airway epithelial
Recently Published Documents


TOTAL DOCUMENTS

4467
(FIVE YEARS 1013)

H-INDEX

121
(FIVE YEARS 17)

2022 ◽  
Vol 19 ◽  
pp. 77-87
Author(s):  
Takeshi Tada ◽  
Hiroe Ohnishi ◽  
Norio Yamamoto ◽  
Fumihiko Kuwata ◽  
Yasuyuki Hayashi ◽  
...  

2022 ◽  
Vol 3 (1) ◽  
pp. 01-13
Author(s):  
Nightingale Syabbalo

Asthma is a common chronic airway disease affecting about 358 million people worldwide, and an estimated 7 million children globally. Approximately 10% of patients with asthma have severe refractory disease, which is difficult to control on high doses of inhaled corticosteroids and other modifiers. Among these, are patients with severe neutrophilic asthma. Neutrophilic asthma is a severe phenotype of asthma, characterized by frequent exacerbations, persistent airway obstruction, and poor lung function. Immunopathologically, it is characterized by the presence of high levels of neutrophils in the airways and lungs. Interleukin-17 produced by Th17 cells, plays a key role in the pathogenesis of neutrophilic asthma by expressing the secretion of chemoattractant cytokines and chemokines for the recruitment, and activation of neutrophils. Interleukin-8 is a powerful chemoattractant and activator of neutrophils. Activated neutrophils produce an oxidative burst, releasing multiple reactive oxygen species, proteinases, cytokines, which cause airway epithelial cell injury, inflammation, airway hyperresponsiveness, and remodeling. Furthermore, exasperated neutrophils due to viral, bacterial or fungal infections, and chemical irritants can release extracellular nucleic acids (DNA), designated as NETs (neutrophil extracellular traps), which are more toxic to the airway epithelial cells, and orchestrate airway inflammation, and release alarmin cytokines. Dysregulated NETs formation is associated with severe asthma. Most patients with neutrophilic asthma are unresponsive to the standard of care, including high dose inhaled corticosteroids, and to targeted biologics, such as mepolizumab, and dupilumab, which are very effective in treating eosinophilic asthma. There is unmet need to explore for novel biologics for the treatment of neutrophilic asthma, and in refining therapies, such as bronchial thermoplasty.


2022 ◽  
Author(s):  
Wenjing Zou ◽  
maozhu xu ◽  
Jie Hu ◽  
Lili Yang ◽  
Gang Gen ◽  
...  

Abstract Backgroud: The chromatin remodeling factor Brg1 (Brahma-related gene 1) is an important nuclear protein that promotes the transcriptional activation or inhibition of target genes by regulating ATP hydrolysis to generate energy which rearranges the position of nucleosomes and the interaction of histone DNA. In this study, we explored the effect of Brg1 on airway mucus hypersecretion in asthma.Methods: Six-to-eight-week-old female wild-type C57BL/6 mice (wild-type, WT) and type II alveolar epithelial cells (AECIIs) specifically knockout Brg1 mice (Brg1fl/fl) were selected as the experimental subjects. The asthma group was established with house dust mite (HDM), and the control group was treated with normal saline (n=10). Wright's staining was used to detect inflammatory cells in bronchoalveolar lavage fluid (BALF). Invasive lung function was used to assess the airway compliance. Hematoxylin and eosin and periodic acid-schiff staining were used to detect mucus secretion. The virus was used to knock down the Brg1 gene in the bronchial epithelial cell line (16HBE) and stimulated with HDM. Immunohistochemistry was used to measure mucin glycoprotein 5AC (MUC5AC) protein expression in the airway epithelium and 16HBE cells. Western blotting was used to detect the expression of the MUC5AC and JAK1/2-STAT6 signaling pathways in mouse lung tissue and 16HBE. Co-immunoprecipitation (Co-IP) and Chromatin Immunoprecipitation (CHIP) were used to detect whether Brg1 could regulate the JAK1/2-STAT6 signaling pathway.Results: Specifically, knocking out the Brg1 gene in AECIIs can reduce airway inflammation, airway compliance, and mucus hypersecretion in asthma. Knockdown of the Brg1 gene can simultaneously reduce Interleukin-13 (IL-13) and the expression of MUC5AC protein in airway epithelial cells and the activation of the JAK1/2-STAT6 signaling pathway. The results of Co-IP and CHIP showed that Brg1 could bind to the JAK1/2 promoter region, regulating the activity of the JAK1/2-STAT6 pathway affects airway mucus secretion in asthma.Conclusion: Brg1 gene knockout in airway epithelial cells can reduce asthmatic airway mucus hypersecretion and the expression of MUC5AC protein in airway epithelial cells partly by inhibiting the activation of the JAK1/2-STAT6 signaling pathway.


2022 ◽  
Author(s):  
Wenguang Yin ◽  
Andreas Liontos ◽  
Janine Koepke ◽  
Maroua Ghoul ◽  
Luciana Mazzocchi ◽  
...  

The tracheal epithelium is a primary target for pulmonary diseases as it provides a conduit for air flow between the environment and the lung lobes. The cellular and molecular mechanisms underlying airway epithelial cell proliferation and differentiation remain poorly understood. Hedgehog (Hh) signaling orchestrates communication between epithelial and mesenchymal cells in the lung, where it modulates stromal cell proliferation, differentiation and signaling back to the epithelium. Here, we reveal a new, autocrine function of Hh signaling in airway epithelial cells. Epithelial cell depletion of the ligand Sonic hedgehog (SHH) or its effector Smoothened (SMO) causes defects in both epithelial cell proliferation and differentiation. In cultured primary human airway epithelial cells, Hh signaling inhibition also hampers cell proliferation and differentiation. Epithelial Hh function is mediated, at least in part, through transcriptional activation as Hh signaling inhibition leads to downregulation of cell-type specific transcription factor genes in both the mouse trachea and human airway epithelial cells. These results provide new insights into the role of Hh signaling in epithelial cell proliferation and differentiation during airway development.


