1
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
2
|
Tiotiu A, Steiropoulos P, Novakova S, Nedeva D, Novakova P, Chong-Neto H, Fogelbach GG, Kowal K. Airway Remodeling in Asthma: Mechanisms, Diagnosis, Treatment, and Future Directions. Arch Bronconeumol 2025; 61:31-40. [PMID: 39368875 DOI: 10.1016/j.arbres.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024]
Abstract
Airway remodeling (AR) with chronic inflammation, are key features in asthma pathogenesis. AR characterized by structural changes in the bronchial wall is associated with a specific asthma phenotype with poor clinical outcomes, impaired lung function and reduced treatment response. Most studies focus on the role of inflammation, while understanding the mechanisms driving AR is crucial for developing disease-modifying therapeutic strategies. This review paper summarizes current knowledge on the mechanisms underlying AR, diagnostic tools, and therapeutic approaches. Mechanisms explored include the role of the resident cells and the inflammatory cascade in AR. Diagnostic methods such as bronchial biopsy, lung function testing, imaging, and possible biomarkers are described. The effectiveness on AR of different treatments of asthma including corticosteroids, leukotriene modifiers, bronchodilators, macrolides, biologics, and bronchial thermoplasty is discussed, as well as other possible therapeutic options. AR poses a significant challenge in asthma management, contributing to disease severity and treatment resistance. Current therapeutic approaches target mostly airway inflammation rather than smooth muscle cell dysfunction and showed limited benefits on AR. Future research should focus more on investigating the mechanisms involved in AR to identify novel therapeutic targets and to develop new effective treatments able to prevent irreversible structural changes and improve long-term asthma outcomes.
Collapse
Affiliation(s)
- Angelica Tiotiu
- Department of Pulmonology, University Hospital Saint-Luc, Brussels, Belgium; Pole Pneumology, ENT, and Dermatology - LUNS, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.
| | - Paschalis Steiropoulos
- Department of Pulmonology, Medical School, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Silviya Novakova
- Department of Allergology, University Hospital "Sv. Georgi" Plovdiv, Bulgaria
| | - Denislava Nedeva
- Clinic of Asthma and Allergology, UMBAL Alexandrovska, Medical University Sofia, Sofia, Bulgaria
| | - Plamena Novakova
- Department of Allergy, Medical University Sofia, Sofia, Bulgaria
| | - Herberto Chong-Neto
- Division of Allergy and Immunology, Complexo Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, Brazil
| | | | - Krzysztof Kowal
- Department of Experimental Allergology and Immunology and Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
3
|
Lu Q, Liu Z, He W, Chu X. Retracted article: Protective effects of ulinastatin on rats with acute lung injury induced by lipopolysaccharide. Bioengineered 2024; 15:1987083. [PMID: 34637694 PMCID: PMC10813561 DOI: 10.1080/21655979.2021.1987083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022] Open
Abstract
Qitong Lu, Zhiyong Liu, Wei He and Xin Chu. Protective effects of ulinastatin on rats with acute lung injury induced by lipopolysaccharide. Bioengineered. 2021 Oct. doi: 10.1080/21655979.2021.1987083.Since publication, significant concerns have been raised about the compliance with ethical policies for human research and the integrity of the data reported in the article.When approached for an explanation, the authors provided some original data but were not able to provide all the necessary supporting information. As verifying the validity of published work is core to the scholarly record's integrity, we are retracting the article. All authors listed in this publication have been informed.We have been informed in our decision-making by our editorial policies and the COPE guidelines. The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted.'
Collapse
Affiliation(s)
- Qitong Lu
- Department of Cardiothoracic Surgery, Zhongda Hospital, Southeast University, Nanjing, P. R. China
| | - Zhiyong Liu
- Department of Cardiothoracic Surgery, Zhongda Hospital, Southeast University, Nanjing, P. R. China
| | - Wei He
- Department of Cardiothoracic Surgery, Zhongda Hospital, Southeast University, Nanjing, P. R. China
| | - Xin Chu
- Department of Cardiothoracic Surgery, Zhongda Hospital, Southeast University, Nanjing, P. R. China
| |
Collapse
|
4
|
Zhang B, Guan Y, Zeng D, Wang R. Arginine methylation and respiratory disease. Transl Res 2024; 272:140-150. [PMID: 38453053 DOI: 10.1016/j.trsl.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Arginine methylation, a vital post-translational modification, plays a pivotal role in numerous cellular functions such as signal transduction, DNA damage response and repair, regulation of gene transcription, mRNA splicing, and protein interactions. Central to this modification is the role of protein arginine methyltransferases (PRMTs), which have been increasingly recognized for their involvement in the pathogenesis of various respiratory diseases. This review begins with an exploration of the biochemical underpinnings of arginine methylation, shedding light on the intricate molecular regulatory mechanisms governed by PRMTs. It then delves into the impact of arginine methylation and the dysregulation of arginine methyltransferases in diverse pulmonary disorders. Concluding with a focus on the therapeutic potential and recent advancements in PRMT inhibitors, this article aims to offer novel perspectives and therapeutic avenues for the management and treatment of respiratory diseases.
Collapse
Affiliation(s)
- Binbin Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, PR China
| | - Youhong Guan
- Department of Infectious Diseases, Hefei Second People's Hospital, Hefei 230001, Anhui Province, PR China
| | - Daxiong Zeng
- Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215006, Jiangsu Province, PR China.
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, PR China.
| |
Collapse
|
5
|
Aluksanasuwan S, Somsuan K, Ngoenkam J, Chiangjong W, Rongjumnong A, Morchang A, Chutipongtanate S, Pongcharoen S. Knockdown of heat shock protein family D member 1 (HSPD1) in lung cancer cell altered secretome profile and cancer-associated fibroblast induction. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119736. [PMID: 38663552 DOI: 10.1016/j.bbamcr.2024.119736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/02/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The crosstalk between lung cancer cells and cancer-associated fibroblast (CAF) is pivotal in cancer progression. Heat shock protein family D member 1 (HSPD1) is a potential prognostic biomarker associated with the tumor microenvironment in lung adenocarcinoma (LUAD). However, the role of HSPD1 in CAF activation remains unclear. This study established stable HSPD1-knockdown A549 lung cancer cells using a lentivirus-mediated shRNA transduction. A targeted label-free proteomic analysis identified six significantly altered secretory proteins in the shHSPD1-A549 secretome compared to shControl-A549. Functional enrichment analysis highlighted their involvement in cell-to-cell communication and immune responses within the tumor microenvironment. Additionally, most altered proteins exhibited positive correlations and significant prognostic impacts on LUAD patient survival. Investigations on the effects of lung cancer secretomes on lung fibroblast WI-38 cells revealed that the shControl-A549 secretome stimulated fibroblast proliferation, migration, and CAF marker expression. These effects were reversed upon the knockdown of HSPD1 in A549 cells. Altogether, our findings illustrate the role of HSPD1 in mediating CAF induction through secretory proteins, potentially contributing to the progression and aggressiveness of lung cancer.
Collapse
Affiliation(s)
- Siripat Aluksanasuwan
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand.
| | - Keerakarn Somsuan
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Artitaya Rongjumnong
- Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Atthapan Morchang
- School of Medicine, Mae Fah Luang University, Chiang Rai 57100, Thailand; Cancer and Immunology Research Unit (CIRU), Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Somchai Chutipongtanate
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0056, USA
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand.
