1
|
Chen H, Liu J, Zhang J, Chen Y, Wang Y, Qiu Y, Hu H, Chai L, Zhang Q, Wang Q, Li M. USP22/BRD4 mediated hedgehog pathway activation contributes to airway remodeling in asthma. Int Immunopharmacol 2025; 153:114538. [PMID: 40132456 DOI: 10.1016/j.intimp.2025.114538] [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: 01/08/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Bromodomain-containing protein 4 (BRD4) is recognized as a member of the bromodomain and extraterminal (BET) family that is involved in the airway inflammation and airway remodeling of asthma. However, the underlying mechanisms of BRD4 in airway smooth muscle cells (ASMCs) proliferation and airway remodeling remain unclear. Primary cultured rat ASMCs and ovalbumin (OVA)-induced rat asthma models were applied to address these issues in the present study. We showed that transforming growth factor β1 (TGF-β1) increased the protein expression of ubiquitin specific peptidase 22 (USP22), which deubiquitinated BRD4 therefore increased its expression, and then resulted in the upregulation of glioma-associated oncogene homolog 1 (GLI1) and osteopontin (OPN) leading to ASMCs proliferation. We further confirmed that induction of TGF-β1 sequentially upregulated USP22, BRD4, GLI1 and OPN leading to airway remodeling in OVA-induced rat asthma models, targeting TGF-β1/USP22/BRD4/GLI1/OPN pathway axis effectively attenuated airway remodeling and asthma development. Our study provides novel sights to understand the role of TGF-β1/USP22/BRD4/GLI1/OPN axis in airway remodeling, and targeting this pathway might have potential value for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Huan Chen
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jin Liu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jia Zhang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuqian Chen
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yan Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuanjie Qiu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Huizhong Hu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Limin Chai
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qianqian Zhang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qingting Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Manxiang Li
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
2
|
Zhang M, Qin Z, Huang C, Liang B, Zhang X, Sun W. The gut microbiota modulates airway inflammation in allergic asthma through the gut-lung axis related immune modulation: A review. BIOMOLECULES & BIOMEDICINE 2025; 25:727-738. [PMID: 39465678 PMCID: PMC11959394 DOI: 10.17305/bb.2024.11280] [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: 09/09/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
The human gut microbiota is a vast and complex microbial community. According to statistics, the number of bacteria residing in the human intestinal tract is approximately ten times that of total human cells, with over 1000 different species. The interaction between the gut microbiota and various organ tissues plays a crucial role in the pathogenesis of local and systemic diseases, exerting a significant influence on disease progression. The relationship between the gut microbiota and intestinal diseases, along with its connection to the pulmonary immune environment and the development of lung diseases, is commonly referred to as the "gut-lung axis." The incidence of bronchial asthma is rising globally. With ongoing research on gut microbiota, it is widely believed that intestinal microorganisms and their metabolic products directly or indirectly participate in the occurrence and development of asthma. Based on the gut-lung axis, this review examines recent research suggesting that the intestinal microbiota can influence the occurrence and progression of allergic asthma through the modulation of cytokine immune balance and mucosal integrity. Though the precise immune pathways or microbial species influencing asthma through the gut-lung axis are still under exploration, summarizing the immune modulation through the gut-lung axis in allergic asthma may provide insights for the clinical management of the condition.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Gastroenterology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Ziwen Qin
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chuanjun Huang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Bin Liang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiuqing Zhang
- Department of Radiology, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| | - Weitao Sun
- Department of Respiratory Medicine, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| |
Collapse
|
3
|
Latayan J, Akkenapally SV, Madala SK. Emerging Concepts in Cytokine Regulation of Airway Remodeling in Asthma. Immunol Rev 2025; 330:e70020. [PMID: 40116139 PMCID: PMC11926778 DOI: 10.1111/imr.70020] [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: 08/24/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Asthma, a chronic respiratory condition that has seen a dramatic rise in prevalence over the past few decades, now affects more than 300 million people globally and imposes a significant burden on healthcare systems. The key pathological features of asthma include inflammation, airway hyperresponsiveness, mucus cell metaplasia, smooth muscle hypertrophy, and subepithelial fibrosis. Cytokines released by lung epithelial cells, stromal cells, and immune cells during asthma are critical to pathological tissue remodeling in asthma. Over the past few decades, researchers have made great strides in understanding key cells involved in asthma and the cytokines that they produce. Epithelial cells as well as many adaptive and innate immune cells are activated by environmental signals to produce cytokines, namely, type 2 cytokines (IL-4, IL-5, IL-13), IFN-γ, IL-17, TGF-β, and multiple IL-6 family members. However, the precise mechanisms through which these cytokines contribute to airway remodeling remain elusive. Additionally, multiple cell types can produce the same cytokines, making it challenging to decipher how specific cell types and cytokines uniquely contribute to asthma pathogenesis. This review highlights recent advances and provides a comprehensive overview of the key cells involved in the production of cytokines and how these cytokines modulate airway remodeling in asthma.
Collapse
Affiliation(s)
- Jana Latayan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
- Immunology Graduate ProgramUniversity of CincinnatiCincinnatiOhioUSA
| | - Santhoshi V. Akkenapally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| | - Satish K. Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| |
Collapse
|
4
|
Gonzales G, Malka R, Bizios R, Dion GR, Guda T. Burn inhalation injury and intubation with dexamethasone-eluting endotracheal tubes modulate local microbiome and alter airway inflammation. Front Bioeng Biotechnol 2025; 13:1524013. [PMID: 40078791 PMCID: PMC11897493 DOI: 10.3389/fbioe.2025.1524013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Background Inhalation injuries, caused by exposure to extreme heat and chemical irritants, lead to complications with speaking, swallowing, and breathing. This study investigates the effects of thermal injury and endotracheal tube (ETT) placement on the airway microbiome and inflammatory response. A secondary aim is to assess the impact of localized dexamethasone delivery via a drug-eluting ETT to reduce laryngeal scarring. Methods Inhalation injury was developed in swine by administering heated air (150°C-160°C) under endoscopic visualization. Following injury, segments of regular or dexamethasone-loaded endotracheal tubes (ETTs) were placed in the injured airways for 3 or 7 days. Computed tomography (CT) scans were used to assess airway narrowing post-injury. Biofilm formation on the ETTs was investigated using micro-CT and microscopy. The airway microbiome was analyzed via 16S rRNA sequencing. Inflammatory markers were quantified using an immunoassay and macrophage populations in laryngeal tissue were assessed with CD86 and CD206 staining. Tracheal tissues were also histologically examined for epithelial thickness, collagen area, and mucin production. Results CT scans confirmed airway narrowing post-injury, particularly around ETT sites. Biofilm formation was more extensive on dexamethasone-coated ETTs at later timepoints. Beta diversity analysis revealed significant shifts in microbial composition related to ETT type (R2 = 0.04, p < 0.05) and duration of placement (R2 = 0.22, p < 0.05). Differential abundance analysis demonstrated significant positive log fold changes in genera such as Bergeriella, Peptostreptococcus, and Bacteriodes with thermal injury over time. Inflammatory markers IFN-γ, IL-4, and IL-1β were elevated in dexamethasone-ETT groups at 3 days, then decreased by 7 days. Macrophage markers CD86 and CD206 were significantly greater in dexamethasone groups compared to regular ETT groups at 7 days (p = 0.002 and p = 0.0213, respectively). Epithelial thickness was significantly greater with regular ETT placement compared to dexamethasone ETT placement in the burn-injured airway at 3 days (p = 0.027). Conclusion Thermal inhalation injury and ETT placement significantly impact airway inflammation, structural integrity, and microbiome composition. Dexamethasone-eluting ETTs, intended to reduce inflammation, increased biofilm formation and elevated cytokine levels, suggesting complex interactions between the drug coating and the host immune response. The airway microbiome shifted significantly with specific taxa thriving in the inflamed environment.
Collapse
Affiliation(s)
- Gabriela Gonzales
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Ronit Malka
- Department of Otolaryngology – Head and Neck Surgery, Brooke Army Medical Center JBSA Fort Sam Houston, San Antonio, TX, United States
| | - Rena Bizios
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Gregory R. Dion
- Department of Otolaryngology – Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, United States
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
5
|
Yadav V, Pandey V, Gaglani P, Srivastava A, Soni, Subhashini. SIRT-2 inhibition by AK-7 orchestrates fibrotic cascades in airways through neuroimmune interaction via TRPA1, TRPM8 and TGF-β signalling. Biochem Pharmacol 2025; 232:116689. [PMID: 39617211 DOI: 10.1016/j.bcp.2024.116689] [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: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024]
Abstract
Chronic obstructive pulmonary diseases (COPD) is characterized by airflow limitation, chronic inflammation and airway remodeling (AR) in airways and lung parenchyma. AR, a lung response, involves mucus production, airflow issues, and structural changes. It is exacerbated by neurogenic inflammation from activated sensory nerves, highlighting the interplay between neuronal and immune regulation in COPD. Sirtuins play a crucial role in lung remodeling, with SIRT-2 being the least studied. Present study explores how SIRT-2 regulates neurogenic inflammation and fibrosis in experimental BALB/c mice with cigarette smoke-induced COPD. Mice from each group, except the control, were exposed to CS for 60 days and AK-7 (100ug/kg and 200ug/kg) was administered intranasally. The study evaluated lung injury and inflammation marked by increased Cortisol, ACTH, COX-2 and LDH in COPD group with its attenuation by SIRT-2 inhibition. Additionally, CS exposure exhibited neurogenic inflammation represented by activated TPRV1 and TRPM8, elevated neuromediators levels (dopamine, acetylcholine, substance P, serotonin) and their respective receptors which were mitigated by AK-7. CS exposure enhanced fibrosis by targeting the fibrotic cascade, enhancing MMP-9, total collagen, hydroxyproline, and upregulating αSMA, MUC5AC, TGF-β, PKA, GATA-3, FOXO3, and STAT-6. SIRT-2 inhibition effectively reversed all these factors suppressing fibrosis further supported by downregulated SIRT-2 expression and histopathological studies where collagen deposition and mucus production were also attenuated by AK-7. Molecular docking revealed strong binding affinity of certain protein such as COX-2, D5DR and 5HT with AK-7. Overall, targeting SIRT-2 to modulate neuro-immune interplay presents a promising therapeutic approach for addressing AR in COPD.
Collapse
Affiliation(s)
- Vandana Yadav
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Vinita Pandey
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Pratikkumar Gaglani
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Atul Srivastava
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Soni
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Subhashini
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
6
|
Elsheikh AA, Shalaby AM, Alabiad MA, Abd-Almotaleb NA, Khayal EES. Perfluorooctanoic acid induced lung toxicity via TGF-β1/Smad pathway, crosstalk between airway hyperresponsiveness and fibrosis: withdrawal impact. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2025; 32:4989-5007. [PMID: 39900883 DOI: 10.1007/s11356-025-36005-2] [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: 08/31/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025]
Abstract
Perfluorooctanoic acid (PFOA) is an environmental persistent agent to which humans are exposed daily through food and water. This study investigated the lung toxic effects induced by ingested PFOA (30 mg/kg/day) for 8 weeks in adult male rats and the impact following 8 weeks of its withdrawal. PFOA increased MDA and reduced TAC inducing oxidative stress. It induced airway hyperresponsiveness (AHR) via increased bronchoalveolar lavage fluid (BALF) IL-4, IL-5, IL-13, IL-9, eosinophil count, TNF-α, and IL-1ß; reduced IL-12; increased serum IgE; and increased urocortin expression in lung tissues. Moreover, it induced pulmonary fibrosis via increased serum KL-6, and SFTP-D, altered pulmonary structure, and increased deposition of collagen fibers in lung tissues. Furthermore, it increased TGF-β1, Smad2, and Smad3 and reduced Smad7 gene expression in lung tissues. These gene alterations were positively correlated with AHR and fibrosis-related factors. The recovered lung upon PFOA withdrawal showed complete resolution of oxidative stress and slight amelioration of other studying parameters. Exposure to PFOA induced lung toxicity by disrupting the TGF-β1/Smad signaling pathway, which acts as a crosstalk between AHR and fibrosis. Additionally, PFOA altered pulmonary architecture, triggered inflammation, and caused oxidative stress. The lung exhibited partial alleviation upon recovery.
