonist) or GsMTx4 (PZ1 inhibitor). Collagen I, fibronectin, and α-SMA expression was increased by stretch and Yoda1, whereas pretreatment with GsMTx4 or knockdown of PZ1 by siRNA blunted this effect. Acute stretch in the presence and absence of Yoda1 demonstrated activation of the ERK pathway but not Smad. Measurement of [Ca<sup>2+</sup>]<sub>i</sub> responses to histamine showed significantly greater responses following stretch, effects that were blunted by knockdown of PZ1. Our findings identify an essential role for PZ1 in mechanical stretch-induced production of ECM mediated by ERK phosphorylation and Ca<sup>2+</sup> influx in lung fibroblasts. Targeting PZ channels in fibroblasts may constitute a novel approach to ameliorate airway remodeling by decreasing ECM deposition.<b>NEW & NOTEWORTHY</b> The lung is an inherently mechanosensitive organ that can respond to mechanical forces in adaptive or maladaptive ways, including via remodeling resulting in increased fibrosis. We explored the mechanisms that link mechanical forces to remodeling using human lung fibroblasts. We found that mechanosensitive Piezo channels increase with stretch and mediate extracellular matrix formation and the fibroblast-to-myofibroblast transition that occurs with stretch. Our data highlight the importance of Piezo channels in lung mechanotransduction toward remodeling.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142071810","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LOXL2 inhibition ameliorates pulmonary artery remodeling in pulmonary hypertension. 抑制 LOXL2 可改善肺动脉高压的肺动脉重塑。
IF 3.6 2区 医学 Q1 PHYSIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-07-16 DOI: 10.1152/ajplung.00327.2023
Jochen Steppan, Huilei Wang, Kavitha Nandakumar, Mahin Gadkari, Alan Poe, Lydia Pak, Travis Brady, Dan E Berkowitz, Larissa A Shimoda, Lakshmi Santhanam

Conduit pulmonary arterial stiffening and the resultant increase in pulmonary vascular impedance have emerged as an important underlying driver of pulmonary arterial hypertension (PAH). Given that matrix deposition is central to vascular remodeling, we evaluated the role of the collagen cross-linking enzyme lysyl oxidase like 2 (LOXL2) in this study. Human pulmonary artery smooth muscle cells (PASMCs) subjected to hypoxia showed increased LOXL2 secretion. LOXL2 activity and expression were markedly higher in primary PASMCs isolated from the pulmonary arteries of the rat Sugen 5416 + hypoxia (SuHx) model of severe pulmonary hypertension (PH). Similarly, LOXL2 protein and mRNA levels were increased in the pulmonary arteries (PA) and lungs of rats with PH (SuHx and monocrotaline (MCT) models). Pulmonary arteries (PAs) isolated from the rats with PH exhibited hypercontractility to phenylephrine and attenuated vasorelaxation elicited by acetylcholine, indicating severe endothelial dysfunction. Tensile testing revealed a significant increase in PA stiffness in PH. Treatment with PAT-1251, a novel small-molecule LOXL2 inhibitor, improved active and passive properties of the PA ex vivo. There was an improvement in right heart function as measured by right ventricular pressure volume loops in vivo with PAT-1251. Importantly, PAT-1251 treatment ameliorated PH, resulting in improved pulmonary artery pressures, right ventricular remodeling, and survival. Hypoxia-induced LOXL2 activation is a causal mechanism in pulmonary artery stiffening in PH and pulmonary artery mechanical and functional decline. LOXL2 inhibition with PAT-1251 could be a promising approach to improve pulmonary artery pressures, right ventricular elastance, cardiac relaxation, and survival in PAH.NEW & NOTEWORTHY Pulmonary arterial stiffening contributes to the progression of PAH and the deterioration of right heart function. This study shows that LOXL2 is upregulated in rat models of PH. LOXL2 inhibition halts pulmonary vascular remodeling and improves PA contractility, endothelial function, and PA pressure, resulting in prolonged survival. Thus, LOXL2 is an important mediator of PA remodeling and stiffening in PH and a promising target to improve PA pressures and survival in PH.