| |
Collapse
|
6
|
Wang T, Fu P, Huang W, Long L, Long F, Liu S. Bronchial thermoplasty decreases airway remodeling by inhibiting autophagy via the AMPK/mTOR signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:730-739. [PMID: 38655617 PMCID: PMC11177112 DOI: 10.3724/abbs.2024028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/20/2023] [Indexed: 04/26/2024] Open
Abstract
Bronchial thermoplasty (BT), an effective treatment for severe asthma, requires heat to reach the airway to reduce the mass of airway smooth muscle cells (ASMCs). Autophagy is involved in the pathological process of airway remodeling in patients with asthma. However, it remains unclear whether autophagy participates in controlling airway remodeling induced by BT. In this study, we aim to elucidate the autophagy-mediated molecular mechanisms in BT. Our study reveal that the number of autophagosomes and the level of alpha-smooth muscle actin (α-SMA) fluorescence are significantly decreased in airway biopsy tissues after BT. As the temperature increased, BT causes a decrease in cell proliferation and a concomitant increase in the apoptosis of human airway smooth muscle cells (HASMCs). Furthermore, increase in temperature significantly downregulates cellular autophagy, autophagosome accumulation, the LC3II/LC3I ratio, and Beclin-1 expression, upregulates p62 expression, and inhibits the AMPK/mTOR pathway. Furthermore, cotreatment with AICAR (an AMPK agonist) or RAPA (an mTOR antagonist) abolishes the inhibition of autophagy and attenuates the increase in the apoptosis rate of HASMCs induced by the thermal effect. Therefore, we conclude that BT decreases airway remodeling by blocking autophagy induced by the AMPK/mTOR signaling pathway in HASMCs.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pulmonary and Critical Care MedicineUniversity of Chinese Academy of Sciences Shenzhen HospitalShenzhen518106China
- Department of Pulmonary and Critical Care Medicinethe First Affiliated Hospital of Jinan UniversityGuangzhou510630China
| | - Peng Fu
- Department of Pulmonary and Critical Care MedicineUniversity of Chinese Academy of Sciences Shenzhen HospitalShenzhen518106China
| | - Wenting Huang
- Department of Pulmonary and Critical Care MedicineUniversity of Chinese Academy of Sciences Shenzhen HospitalShenzhen518106China
| | - Liang Long
- Department of Pulmonary and Critical Care MedicineUniversity of Chinese Academy of Sciences Shenzhen HospitalShenzhen518106China
| | - Fa Long
- Department of Pulmonary and Critical Care MedicineUniversity of Chinese Academy of Sciences Shenzhen HospitalShenzhen518106China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicinethe First Affiliated Hospital of Jinan UniversityGuangzhou510630China
| |
Collapse
|
7
|
Zhou S, Zhang Q, Yang H, Zhu Y, Hu X, Wan G, Yu L. Targeting type I PRMTs as promising targets for the treatment of pulmonary disorders: Asthma, COPD, lung cancer, PF, and PH. Life Sci 2024; 342:122538. [PMID: 38428571 DOI: 10.1016/j.lfs.2024.122538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
Pulmonary disorders, including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), pulmonary hypertension (PH), and lung cancer, seriously impair the quality of lives of patients. A deeper understanding of the occurrence and development of the above diseases may inspire new strategies to remedy the scarcity of treatments. Type I protein arginine methyltransferases (PRMTs) can affect processes of inflammation, airway remodeling, fibroblast proliferation, mitochondrial mass, and epithelial dysfunction through substrate methylation and non-enzymatic activity, thus affecting the occurrence and development of asthma, COPD, lung cancer, PF, and PH. As potential therapeutic targets, inhibitors of type I PRMTs are developed, moreover, representative compounds such as GSK3368715 and MS023 have also been used for early research. Here, we collated structures of type I PRMTs inhibitors and compared their activity. Finally, we highlighted the physiological and pathological associations of type I PRMTs with asthma, COPD, lung cancer, PF, and PH. The developing of type I PRMTs modulators will be beneficial for the treatment of these diseases.
Collapse
Affiliation(s)
- Shuyan Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiangsheng Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Honglin Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongxia Zhu
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guoquan Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luoting Yu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Gagnon PA, Klein M, De Vos J, Biardel S, Côté A, Godbout K, Laviolette M, Laprise C, Assou S, Chakir J. S100A alarmins and thymic stromal lymphopoietin (TSLP) regulation in severe asthma following bronchial thermoplasty. Respir Res 2023; 24:294. [PMID: 37996952 PMCID: PMC10668474 DOI: 10.1186/s12931-023-02604-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
RATIONALE Severe asthma affects a small proportion of asthmatics but represents a significant healthcare challenge. Bronchial thermoplasty (BT) is an interventional treatment approach preconized for uncontrolled severe asthma after considering biologics therapy. It was showed that BT long-lastingly improves asthma control. These improvements seem to be related to the ability of BT to reduce airway smooth muscle remodeling, reduce the number of nerve fibers and to modulate bronchial epithelium integrity and behavior. Current evidence suggest that BT downregulates epithelial mucins expression, cytokine production and metabolic profile. Despite these observations, biological mechanisms explaining asthma control improvement post-BT are still not well understood. OBJECTIVES To assess whether BT affects gene signatures in bronchial epithelial cells (BECs). METHODS In this study we evaluated the transcriptome of cultured bronchial epithelial cells (BECs) of severe asthmatics obtained pre- and post-BT treatment using microarrays. We further validated gene and protein expressions in BECs and in bronchial biopsies with immunohistochemistry pre- and post-BT treatment. MEASUREMENTS AND MAIN RESULTS Transcriptomics analysis revealed that a large portion of differentially expressed genes (DEG) was involved in anti-viral response, anti-microbial response and pathogen induced cytokine storm signaling pathway. S100A gene family stood out as five members of this family where consistently downregulated post-BT. Further validation revealed that S100A7, S100A8, S100A9 and their receptor (RAGE, TLR4, CD36) expressions were highly enriched in severe asthmatic BECs. Further, these S100A family members were downregulated at the gene and protein levels in BECs and in bronchial biopsies of severe asthmatics post-BT. TLR4 and CD36 protein expression were also reduced in BECs post-BT. Thymic stromal lymphopoietin (TSLP) and human β-defensin 2 (hBD2) were significantly decreased while no significant change was observed in IL-25 and IL-33. CONCLUSIONS These data suggest that BT might improve asthma control by downregulating epithelial derived S100A family expression and related downstream signaling pathways.
Collapse
Affiliation(s)
- Pierre-Alexandre Gagnon
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Martin Klein
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - John De Vos
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Sabrina Biardel
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Andréanne Côté
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Krystelle Godbout
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Michel Laviolette
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Catherine Laprise
- Département des Sciences Fondamentales, Université du Québec à Chicoutimi (UQAC), Saguenay, QC, Canada
| | - Said Assou
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Jamila Chakir
- Centre de Recherche, Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval (IUCPQ-UL), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada.
| |
Collapse
|
9
|
Henry C, Biardel S, Boucher M, Godbout K, Chakir J, Côté A, Laviolette M, Bossé Y. Bronchial thermoplasty attenuates bronchodilator responsiveness. Respir Med 2023; 217:107340. [PMID: 37422022 DOI: 10.1016/j.rmed.2023.107340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/31/2023] [Accepted: 06/25/2023] [Indexed: 07/10/2023]
Abstract
INTRODUCTION Bronchial thermoplasty is an effective intervention to improve respiratory symptoms and to reduce the rate of exacerbations in uncontrolled severe asthma. A reduction in airway smooth muscle is arguably the most widely discussed mechanisms accounting for these clinical benefits. Yet, this smooth muscle reduction should also translate into an impaired response to bronchodilator drugs. This study was designed to address this question. METHODS Eight patients with clinical indication for thermoplasty were studied. They were uncontrolled severe asthmatics despite optimal environmental control, treatment of comorbidities, and the use of high-dose inhaled corticosteroids and long-acting β2-agonists. Lung function measured by spirometry and respiratory mechanics measured by oscillometry were examined pre- and post-bronchodilator (salbutamol, 400 μg), both before and at least 1 year after thermoplasty. RESULTS Consistent with previous studies, thermoplasty yielded no benefits in terms of baseline lung function and respiratory mechanics, despite improving symptoms based on two asthma questionnaires (ACQ-5 and ACT-5). The response to salbutamol was also not affected by thermoplasty based on spirometric readouts, including forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC), and FEV1/FVC ratio. However, a significant interaction was observed between thermoplasty and salbutamol for two oscillometric readouts, namely reactance at 5 Hz (Xrs5) and reactance area (Ax), showing an attenuated response to salbutamol after thermoplasty. CONCLUSIONS Thermoplasty attenuates the response to a bronchodilator. We argue that this result is a physiological proof of therapeutic efficacy, consistent with the well-described effect of thermoplasty in reducing the amount of airway smooth muscle.
Collapse
Affiliation(s)
- Cyndi Henry
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Sabrina Biardel
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Magali Boucher
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Krystelle Godbout
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Jamila Chakir
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Andréanne Côté
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Michel Laviolette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada
| | - Ynuk Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ) - Université Laval, Québec, Canada.
| |
Collapse
|
10
|
Liao X, Gao S, Xie F, Wang K, Wu X, Wu Y, Gao W, Wang M, Sun J, Liu D, Xu W, Li Q. An underlying mechanism behind interventional pulmonology techniques for refractory asthma treatment: Neuro-immunity crosstalk. Heliyon 2023; 9:e20797. [PMID: 37867902 PMCID: PMC10585236 DOI: 10.1016/j.heliyon.2023.e20797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/11/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
Asthma is a common disease that seriously threatens public health. With significant developments in bronchoscopy, different interventional pulmonology techniques for refractory asthma treatment have been developed. These technologies achieve therapeutic purposes by targeting diverse aspects of asthma pathophysiology. However, even though these newer techniques have shown appreciable clinical effects, their differences in mechanisms and mutual commonalities still deserve to be carefully explored. Therefore, in this review, we summarized the potential mechanisms of bronchial thermoplasty, targeted lung denervation, and cryoablation, and analyzed the relationship between these different methods. Based on available evidence, we speculated that the main pathway of chronic airway inflammation and other pathophysiologic processes in asthma is sensory nerve-related neurotransmitter release that forms a "neuro-immunity crosstalk" and amplifies airway neurogenic inflammation. The mechanism of completely blocking neuro-immunity crosstalk through dual-ablation of both efferent and afferent fibers may have a leading role in the clinical efficacy of interventional pulmonology in the treatment of asthma and deserves further investigation.