Collapse
Affiliation(s)
- Arwa A Elsheikh
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amany Mohamed Shalaby
- Histology and Cell Biology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed Ali Alabiad
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Noha Ali Abd-Almotaleb
- Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman El-Sayed Khayal
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
7
|
Derakhshan T, Hollers E, Perniss A, Ryan T, McGill A, Hacker J, Bergmark RW, Bhattacharyya N, Lee SE, Maxfield AZ, Roditi RE, Bankova L, Buchheit KM, Laidlaw TM, Boyce JA, Dwyer DF. Human intraepithelial mast cell differentiation and effector function are directed by TGF-β signaling. J Clin Invest 2025; 135:e174981. [PMID: 39744949 PMCID: PMC11684804 DOI: 10.1172/jci174981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mast cells (MCs) expressing a distinctive protease phenotype (MCTs) selectively expand within the epithelium of human mucosal tissues during type 2 (T2) inflammation. While MCTs are phenotypically distinct from subepithelial MCs (MCTCs), signals driving human MCT differentiation and this subset's contribution to inflammation remain unexplored. Here, we have identified TGF-β as a key driver of the MCT transcriptome in nasal polyps. We found that short-term TGF-β signaling alters MC cell surface receptor expression and partially recapitulated the in vivo MCT transcriptome, while TGF-β signaling during MC differentiation upregulated a larger number of MCT-associated transcripts. TGF-β inhibited the hallmark MCTC proteases chymase and cathepsin G at both the transcript and protein level, allowing selective in vitro differentiation of MCTs for functional study. We identified discrete differences in effector phenotype between in vitro-derived MCTs and MCTCs, with MCTs exhibiting enhanced proinflammatory lipid mediator generation and a distinct cytokine, chemokine, and growth factor production profile in response to both innate and adaptive stimuli, recapitulating functional features of their tissue-associated counterpart MC subsets. Thus, our findings support a role for TGF-β in promoting human MCT differentiation and identified a discrete contribution of this cell type to T2 inflammation.
Collapse
Affiliation(s)
- Tahereh Derakhshan
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Eleanor Hollers
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Alex Perniss
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Alanna McGill
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jonathan Hacker
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Regan W. Bergmark
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Neil Bhattacharyya
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Stella E. Lee
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Alice Z. Maxfield
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Rachel E. Roditi
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Lora Bankova
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen M. Buchheit
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Tanya M. Laidlaw
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua A. Boyce
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel F. Dwyer
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
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
|
9
|
Dehghani A, Wang L, Garssen J, Styla E, Leusink-Muis T, Van Ark I, Folkerts G, Van Bergenhenegouwen J, Braber S. Synbiotics, a promising approach for alleviating exacerbated allergic airway immune responses in offspring of a preclinical murine pollution model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 112:104591. [PMID: 39577477 DOI: 10.1016/j.etap.2024.104591] [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: 07/07/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Exposure to pollutants like environmental cigarette smoke (CS) poses a major global health risk, affecting individuals from an early age. Therefore, this study explores how postnatal synbiotic supplementation affects allergic asthma symptoms in house-dust-mite (HDM)- challenged offspring maternally exposed to CS. In HDM-allergic offspring of CS-exposed dams, lung resistance was elevated, but synbiotic supplementation effectively reduced this resistance. Elevated eosinophil BALF counts following HDM challenge were intensified in pups maternally exposed to CS. Similarly, Th2 cell activation and serum IgE and IgG1 levels were more pronounced in HDM-allergic offspring of CS-exposed mothers. Synbiotics reduced eosinophil numbers and serum IgE and IgG1, and tended to decrease Th2 cell infiltration and activation. Synbiotics promoted beneficial gut bacteria like Bifidobacterium and Akkermansia. In conclusion, early-life synbiotic intervention mitigated allergic asthma associated with maternal air pollution exposure, highlighting the potential of synbiotics for clinical evaluation as a strategy to prevent allergy development in offspring.
Collapse
Affiliation(s)
- Ali Dehghani
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Department of Public and Occupational Health, Amsterdam University of Medical Science, Amsterdam, Netherlands
| | - Lei Wang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Danone Nutricia Research, Utrecht, Netherlands
| | - Eirini Styla
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ingrid Van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jeroen Van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Danone Nutricia Research, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Danone Nutricia Research, Utrecht, Netherlands.
| |
Collapse
|
10
|
Ullah MA, Rittchen S, Li J, Curren BF, Namubiru P, Ahmed T, Howard DR, Rahman MM, Sikder MAA, Rashid RB, Collinson N, Lor M, Smythe ML, Phipps S. Dual therapy with corticosteroid ablates the beneficial effect of DP2 antagonism in chronic experimental asthma. Nat Commun 2024; 15:10253. [PMID: 39592603 PMCID: PMC11599388 DOI: 10.1038/s41467-024-54670-8] [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: 11/26/2023] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Prostaglandin D2 (PGD2) signals via the DP1 and DP2 receptors. In Phase II trials, DP2 antagonism decreased airway inflammation and airway smooth muscle (ASM) area in moderate-to-severe asthma patients. However, in Phase III, DP2 antagonism failed to lower the rate of exacerbations, and DP2 as a target was shelved. Here, using a preclinical model of chronic experimental asthma, we demonstrate that rhinovirus-induced exacerbations increase PGD2 release, mucus production, transforming growth factor (TGF)-β1 and type-2 inflammation. DP2 antagonism or DP1 agonism ablates these phenotypes, increases epithelial EGF expression and decreases ASM area via increased IFN-γ. In contrast, dual DP1-DP2 antagonism or dual corticosteroid/DP2 antagonism, which attenuates endogenous PGD2, prevented ASM resolution. We demonstrate that DP2 antagonism resolves ASM remodelling via PGD2/DP1-mediated upregulation of IFN-γ expression, and that dual DP2 antagonism/corticosteroid therapy, as often occurred in the human trials, impairs the efficacy of DP2 antagonism by suppressing endogenous PGD2 and IFN-γ production.
Collapse
Affiliation(s)
- Md Ashik Ullah
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Sonja Rittchen
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, 8010, Austria
| | - Jia Li
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Bodie F Curren
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Patricia Namubiru
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Tufael Ahmed
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Daniel R Howard
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Muhammed Mahfuzur Rahman
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Md Al Amin Sikder
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ridwan B Rashid
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Natasha Collinson
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Mary Lor
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Mark L Smythe
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Simon Phipps
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia.
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| |
Collapse
|
11
|
Salles-Dias LP, Brandao-Rangel MAR, Cristina-Rosa A, Morais-Felix RT, Oliveira-Freitas S, Oliveira LVF, Moraes-Ferreira R, Bachi ALL, Coutinho ET, Frison CR, Abbasi A, Melamed D, Vieira RP. Functional analysis of airway remodeling is related with fibrotic mediators in asthmatic children. J Asthma 2024; 61:1284-1293. [PMID: 38577973 DOI: 10.1080/02770903.2024.2338862] [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: 08/28/2023] [Revised: 03/14/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Asthmatic children present variable degrees of airway inflammation, remodeling, and resistance, which correlate with disease control and severity. The chronic inflammatory process of the airway triggers airway remodeling, which reflects the degree of airway resistance. Pro-inflammatory and pro-fibrotic mediators are centrally involved in this process. OBJECTIVE To investigate whether the levels of pulmonary and systemic pro-inflammatory and pro-fibrotic mediators present a correlation with the resistance of the respiratory system and of the proximal and distal airways. METHODS 39 Asthmatic children (persistent mild and moderate) and 39 non-asthmatic children (both between 6 and 13 years old) were evaluated for anthropometric characteristics, lung function and mechanics, and pulmonary and systemic immune responses. RESULTS Asthmatic children showed an increased number of blood eosinophils (p < 0.04), basophils (p < 0.04), monocytes (p < 0.002) and lymphocytes (p < 0.03). In addition, asthmatic children showed impaired lung function, as demonstrated by FEV1 (p < 0.0005) and FEV1/FVC (p < 0.004), decreased total resistance of the respiratory system (R5Hz; p < 0.009), increased resistance of the proximal airways (R20Hz; p < 0.02), increased elastance (Z5Hz; p < 0.02) and increased reactance (X5Hz; p < 0.002) compared to non-asthmatic children. Moreover, the following inflammatory factors were significantly higher in asthmatic than non-asthmatic children: GM-CSF in the breath condensate (BC) (p < 0.0001) and in the serum (p < 0.0001); TGF-beta in the BC (p < 0.0001) and in the serum (p < 0.004); IL-5 in the BC (p < 0.02) and in the serum (p < 0.01); IL-4 in the serum (p < 0.0002). CONCLUSIONS Impulse oscillometry is a sensitive method to detect airway resistance in persistent mild and moderate asthmatic children, an event followed by increased levels of pro-inflammatory and pro-fibrotic mediators.
Collapse
Affiliation(s)
- Lucas P Salles-Dias
- Post-graduate Program in Bioengineering, Universidade Brasil, São Paulo, SP, Brazil
| | - Maysa A R Brandao-Rangel
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Amanda Cristina-Rosa
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Rayssa T Morais-Felix
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | | | - Luis V F Oliveira
- Post-graduate Program in Human Movement and Rehabilitation and in Pharmaceutical Sciences, Evangelical University of Goias (UniEvangélica), Anápolis, GO, Brazil
| | - Renilson Moraes-Ferreira
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Andre L L Bachi
- Post-graduation Program in Health Science, University of Santo Amaro, Rua Isabel Schmidt, São Paulo, SP, Brazil
| | - Elisa T Coutinho
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Claudio R Frison
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Asghar Abbasi
- Division of Respiratory & Critical Care Physiology & Medicine, Lundquist Institute for Biomedical Innovation at Harbor - UCLA Medical Center, Torrance, CA, USA
| | | | - Rodolfo P Vieira
- Post-graduate Program in Bioengineering, Universidade Brasil, São Paulo, SP, Brazil
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
- Post-graduate Program in Human Movement and Rehabilitation and in Pharmaceutical Sciences, Evangelical University of Goias (UniEvangélica), Anápolis, GO, Brazil
| |
Collapse
|
12
|
Gohal G, Moni SS, Bakkari MA, Elmobark ME. A Review on Asthma and Allergy: Current Understanding on Molecular Perspectives. J Clin Med 2024; 13:5775. [PMID: 39407835 PMCID: PMC11476424 DOI: 10.3390/jcm13195775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Asthma, a complex disease characterized by persistent airway inflammation, remains an urgent global health concern. We explored the critical role of allergic biomarkers and dysregulated immune system in asthma through an extensive literature review in databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. This review summarizes the growing data on the pivotal role of allergic biomarkers and dysregulated immune system in the development and evolution of asthma. Recent studies have uncovered several biomarkers that elucidate intrinsic allergic mechanisms in individuals with asthma. This article highlights these biomarkers' potential in predicting asthma onset, assessing its intensity, guiding therapeutic interventions, and tracking disease progression. We also explore the innovative therapeutic prospects arising from the convergence of allergy and dysregulated immune system in asthma and emphasize the potential for precision medicine approaches. Understanding allergic biomarkers intertwined with a dysregulated immune system heralds a new era in asthma treatment and points to improved and individualized treatment modalities.