背景:导管肺动脉僵化和由此导致的肺血管阻抗增加已成为肺动脉高压(PAH)的一个重要潜在驱动因素。鉴于基质沉积是血管重塑的核心,我们在本研究中评估了胶原交联酶类赖氨酸氧化酶 2(LOXL2)的作用:缺氧状态下的人肺动脉平滑肌细胞(PASMCs)的 LOXL2 分泌增加。从大鼠 Sugen5416 + 缺氧(SuHx)重度 PH 模型的肺动脉中分离出的原代 PASMCs 中,LOXL2 的活性和表达量明显升高。同样,LOXL2 蛋白和 mRNA 水平在 PH 大鼠的肺动脉(PA)和肺(SuHx 和一缩醛(MCT)模型)中也有所增加。从患有 PH 的大鼠体内分离出的肺动脉(PA)对苯肾上腺素表现出过度收缩性,乙酰胆碱引起的血管舒张减弱,表明内皮功能严重失调。拉伸试验显示,PH 大鼠的 PA 硬度显著增加。使用新型小分子 LOXL2 抑制剂 PAT-1251 治疗后,体内 PA 的主动和被动特性均有所改善。使用 PAT-1251 治疗后,体内右心室压力容积环测量的右心功能也有所改善。重要的是,PAT-1251治疗可改善PH,从而改善肺动脉压力、右心室重塑和存活率:结论:缺氧诱导的 LOXL2 激活是导致 PH 中肺动脉僵化以及肺动脉机械和功能衰退的原因之一。用PAT-1251抑制LOXL2可能是改善PAH患者肺动脉压力、右心室弹性、心脏松弛和存活率的有效方法。
{"title":"LOXL2 inhibition ameliorates pulmonary artery remodeling in pulmonary hypertension.","authors":"Jochen Steppan, Huilei Wang, Kavitha Nandakumar, Mahin Gadkari, Alan Poe, Lydia Pak, Travis Brady, Dan E Berkowitz, Larissa A Shimoda, Lakshmi Santhanam","doi":"10.1152/ajplung.00327.2023","DOIUrl":"10.1152/ajplung.00327.2023","url":null,"abstract":"<p><p>Conduit pulmonary arterial stiffening and the resultant increase in pulmonary vascular impedance have emerged as an important underlying driver of pulmonary arterial hypertension (PAH). Given that matrix deposition is central to vascular remodeling, we evaluated the role of the collagen cross-linking enzyme lysyl oxidase like 2 (LOXL2) in this study. Human pulmonary artery smooth muscle cells (PASMCs) subjected to hypoxia showed increased LOXL2 secretion. LOXL2 activity and expression were markedly higher in primary PASMCs isolated from the pulmonary arteries of the rat Sugen 5416 + hypoxia (SuHx) model of severe pulmonary hypertension (PH). Similarly, LOXL2 protein and mRNA levels were increased in the pulmonary arteries (PA) and lungs of rats with PH (SuHx and monocrotaline (MCT) models). Pulmonary arteries (PAs) isolated from the rats with PH exhibited hypercontractility to phenylephrine and attenuated vasorelaxation elicited by acetylcholine, indicating severe endothelial dysfunction. Tensile testing revealed a significant increase in PA stiffness in PH. Treatment with PAT-1251, a novel small-molecule LOXL2 inhibitor, improved active and passive properties of the PA ex vivo. There was an improvement in right heart function as measured by right ventricular pressure volume loops in vivo with PAT-1251. Importantly, PAT-1251 treatment ameliorated PH, resulting in improved pulmonary artery pressures, right ventricular remodeling, and survival. Hypoxia-induced LOXL2 activation is a causal mechanism in pulmonary artery stiffening in PH and pulmonary artery mechanical and functional decline. LOXL2 inhibition with PAT-1251 could be a promising approach to improve pulmonary artery pressures, right ventricular elastance, cardiac relaxation, and survival in PAH.<b>NEW & NOTEWORTHY</b> Pulmonary arterial stiffening contributes to the progression of PAH and the deterioration of right heart function. This study shows that LOXL2 is upregulated in rat models of PH. LOXL2 inhibition halts pulmonary vascular remodeling and improves PA contractility, endothelial function, and PA pressure, resulting in prolonged survival. Thus, LOXL2 is an important mediator of PA remodeling and stiffening in PH and a promising target to improve PA pressures and survival in PH.</p>","PeriodicalId":7593,"journal":{"name":"American journal of physiology. Lung cellular and molecular physiology","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
American journal of physiology. Lung cellular and molecular physiology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1