Collapse
Affiliation(s)
- Ximing Liao
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shaoyong Gao
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fengyang Xie
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun Wang
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaodong Wu
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yin Wu
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Gao
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Muyun Wang
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiaxing Sun
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dongchen Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, China
| | - Wujian Xu
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Li
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Wang T, Fu P, Long F, Liu S, Hu S, Wang Q, Huang Z, Long L, Huang W, Hu F, Gan J, Dong H, Yan G. Research on the effectiveness and safety of bronchial thermoplasty in patients with chronic obstructive pulmonary disease. Eur J Med Res 2023; 28:331. [PMID: 37689769 PMCID: PMC10492361 DOI: 10.1186/s40001-023-01319-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 08/27/2023] [Indexed: 09/11/2023] Open
Abstract
OBJECTIVES To investigate the clinical efficacy and safety of bronchial thermoplasty (BT) in treating patients with chronic obstructive pulmonary disease (COPD). METHODS Clinical data of 57 COPD patients were randomized into the control (n = 29, conventional inhalation therapy) or intervention group (n = 28, conventional inhalation therapy plus BT). Primary outcomes were differences in clinical symptom changes, pulmonary function-related indicators, modified Medical Research Council (mMRC), 6-min walk test (6MWT), COPD assessment test (CAT) score and acute exacerbation incidence from baseline to an average of 3 and 12 months. Safety was assessed by adverse events. RESULTS FEV1, FEV1(%, predicted) and FVC in both groups improved to varying degrees post-treatment compared with those pre-treatment (P < 0.05). The Intervention group showed greater improving amplitudes of FEV1 (Ftime × between groups = 21.713, P < 0.001) and FEV1(%, predicted) (Ftime × between groups = 31.216, P < 0.001) than the control group, and there was no significant difference in FVC variation trend (Ftime × between groups = 1.705, P = 0.193). mMRC, 6MWT and CAT scores of both groups post-treatment improved to varying degrees (Ps < 0.05), but the improving amplitudes of mMRC (Ftime × between groups = 3.947, P = 0.025), 6MWT (Ftime × between groups = 16.988, P < 0.001) and CAT score (Ftime × between groups = 16.741, P < 0.001) in the intervention group were greater than the control group. According to risk assessment of COPD acute exacerbation, the proportion of high-risk COPD patients with acute exacerbation in the control and intervention groups at 1 year post-treatment (100% vs 65%, 100% vs 28.6%), inpatient proportion (100% vs 62.1%; 100% vs 28.6%), COPD acute exacerbations [3.0 (2.50, 5.0) vs 1.0 (1.0, 2.50); 3.0(3.0, 4.0) vs 0 (0, 1.0)] and hospitalizations [2.0 (2.0, 3.0) vs 1.0 (0, 2.0); 2.0 (2.0, 3.0) vs 0 (0, 1.0)] were significantly lower than those pre-treatment (P < 0.05). Besides, data of the intervention group were significantly lower than the control group at each timepoint after treatment (P < 0.05). CONCLUSIONS Combined BT therapy is superior to conventional medical treatment in improving lung function and quality of life of COPD patients, and it also significantly reduces the COPD exacerbation risk without causing serious adverse events.
Collapse
Affiliation(s)
- Tao Wang
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
- The First Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
| | - Peng Fu
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Fa Long
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China.
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, People's Republic of China.
| | - Siyu Hu
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Qiongping Wang
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
- The First Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, People's Republic of China
| | - Zhihui Huang
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Liang Long
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Wenting Huang
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Fengbo Hu
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Jingfan Gan
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Hongbo Dong
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| | - Guomei Yan
- University of Chinese Academy of Sciences Shenzhen Hospital, No. 4253, Songbai Road, Guangming District, Shenzhen, 518106, People's Republic of China
| |
Collapse
|
12
|
Schleich F, Bougard N, Moermans C, Sabbe M, Louis R. Cytokine-targeted therapies for asthma and COPD. Eur Respir Rev 2023; 32:32/168/220193. [PMID: 37076177 PMCID: PMC10113955 DOI: 10.1183/16000617.0193-2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/23/2023] [Indexed: 04/21/2023] Open
Abstract
Asthma affects over 300 million people worldwide and its prevalence is increasing. COPD is the third leading cause of death globally. Asthma and COPD are complex inflammatory diseases of the airways in which impaired host defences lead to increased susceptibility to pathogens, pollutants and allergens. There is a constant interplay between host and the environment. Environmental exposures can alter the lung microbiome and influence the development of sensitisation by disrupting normal immunoregulation. The underlying airway inflammation in severe asthma is heterogeneous, with upregulation of type 2 cytokines in most cases but increased neutrophilic inflammation and activated T-helper 17 mediated immunity in others. COPD may also comprise several different phentoypes that are driven by different molecular mechanisms or endotypes. This disease heterogeneity is affected by comorbidities, treatments and environmental exposures. Recent intervention trials have shed light on the pathways beyond type 2 inflammation that can lead to beneficial outcomes versus potentially deleterious effects. We have made a great deal of progress over the last 10 years in terms of immunology and the pathophysiology of asthma and this has led to the development of novel treatments and major improvements in severe asthma outcomes. In COPD, however, no targeted treatments have demonstrated great improvements. This article reviews the mechanism of action and efficacy of the available biologics in asthma and COPD.
Collapse
Affiliation(s)
- Florence Schleich
- Respiratory Medicine, CHU of Liege, Belgium
- GIGA I3, University of Liege, Belgium
| | | | | | - Mare Sabbe
- Respiratory Medicine, CHU of Liege, Belgium
| | - Renaud Louis
- Respiratory Medicine, CHU of Liege, Belgium
- GIGA I3, University of Liege, Belgium
| |
Collapse
|
13
|
Stolz D, Matera MG, Rogliani P, van den Berge M, Papakonstantinou E, Gosens R, Singh D, Hanania N, Cazzola M, Maitland-van der Zee AH, Fregonese L, Mathioudakis AG, Vestbo J, Rukhadze M, Page CP. Current and future developments in the pharmacology of asthma and COPD: ERS seminar, Naples 2022. Breathe (Sheff) 2023; 19:220267. [PMID: 37377851 PMCID: PMC10292790 DOI: 10.1183/20734735.0267-2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/28/2023] [Indexed: 06/29/2023] Open
Abstract
Pharmacological management of airway obstructive diseases is a fast-evolving field. Several advances in unravelling disease mechanisms as well as intracellular and molecular pathways of drug action have been accomplished. While the clinical translation and implementation of in vitro results to the bedside remains challenging, advances in comprehending the mechanisms of respiratory medication are expected to assist clinicians and scientists in identifying meaningful read-outs and designing clinical studies. This European Respiratory Society Research Seminar, held in Naples, Italy, 5-6 May 2022, focused on current and future developments of the drugs used to treat asthma and COPD; on mechanisms of drug action, steroid resistance, comorbidities and drug interactions; on prognostic and therapeutic biomarkers; on developing novel drug targets based on tissue remodelling and regeneration; and on pharmacogenomics and emerging biosimilars. Related European Medicines Agency regulations are also discussed, as well as the seminar's position on the above aspects.
Collapse
Affiliation(s)
- Daiana Stolz
- Clinic of Pulmonary Medicine, Department of Internal Medicine, Medical Center University of Freiburg, Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel, Basel, Switzerland
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, and Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine, Department of Internal Medicine, Medical Center University of Freiburg, Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel, Basel, Switzerland
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD, and Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Nicola Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | | | | | - Alexander G. Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Maia Rukhadze
- Center of Allergy and Immunology, Teaching University Geomedi LLC, Tbilisi, Georgia
| | - Clive P. Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
14
|
Li A, Lee P. Which Endoscopic Procedure to Use and in What Patient? Valves, Coils, Foam, and Heat in COPD and Asthma. Pulm Ther 2022; 9:49-69. [PMID: 36534323 PMCID: PMC9931990 DOI: 10.1007/s41030-022-00208-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/10/2022] [Indexed: 12/23/2022] Open
Abstract
Despite the latest developments in therapeutic agents targeting airway endotypes, a significant proportion of patients with asthma and chronic obstructive pulmonary disease (COPD) remain symptomatic. Endoscopic therapies have a complementary role in the management of these airway diseases. The sustained efficacy of bronchial thermoplasty (BT) among patients with asthma over 10 years has been encouraging, as it has been shown to improve symptom control and reduce hospital admissions and exacerbations. Studies suggest that BT helps ameliorate airway inflammation and reduce airway smooth muscle thickness. While studies suggest that it is as effective as biologic agents, its role in the management of severe asthma has yet to be clearly defined and GINA 2022 still suggests limiting its use to patients with characteristics of the various populations studied. Conversely, bronchoscopic lung volume reduction has shown promise among patients with advanced COPD. Rigorous patient selection is important. Patients with minimal collateral ventilation (CV) and higher heterogeneity index have shown to benefit the most from endobronchial valve (EBV) therapy. For those with ongoing CV, endobronchial coils would be more appropriate. Both therapeutic modalities have demonstrated improved quality of life, effort tolerance, and lung function indices among appropriately selected patients. The emerging evidence suggests that endoscopic procedures among airway disease still have a substantial role to play despite the development of new therapeutic options.