Collapse
Affiliation(s)
- Gassem Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Sivakumar S. Moni
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
13
|
Veisman I, Massey WJ, Goren I, Liu W, Chauhan G, Rieder F. Muscular hyperplasia in Crohn's disease strictures: through thick and thin. Am J Physiol Cell Physiol 2024; 327:C671-C683. [PMID: 38912732 PMCID: PMC11427014 DOI: 10.1152/ajpcell.00307.2024] [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: 05/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Fibrostenosing Crohn's disease (CD) represents a challenging clinical condition characterized by the development of symptomatic strictures within the gastrointestinal tract. Despite therapeutic advancements in managing inflammation, the progression of fibrostenotic complications remains a significant concern, often necessitating surgical intervention. Recent investigations have unveiled the pivotal role of smooth muscle cell hyperplasia in driving luminal narrowing and clinical symptomatology. Drawing parallels to analogous inflammatory conditions affecting other organs, such as the airways and blood vessels, sheds light on common underlying mechanisms of muscular hyperplasia. This review synthesizes current evidence to elucidate the mechanisms underlying smooth muscle cell proliferation in CD-associated strictures, offering insights into potential therapeutic targets. By highlighting the emerging significance of muscle thickening as a novel therapeutic target, this review aims to inform future research endeavors and clinical strategies with the goal to mitigate the burden of fibrostenotic complications in CD and other conditions.
Collapse
Affiliation(s)
- Ido Veisman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - William J Massey
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Idan Goren
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Weiwei Liu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States
- Cleveland Clinic Program for Global Translational Inflammatory Bowel Diseases (GRID), Cleveland, Ohio, United States
| |
Collapse
|
14
|
Yuan Q, Jia X, Wang M, Chen Z, Xu T, Zhang X, Liu Y, Wang Z, Yang C, Zhang M, Zhang W, Huang M, Ji N. LincR-PPP2R5C Deficiency Alleviates Airway Remodeling by Inhibiting Epithelial-Mesenchymal Transition Through the PP2A/TGF-β1 Signaling Pathway in Chronic Experimental Allergic Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:422-433. [PMID: 39155740 PMCID: PMC11331192 DOI: 10.4168/aair.2024.16.4.422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/13/2024] [Accepted: 02/21/2024] [Indexed: 08/20/2024]
Abstract
Airway remodeling is a key characteristic of allergic asthma. Epithelial-mesenchymal transition (EMT) induced by various factors, particularly transforming growth factor (TGF)-β1, orchestrates airway remodeling. Protein phosphatase 2A (PP2A), an important serine-threonine phosphatase, is involved in TGF-β1 production and EMT. Long noncoding RNAs (lncRNAs) have emerged as novel players in regulating EMT. Here, we aimed to explore the effects and mechanisms of action of lincR-PPP2R5C, a lncRNA that affects PP2A activity, on airway remodeling in a mouse model of chronic allergic asthma. LincR-PPP2R5C knockout (KO) alleviated inflammatory responses in house dust mite (HDM)-induced chronic allergic asthma. Moreover, airway remodeling and EMT were reduced in lung tissues of lincR-PPP2R5C KO mice. HDM extract induced EMT in airway epithelial cells, which was decreased following lincR-PPP2R5C KO. Mechanistically, lincR-PPP2R5C deficiency enhanced PP2A activity, which inhibited TGF-β1 production in epithelial cells. In conclusion, lincR-PPP2R5C deficiency prevented HDM-induced airway remodeling in mice by reversing EMT, which was mediated by the PP2A/TGF-β1 signaling pathway. Thus, lncRNAs, i.e., lincR-PPP2R5C, may be potential targets to prevent airway remodeling in allergic asthma.
Collapse
Affiliation(s)
- Qi Yuan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Jia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xijie Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanan Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Yang
- NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China.
| | - Wei Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Ma Y, Ye S, Sun K, Gu Y. Effect of curcumin nanoparticles on proliferation and migration of mouse airway smooth muscle cells and airway inflammatory infiltration. Front Pharmacol 2024; 15:1344333. [PMID: 38708080 PMCID: PMC11066239 DOI: 10.3389/fphar.2024.1344333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/02/2024] [Indexed: 05/07/2024] Open
Abstract
Curcumin (CUR) possesses the capability to inhibit various inflammatory factors, exert anti-inflammatory effects, and alleviate asthma attacks; however, its hydrophobicity and instability significantly impede its clinical application. In this study, we synthesized CUR-loaded nanoparticles (CUR-NPs) and evaluated their impact on the proliferation, migration, and inflammatory infiltration of mouse airway smooth muscle cells (ASMCs), while investigating their underlying mechanisms. To achieve this objective, ASMCs were isolated from BALB/c mice and subjected to TGF-β1-induced cell proliferation and migration. Our findings demonstrate that CUR-NPs effectively regulate the release of CUR within cells with superior intracellular uptake compared to free CUR. The CCK-8 assay results indicate that the blank carrier does not exhibit any cytotoxic effects on cells, thus rendering the impact of the carrier itself negligible. The TGF-β1 group exhibited a significant increase in cell proliferation, whereas treatment with CUR-NPs significantly suppressed TGF-β1-induced cell proliferation. The findings from both the cell scratch assay and transwell assay demonstrated that TGF-β1 substantially enhanced cell migration, while CUR-NPs treatment effectively attenuated TGF-β1-induced cell migration. The Western blot analysis demonstrated a substantial increase in the expression levels of TGF-β1, p-STAT3, and CTGF in ASMCs following treatment with TGF-β1 when compared to the control group. Nevertheless, this effect was effectively counteracted upon administration of CUR-NPs. Furthermore, an asthma mouse model was successfully established and CUR-NPs were administered through tail vein injection. The serum levels of TGF-β1 and the expression levels of TGF-β1, p-STAT3, and CTGF proteins in the lung tissue of mice in the model group exhibited significant increases compared to those in the control group. However, CUR-NPs treatment effectively attenuated this change. Our research findings suggest that CUR-NPs possess inhibitory effects on ASMC proliferation, migration, and inflammatory infiltration by suppressing activation of the TGF-β1/p-STAT3/CTGF signaling pathway, thereby facilitating inhibition of airway remodeling.
Collapse
Affiliation(s)
- Yucong Ma
- Department of Pediatric Respiration, Children’s Medical Center, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Suping Ye
- Department of Reparatory and Critical Care Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Kunpeng Sun
- Department of Reparatory and Critical Care Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Yue Gu
- Department of Reparatory and Critical Care Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
16
|
Goyal RK, Rattan S. Role of mechanoregulation in mast cell-mediated immune inflammation of the smooth muscle in the pathophysiology of esophageal motility disorders. Am J Physiol Gastrointest Liver Physiol 2024; 326:G398-G410. [PMID: 38290993 PMCID: PMC11213482 DOI: 10.1152/ajpgi.00258.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/01/2024]
Abstract
Major esophageal disorders involve obstructive transport of bolus to the stomach, causing symptoms of dysphagia and impaired clearing of the refluxed gastric contents. These may occur due to mechanical constriction of the esophageal lumen or loss of relaxation associated with deglutitive inhibition, as in achalasia-like disorders. Recently, immune inflammation has been identified as an important cause of esophageal strictures and the loss of inhibitory neurotransmission. These disorders are also associated with smooth muscle hypertrophy and hypercontractility, whose cause is unknown. This review investigated immune inflammation in the causation of smooth muscle changes in obstructive esophageal bolus transport. Findings suggest that smooth muscle hypertrophy occurs above the obstruction and is due to mechanical stress on the smooth muscles. The mechanostressed smooth muscles release cytokines and other molecules that may recruit and microlocalize mast cells to smooth muscle bundles, so that their products may have a close bidirectional effect on each other. Acting in a paracrine fashion, the inflammatory cytokines induce genetic and epigenetic changes in the smooth muscles, leading to smooth muscle hypercontractility, hypertrophy, and impaired relaxation. These changes may worsen difficulty in the esophageal transport. Immune processes differ in the first phase of obstructive bolus transport, and the second phase of muscle hypertrophy and hypercontractility. Moreover, changes in the type of mechanical stress may change immune response and effect on smooth muscles. Understanding immune signaling in causes of obstructive bolus transport, type of mechanical stress, and associated smooth muscle changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.NEW & NOTEWORTHY Esophageal disorders such as esophageal stricture or achalasia, and diffuse esophageal spasm are associated with smooth muscle hypertrophy and hypercontractility, above the obstruction, yet the cause of such changes is unknown. This review suggests that smooth muscle obstructive disorders may cause mechanical stress on smooth muscle, which then secretes chemicals that recruit, microlocalize, and activate mast cells to initiate immune inflammation, producing functional and structural changes in smooth muscles. Understanding the immune signaling in these changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.
Collapse
Affiliation(s)
- Raj K Goyal
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Boston Healthcare System, West Roxbury, Massachusetts, United States
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts, United States
| | - Satish Rattan
- Department of Medicine, Division of Gastroenterology and Hepatology, Sidney Kummel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
17
|
Abud EM, White AA. Mast Cells in Aspirin-Exacerbated Respiratory Disease. Curr Allergy Asthma Rep 2024; 24:73-80. [PMID: 38217825 DOI: 10.1007/s11882-024-01125-1] [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] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
PURPOSE OF REVIEW Aspirin-exacerbated respiratory disease (AERD) is a syndrome of high type 2 inflammation and is known to critically involve mast cell activation. The mast cell is an important cell in the baseline inflammatory processes in the upper and lower airway by maintaining and amplifying type 2 inflammation. But it also is prominent in the hypersensitivity reaction to COX-1 inhibition which defines this condition. RECENT FINDINGS Recent work highlights the mast cell as a focal point in AERD pathogenesis. Using AERD as a specific model of both high type 2 asthma and chronic sinusitis, the role of mast cell activity can be better understood in other aspects of airway inflammation. Further dissecting out the mechanism of COX-1-mediated mast cell activation in AERD will be an important next phase in our understanding of NSAID-induced hypersensitivity as well as AERD pathophysiology.
Collapse
Affiliation(s)
- Edsel M Abud
- Division of Allergy, Asthma, and Immunology, Scripps Clinic, San Diego, USA
- Scripps Research Translational Institute, Scripps Research, San Diego, USA
| | - Andrew A White
- Division of Allergy, Asthma, and Immunology, Scripps Clinic, San Diego, USA.
| |
Collapse
|
18
|
Škrgat S, Harlander M, Janić M. Obesity and Insulin Resistance in Asthma Pathogenesis and Clinical Outcomes. Biomedicines 2024; 12:173. [PMID: 38255279 PMCID: PMC10813771 DOI: 10.3390/biomedicines12010173] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Common inflammatory ground links obesity, insulin resistance, and asthma. As recognition of their interplay, one worsening the natural course of the other, is recognised, questions remain about how to adequately address them altogether to improve clinical outcomes. The present manuscript sheds light on the problem, describing possible pathophysiological links, clinical views, and therapeutic challenges, raising questions about what remains to be done, and calling for multidisciplinary treatment of these patients to detect diseases early and adequately address them before they become full-blown and deteriorate their health and quality of life.
Collapse
Affiliation(s)
- Sabina Škrgat
- Department of Pulmonary Diseases and Allergy, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
- Medical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Matevž Harlander
- Department of Pulmonary Diseases and Allergy, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
- Medical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Miodrag Janić
- Medical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
- Clinical Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
19
|
Giombi F, Pace GM, Pirola F, Cerasuolo M, Ferreli F, Mercante G, Spriano G, Canonica GW, Heffler E, Ferri S, Puggioni F, Paoletti G, Malvezzi L. Airways Type-2 Related Disorders: Multiorgan, Systemic or Syndemic Disease? Int J Mol Sci 2024; 25:730. [PMID: 38255804 PMCID: PMC10815382 DOI: 10.3390/ijms25020730] [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: 12/14/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Chronic rhinosinusitis (CRS) has recently undergone a significant paradigm shift, moving from a phenotypical classification towards an "endotype-based" definition that places more emphasis on clinical and therapeutic aspects. Similar to other airway diseases, like asthma, most cases of CRS in developed countries exhibit a dysregulated type-2 immune response and related cytokines. Consequently, the traditional distinction between upper and lower airways has been replaced by a "united airway" perspective. Additionally, type-2 related disorders extend beyond respiratory boundaries, encompassing conditions beyond the airways, such as atopic dermatitis. This necessitates a multidisciplinary approach. Moreover, consideration of possible systemic implications is crucial, particularly in relation to sleep-related breathing diseases like Obstructive Sleep Apnoea Syndrome (OSAS) and the alteration of systemic inflammatory mediators such as nitric oxide. The trends in epidemiological, economic, and social burden are progressively increasing worldwide, indicating syndemic characteristics. In light of these insights, this narrative review aims to present the latest evidence on respiratory type-2 related disorders, with a specific focus on CRS while promoting a comprehensive perspective on the "united airways". It also introduces a novel concept: viewing these conditions as a multiorgan, systemic, and syndemic disease.