Author(s):  
Ruobing Wang ◽  
Adam J. Hume ◽  
Mary Lou Beermann ◽  
Chantelle Simone-Roach ◽  
Jonathan Lindstrom-Vautrin ◽  
...  

There is an urgent need to understand how SARS-CoV-2 infects the airway epithelium and in a subset of individuals leads to severe illness or death. Induced pluripotent stem cells (iPSCs) provide a near limitless supply of human cells that can be differentiated into cell types of interest, including airway epithelium, for disease modeling. We present a human iPSC-derived airway epithelial platform, composed of the major airway epithelial cell types, that is permissive to SARS-CoV-2 infection. Subsets of iPSC-airway cells express the SARS-CoV-2 entry factors ACE2 and TMPRSS2. Multiciliated cells are the primary initial target of SARS-CoV-2 infection. Upon infection with SARS-CoV-2, iPSC-airway cells generate robust interferon and inflammatory responses and treatment with remdesivir or camostat methylate causes a decrease in viral propagation and entry, respectively. In conclusion, iPSC-derived airway cells provide a physiologically relevant in vitro model system to interrogate the pathogenesis of, and develop treatment strategies for, COVID-19 pneumonia.


2022 ◽  
Author(s):  
Min-yan Li ◽  
Yan-qin Qin ◽  
Jian-sheng Li ◽  
Peng Zhao ◽  
Yan-ge Tian ◽  
...  

Abstract Background: Effective-component compatibility of Bufei Yishen formula Ⅲ (ECC-BYF Ⅲ) shows positive effects on stable chronic obstructive pulmonary disease (COPD).Purpose: To investigate the mechanisms of ECC-BYF Ⅲ on COPD rats from the aspect of airway epithelial cell senescence.Methods: COPD model rats were treated with ECC-BYF Ⅲ for 8 weeks and the efficacy was evaluated. Cigarette smoke extract (CSE) induced senescence model of airway epithelial cells were treated with ECC-BYF Ⅲ, the related enzymes and proteins involved in oxidative stress and mitophagy were detected.Results: ECC-BYF Ⅲ markedly rescued pulmonary function and histopathological changes, which might be associated with the amelioration of lung senescence, including reduction of malondialdehyde (MDA) and tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and matrix metalloproteinase (MMP)-9, increase of the level of total superoxide dismutase (T-SOD), and decease of p21 level in airway. Furthermore, ECC-BYF Ⅲ suppressed p16, p21 expressions and senescence-associated β-galactosidase (SA-β-Gal) in CSE-induced airway epithelial cells. Moreover, ECC-BYF Ⅲ upregulated the mitophagy-related proteins, including co-localization of TOM20 and LC3B, PINK1, PARK2, and improved mitochondrial function with upregulating mitochondrial mitofusin (Mfn)2 and reducing dynamin-related protein 1 (Drp1) expression. ECC-BYF Ⅲ enhanced the activities of T-SOD and GSH-PX by up-regulating Nrf2, thus inhibiting oxidative stress. After intervention with Nrf2 inhibitor, the regulation effects of ECC-BYF Ⅲ on oxidative stress, mitophagy and senescence in airway epithelial cells were significantly suppressed.Conclusions: ECC-BYF Ⅲ exerts beneficial effects on COPD rats by ameliorating airway epithelial cell senescence, which is mediated by inhibiting oxidative stress and subsequently enhancing mitophagy through activation of Nrf2 signaling.


2022 ◽  
Author(s):  
Kelsey E. Huntington ◽  
Lindsey Carlsen ◽  
Eui-Young So ◽  
Matthias Piesche ◽  
Olin Liang ◽  
...  

As COVID-19 continues to pose major risk for vulnerable populations including the elderly, immunocompromised, patients with cancer, and those with contraindications to vaccination, novel treatment strategies are urgently needed. SARS-CoV-2 infects target cells via RGD-binding integrins either independently or as a co-receptor with surface receptor angiotensin-converting enzyme 2 (ACE2). We used pan-integrin inhibitor GLPG-0187 to demonstrate blockade of SARS-CoV-2 pseudovirus infection of target cells. Omicron pseudovirus infected normal human small airway epithelial (HSAE) cells significantly less than D614G or Delta variant pseudovirus, and GLPG-0187 effectively blocked SARS-CoV-2 pseudovirus infection in a dose-dependent manner across multiple viral variants. GLPG-0187 inhibited Omicron and Delta pseudovirus infection of HSAE cells more significantly than other variants. Pre-treatment of HSAE cells with MEK inhibitor (MEKi) VS-6766 enhanced inhibition of pseudovirus infection by GLPG-0187. Because integrins activate TGF-beta; signaling, we compared plasma levels of active and total TGF-beta; in COVID-19+ patients. Plasma TGF-beta1 levels correlated with age, race, and number of medications upon presentation with COVID-19, but not with sex. Total plasma TGF-beta1 levels correlated with activated TGF-beta1 levels. In our preclinical studies, Omicron infects lower airway lung cells less efficiently than other COVID-19 variants. Moreover, inhibition of integrin signaling prevents SARS-CoV-2 Delta and Omicron pseudovirus infectivity, and may mitigate COVID-19 severity through decreased TGF-beta1 activation. This therapeutic strategy may be further explored through clinical testing in vulnerable and unvaccinated populations.


Sign in / Sign up

Export Citation Format

Share Document