Collapse
Affiliation(s)
- Andrew Li
- Respiratory and Critical Care Medicine, National University Hospital, 1E Kent Ridge Road, Singapore, 119228, Singapore. .,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Respiratory Service, Department of Medicine, Woodlands Health, Singapore, Singapore.
| | - Pyng Lee
- Respiratory and Critical Care Medicine, National University Hospital, 1E Kent Ridge Road, Singapore, 119228 Singapore ,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Chen J, Li S, Huang Z, Cao C, Wang A, He Q. METTL3 suppresses anlotinib sensitivity by regulating m 6A modification of FGFR3 in oral squamous cell carcinoma. Cancer Cell Int 2022; 22:295. [PMID: 36167542 PMCID: PMC9516809 DOI: 10.1186/s12935-022-02715-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/09/2022] [Indexed: 12/01/2022] Open
Abstract
Background N6-methyladenosine (m6A) is an abundant nucleotide modification in mRNA, but there were few studies on its role in cancer drug sensitivity and resistance. Anlotinib has been proved to have effective antitumor effects in oral squamous cell carcinoma (OSCC) in our previous study. Here, we sought to investigate the treatment target of anlotinib and the function and mechanisms of m6A modification in regulating anlotinib effect in OSCC. Methods Anlotinib treatment in a dose-dependent manner, western blotting, qRT-PCR and cell lost-of-function assays were used to study the treatment target of anlotinib in OSCC. RNA m6A dot blot assays, the m6A MeRIP-seq and MeRIP-qPCR, RNA and protein stability assays were used to explore the m6A modification of the treatment target of anlotinib. Cell lost-of-function assays after METTL3 depletion were conducted to investigate the effect of m6A modification level on the therapeutic effect of anlotinib in OSCC. Patient-derived tumor xenograft (PDX) models and immunohistochemistry staining were performed to study the relationship of METTL3 and antitumor sensitivity of anlotinib in vivo. Results Anlotinib targeted FGFR3 in the treatment of OSCC and inhibited tumor cell proliferation and promoted apoptosis by inactivating the FGFR3/AKT/mTOR signaling pathway. METTL3 was identified to target and modify FGFR3 m6A methylation and then decrease the stability of mRNA. METTL3 expression level was related to the anlotinib sensitivity in OSCC cells in vitro and METTL3 knockdown promoted anlotinib sensitivity of OSCC cells by inhibiting the FGFR3 expression. PDX models samples furthermore showed that METTL3 and FGFR3 levels were tightly correlated with the anlotinib efficacy in OSCC. Conclusions In summary, our work revealed that FGFR3 was served as the treatment target of anlotinib and METTL3-mediated FGFR3 m6A modification played a critical function in the anlotinib sensitivity in OSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02715-7.
Collapse
Affiliation(s)
- Jie Chen
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.,Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510055, China
| | - Shuai Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.,Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - Zhexun Huang
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Congyuan Cao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Anxun Wang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Qianting He
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
16
|
Sesé L, Mahay G, Barnig C, Guibert N, Leroy S, Guilleminault L. [Markers of severity and predictors of response to treatment in severe asthma]. Rev Mal Respir 2022; 39:740-757. [PMID: 36115752 DOI: 10.1016/j.rmr.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/19/2022] [Indexed: 10/14/2022]
Abstract
Asthma is a multifactorial disease with complex pathophysiology. Knowledge of its immunopathology and inflammatory mechanisms is progressing and has led to the development over recent years of increasingly targeted therapeutic strategies. The objective of this review is to pinpoint the different predictive markers of asthma severity and therapeutic response. Obesity, nasal polyposis, gastroesophageal reflux disease and intolerance to aspirin have all been considered as clinical markers associated with asthma severity, as have functional markers such as bronchial obstruction, low FEV1, small daily variations in FEV1, and high FeNO. While sinonasal polyposis and allergic comorbidities are associated with better response to omalizumab, nasal polyposis or long-term systemic steroid use are associated with better response to antibodies targeting the IL5 pathway. Elevated total IgE concentrations and eosinophil counts are classic biological markers regularly found in severe asthma. Blood eosinophils are predictive biomarkers of response to anti-IgE, anti-IL5, anti-IL5R and anti-IL4R biotherapies. Dupilumab is particularly effective in a subgroup of patients with marked type 2 inflammation (long-term systemic corticosteroid therapy, eosinophilia≥150/μl or FENO>20 ppb). Chest imaging may help to identify severe patients by seeking out bronchial wall thickening and bronchial dilation. Study of the patient's environment is crucial insofar as exposure to tobacco, dust mites and molds, as well as outdoor and indoor air pollutants (cleaning products), can trigger asthma exacerbation. Wider and more systematic use of markers of severity or response to treatment could foster increasingly targeted and tailored approaches to severe asthma.
Collapse
Affiliation(s)
- L Sesé
- AP-HP, service de physiologie, hôpital Avicenne, Bobigny, France
| | - G Mahay
- Service de pneumologie, oncologie thoracique et soins intensifs respiratoires, CHU Rouen, Rouen, France
| | - C Barnig
- INSERM, EFS BFC, LabEx LipSTIC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, Besançon, France; Service de pneumologie, oncologie thoracique et allergologie respiratoire, CHRU Besançon, Besançon, France
| | - N Guibert
- AP-HP, service de physiologie, hôpital Avicenne, Bobigny, France
| | - S Leroy
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, CNRS UMR 7275-FHU OncoAge, service de pneumologie oncologie thoracique et soins intensifs respiratoires, CHU de Nice, hôpital Pasteur, Nice, France
| | - L Guilleminault
- AP-HP, service de physiologie, hôpital Avicenne, Bobigny, France; Institut Toulousain des maladies infectieuses et inflammatoires (Infinity) inserm UMR1291-CNRS UMR5051-université Toulouse III, CRISALIS F-CRIN, Toulouse, France.
| |
Collapse
|
17
|
Hassoun D, Rose L, Blanc FX, Magnan A, Loirand G, Sauzeau V. Bronchial smooth muscle cell in asthma: where does it fit? BMJ Open Respir Res 2022; 9:9/1/e001351. [PMID: 36109087 PMCID: PMC9478857 DOI: 10.1136/bmjresp-2022-001351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/04/2022] [Indexed: 11/04/2022] Open
Abstract
Asthma is a frequent respiratory condition whose pathophysiology relies on altered interactions between bronchial epithelium, smooth muscle cells (SMC) and immune responses. Those leads to classical hallmarks of asthma: airway hyper-responsiveness, bronchial remodelling and chronic inflammation. Airway smooth muscle biology and pathophysiological implication in asthma are now better understood. Precise deciphering of intracellular signalling pathways regulating smooth muscle contraction highlighted the critical roles played by small GTPases of Rho superfamily. Beyond contractile considerations, active involvement of airway smooth muscle in bronchial remodelling mechanisms is now established. Not only cytokines and growth factors, such as fibroblats growth factor or transforming growth factor-β, but also extracellular matrix composition have been demonstrated as potent phenotype modifiers for airway SMC. Although basic science knowledge has grown significantly, little of it has translated into improvement in asthma clinical practice. Evaluation of airway smooth muscle function is still limited to its contractile activity. Moreover, it relies on tools, such as spirometry, that give only an overall assessment and not a specific one. Interesting technics such as forced oscillometry or specific imagery (CT and MRI) give new perspectives to evaluate other aspects of airway muscle such as bronchial remodelling. Finally, except for the refinement of conventional bronchodilators, no new drug therapy directly targeting airway smooth muscle proved its efficacy. Bronchial thermoplasty is an innovative and efficient therapeutic strategy but is only restricted to a small proportion of severe asthmatic patients. New diagnostic and therapeutic strategies specifically oriented toward airway smooth muscle are needed to improve global asthma care.