Collapse
Affiliation(s)
- Francesco Giombi
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Gian Marco Pace
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Francesca Pirola
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
| | - Michele Cerasuolo
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Otorhinolaryngology Head & Neck Surgery Unit, Casa di Cura Humanitas San Pio X, Via Francesco Nava 31, 20159 Milan, Italy
| | - Fabio Ferreli
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Giuseppe Mercante
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Giuseppe Spriano
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Sebastian Ferri
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Francesca Puggioni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Giovanni Paoletti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Luca Malvezzi
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Otorhinolaryngology Head & Neck Surgery Unit, Casa di Cura Humanitas San Pio X, Via Francesco Nava 31, 20159 Milan, Italy
| |
Collapse
|
20
|
Zhan W, Luo W, Zhang Y, Xiang K, Chen X, Shen S, Huang C, Xu T, Ding W, Chen Y, Lin M, Pan X, Lai K. Sputum Transcriptomics Reveals FCN1+ Macrophage Activation in Mild Eosinophilic Asthma Compared to Non-Asthmatic Eosinophilic Bronchitis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:55-70. [PMID: 38262391 PMCID: PMC10823142 DOI: 10.4168/aair.2024.16.1.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/08/2023] [Accepted: 08/05/2023] [Indexed: 01/25/2024]
Abstract
PURPOSE Eosinophilic asthma (EA) and non-asthmatic eosinophilic bronchitis (EB) share similar eosinophilic airway inflammation. Unlike EA, EB did not present airway hyperresponsiveness or airflow obstruction. We aimed to compare the mechanism underlying the different manifestations between EA and EB via sputum transcriptomics analysis. METHODS Induced-sputum cells from newly physician-diagnosed EA, EB patients, and healthy controls (HCs) were collected for RNA sequencing. RESULTS Bulk RNA sequencing was performed using sputum cells from patients with EA (n = 18), EB (n = 15) and HCs (n = 28). Principal component analysis revealed similar gene expression patterns in EA and EB. The most differentially expressed genes in EB compared with HC were also shared by EA, including IL4, IL5 IL13, CLC, CPA3, and DNASE1L3. However, gene set enrichment analysis showed that the signatures regulating macrophage activation were enriched in EA compared to EB. Sputum cells were profiled using single-cell RNA sequencing. FABP4+ macrophages, SPP1+ macrophages, FCN1+ macrophages, dendritic cells, T cells, B cells, mast cells, and epithelial cells were identified based on gene expression profiling. Analysis of cell-cell communication revealed that interactions between FCN1+ macrophages and other cells were higher in EA than in EB. A wealth of transforming growth factor beta (TGF-β) and vascular endothelial growth factor (VEGF) interactions between FCN1+ macrophages and other cells have been shown in EA. The gene expression levels of EREG, TGFBI, and VEGFA in FCN1+ macrophages of EA were significantly higher than those of EB. Furthermore, signatures associated with the response to TGF-β, cellular response to VEGF stimulus and developmental cell growth were enriched in FCN1+ macrophages of EA compared to those of EB. CONCLUSIONS FCN1+ macrophage activation associated with airway remodeling processes was upregulated in EA compared to that in EB, which may contribute to airway hyperresponsiveness and airflow obstruction.
Collapse
Affiliation(s)
- Wenzhi Zhan
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Luo
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yulong Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Keheng Xiang
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomei Chen
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuirong Shen
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chuqing Huang
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tingting Xu
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenbin Ding
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuehan Chen
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingtong Lin
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Kefang Lai
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
21
|
Pluangnooch P, Soontrapa K, Pudgerd A, Sridurongrit S. Expression of constitutively active TβRI leads to attenuation of ovalbumin-induced allergic airway inflammation associated with augmented M2 polarization of alveolar macrophage. Respir Investig 2024; 62:90-97. [PMID: 38007853 DOI: 10.1016/j.resinv.2023.10.005] [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: 06/17/2023] [Revised: 09/14/2023] [Accepted: 10/14/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Transforming growth factor-β (Tgf-β) plays an important role in the pathogenesis of asthma through the regulation of T cells and airway epithelium. Its functions in alveolar macrophage (AM) during allergic airway inflammation remain unknown. METHODS A murine asthma model was induced with ovalbumin (ova) in TβRICA/Fsp1-Cre transgenic mice expressing constitutively active Tgf-β receptor type I (TβRICA) under the control of Fsp1-Cre transgene. Cells in the bronchoalveolar lavage (BAL) were collected to study immune cell infiltration in the lungs. Cytokine levels in BAL fluid were measured by enzyme-linked immunoassay (ELISA). Lungs were sectioned and stained with hematoxylin and eosin, periodic acid-Schiff, and trichrome for histopathologic evaluation. AMs were assessed by flow cytometry and were sorted for quantitative polymerase chain reaction analysis. RESULTS Our data indicated that TβRICA transcripts were induced in AMs of TβRICA/Fsp1-Cre mice. Following the ova challenges, TβRICA/Fsp1-Cre mice exhibited reduced cellular infiltration of the airway, reduced pulmonary fibrosis, and reduced bronchial mucus secretion as compared to ova-challenged wild-type mice. An alternatively activated macrophage (M2) polarization was significantly elevated in the lungs of ova-challenged TβRICA/Fsp1-Cre mice as reflected by increased numbers of AMs expressing M2 subtype marker, CD163, in the lungs and enhanced expression of CCR2 and CD206 in AMs. Moreover, TβRICA/Fsp1-Cre AMs showed augmented expression of transcription factors, Foxo1, and IRF4, which are known to be positive regulators for M2 polarization. CONCLUSIONS Expression of TβRICA in AMs promoted M2 polarization and ameliorated allergic airway inflammation in an ova-induced asthma mouse model.
Collapse
Affiliation(s)
- Panwadee Pluangnooch
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Arnon Pudgerd
- Division of Anatomy, School of Medical Science, University of Phayao, Phayao 56000, Thailand
| | - Somyoth Sridurongrit
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok 10400, Thailand.
| |
Collapse
|
22
|
Dai B, Liu S, Shen W, Chen L, Zhou Q, Han L, Zhang Q, Shan L. Role of SYVN1 in the control of airway remodeling in asthma protection by promoting SIRT2 ubiquitination and degradation. Biol Res 2023; 56:64. [PMID: 38041162 PMCID: PMC10693155 DOI: 10.1186/s40659-023-00478-7] [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: 05/22/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Asthma is a heterogenous disease that characterized by airway remodeling. SYVN1 (Synoviolin 1) acts as an E3 ligase to mediate the suppression of endoplasmic reticulum (ER) stress through ubiquitination and degradation. However, the role of SYVN1 in the pathogenesis of asthma is unclear. RESULTS In the present study, an ovalbumin (OVA)-induced murine model was used to evaluate the effect of SYVN1 on asthma. An increase in SYVN1 expression was observed in the lungs of mice after OVA induction. Overexpression of SYVN1 attenuated airway inflammation, goblet cell hyperplasia and collagen deposition induced by OVA. The increased ER stress-related proteins and altered epithelial-mesenchymal transition (EMT) markers were also inhibited by SYVN1 in vivo. Next, TGF-β1-induced bronchial epithelial cells (BEAS-2B) were used to induce EMT process in vitro. Results showed that TGF-β1 stimulation downregulated the expression of SYVN1, and SYVN1 overexpression prevented ER stress response and EMT process in TGF-β1-induced cells. In addition, we identified that SYVN1 bound to SIRT2 and promoted its ubiquitination and degradation. SIRT2 overexpression abrogated the protection of SYVN1 on ER stress and EMT in vitro. CONCLUSIONS These data suggest that SYVN1 suppresses ER stress through the ubiquitination and degradation of SIRT2 to block EMT process, thereby protecting against airway remodeling in asthma.
Collapse
Affiliation(s)
- Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Si Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Wenxin Shen
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Li Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Qianlan Zhou
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Lina Han
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Qinzhen Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China
| | - Lishen Shan
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
23
|
Wang J, Jian Q, Yan K, Yang J, Yan L, Cheng W. m 6A-modified miR-143-3p inhibits epithelial mesenchymal transition in bronchial epithelial cells and extracellular matrix production in lung fibroblasts by targeting Smad3. Pulm Pharmacol Ther 2023; 83:102251. [PMID: 37666296 DOI: 10.1016/j.pupt.2023.102251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/11/2023] [Accepted: 08/27/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Airway epithelial cells epithelial mesenchymal transition (EMT) and lung fibroblasts extracellular matrix (ECM) production are the key steps in airway remodeling. Our previous study demonstrated that miR-143-3p has the ability to impede airway smooth muscle cell proliferation and ECM deposition. However, the function of miR-143-3p in airway epithelial cells and lung fibroblasts remains unclear. METHODS Cell viability was determined using MTT method, while cell migration was evaluated through scratch assay. EMT and ECM proteins were detected by western blot, RT-qPCR, and ELISA. To determine the level of miR-143-3p m6A methylation, we employed the meRIP-qPCR assay. Additionally, the binding of miR-143-3p with Smad3 were projected by bioinformatics and validated by dual luciferase reporter assays. RESULTS It was discovered that the expression of miR-143-3p were lower in both asthma patients and TGF-β1-treated human bronchial epithelial 16HBE cells and human lung fibroblast HPF cells. Upregulation of miR-143-3p restrained 16HBE cell migration, and decreased EMT mesenchymal markers and increased epithelial markers. And upregulation of miR-143-3p impaired cell viability and ECM protein production in HPF cells. Mechanistically, interfering with METTL3 resulted in decreased m6A modification of miR-143-3p and led to lower levels of miR-143-3p. Moreover, miR-143-3p were verified to directly target and downregulate Smad3. Upregulation of Smad3 attenuated the effects of miR-143-3p on cell EMT and ECM production. CONCLUSION MiR-143-3p inhibits airway epithelial cell EMT as well as lung fibroblast ECM production by downregulating Smad3. Therefore, miR-143-3p may be a promising target to reduce airway remodeling in asthma.
Collapse
Affiliation(s)
- Jing Wang
- Department of Respiratory and Asthma, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Qiang Jian
- Department of Emergency, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Kun Yan
- Department of General Surgery, 2nd Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jiao Yang
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Liping Yan
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Wei Cheng
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China.
| |
Collapse
|
24
|
Wang Z. Role of transforming growth factor-β in airway remodelling in bronchiolitis obliterans. Growth Factors 2023; 41:192-209. [PMID: 37487145 DOI: 10.1080/08977194.2023.2239356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Airway remodelling is the main pathological mechanism of bronchiolitis obliterans (BO). Several studies have found that transforming growth factor-β (TGF-β) expression is increased in BO during airway remodelling, where it plays an important role in various biological processes by binding to its receptor complex to activate multiple signalling proteins and pathways. This review examines the role of TGF-β in airway remodelling in BO and its potential as a therapeutic target, highlighting the mechanisms of TGF-β activation and signalling, cellular targets of TGF-β actions, and research progress in TGF-β signalling and TGF-β-mediated processes.