Collapse
Affiliation(s)
- Dorian Hassoun
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Lindsay Rose
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, Pays de la Loire, France
| | - François-Xavier Blanc
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Antoine Magnan
- INRAe, UMR 0892, Hôpital Foch, Suresnes, France.,Université Versailles-Saint-Quentin-en-Yvelines Paris-Saclay, Versailles, France
| | - Gervaise Loirand
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, Pays de la Loire, France
| | - Vincent Sauzeau
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, Pays de la Loire, France
| |
Collapse
|
18
|
Facciolongo N, Bonacini M, Galeone C, Ruggiero P, Menzella F, Ghidoni G, Piro R, Scelfo C, Catellani C, Zerbini A, Croci S. Bronchial thermoplasty in severe asthma: a real-world study on efficacy and gene profiling. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2022; 18:39. [PMID: 35534846 PMCID: PMC9087992 DOI: 10.1186/s13223-022-00680-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022]
Abstract
Background Bronchial thermoplasty (BT) is an effective treatment in severe asthma. How to select patients who more likely benefit from BT is an unmet clinical need. Moreover, mechanisms of BT efficacy are still largely unknown. We sought to determine BT efficacy and to identify potential mechanisms of response. Methods This retrospective cohort study evaluated clinical outcomes in 27 patients with severe asthma: 13 with T2-high and 14 with T2-low endotype. Expression levels of 20 genes were compared by real-time PCR in bronchial biopsies performed at the third BT session versus baseline. Clinical response was measured based on Asthma Control Questionnaire (ACQ) score < 1.5, asthma exacerbations < 2, oral corticosteroids reduction of at least 50% at 12 months post-BT. Patients were classified as responders when they had at least 2 of 3 outcome measures. Results 81% of patients were defined as responders. BT induced a reduction in alpha smooth muscle actin (ACTA2) and an increase in CD68, fibroblast activation protein-alpha (FAP), alpha-1 and alpha-2 type I collagen (COL1A1, COL1A2) gene expression in the majority of patients. A higher reduction in ubiquitin carboxy-terminal-hydrolase L1 (PGP9.5) mRNA correlated with a better response based on Asthma Quality of Life Questionnaire (AQLQ). Lower changes in CD68 and FAP mRNAs correlated with a better response based on ACQ. Lower levels of occludin (OCLN), CD68, connective tissue growth factor (CTGF), higher levels of secretory leukocyte protease inhibitor (SLPI) and lower changes in CD68 and CTGF mRNAs were observed in patients who had less than 2 exacerbations post-BT. Lower levels of COL1A2 at baseline were observed in patients who had ACQ < 1.5 at 12 months post-BT. Conclusions BT is effective irrespective of the asthma endotypes and seems associated with airway remodelling. Quantification of OCLN, CD68, CTGF, SLPI, COL1A2 mRNAs could be useful to identify patients with better results. Trial registration: The study protocol was approved by the Local Ethics Committee (Azienda USL-IRCCS of Reggio Emilia—Comitato Etico Area Vasta Nord of Emilia Romagna; protocol number: 2019/0014076) and all the patients provided written informed consent before participating in the study. Supplementary Information The online version contains supplementary material available at 10.1186/s13223-022-00680-4.
Collapse
Affiliation(s)
- Nicola Facciolongo
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Martina Bonacini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Carla Galeone
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Patrizia Ruggiero
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Menzella
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy. .,Department of Medical Specialties, Pneumology Unit, Arcispedale Santa Maria Nuova, Azienda Unità Sanitaria Locale-IRCCS, 42123, Reggio Emilia, Italy.
| | - Giulia Ghidoni
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Roberto Piro
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Chiara Scelfo
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Chiara Catellani
- Pneumology Unit, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Zerbini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Stefania Croci
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
19
|
Carr TF, Peters MC. Novel potential treatable traits in asthma: Where is the research taking us? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2022; 1:27-36. [PMID: 37780590 PMCID: PMC10509971 DOI: 10.1016/j.jacig.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 10/03/2023]
Abstract
Asthma is a complex, heterogeneous disease in which the underlying mechanisms are not fully understood. Patients are often grouped into phenotypes (based on clinical, biologic, and physiologic characteristics) and endotypes (based on distinct genetic or molecular mechanisms). Recently, patients with asthma have been broadly split into 2 phenotypes based on their levels of type 2 inflammation: type 2 and non-type 2 asthma. However, this approach is likely oversimplified, and our understanding of the non-type 2 mechanisms in asthma remains extremely limited. A better understanding of asthma phenotypes and endotypes may assist in development of drugs for new therapeutic targets in asthma. One approach is to identify "treatable traits," which are specific patient characteristics related to phenotypes and endotypes that can be targeted by therapies. This review will focus on emerging treatable traits in asthma and aim to describe novel patient subgroups and endotypes that may represent the next step in the search for new therapeutic approaches.
Collapse
Affiliation(s)
- Tara F. Carr
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Michael C. Peters
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, Calif
| |
Collapse
|
20
|
Jendzjowsky N, Laing A, Malig M, Matyas J, de Heuvel E, Dumonceaux C, Dumoulin E, Tremblay A, Leigh R, Chee A, Kelly MM. Long-term modulation of airway remodelling in severe asthma following bronchial thermoplasty. Eur Respir J 2022; 59:2100622. [PMID: 34049950 DOI: 10.1183/13993003.00622-2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/16/2021] [Indexed: 11/05/2022]
Abstract
RationaleBronchial thermoplasty is a mechanical therapeutic intervention that has been advocated as an effective treatment option for severe asthma. The mechanism is promoted as being related to the attenuation of airway smooth muscle which has been shown to occur in the short-term. However, long-term studies of the effects of bronchial thermoplasty on airway remodelling are few, with only limited assessment of airway remodelling indices.ObjectivesTo evaluate the effect of bronchial thermoplasty on 1) airway epithelial and smooth muscle cells in culture and 2) airway remodelling in patients with severe asthma who have been prescribed bronchial thermoplasty up to 12 months post-treatment.MethodsThe distribution of heat within the airway by bronchial thermoplasty was assessed in a porcine model. Culture of human airway smooth muscle cells and bronchial epithelial cells evaluated the impact of thermal injury. Histological evaluation and morphometric assessment were performed on bronchial biopsies obtained from severe asthma patients at baseline, 6 weeks and 12 months following bronchial thermoplasty.ResultsBronchial thermoplasty resulted in heterogeneous heating of the airway wall. Airway smooth muscle cell cultures sustained thermal injury, whilst bronchial epithelial cells were relatively resistant to heat. Airway smooth muscle and neural bundles were significantly reduced at 6 weeks and 12 months post-treatment. At 6 weeks post-treatment, submucosal collagen was reduced and vessel density increased, with both indices returning to baseline at 12 months. Goblet cell numbers, submucosal gland area and sub-basement membrane thickness were not significantly altered at any time point examined.ConclusionsBronchial thermoplasty primarily affects airway smooth muscle and nerves with the effects still present at 12 months post-treatment.
Collapse
Affiliation(s)
- Nicholas Jendzjowsky
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Authors contributed equally
| | - Austin Laing
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Authors contributed equally
| | - Michelle Malig
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - John Matyas
- Dept of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Elaine de Heuvel
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Curtis Dumonceaux
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Elaine Dumoulin
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alain Tremblay
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Richard Leigh
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Alex Chee
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Authors contributed equally
| | - Margaret M Kelly
- Dept of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Authors contributed equally
| |
Collapse
|
21
|
Fang L, Roth M. Airway Wall Remodeling in Childhood Asthma-A Personalized Perspective from Cell Type-Specific Biology. J Pers Med 2021; 11:jpm11111229. [PMID: 34834581 PMCID: PMC8625708 DOI: 10.3390/jpm11111229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/16/2022] Open
Abstract
Airway wall remodeling is a pathology occurring in chronic inflammatory lung diseases including asthma, chronic obstructive pulmonary disease, and fibrosis. In 2017, the American Thoracic Society released a research statement highlighting the gaps in knowledge and understanding of airway wall remodeling. The four major challenges addressed in this statement were: (i) the lack of consensus to define “airway wall remodeling” in different diseases, (ii) methodologic limitations and inappropriate models, (iii) the lack of anti-remodeling therapies, and (iv) the difficulty to define endpoints and outcomes in relevant studies. This review focuses on the importance of cell-cell interaction, especially the bronchial epithelium, in asthma-associated airway wall remodeling. The pathology of “airway wall remodeling” summarizes all structural changes of the airway wall without differentiating between different pheno- or endo-types of asthma. Indicators of airway wall remodeling have been reported in childhood asthma in the absence of any sign of inflammation; thus, the initiation event remains unknown. Recent studies have implied that the interaction between the epithelium with immune cells and sub-epithelial mesenchymal cells is modified in asthma by a yet unknown epigenetic mechanism during early childhood.
Collapse
|
22
|
IPF-Fibroblast Erk1/2 Activity Is Independent from microRNA Cluster 17-92 but Can Be Inhibited by Treprostinil through DUSP1. Cells 2021; 10:cells10112836. [PMID: 34831059 PMCID: PMC8616195 DOI: 10.3390/cells10112836] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/25/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive terminal lung disease, and therapies aim to block fibrosis. Fibroblast proliferation is controlled by C/EBP-β, microRNA cluster 17-92 (miR17-92), and Erk1/2 mitogen-activated protein kinase. This study assessed the role of miR17-92 in IPF-fibroblast proliferation and its modification by treprostinil. Fibroblasts were isolated from eight IPF patients, five interstitial lung fibrosis patients, and seven control lungs. Fibroblasts were stimulated with TGF-β1 over 24 h. The miR17-92 expression was analyzed by RT-qPCR, and protein expression by Western blotting. TGF-β1 upregulated C/EBP-β in all fibroblasts, which was reduced by treprostinil in control-fibroblasts, but not in IPF-fibroblasts. Compared to controls, the guide strands miR-19a-3p, miR-19b-3p, miR-20a-5p, and miR-92a-3p, as well as the passenger strands miR-17-3p, miR-18-3p, miR-19a-1-5p, and miR-92a-5p were significantly increased in IPF-fibroblasts. In controls, TGF-β1 and treprostinil significantly reduced specific miR17-92 members. IPF-fibroblast proliferation was inhibited by treprostinil through increased expression of the Erk1/2 inhibitor DUSP1. These data suggest that proliferation control via miR17-92 and C/EBP-β is disrupted in IPF-fibroblasts. Therefore, the inhibition of early stages of signaling cascades or specific mitogen receptors might be less effective. However, the increased proliferation is sensitive to Erk1/2 inhibition by treprostinil-induced DUSP1.
Collapse
|
23
|
Bronchial thermoplasty: The heat that reprograms the airways? J Allergy Clin Immunol 2021; 148:1167-1169. [PMID: 34509527 DOI: 10.1016/j.jaci.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
|
24
|
Zhang M, Fang L, Zhou L, Molino A, Valentino MR, Yang S, Zhang J, Li Y, Roth M. MAPK15-ULK1 signaling regulates mitophagy of airway epithelial cell in chronic obstructive pulmonary disease. Free Radic Biol Med 2021; 172:541-549. [PMID: 34224814 DOI: 10.1016/j.freeradbiomed.2021.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/23/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
Airway epithelial mitochondrial oxidative stress and damage is an important pathology in chronic obstructive pulmonary disease (COPD). Mitophagy involves MAPK15-ULK1 signaling, the role of which is unknown in COPD. This study investigated MAPK15-ULK1 signaling in airway epithelial cells of COPD patients and its activation by cigarette smoke extract (CSE) in isolated human airway epithelial cells. Significant increased phosphorylation of MAPK15 and ULK1 (Ser555) was detected in the airway epithelium of COPD patients. This pathology was maintained in isolated primary COPD-epithelial cells. Compared to control cells, the protein expression of Beclin1 and the ratio of LC3II to LC3I were both significantly increased in COPD-epithelial cells. In human airway epithelial cells, CSE significantly increased the phosphorylation of MAPK15, ULK1 (Ser555), the expression of Beclin1, and the LC3II/LC3I ratio in a concentration- and time-dependent manner. Transfection with MAPK15 siRNA significantly inhibited the CSE-induced ULK1 (Ser555) phosphorylation in airway epithelial cells. Silencing of MAPK15 or ULK1 significantly reduced CSE-induced mitophagy and mitochondrial oxidative stress, thereby improving cell viability. In summary, cigarette smoke activated MAPK15-ULK1 signaling, thereby promoting mitophagy and mitochondrial oxidative stress in airway epithelial cells. This signaling pathway is activated in COPD-epithelial cells and therefore might present a novel therapeutic target for COPD.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Pulmonary Cell Research & Clinic of Respiratory Medicine, Dept. Biomedicine University of Basel & University Hospital of Basel, CH-4031, Basel, Switzerland
| | - Lei Fang
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Dept. Biomedicine University of Basel & University Hospital of Basel, CH-4031, Basel, Switzerland
| | - Liang Zhou
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Dept. Biomedicine University of Basel & University Hospital of Basel, CH-4031, Basel, Switzerland
| | - Antonio Molino
- Department of Respiratory Diseases, University of Naples, Federico II, Naples, Italy
| | | | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yali Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Michael Roth
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Dept. Biomedicine University of Basel & University Hospital of Basel, CH-4031, Basel, Switzerland.
| |
Collapse
|
25
|
Ano S, Kikuchi N, Matsuyama M, Nakajima M, Kondo Y, Masuda M, Osawa H, Ishii Y, Hizawa N. Transcriptome genetic differences between responders and non-responders before bronchial thermoplasty. J Asthma 2021; 59:1641-1651. [PMID: 34143700 DOI: 10.1080/02770903.2021.1945088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Bronchial thermoplasty (BT) is an endoscopic therapy used for the treatment of refractory asthma. Some predictive factors, for example the number of activations and severity of disease at baseline, have been used to determine the effectiveness of BT in treating patients with asthma. The aim of the present study was to comprehensively analyze RNA samples from the airway bronchial tissues of patients with severe asthma treated by BT, and to characterize each patient as a BT responder or non-responder. METHODS Eight patients with severe asthma scheduled to undergo BT and bronchus biopsies were recruited before the procedures were conducted. Extracted RNA samples from bronchial tissues were sequenced and differential gene expression analysis was carried out.Results/discussion: Subjects with Asthma Quality of Life Questionnaire score changes ≥0.5 for a period of 12 months were considered BT responders. Non-responders had score changes <0.5 for 12 months. Histopathology findings were similar to those reported previously, and no significant differences in the expression of α-smooth muscle actin and protein gene product 9.5 were observed between responders and non-responders. Transcriptome analysis at baseline identified 67 genes that were differentially expressed between responders and non-responders, including SLPI, MMP3, and MUC19, which were upregulated in responders. Although the differentially expressed gene products may have conflicting effects, genes in the airway epithelium and extracellular matrix of patients with severe asthma may determine the BT response. Our results identified possible transcriptomic changes that could be used to identify BT responders.
Collapse
Affiliation(s)
- Satoshi Ano
- The Department of Respiratory Medicine, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan
| | - Norihiro Kikuchi
- The Department of Respiratory Medicine, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan
| | - Masashi Matsuyama
- The Department of Respiratory Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masayuki Nakajima
- The Department of Respiratory Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuzuru Kondo
- The Department of Diagnostic Pathology, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan
| | - Michiko Masuda
- The Department of Respiratory Medicine, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan
| | - Hajime Osawa
- The Department of Respiratory Medicine, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan.,The Department of Respiratory Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukio Ishii
- The Department of Respiratory Medicine, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan.,The Department of Respiratory Medicine, University of Tsukuba, Tsukuba, Japan
| | - Nobuyuki Hizawa
- The Department of Respiratory Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
26
|
Papakonstantinou E, Koletsa T, Zhou L, Fang L, Roth M, Karakioulaki M, Savic S, Grize L, Tamm M, Stolz D. Bronchial thermoplasty in asthma: an exploratory histopathological evaluation in distinct asthma endotypes/phenotypes. Respir Res 2021; 22:186. [PMID: 34183014 PMCID: PMC8240300 DOI: 10.1186/s12931-021-01774-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 11/24/2022] Open
Abstract
Background Bronchial thermoplasty regulates structural abnormalities involved in airway narrowing in asthma. In the present study we aimed to investigate the effect of bronchial thermoplasty on histopathological bronchial structures in distinct asthma endotypes/phenotypes.
Methods Endobronchial biopsies (n = 450) were collected from 30 patients with severe uncontrolled asthma before bronchial thermoplasty and after 3 sequential bronchial thermoplasties. Patients were classified based on blood eosinophils, atopy, allergy and smoke exposure. Tissue sections were assessed for histopathological parameters and expression of heat-shock proteins and glucocorticoid receptor. Proliferating cells were determined by Ki67-staining. Results In all patients, bronchial thermoplasty improved asthma control (p < 0.001), reduced airway smooth muscle (p = 0.014) and increased proliferative (Ki67 +) epithelial cells (p = 0.014). After bronchial thermoplasty, airway smooth muscle decreased predominantly in patients with T2 high asthma endotype. Epithelial cell proliferation was increased after bronchial thermoplasty in patients with low blood eosinophils (p = 0.016), patients with no allergy (p = 0.028) and patients without smoke exposure (p = 0.034).
In all patients, bronchial thermoplasty increased the expression of glucocorticoid receptor in epithelial cells (p = 0.018) and subepithelial mesenchymal cells (p = 0.033) and the translocation of glucocorticoid receptor in the nucleus (p = 0.036). Furthermore, bronchial thermoplasty increased the expression of heat shock protein-70 (p = 0.002) and heat shock protein-90 (p = 0.001) in epithelial cells and decreased the expression of heat shock protein-70 (p = 0.009) and heat shock protein-90 (p = 0.002) in subepithelial mesenchymal cells. The effect of bronchial thermoplasty on the expression of heat shock proteins -70 and -90 was distinctive across different asthma endotypes/phenotypes. Conclusions Bronchial thermoplasty leads to a diminishment of airway smooth muscle, to epithelial cell regeneration, increased expression and activation of glucocorticoid receptor in the airways and increased expression of heat shock proteins in the epithelium. Histopathological effects appear to be distinct in different endotypes/phenotypes indicating that the beneficial effects of bronchial thermoplasty are achieved by diverse molecular targets associated with asthma endotypes/phenotypes. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01774-0.