Collapse
Affiliation(s)
- Ziwei Wang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Siddiqui S, Bachert C, Bjermer L, Buchheit KM, Castro M, Qin Y, Rupani H, Sagara H, Howarth P, Taillé C. Eosinophils and tissue remodeling: Relevance to airway disease. J Allergy Clin Immunol 2023; 152:841-857. [PMID: 37343842 DOI: 10.1016/j.jaci.2023.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/15/2023] [Accepted: 06/02/2023] [Indexed: 06/23/2023]
Abstract
The ability of human tissue to reorganize and restore its existing structure underlies tissue homeostasis in the healthy airways, but in disease can persist without normal resolution, leading to an altered airway structure. Eosinophils play a cardinal role in airway remodeling both in health and disease, driving epithelial homeostasis and extracellular matrix turnover. Physiological consequences associated with eosinophil-driven remodeling include impaired lung function and reduced bronchodilator reversibility in asthma, and obstructed airflow in chronic rhinosinusitis with nasal polyps. Given the contribution of airway remodeling to the development and persistence of symptoms in airways disease, targeting remodeling is an important therapeutic consideration. Indeed, there is early evidence that eosinophil attenuation may reduce remodeling and disease progression in asthma. This review provides an overview of tissue remodeling in both health and airway disease with a particular focus on eosinophilic asthma and chronic rhinosinusitis with nasal polyps, as well as the role of eosinophils in these processes and the implications for therapeutic interventions. Areas for future research are also noted, to help improve our understanding of the homeostatic and pathological roles of eosinophils in tissue remodeling, which should aid the development of targeted and effective treatments for eosinophilic diseases of the airways.
Collapse
Affiliation(s)
- Salman Siddiqui
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Claus Bachert
- Department of Otorhinolaryngology-Head and Neck Surgery, University Hospital of Münster, Münster, Germany; First Affiliated Hospital, Sun Yat-Sen University, International Airway Research Center, Guangzhou, China; Division of Ear, Nose, and Throat Diseases, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden; Upper Airways Research Laboratory, Faculty of Medicine, Ghent University, Ghent, Belgium
| | - Leif Bjermer
- Department of Clinical Sciences, Respiratory Medicine, and Allergology, Lund University, Lund, Sweden
| | - Kathleen M Buchheit
- Jeff and Penny Vinik Center for Allergic Diseases Research, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Mario Castro
- Division of Pulmonary, Critical Care Medicine, University of Kansas School of Medicine, Kansas City, NC
| | - Yimin Qin
- Global Medical Affairs, Global Specialty and Primary Care, GlaxoSmithKline, Research Triangle Park, NC
| | - Hitasha Rupani
- Department of Respiratory Medicine, University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Hironori Sagara
- Department of Medicine, Division of Respiratory Medicine and Allergology, Showa University, School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Peter Howarth
- Global Medical, Global Specialty and Primary Care, GlaxoSmithKline, Brentford, Middlesex, United Kingdom
| | - Camille Taillé
- Pneumology Department, Reference Center for Rare Pulmonary Diseases, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unit 1152, University of Paris Cité, Paris, France
| |
Collapse
|
26
|
Xu T, Wu Z, Yuan Q, Zhang X, Liu Y, Wu C, Song M, Wu J, Jiang J, Wang Z, Chen Z, Zhang M, Huang M, Ji N. Proline is increased in allergic asthma and promotes airway remodeling. JCI Insight 2023; 8:e167395. [PMID: 37432745 PMCID: PMC10543727 DOI: 10.1172/jci.insight.167395] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
Proline and its synthesis enzyme pyrroline-5-carboxylate reductase 1 (PYCR1) are implicated in epithelial-mesenchymal transition (EMT), yet how proline and PYCR1 function in allergic asthmatic airway remodeling via EMT has not yet been addressed to our knowledge. In the present study, increased levels of plasma proline and PYCR1 were observed in patients with asthma. Similarly, proline and PYCR1 in lung tissues were high in a murine allergic asthma model induced by house dust mites (HDMs). Pycr1 knockout decreased proline in lung tissues, with reduced airway remodeling and EMT. Mechanistically, loss of Pycr1 restrained HDM-induced EMT by modulating mitochondrial fission, metabolic reprogramming, and the AKT/mTORC1 and WNT3a/β-catenin signaling pathways in airway epithelial cells. Therapeutic inhibition of PYCR1 in wild-type mice disrupted HDM-induced airway inflammation and remodeling. Deprivation of exogenous proline relieved HDM-induced airway remodeling to some extent. Collectively, this study illuminates that proline and PYCR1 involved with airway remodeling in allergic asthma could be viable targets for asthma treatment.
Collapse
Affiliation(s)
- Tingting Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenzhen Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Yuan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xijie Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanan Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chaojie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meijuan Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingxian Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Jiangsu Province Engineering Research Center of Antibody Drug, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Dy ABC, Girkin J, Marrocco A, Collison A, Mwase C, O'Sullivan MJ, Phung TKN, Mattes J, Koziol-White C, Gern JE, Bochkov YA, Bartlett NW, Park JA. Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models. Respir Res 2023; 24:205. [PMID: 37598152 PMCID: PMC10440034 DOI: 10.1186/s12931-023-02510-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Rhinovirus (RV) infection of airway epithelial cells triggers asthma exacerbations, during which airway smooth muscle (ASM) excessively contracts. Due to ASM contraction, airway epithelial cells become mechanically compressed. We previously reported that compressed human bronchial epithelial (HBE) cells are a source of endothelin-1 (ET-1) that causes ASM contraction. Here, we hypothesized that epithelial sensing of RV by TLR3 and epithelial compression induce ET-1 secretion through a TGF-β receptor (TGFβR)-dependent mechanism. METHODS To test this, we used primary HBE cells well-differentiated in air-liquid interface culture and two mouse models (ovalbumin and house dust mite) of allergic airway disease (AAD). HBE cells were infected with RV-A16, treated with a TLR3 agonist (poly(I:C)), or exposed to compression. Thereafter, EDN1 (ET-1 protein-encoding gene) mRNA expression and secreted ET-1 protein were measured. We examined the role of TGFβR in ET-1 secretion using either a pharmacologic inhibitor of TGFβR or recombinant TGF-β1 protein. In the AAD mouse models, allergen-sensitized and allergen-challenged mice were subsequently infected with RV. We then measured ET-1 in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness (AHR) following methacholine challenge. RESULTS Our data reveal that RV infection induced EDN1 expression and ET-1 secretion in HBE cells, potentially mediated by TLR3. TGFβR activation was partially required for ET-1 secretion, which was induced by RV, poly(I:C), or compression. TGFβR activation alone was sufficient to increase ET-1 secretion. In AAD mouse models, RV induced ET-1 secretion in BALF, which positively correlated with AHR. CONCLUSIONS Our data provide evidence that RV infection increased epithelial-cell ET-1 secretion through a TGFβR-dependent mechanism, which contributes to bronchoconstriction during RV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Alane Blythe C Dy
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Jason Girkin
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Antonella Marrocco
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Adam Collison
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Chimwemwe Mwase
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Michael J O'Sullivan
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Thien-Khoi N Phung
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA
| | - Joerg Mattes
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | | | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Nathan W Bartlett
- College of Health, Medicine and Wellbeing, University of Newcastle and Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Jin-Ah Park
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Ave, Boston, MA, SPH1-315, USA.
| |
Collapse
|
28
|
Raby KL, Michaeloudes C, Tonkin J, Chung KF, Bhavsar PK. Mechanisms of airway epithelial injury and abnormal repair in asthma and COPD. Front Immunol 2023; 14:1201658. [PMID: 37520564 PMCID: PMC10374037 DOI: 10.3389/fimmu.2023.1201658] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
The airway epithelium comprises of different cell types and acts as a physical barrier preventing pathogens, including inhaled particles and microbes, from entering the lungs. Goblet cells and submucosal glands produce mucus that traps pathogens, which are expelled from the respiratory tract by ciliated cells. Basal cells act as progenitor cells, differentiating into different epithelial cell types, to maintain homeostasis following injury. Adherens and tight junctions between cells maintain the epithelial barrier function and regulate the movement of molecules across it. In this review we discuss how abnormal epithelial structure and function, caused by chronic injury and abnormal repair, drives airway disease and specifically asthma and chronic obstructive pulmonary disease (COPD). In both diseases, inhaled allergens, pollutants and microbes disrupt junctional complexes and promote cell death, impairing the barrier function and leading to increased penetration of pathogens and a constant airway immune response. In asthma, the inflammatory response precipitates the epithelial injury and drives abnormal basal cell differentiation. This leads to reduced ciliated cells, goblet cell hyperplasia and increased epithelial mesenchymal transition, which contribute to impaired mucociliary clearance and airway remodelling. In COPD, chronic oxidative stress and inflammation trigger premature epithelial cell senescence, which contributes to loss of epithelial integrity and airway inflammation and remodelling. Increased numbers of basal cells showing deregulated differentiation, contributes to ciliary dysfunction and mucous hyperproduction in COPD airways. Defective antioxidant, antiviral and damage repair mechanisms, possibly due to genetic or epigenetic factors, may confer susceptibility to airway epithelial dysfunction in these diseases. The current evidence suggests that a constant cycle of injury and abnormal repair of the epithelium drives chronic airway inflammation and remodelling in asthma and COPD. Mechanistic understanding of injury susceptibility and damage response may lead to improved therapies for these diseases.
Collapse
Affiliation(s)
- Katie Louise Raby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - James Tonkin
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - Pankaj Kumar Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, United Kingdom
| |
Collapse
|
29
|
Fang Y, Jin W, Guo Z, Hao J. Quercetin Alleviates Asthma-Induced Airway Inflammation and Remodeling through Downregulating Periostin via Blocking TGF-β1/Smad Pathway. Pharmacology 2023; 108:432-443. [PMID: 37343534 DOI: 10.1159/000530703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/10/2023] [Indexed: 06/23/2023]
Abstract
INTRODUCTION The aim of the study was to discuss whether the anti-asthmatic effect of quercetin is related to periostin and the downstream molecular pathway of quercetin's anti-asthmatic effect. METHODS We constructed asthmatic mice, sensitized by ovalbumin, and administrated different treatments into mice according to the experimental design. In this study, we mainly observed the inflammatory response, airway fibrosis, and airway hyperresponsiveness in asthmatic mice. Pathological stains (H&E, PAS, and Masson) were performed. We also detected the inflammation factors and fibrosis-related cytokines by enzyme-linked immunosorbent serologic assay. In addition, we also explored the level of periostin by enzyme-linked immunosorbent serologic assay and Western blot. At the same time, TGF-β1/Smad pathway was also determined by Western blot. RESULTS A high expression of periostin was found in asthmatic mice, and quercetin decreases periostin content in bronchoalveolar lavage fluid. Quercetin and OC-20 inhibit airway inflammation response, airway fibrosis, and airway hyperreactivity. Quercetin downregulated TGF-β1/Smad pathway in the lung tissues of asthmatic mice. Anti-asthma role of quercetin is related to periostin. Then deeper mechanical study revealed that inhibiting TGF-β1 could improve asthmatic symptoms, and quercetin exerted the protective effect on asthmatic mice through inhibition of TGF-β1/Smad pathway. CONCLUSION Quercetin provided a protective role against asthma via periostin, manifested by mild inflammatory infiltration, reduced goblet cell proliferation, and reduced airway fibrosis. TGF-β1/Smad pathway is an important transduction system, participating in the protective effect of quercetin on asthma.
Collapse
Affiliation(s)
- Yanni Fang
- Department of Pediatrics, Yantaishan Hospital, Yantai, China
| | - Wenwen Jin
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, China
| | - Zhen Guo
- Department of Pediatrics, Yantaishan Hospital, Yantai, China
| | - Jumei Hao
- Department of Pediatrics, Yantaishan Hospital, Yantai, China
| |
Collapse
|
30
|
Abdelgied M, Uhl K, Chen OG, Schultz C, Tripp K, Peraino AM, Paithankar S, Chen B, Tamae Kakazu M, Castillo Bahena A, Jager TE, Lawson C, Chesla DW, Pestov N, Modyanov NN, Prokop J, Neubig RR, Uhal BD, Girgis RE, Li X. Targeting ATP12A, a Nongastric Proton Pump α Subunit, for Idiopathic Pulmonary Fibrosis Treatment. Am J Respir Cell Mol Biol 2023; 68:638-650. [PMID: 36780662 PMCID: PMC10257074 DOI: 10.1165/rcmb.2022-0264oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/13/2023] [Indexed: 02/15/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a pathological condition of unknown etiology that results from injury to the lung and an ensuing fibrotic response that leads to the thickening of the alveolar walls and obliteration of the alveolar space. The pathogenesis is not clear, and there are currently no effective therapies for IPF. Small airway disease and mucus accumulation are prominent features in IPF lungs, similar to cystic fibrosis lung disease. The ATP12A gene encodes the α-subunit of the nongastric H+, K+-ATPase, which functions to acidify the airway surface fluid and impairs mucociliary transport function in patients with cystic fibrosis. It is hypothesized that the ATP12A protein may play a role in the pathogenesis of IPF. The authors' studies demonstrate that ATP12A protein is overexpressed in distal small airways from the lungs of patients with IPF compared with normal human lungs. In addition, overexpression of the ATP12A protein in mouse lungs worsened bleomycin induced experimental pulmonary fibrosis. This was prevented by a potassium competitive proton pump blocker, vonoprazan. These data support the concept that the ATP12A protein plays an important role in the pathogenesis of lung fibrosis. Inhibition of the ATP12A protein has potential as a novel therapeutic strategy in IPF treatment.
Collapse
Affiliation(s)
| | - Katie Uhl
- Department of Pediatrics and Human Development and
| | | | - Chad Schultz
- Department of Pediatrics and Human Development and
| | - Kaylie Tripp
- Department of Pediatrics and Human Development and
| | | | | | - Bin Chen
- Department of Pediatrics and Human Development and
- Department of Pharmacology and Toxicology and
| | - Maximiliano Tamae Kakazu
- Department of Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
- Division of Pulmonary and Critical Care Medicine
| | | | - Tara E. Jager
- Richard Devos Heart and Lung Transplant Program, Spectrum Health, Grand Rapids, Michigan
| | - Cameron Lawson
- Richard Devos Heart and Lung Transplant Program, Spectrum Health, Grand Rapids, Michigan
| | | | - Nikolay Pestov
- Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, College of Medicine, University of Toledo, Health Science Campus, Toledo, Ohio
| | - Nikolai N. Modyanov
- Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, College of Medicine, University of Toledo, Health Science Campus, Toledo, Ohio
| | - Jeremy Prokop
- Department of Pediatrics and Human Development and
- Department of Pharmacology and Toxicology and
| | | | - Bruce D. Uhal
- Department of Physiology, Michigan State University, East Lansing, Michigan; and
| | - Reda E. Girgis
- Department of Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
- Division of Pulmonary and Critical Care Medicine
- Richard Devos Heart and Lung Transplant Program, Spectrum Health, Grand Rapids, Michigan
| | - Xiaopeng Li
- Department of Pediatrics and Human Development and
| |
Collapse
|
31
|
Wang J, Xian M, Cao H, Wu L, Zhou L, Ma Y, Fan L, Lin L, Li G, Huang Q, Huang SK, Xiao X. Prophylactic and therapeutic potential of magnolol-loaded PLGA-PEG nanoparticles in a chronic murine model of allergic asthma. Front Bioeng Biotechnol 2023; 11:1182080. [PMID: 37214308 PMCID: PMC10192565 DOI: 10.3389/fbioe.2023.1182080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Magnolol is a chemically defined and active polyphenol extracted from magnolia plants possessing anti-allergic activity, but its low solubility and rapid metabolism dramatically hinder its clinical application. To improve the therapeutic effects, magnolol-encapsulated polymeric poly (DL-lactide-co-glycolide)-poly (ethylene glycol) (PLGA-PEG) nanoparticles were constructed and characterized. The prophylactic and therapeutic efficacy in a chronic murine model of OVA-induced asthma and the mechanisms were investigated. The results showed that administration of magnolol-loaded PLGA-PEG nanoparticles significantly reduced airway hyperresponsiveness, lung tissue eosinophil infiltration, and levels of IL-4, IL-13, TGF-β1, IL-17A, and allergen-specific IgE and IgG1 in OVA-exposed mice compared to their empty nanoparticles-treated mouse counterparts. Magnolol-loaded PLGA-PEG nanoparticles also significantly prevented mouse chronic allergic airway mucus overproduction and collagen deposition. Moreover, magnolol-encapsulated PLGA-PEG nanoparticles showed better therapeutic effects on suppressing allergen-induced airway hyperactivity, airway eosinophilic inflammation, airway collagen deposition, and airway mucus hypersecretion, as compared with magnolol-encapsulated poly (lactic-co-glycolic acid) (PLGA) nanoparticles or magnolol alone. These data demonstrate the protective effect of magnolol-loaded PLGA-PEG nanoparticles against the development of allergic phenotypes, implicating its potential usefulness for the asthma treatment.
Collapse
Affiliation(s)
- Junyi Wang
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Mo Xian
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Cao
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lei Wu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Libo Zhou
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yihe Ma
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Long Fan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Lin Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Qinmiao Huang
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shau-Ku Huang
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaojun Xiao
- Shenzhen Key Laboratory of Allergy and Immunology, Guangdong Provincial Standardization Allergen Engineering Research Center, State Key Laboratory of Respiratory Disease Shenzhen University Division, Institute of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
32
|
Tjong J, Pendlmayr S, Barter J, Chen J, Maksym GN, Quinn TA, Frampton JP. Cell-contact-mediated assembly of contractile airway smooth muscle rings. Biomed Mater 2023; 18. [PMID: 36801856 DOI: 10.1088/1748-605x/acbd09] [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: 04/29/2022] [Accepted: 02/17/2023] [Indexed: 02/19/2023]
Abstract
Microtissues in the shape of toroidal rings provide an ideal geometry to better represent the structure and function of the airway smooth muscle present in the small airways, and to better understand diseases such as asthma. Here, polydimethylsiloxane devices consisting of a series of circular channels surrounding central mandrels are used to form microtissues in the shape of toroidal rings by way of the self-aggregation and -assembly of airway smooth muscle cell (ASMC) suspensions. Over time, the ASMCs present in the rings become spindle-shaped and axially align along the ring circumference. Ring strength and elastic modulus increase over 14 d in culture, without significant changes in ring size. Gene expression analysis indicates stable expression of mRNA for extracellular matrix-associated proteins, including collagen I and lamininsα1 andα4 over 21 d in culture. Cells within the rings respond to TGF-β1 treatment, leading to dramatic decreases in ring circumference, with increases in mRNA and protein levels for extracellular matrix and contraction-associated markers. These data demonstrate the utility of ASMC rings as a platform for modeling diseases of the small airways such as asthma.
Collapse
Affiliation(s)
- Jonathan Tjong
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada
| | - Stefan Pendlmayr
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada
| | - Jena Barter
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada.,Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Canada
| | - Julie Chen
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada
| | - Geoffrey N Maksym
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada.,Department of Physics & Atmospheric Science, Dalhousie University, Halifax, Canada
| | - T Alexander Quinn
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada.,Department of Physiology & Biophysics, Dalhousie University, Halifax, Canada
| | - John P Frampton
- School of Biomedical Engineering, Dalhousie University, Halifax, Canada.,Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Canada
| |
Collapse
|
33
|
Blood Inflammatory-like and Lung Resident-like Eosinophils Affect Migration of Airway Smooth Muscle Cells and Their ECM-Related Proliferation in Asthma. Int J Mol Sci 2023; 24:ijms24043469. [PMID: 36834879 PMCID: PMC9958882 DOI: 10.3390/ijms24043469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Airway remodeling is a hallmark feature of asthma, and one of its key structural changes is increased airway smooth muscle (ASM) mass and disturbed extracellular matrix (ECM) homeostasis. Eosinophil functions in asthma are broadly defined; however, we lack knowledge about eosinophil subtypes' interaction with lung structural cells and their effect on the airway's local microenvironment. Therefore, we investigated the effect of blood inflammatory-like eosinophils (iEOS-like) and lung resident-like eosinophils (rEOS-like) on ASM cells via impact on their migration and ECM-related proliferation in asthma. A total of 17 non-severe steroid-free allergic asthma (AA), 15 severe eosinophilic asthma (SEA) patients, and 12 healthy control subjects (HS) were involved in this study. Peripheral blood eosinophils were enriched using Ficoll gradient centrifugation and magnetic separation, subtyped by using magnetic separation against CD62L. ASM cell proliferation was assessed by AlamarBlue assay, migration by wound healing assay, and gene expression by qRT-PCR analysis. We found that blood iEOS-like and rEOS-like cells from AA and SEA patients' upregulated genes expression of contractile apparatus proteins, COL1A1, FN, TGF-β1 in ASM cells (p < 0.05), and SEA eosinophil subtypes demonstrated the highest effect on sm-MHC, SM22, and COL1A1 gene expression. Moreover, AA and SEA patients' blood eosinophil subtypes promoted migration of ASM cells and their ECM-related proliferation, compared with HS (p < 0.05) with the higher effect of rEOS-like cells. In conclusion, blood eosinophil subtypes may contribute to airway remodeling by upregulating contractile apparatus and ECM component production in ASM cells, further promoting their migration and ECM-related proliferation, with a stronger effect of rEOS-like cells and in SEA.
Collapse
|
34
|
Hahn K, Sundar IK. Current Perspective on the Role of the Circadian Clock and Extracellular Matrix in Chronic Lung Diseases. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2455. [PMID: 36767821 PMCID: PMC9915635 DOI: 10.3390/ijerph20032455] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
The circadian clock is a biochemical oscillator that rhythmically regulates physiological and behavioral processes such as inflammation, immunity, and metabolism in mammals. Circadian clock disruption is a key driver for chronic inflammatory as well as fibrotic lung diseases. While the mechanism of circadian clock regulation in the lung has been minimally explored, some evidence suggests that the transforming growth factor β (TGFβ) signaling pathway and subsequent extracellular matrix (ECM) accumulation in the lung may be controlled via a clock-dependent mechanism. Recent advancements in this area led us to believe that pharmacologically targeting the circadian clock molecules may be a novel therapeutic approach for treating chronic inflammatory lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). Here, we update the current perspective on the circadian clock role in TGFβ1 signaling and extracellular matrix production during chronic lung diseases.
Collapse
Affiliation(s)
- Kameron Hahn
- Department of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Isaac Kirubakaran Sundar
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
35
|
Bai F, Chen Z, Xu S, Han L, Zeng X, Huang S, Zhu Z, Zhou L. Wogonin attenuates neutrophilic inflammation and airway smooth muscle proliferation through inducing caspase-dependent apoptosis and inhibiting MAPK/Akt signaling in allergic airways. Int Immunopharmacol 2022; 113:109410. [DOI: 10.1016/j.intimp.2022.109410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022]
|
36
|
Saad EA, Elsaid AM, Shoaib RMS, Megahed KF, Elsharawy AN. MUC7 VNTR polymorphism and association with bronchial asthma in Egyptian children. Sci Rep 2022; 12:18910. [PMID: 36344553 PMCID: PMC9640678 DOI: 10.1038/s41598-022-21631-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
Overproduction of mucins in the airways donates largely to airway blockage in asthma patients. Glycoprotein MUC7 plays a role in the clearance of bacteria and has anti-candidacidal criteria. Our goal was to investigate the association between the MUC7 variable number of tandem repeats (VNTR) polymorphism and bronchial asthma among Egyptian children. The MUC7 VNTR polymorphism was investigated among 100 children with bronchial asthma and 100 healthy controls using polymerase chain reaction (PCR) method. Serum levels of immunoglobulin E (IgE), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta1 (TGF-β1) were assessed by enzyme-linked immunosorbent assay (ELISA) technique. The frequencies of 6*5 genotype, 5*5 genotype, (6*5 + 5*5) genotypes, and MUC7*5 allele of the MUC7 VNTR variant were significantly lower among asthmatic patients than controls (p < 0.015, OR = 0.39, 95% CI = 0.19-0.81; p = 0.03, OR = 0.18, 95% CI = 0.04-0.86; p < 0.001, OR = 0.29, 95% CI = 0.15-0.58; p < 0.001, OR = 0.3, 95% CI = 0.17-0.55, respectively). The (6*5 + 5*5) genotypes of the MUC7 VNTR variant were not associated with the clinical manifestations and serum levels of IgE, TNF-α, and TGF-β1 among asthmatic patients (p ˃ 0.05). In conclusion, the (6*5 + 5*5) genotypes of the MUC7 VNTR variant may have a protective role for bronchial asthma in Egyptian children.