Collapse
Affiliation(s)
- Eleni Papakonstantinou
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.,Laboratory of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Triantafyllia Koletsa
- Department of Pathology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Liang Zhou
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Lei Fang
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Michael Roth
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Meropi Karakioulaki
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Spasenija Savic
- Department of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Leticia Grize
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Michael Tamm
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel and Department of Biomedicine, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
| |
Collapse
|
27
|
TSLP-induced collagen type-I synthesis through STAT3 and PRMT1 is sensitive to calcitriol in human lung fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119083. [PMID: 34147561 DOI: 10.1016/j.bbamcr.2021.119083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022]
Abstract
Airway wall remodeling, a main pathology of asthma was linked to vitamin-D deficiency and protein arginine methyltransferase-1 (PRMT1) expression in sub-epithelial cell layers. Calcitriol reduced remodeling in asthma model, but its mode of action is unclear. This study assessed the effect of calcitriol on PRMT1-dependent fibroblast remodeling in human lung fibroblasts, and allergen-induced asthma in E3-rats. Fibroblasts were activated with thymic stromal lymphopoietin (TLSP); asthma was induced by ovalbumin inhalation in rats. The airway structure was assessed by immunohistology. Protein expression in fibroblasts and activation of the mitogen activated protein kinases were detected by Western-blotting. Transcription factor activation was determined by luciferase reporter assay. PRMT1 action was blocked by siRNA and PRMT-inhibition. Ovalbumin upregulated the expression of TSLP, PRMT1, matrix metallopro-teinase-1 (MMP1), interleukin-25, and collagen type-I in sub-epithelial fibroblasts. In isolated fibroblasts, TSLP induced the same proteins, which were blocked by inhibition of Erk1/2 and p38. TLSP induced PRMT1 through activation of signal transducer and activator of transcription-3. PRMT1 inhibition reduced collagen type-I expression and suppressed MMP1. In fibroblasts, calcitriol supplementation over 12 days prevented TSLP-induced remodeling by blocking the PRMT1 levels. Interestingly, short-term calcitriol treatment had no such effect. The data support the beneficial role of calcitriol in asthma therapy.
Collapse
|
28
|
Mast AE, Wolberg AS, Gailani D, Garvin MR, Alvarez C, Miller JI, Aronow B, Jacobson D. SARS-CoV-2 suppresses anticoagulant and fibrinolytic gene expression in the lung. eLife 2021; 10:e64330. [PMID: 33683204 PMCID: PMC8049742 DOI: 10.7554/elife.64330] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/06/2021] [Indexed: 12/13/2022] Open
Abstract
Extensive fibrin deposition in the lungs and altered levels of circulating blood coagulation proteins in COVID-19 patients imply local derangement of pathways that limit fibrin formation and/or promote its clearance. We examined transcriptional profiles of bronchoalveolar lavage fluid (BALF) samples to identify molecular mechanisms underlying these coagulopathies. mRNA levels for regulators of the kallikrein-kinin (C1-inhibitor), coagulation (thrombomodulin, endothelial protein C receptor), and fibrinolytic (urokinase and urokinase receptor) pathways were significantly reduced in COVID-19 patients. While transcripts for several coagulation proteins were increased, those encoding tissue factor, the protein that initiates coagulation and whose expression is frequently increased in inflammatory disorders, were not increased in BALF from COVID-19 patients. Our analysis implicates enhanced propagation of coagulation and decreased fibrinolysis as drivers of the coagulopathy in the lungs of COVID-19 patients.
Collapse
Affiliation(s)
- Alan E Mast
- Versiti Blood Research Institute, Department of Cell Biology Neurobiology and Anatomy Medical College of WisconsinMilwaukeeUnited States
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research CenterChapel HillUnited States
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashvilleUnited States
| | - Michael R Garvin
- Oak Ridge National Laboratory, Biosciences DivisionOak RidgeUnited States
| | - Christiane Alvarez
- Oak Ridge National Laboratory, Biosciences DivisionOak RidgeUnited States
| | - J Izaak Miller
- Oak Ridge National Laboratory, Biosciences DivisionOak RidgeUnited States
| | - Bruce Aronow
- University of Tennessee Knoxville, The Bredesen Center for Interdisciplinary Research and Graduate EducationKnoxvilleUnited States
- Biomedical Informatics, Cincinnati Children’s Hospital Research FoundationCincinnatiUnited States
- University of CincinnatiCincinnatiUnited States
| | - Daniel Jacobson
- Oak Ridge National Laboratory, Biosciences DivisionOak RidgeUnited States
- University of Tennessee Knoxville, The Bredesen Center for Interdisciplinary Research and Graduate EducationKnoxvilleUnited States
- University of Tennessee Knoxville, Department of PsychologyKnoxvilleUnited States
| |
Collapse
|
29
|
Fang L, Li J, Papakonstantinou E, Karakioulaki M, Sun Q, Schumann D, Tamm M, Stolz D, Roth M. Secreted heat shock proteins control airway remodeling: Evidence from bronchial thermoplasty. J Allergy Clin Immunol 2021; 148:1249-1261.e8. [PMID: 33675818 DOI: 10.1016/j.jaci.2021.02.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Increased airway smooth muscle mass is a key pathology in asthma. Bronchial thermoplasty is a treatment for severe asthma based on selective heating of the airways that aims to reduce the mass of airway smooth muscle cells (ASMCs), and thereby bronchoconstriction. However, short heat exposure is insufficient to explain the long-lasting effect, and heat shock proteins (HSPs) have been suggested to play a role. OBJECTIVE We sought to determine the role of HSP70 and HSP90 in the control of airway wall remodeling by bronchial thermoplasty. METHODS Bronchoalveolar lavage fluid and endobronchial biopsies of 20 patients with severe asthma were obtained before and after thermoplasty. Isolated epithelial cells and ASMCs were exposed to 65oC for 10 seconds, mimicking thermoplasty. Proteins were determined by immunohistochemistry, Western blotting, immunofluorescence, and ELISA; proliferation by cell counts and antigen Ki67 (MKI67) expression. RESULTS Thermoplasty significantly increased the expression of HSP70 and HSP90 in the epithelium and bronchoalveolar lavage fluid. In ASMCs, thermoplasty reduced both HSPs. These cell-type-specific effects were detectable even 1 month after thermoplasty in tissue sections. In epithelial cells, ex vivo exposure to heat (65oC, 10 seconds) increased the expression and secretion of HSP70 and HSP90. In addition, epithelial cell proliferation was upregulated by heat or treatment with human recombinant HSP70 or HSP90. In ASMCs, heat exposure or exogenous HSPs reduced proliferation and differentiation. In both cell types, HSP70 and HSP90 activated the signaling cascade of serine/threonine-protein kinase →mammalian target of rapamycin→ribosomal protein S6 kinase 1 and CCAAT/enhancer binding protein-β→protein arginine methyltransferase 1→ mitochondria activity. CONCLUSIONS Epithelial cell-derived HSP70 and HSP90 improve the function of epithelial cells, but block ASMC remodeling.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland
| | - Junling Li
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland; The affiliated Dongguan Shilong People's Hospital of Southern Medical University, Guangdong, China
| | - Eleni Papakonstantinou
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland
| | - Meropi Karakioulaki
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland
| | - Qingzhu Sun
- College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi, China
| | - Desiree Schumann
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland
| | - Michael Tamm
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland
| | - Daiana Stolz
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research & Clinic of Respiratory Medicine, Department of Biomedicine, University of Basel & University Hospital of Basel, Basel, Switzerland.
| |
Collapse
|
30
|
Héluain V, Guilleminault L, Hermant C, Plat G, Didier A, Guibert N. [Bronchial thermoplasty for severe asthma]. Rev Mal Respir 2021; 38:289-296. [PMID: 33531186 DOI: 10.1016/j.rmr.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/13/2020] [Indexed: 11/29/2022]
Abstract
Bronchial thermoplasty has been developed over the past fifteen years and is the first endoscopic technique approved in the management of severe asthma. This procedure uses radiofrequency applied to the airway wall to target bronchial smooth muscle. Patients treated in randomized controlled trials have experienced significant decreases in the use of rescue medications, urgent care visits, and exacerbations rate. The lack of reliable predictive markers of response to this expensive, minimally-invasive technique currently makes it a last-line treatment option. We review the principles and supposed mechanisms of action of this treatment, the results from the main trials and clinical registry data and discuss the place of bronchial thermoplasty in the current management of severe asthma. We also discuss perspectives to better characterize the mechanisms of action and identify the responder phenotype, the main challenge of current studies.