Collapse
Affiliation(s)
- Entsar A. Saad
- grid.462079.e0000 0004 4699 2981Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517 Egypt
| | - Afaf M. Elsaid
- grid.10251.370000000103426662Genetics Unit, Children Hospital, Mansoura University, Mansoura, Egypt
| | - Rasha M. S. Shoaib
- grid.510451.4Food and Dairy Sciences and Technology Department, Faculty of Environmental Agricultural Sciences, Arish University, Arish, North Sinai Egypt
| | - Khaled F. Megahed
- grid.10251.370000000103426662Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amal N. Elsharawy
- grid.462079.e0000 0004 4699 2981Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517 Egypt
| |
Collapse
|
37
|
Chen XY, Zhu XJ, Chen M, Lu MP, Wang ML, Yin M, Chen RX, Wu ZF, Bu DY, Zhang ZD, Cheng L. GARP Polymorphisms Associated with Susceptibility to House Dust Mite-Sensitized Persistent Allergic Rhinitis in a Chinese Population. J Asthma Allergy 2022; 15:1369-1381. [PMID: 36196093 PMCID: PMC9527031 DOI: 10.2147/jaa.s366815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Genetic variants in GARP (also known as LRRC32) have been reported to have significant associations with asthma and eczema in special populations, but little is known about allergic rhinitis. This study purposes to evaluate the association of single nucleotide polymorphisms (SNPs) in GARP with house dust mite (HDM)-sensitized persistent allergic rhinitis (PER) in a population of Han Chinese. Methods In this hospital-based case–control study, 534 HDM-sensitized PER patients and 451 healthy controls were recruited from East China. In this population, six SNPs in GARP were identified. Serum total and specific IgE levels were measured with ImmunoCAP. Secondary structure and minimum free energy were predicted by RNAfold. Results rs79525962 was associated with the risk of HDM-sensitized PER (P < 0.05). The individuals with CT+TT genotype demonstrated a higher risk of HDM-sensitized PER than those with CC genotype (adjusted OR = 1.393, 95% CI = 1.019–1.904). The homozygous genotype CC of rs3781699 rendered a lower risk of HDM-sensitized PER than the wild-type genotype AA (adjusted OR = 0.646, 95% CI = 0.427–0.976); however, the genotype and allele frequencies of rs3781699 demonstrated no associations with HDM-sensitized PER (P > 0.05). rs79525962 increased the risk of HDM-sensitized PER in the subgroup aged ≥16 years (adjusted OR = 1.745, 95% CI = 1.103–2.760), and this high risk was also found in the females (adjusted OR = 1.708, 95% CI = 1.021–2.856). The G-C haplotype of rs1320646-rs3781699 rendered a lower risk of HDM-sensitized PER than the common haplotype G-A (adjusted OR = 0.819, 95% CI = 0.676–0.993). The secondary structure of GARP altered in response to different genotypes of rs79525962 and rs3781699. Conclusion SNP rs79525962 in the GARP gene marks a risk locus of HDM-sensitized PER in Chinese Hans.
Collapse
Affiliation(s)
- Xin-Yuan Chen
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xin-Jie Zhu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Min Chen
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Mei-Ping Lu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Mei-Lin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Min Yin
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
- International Centre for Allergy Research, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ruo-Xi Chen
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zhong-Fei Wu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Dong-Yun Bu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zheng-Dong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Zheng-Dong Zhang, Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People’s Republic of China, Email
| | - Lei Cheng
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
- International Centre for Allergy Research, Nanjing Medical University, Nanjing, People’s Republic of China
- Correspondence: Lei Cheng, Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People’s Republic of China, Email
| |
Collapse
|
38
|
Li H, Yang T, Chen T, Liu Y, Pang Y, Yang L. BRD7 restrains TNF-α-induced proliferation and migration of airway smooth muscle cells by inhibiting notch signaling. Exp Lung Res 2022; 48:199-212. [PMID: 35943053 DOI: 10.1080/01902148.2022.2107730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objective: Bromodomain-containing protein 7 (BRD7) is a key component of the switch/sucrose non-fermentable complex that participates in chromatin remodeling and transcriptional regulation. Although the emerging role of BRD7 in the pathophysiology of various diseases has been observed, its role in asthma remains unknown. Here, we assessed the function of BRD7 as a mediator of airway remodeling in asthma using an in vitro model. Methods: Airway smooth muscle cells (ASMCs) were challenged with tumor necrosis factor-α (TNF-α) to establish an in vitro airway remodeling model. Protein levels were examined using western blotting. Cell proliferation was measured using the cell counting kit-8 and 5-ethynyl-2'-deoxyuridine assays. Cell migration was assessed using a transwell migration assay. Results: Exposure to TNF-α dramatically decreased BRD7 levels in ASMCs. BRD7 remarkably decreased TNF-α-induced proliferation and migration of ASMCs. In contrast, ASMCs with BRD7 deficiency were more sensitive to TNF-α-induced pro-proliferative and pro-migratory effects. Mechanistically, BRD7 could repress the expression of Notch1 and block the Notch pathway in TNF-α-challenged cells. Notably, reactivation of Notch signaling substantially reversed the BRD7 overexpression-mediated effects, whereas restraining Notch signaling abolished BRD7-depletion-mediated effects on TNF-α-challenged cells. Conclusions: BRD7 inhibits the proliferation and migration of ASMCs elicited by TNF-α by downregulating the Notch pathway. This study indicates that BRD7 may exert a suppressive effect on airway remodeling during asthma.
Collapse
Affiliation(s)
- Hong Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Tian Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Tianjun Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Ya Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Yamei Pang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Lan Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
39
|
CHEN J, WANG Y, ZHANG N, XUE X. Shenqihuatan formula reduces inflammation by inhibiting transforming growth factor-beta-stimulated signaling pathway in airway smooth muscle cells. J TRADIT CHIN MED 2022; 42:520-529. [PMID: 35848968 PMCID: PMC9924674 DOI: 10.19852/j.cnki.jtcm.20220519.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
OBJECTIVE To study the effects and mechanism of Shenqihuatan formula (, SQHT) of the transforming growth factor-beta (TGF-β)-stimulated cell processes in airway remodeling. METHODS The current study examined cell viability using a Cell Counting Kit-8 assay. Furthermore, a Transwell assay was conducted to detect the ability of cell migration, and apoptosis was detected via flowcytometry. Western Blot and quantitative real-time polymerase chain reaction (qRT-PCR) were used to determine the expression levels of apoptosis or inflammation-related factors, such as TGF-β, Interleukin-1β (IL-1β), B cell lymphoma 2 (Bcl-2), Bcl-2-Associated X (Bax), Ras homolog gene family, member A (RhoA), recombinant rho associated coiled coil containing protein kinase 1/2 (ROCK1/2), extracellular regulated protein kinases 1/2 (ERK1/2), Snail, and Slug. Finally, the expression levels of matrix metalloproteinase-9 (MMP-9) and Tissue inhibitor of metalloproteinase (TIMP-1) were admeasured by enzyme-linked immuno sorbent assay. RESULTS The results demonstrated that SQHT inhibited the viability and migration, as well as the the F-actin formation and cytoskeletal reorganization of airway smooth muscle cells (ASMCs) stimulated by TGF-β. By monitoring the changes of critical regulators in the presence of the formula, it was observed that the expression levels of TGF-β, IL-1β, Bcl-2, RhoA, ROCK1/2, ERK1/2, Snail, and Slug were markedly suppressed, whereas Bax expression exhibited the opposite effect. Compared with a well-characterized RhoA pathway inhibitor, Fasudil, SQHT generated equivalent or even higher inhibitory effects on these processes in ASMCs. CONCLUSIONS Collectively, these suggested that SQHT can reduce airway inflammation by inhibiting TGF-β-stimulated signaling pathways in ASMCs. These findings may provide a novel remedy for treating ASMC inflammation, which causes thickening and obstruction of the airway in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Jingjing CHEN
- 1 Shanxi Provincial Traditional Chinese Medicine Hospital, Taiyuan 030012, China
| | - Yuanyuan WANG
- 2 Anhui University of Traditional Chinese Medicine, Hefei 230038, China
| | - Nianzhi ZHANG
- 3 Department of Respiratory, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China
- Prof. ZHANG Nianzhi, Department of Respiratory, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China. , Telephone: +86-13505615645
| | - Xiaoming XUE
- 1 Shanxi Provincial Traditional Chinese Medicine Hospital, Taiyuan 030012, China
| |
Collapse
|
40
|
Zhang L, Devanathadesikan Seshadri V, Abdel Aziz Ibrahim I, Han X, Ou L. Tilianin alleviates airway inflammation in ovalbumin-induced allergic asthma in mice through the regulation of Th2 cytokines and TGF–β1/Smad markers. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
41
|
Jakwerth CA, Ordovas-Montanes J, Blank S, Schmidt-Weber CB, Zissler UM. Role of Respiratory Epithelial Cells in Allergic Diseases. Cells 2022; 11:1387. [PMID: 35563693 PMCID: PMC9105716 DOI: 10.3390/cells11091387] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023] Open
Abstract
The airway epithelium provides the first line of defense to the surrounding environment. However, dysfunctions of this physical barrier are frequently observed in allergic diseases, which are tightly connected with pro- or anti-inflammatory processes. When the epithelial cells are confronted with allergens or pathogens, specific response mechanisms are set in motion, which in homeostasis, lead to the elimination of the invaders and leave permanent traces on the respiratory epithelium. However, allergens can also cause damage in the sensitized organism, which can be ascribed to the excessive immune reactions. The tight interaction of epithelial cells of the upper and lower airways with local and systemic immune cells can leave an imprint that may mirror the pathophysiology. The interaction with effector T cells, along with the macrophages, play an important role in this response, as reflected in the gene expression profiles (transcriptomes) of the epithelial cells, as well as in the secretory pattern (secretomes). Further, the storage of information from past exposures as memories within discrete cell types may allow a tissue to inform and fundamentally alter its future responses. Recently, several lines of evidence have highlighted the contributions from myeloid cells, lymphoid cells, stromal cells, mast cells, and epithelial cells to the emerging concepts of inflammatory memory and trained immunity.
Collapse
Affiliation(s)
- Constanze A. Jakwerth
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA;
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| |
Collapse
|
42
|
The Role of Osthole on TGF- β-Induced Lung Epithelium Apoptosis Injury and Epithelial-Mesenchymal Transition-Mediated Airway Remodeling in Pediatric Asthma. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7099097. [PMID: 35368934 PMCID: PMC8970801 DOI: 10.1155/2022/7099097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/19/2021] [Accepted: 01/19/2022] [Indexed: 11/18/2022]
Abstract
Osthole, a coumarin compound derived from Fructus Cnidii, exerts anti-inflammatory effects in an asthma model. But the effect of osthole on epithelial injury and epithelial-mesenchymal transition (EMT) in asthma remains unclear. 16HBE cells were incubated with TGF-β1 with or without osthole in vitro. Ovalbumin (OVA)-induced asthmatic mouse model was established in vivo. Cell counting kit-8 was carried out to evaluate the viability of 16HBE cells. The impact of osthole on TGF-β1-evoked cell apoptosis and EMT process was measured by flow cytometry based on Annexin V-FITC/PI staining, transwell assay, immunofluorescence, and Western blot. The regulatory role of osthole in TGF-β1/Smad and p38, ERK1/2, and JNK MAPK signaling was detected via Western blot. Osthole treatment significantly suppressed TGF-β1-induced 16HBE cell apoptosis, verified by a reduced percentage of apoptotic cells, decreased expression of proapoptotic proteins (cleaved-caspase3 and Bax), and enhanced antiapoptotic factor (Bcl-2) expression. In addition, the promotive impact of TGF-β1 on the migration of 16HBE cells was reversed by osthole, accompanied by elevated E-cadherin expression and reduced Snail and N-cadherin expression. The activation of the Smad2/3 and MAPKs pathway evoked by TGF-β1 was inhibited by osthole in 16HBE cells. We also found that osthole mitigated airway epithelium injury and subepithelial fibrosis in OVA-challenged asthmatic mice in vivo. Osthole could mitigate TGF-β1-induced epithelial cell injury and EMT process by suppressing the activation of MAPK and Smad2/3 pathways separately. Our present study showed a new insight into understanding the underlying mechanism of osthole injury on epithelium injury and subepithelial fibrosis in airway remodeling. Asthma, epithelial injury, epithelial-mesenchymal transition, and airway remodeling are the effects of osthole on airway remodeling.