Collapse
Affiliation(s)
- V Héluain
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, Toulouse, France
| | - L Guilleminault
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, Toulouse, France; Centre de physiopathologie Toulouse Purpan, INSERM U1043, CNRS UMR 5282, université Toulouse III, Toulouse, France; INSERM, F-CRIN, Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS), Toulouse, France
| | - C Hermant
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, Toulouse, France
| | - G Plat
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, Toulouse, France
| | - A Didier
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, Toulouse, France; Centre de physiopathologie Toulouse Purpan, INSERM U1043, CNRS UMR 5282, université Toulouse III, Toulouse, France; INSERM, F-CRIN, Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS), Toulouse, France
| | - N Guibert
- Service de pneumologie, hôpital Larrey, université Paul-Sabatier, CHU Toulouse, Toulouse, France.
| |
Collapse
|
31
|
Zhai W, Sun H, Li Z, Li L, Jin A, Li Y, Chen J, Yang X, Sun Q, Lu S, Roth M. PRMT1 Modulates Processing of Asthma-Related Primary MicroRNAs (Pri-miRNAs) into Mature miRNAs in Lung Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2020; 206:11-22. [PMID: 33239422 DOI: 10.4049/jimmunol.2000887] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/28/2020] [Indexed: 01/07/2023]
Abstract
Protein arginine methyltransferase-1 (PRMT1) is an important epigenetic regulator of cell function and contributes to inflammation and remodeling in asthma in a cell type-specific manner. Disease-specific expression patterns of microRNAs (miRNA) are associated with chronic inflammatory lung diseases, including asthma. The de novo synthesis of miRNA depends on the transcription of primary miRNA (pri-miRNA) transcript. This study assessed the role of PRMT1 on pri-miRNA to mature miRNA process in lung epithelial cells. Human airway epithelial cells, BEAS-2B, were transfected with the PRMT1 expression plasmid pcDNA3.1-PRMT1 for 48 h. Expression profiles of miRNA were determined by small RNA deep sequencing. Comparing these miRNAs with datasets of microarrays from five asthma patients (Gene Expression Omnibus dataset), 12 miRNAs were identified that related to PRMT1 overexpression and to asthma. The overexpression or knockdown of PRMT1 modulated the expression of the asthma-related miRNAs and their pri-miRNAs. Coimmunoprecipitation showed that PRMT1 formed a complex with STAT1 or RUNX1 and thus acted as a coactivator, stimulating the transcription of pri-miRNAs. Stimulation with TGF-β1 promoted the interaction of PRMT1 with STAT1 or RUNX1, thereby upregulating the transcription of two miRNAs: let-7i and miR-423. Subsequent chromatin immunoprecipitation assays revealed that the binding of the PRMT1/STAT1 or PRMT1/RUNX1 coactivators to primary let-7i (pri-let-7i) and primary miR (pri-miR) 423 promoter was critical for pri-let-7i and pri-miR-423 transcription. This study describes a novel role of PRMT1 as a coactivator for STAT1 or RUNX1, which is essential for the transcription of pri-let-7i and pri-miR-423 in epithelial cells and might be relevant to epithelium dysfunction in asthma.
Collapse
Affiliation(s)
- Weiqi Zhai
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haoming Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhi Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Li Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ai Jin
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yuwen Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jian Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China;
| | - Qingzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China; .,Pneumology and Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland; and
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Michael Roth
- Pneumology and Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland; and
| |
Collapse
|
32
|
Qiu M, Wei S, Lai Z, Huang P, Wang Z, Zhong C, Chen Y, Zhang X, Lin X, Zeng Q, Chung KF, Zhang Q, Xie J, Li S. Early radiologic and bronchoscopic changes after bronchial thermoplasty in patients with severe asthma. Exp Ther Med 2020; 20:278. [PMID: 33200003 PMCID: PMC7664609 DOI: 10.3892/etm.2020.9408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 10/06/2020] [Indexed: 11/24/2022] Open
Abstract
Bronchial thermoplasty (BT) is a treatment to reduce the airway smooth muscle mass by delivering radiofrequency thermal energy to the airways. BT is used in patients with severe asthma. The present study reported on cases of pneumothorax directly after BT and retrospectively analyzed early radiologic and bronchoscopic modifications after BT. The clinical data and radiologic and bronchoscopic findings of 12 patients with severe asthma who were subjected to BT between July 2014 and October 2017 were analyzed. A total of 33 chest radiographs were collected within 18-24 h after BT. Radiological abnormalities were observed in 32 radiographs as atelectasis (53.1%), peribronchial consolidations (84.4%), pleural effusion (18.8%), effusion in oblique fissures (3.1%), pleural thickening (6.3%) and pneumothorax (3.1%). Of note, one patient suffered pneumothorax after the third BT session and underwent chest drain insertion, followed by mechanical ventilation at the intensive care unit and multiple bronchoscopic interventions, which revealed extensive phlegm plugs. A total of six patients with worsened symptoms and lobar atelectasis also required bronchoscopic intervention, which revealed that phlegm plugs occluded the bronchus in the treated lobe. No bronchoscopic intervention was required in the remaining five patients. During 16-30 days of follow-up, 95.7% of the findings on chest radiography were resolved. To the best of our knowledge, the present study reported the first case of pneumothorax following BT. Early radiologic modifications such as atelectasis and peribronchial consolidations appear common after BT. However, whether bronchoscopic intervention is required for atelectasis following BT warrants further investigation. Of note, BT should be audited and recorded in detail to ideally contribute to a framework of clinical trials to improve risk-benefit evaluations and the selection of patients likely to benefit from treatment.
Collapse
Affiliation(s)
- Minzhi Qiu
- Department of Respiratory and Critical Care Medicine, Shenzhen Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China.,Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Shushan Wei
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhengdao Lai
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Peikai Huang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Zhiqiang Wang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Changhao Zhong
- Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Yu Chen
- Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiaoxian Zhang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiaofeng Lin
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Qingsi Zeng
- Department of Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College London and Royal Brompton and Harefield NHS Foundation Trust, London SW3 6LY, UK
| | - Qingling Zhang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Jiaxing Xie
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Shiyue Li
- Department of Respiratory and Critical Care Medicine, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
33
|
Fang L, Sun Q, Roth M. Immunologic and Non-Immunologic Mechanisms Leading to Airway Remodeling in Asthma. Int J Mol Sci 2020; 21:ijms21030757. [PMID: 31979396 PMCID: PMC7037330 DOI: 10.3390/ijms21030757] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Asthma increases worldwide without any definite reason and patient numbers double every 10 years. Drugs used for asthma therapy relax the muscles and reduce inflammation, but none of them inhibited airway wall remodeling in clinical studies. Airway wall remodeling can either be induced through pro-inflammatory cytokines released by immune cells, or direct binding of IgE to smooth muscle cells, or non-immunological stimuli. Increasing evidence suggests that airway wall remodeling is initiated early in life by epigenetic events that lead to cell type specific pathologies, and modulate the interaction between epithelial and sub-epithelial cells. Animal models are only available for remodeling in allergic asthma, but none for non-allergic asthma. In human asthma, the mechanisms leading to airway wall remodeling are not well understood. In order to improve the understanding of this asthma pathology, the definition of “remodeling” needs to be better specified as it summarizes a wide range of tissue structural changes. Second, it needs to be assessed if specific remodeling patterns occur in specific asthma pheno- or endo-types. Third, the interaction of the immune cells with tissue forming cells needs to be assessed in both directions; e.g., do immune cells always stimulate tissue cells or are inflamed tissue cells calling immune cells to the rescue? This review aims to provide an overview on immunologic and non-immunologic mechanisms controlling airway wall remodeling in asthma.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
| | - Qinzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China;
| | - Michael Roth
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
- Correspondence: ; Tel.: +41-61-265-2337
| |
Collapse
|
34
|
Rahmawati SF, Gosens R. Hot off the press: downregulation of PRMT1 for long-lasting effects of bronchial thermoplasty. Eur Respir J 2019; 54:54/6/1901898. [PMID: 31801822 DOI: 10.1183/13993003.01898-2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/28/2019] [Indexed: 11/05/2022]
Affiliation(s)
- Siti Farah Rahmawati
- Dept of Molecular Pharmacology and GRIAC Research Institute, University of Groningen, Groningen, The Netherlands.,Dept of Pharmacology-Clinical Pharmacy, School of Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
| | - Reinoud Gosens
- Dept of Molecular Pharmacology and GRIAC Research Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
35
|
Thomson NC. Recent Developments In Bronchial Thermoplasty For Severe Asthma. J Asthma Allergy 2019; 12:375-387. [PMID: 31819539 PMCID: PMC6875488 DOI: 10.2147/jaa.s200912] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Bronchial thermoplasty is approved in many countries worldwide as a non-pharmacological treatment for severe asthma. This review summarizes recent publications on the selection of patients with severe asthma for bronchial thermoplasty, predictors of a beneficial response and developments in the procedure and discusses specific issues about bronchial thermoplasty including effectiveness in clinical practice, mechanism of action, cost-effectiveness, and place in management. RESULTS Bronchial thermoplasty is a treatment option for patients with severe asthma after assessment and management of causes of difficult-to-control asthma, such as nonadherence, poor inhaler technique, comorbidities, under treatment, and other behavioral factors. Patients treated with bronchial thermoplasty in clinical practice have worse baseline characteristics and comparable clinical outcomes to clinical trial data. Bronchial thermoplasty causes a reduction in airway smooth muscle mass although it is uncertain whether this effect explains its efficacy since other mechanisms of action may be relevant, such as alterations in airway epithelial, gland, and/or nerve function; improvements in small airway function; or a placebo effect. The cost-effectiveness of bronchial thermoplasty is greater in countries where the costs of hospitalization and emergency department are high. The place of bronchial thermoplasty in the management of severe asthma is not certain, although some experts propose that bronchial thermoplasty should be considered for patients with severe asthma associated with non-type 2 inflammation or who fail to respond favorably to biologic therapies targeting type 2 inflammation. CONCLUSION Bronchial thermoplasty is a modestly effective treatment for severe asthma after assessment and management of causes of difficult-to-control asthma. Asthma morbidity increases during and shortly after treatment. Follow-up studies provide reassurance on the long-term safety of the procedure. Uncertainties remain about predictors of response, mechanism(s) of action, and place in management of severe asthma.
Collapse
Affiliation(s)
- Neil C Thomson
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|