Collapse
|
43
|
Namvar S, Labram B, Rowley J, Herrick S. Aspergillus fumigatus-Host Interactions Mediating Airway Wall Remodelling in Asthma. J Fungi (Basel) 2022; 8:jof8020159. [PMID: 35205913 PMCID: PMC8879933 DOI: 10.3390/jof8020159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 12/03/2022] Open
Abstract
Asthma is a chronic heterogeneous respiratory condition that is mainly associated with sensitivity to airborne agents such as pollen, dust mite products and fungi. Key pathological features include increased airway inflammation and airway wall remodelling. In particular, goblet cell hyperplasia, combined with excess mucus secretion, impairs clearance of the inhaled foreign material. Furthermore, structural changes such as subepithelial fibrosis and increased smooth muscle hypertrophy collectively contribute to deteriorating airway function and possibility of exacerbations. Current pharmacological therapies focused on airway wall remodelling are limited, and as such, are an area of unmet clinical need. Sensitisation to the fungus, Aspergillus fumigatus, is associated with enhanced asthma severity, bronchiectasis, and hospitalisation. How Aspergillus fumigatus may drive airway structural changes is unclear, although recent evidence points to a central role of the airway epithelium. This review provides an overview of the airway pathology in patients with asthma and fungal sensitisation, summarises proposed airway epithelial cell-fungal interactions and discusses the initiation of a tissue remodelling response. Related findings from in vivo animal models are included given the limited analysis of airway pathology in patients. Lastly, an important role for Aspergillus fumigatus-derived proteases in triggering a cascade of damage-repair events through upregulation of airway epithelial-derived factors is proposed.
Collapse
Affiliation(s)
- Sara Namvar
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (B.L.); (J.R.)
- School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, UK
- Correspondence: (S.N.); (S.H.)
| | - Briony Labram
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (B.L.); (J.R.)
| | - Jessica Rowley
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (B.L.); (J.R.)
| | - Sarah Herrick
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (B.L.); (J.R.)
- Correspondence: (S.N.); (S.H.)
| |
Collapse
|
44
|
Abstract
Asthma is chronic eosinophilic bronchitis with the dominancy of T helper 2 (Th2) inflammation. However, patients with asthma and metabolic dysfunction have pathogenic and pathological differences from those with Th2 inflammation. Metabolic dysfunction, typically presented as metabolic syndrome, has several important clinical components including central obesity, insulin resistance or glucose intolerance, dyslipidemia, and vitamin D deficiency. Data from large epidemiological studies support the significance of these components in the control of asthma and their contribution to airway remodeling, suggesting the presence of an asthma phenotype with metabolic dysfunction. These components are quite interactive with each other, so it is difficult to reveal the individual role of each. It is well known that asthma is difficult to treat in patients with obesity, due in part to inadequate response to inhaled corticosteroids. Additionally, vitamin D deficiency and insulin resistance have been regarded as aggravating factors of asthma control and airway remodeling. Recent clinical and in vivo studies have revealed the specific mechanisms of these components, which may aggravate asthma control and airway remodeling. In this review article, I summarize the recent studies and unmet needs for patients with asthma and metabolic dysfunction.
Collapse
Affiliation(s)
- Jung-Won Park
- Institute for Allergy & Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
45
|
Jacobs I, Ceulemans M, Wauters L, Breynaert C, Vermeire S, Verstockt B, Vanuytsel T. Role of Eosinophils in Intestinal Inflammation and Fibrosis in Inflammatory Bowel Disease: An Overlooked Villain? Front Immunol 2021; 12:754413. [PMID: 34737752 PMCID: PMC8560962 DOI: 10.3389/fimmu.2021.754413] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022] Open
Abstract
Eosinophils are leukocytes which reside in the gastrointestinal tract under homeostatic conditions, except for the esophagus which is normally devoid of eosinophils. Research on eosinophils has primarily focused on anti-helminth responses and type 2 immune disorders. In contrast, the search for a role of eosinophils in chronic intestinal inflammation and fibrosis has been limited. With a shift in research focus from adaptive to innate immunity and the fact that the eosinophilic granules are filled with inflammatory mediators, eosinophils are becoming a point of interest in inflammatory bowel diseases. In the current review we summarize eosinophil characteristics and recruitment as well as the current knowledge on presence, inflammatory and pro-fibrotic functions of eosinophils in inflammatory bowel disease and other chronic inflammatory conditions, and we identify research gaps which should be covered in the future.
Collapse
Affiliation(s)
- Inge Jacobs
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthias Ceulemans
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lucas Wauters
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Christine Breynaert
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Katholieke Universiteit Leuven, Leuven, Belgium
- Department of General Internal Medicine, Allergy and Clinical Immunology, University Hospitals Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
46
|
Liu P, Li S, Tang L. Nerve Growth Factor: A Potential Therapeutic Target for Lung Diseases. Int J Mol Sci 2021; 22:ijms22179112. [PMID: 34502019 PMCID: PMC8430922 DOI: 10.3390/ijms22179112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
Abstract
The lungs play a very important role in the human respiratory system. However, many factors can destroy the structure of the lung, causing several lung diseases and, often, serious damage to people's health. Nerve growth factor (NGF) is a polypeptide which is widely expressed in lung tissues. Under different microenvironments, NGF participates in the occurrence and development of lung diseases by changing protein expression levels and mediating cell function. In this review, we summarize the functions of NGF as well as some potential underlying mechanisms in pulmonary fibrosis (PF), coronavirus disease 2019 (COVID-19), pulmonary hypertension (PH), asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. Furthermore, we highlight that anti-NGF may be used in future therapeutic strategies.
Collapse
Affiliation(s)
- Piaoyang Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China;
| | - Shun Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu 610500, China
- Correspondence: (S.L.); (L.T.)
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China;
- Correspondence: (S.L.); (L.T.)
| |
Collapse
|
47
|
Bluhmki T, Traub S, Müller AK, Bitzer S, Schruf E, Bammert MT, Leist M, Gantner F, Garnett JP, Heilker R. Functional human iPSC-derived alveolar-like cells cultured in a miniaturized 96‑Transwell air-liquid interface model. Sci Rep 2021; 11:17028. [PMID: 34426605 PMCID: PMC8382767 DOI: 10.1038/s41598-021-96565-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
In order to circumvent the limited access and donor variability of human primary alveolar cells, directed differentiation of human pluripotent stem cells (hiPSCs) into alveolar-like cells, provides a promising tool for respiratory disease modeling and drug discovery assays. In this work, a unique, miniaturized 96-Transwell microplate system is described where hiPSC-derived alveolar-like cells were cultured at an air-liquid interface (ALI). To this end, hiPSCs were differentiated into lung epithelial progenitor cells (LPCs) and subsequently matured into a functional alveolar type 2 (AT2)-like epithelium with monolayer-like morphology. AT2-like cells cultured at the physiological ALI conditions displayed characteristics of AT2 cells with classical alveolar surfactant protein expressions and lamellar-body like structures. The integrity of the epithelial barriers between the AT2-like cells was confirmed by applying a custom-made device for 96-parallelized transepithelial electric resistance (TEER) measurements. In order to generate an IPF disease-like phenotype in vitro, the functional AT2-like cells were stimulated with cytokines and growth factors present in the alveolar tissue of IPF patients. The cytokines stimulated the secretion of pro-fibrotic biomarker proteins both on the mRNA (messenger ribonucleic acid) and protein level. Thus, the hiPSC-derived and cellular model system enables the recapitulation of certain IPF hallmarks, while paving the route towards a miniaturized medium throughput approach of pharmaceutical drug discovery.
Collapse
Affiliation(s)
- Teresa Bluhmki
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany.
| | - Stefanie Traub
- Trenzyme GmbH, Byk-Gulden-Str. 2, 78467, Constance, Germany
| | | | - Sarah Bitzer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Eva Schruf
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Marie-Therese Bammert
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Marcel Leist
- In-vitro Toxicology and Biomedicine, University of Konstanz, 78457, Constance, Germany
| | - Florian Gantner
- Department of Translational Medicine and Clinical Pharmacology, C. H. Boehringer Sohn AG & Co. KG, 88397, Biberach an der Riss, Germany
| | - James P Garnett
- Department of Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Ralf Heilker
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| |
Collapse
|
48
|
Xing Q, You Y, Zhao X, Ji J, Yan H, Dong Y, Ren L, Ding Y, Hou S. iTRAQ-Based Proteomics Reveals Gu-Ben-Fang-Xiao Decoction Alleviates Airway Remodeling via Reducing Extracellular Matrix Deposition in a Murine Model of Chronic Remission Asthma. Front Pharmacol 2021; 12:588588. [PMID: 34194321 PMCID: PMC8237094 DOI: 10.3389/fphar.2021.588588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
Airway remodeling is a primary pathological feature of asthma. The current therapy for asthma mainly targets reducing inflammation but not particularly airway remodeling. Therefore, it is worthwhile to develop alternative and more effective therapies to attenuate remodeling. Gu-Ben-Fang-Xiao Decoction (GBFXD) has been used to effectively and safely treat asthma for decades. In this study, GBFXD regulated airway inflammation, collagen deposition, and the molecules relevant to airway remodeling such as Vimentin, α-SMA, hydroxyproline, and E-cadherin in chronic remission asthma (CRA) murine model. Proteomic analysis indicated that the overlapping differentially expressed proteins (DEPs) (Model/Control and GBFXD/Model) were mainly collagens and laminins, which were extracellular matrix (ECM) proteins. In addition, the KEGG analysis showed that GBFXD could regulate pathways related to airway remodeling including ECM-receptor interactions, focal adhesion, and the PI3K/AKT signaling pathway, which were the top three significantly enriched pathways containing the most DEPs for both Model/Control and GBFXD/Model. Further validation research showed that GBFXD regulated reticulon-4 (RTN4) and suppressed the activation of the PI3K/AKT pathway to alleviate ECM proteins deposition. In conclusion, our findings indicate that GBFXD possibly regulate the PI3K/AKT pathway via RTN4 to improve airway remodeling, which provides a new insight into the molecular mechanism of GBFXD for the treatment of CRA.
Collapse
Affiliation(s)
- Qiongqiong Xing
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Yannan You
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Xia Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Jianjian Ji
- Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Hua Yan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Yingmei Dong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Lishun Ren
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Yuanyuan Ding
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| | - Shuting Hou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing, China
| |
Collapse
|
49
|
Li Q, Zhai C, Wang G, Zhou J, Li W, Xie L, Shi Z. Ginsenoside Rh1 attenuates ovalbumin-induced asthma by regulating Th1/Th2 cytokines balance. Biosci Biotechnol Biochem 2021; 85:1809-1817. [PMID: 34057179 DOI: 10.1093/bbb/zbab099] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/08/2021] [Indexed: 12/24/2022]
Abstract
Ginsenoside Rh1 (Rh1) has anti-inflammatory effects in asthma mice, but the underlying mechanism remains unclear. BALB/c mice were sensitized and challenged with ovalbumin (OVA) to construct asthma model. Mice received Rh1 or tiotropium bromide 0.5 h before OVA challenge. Airway morphology and airway remodeling were assessed by HE staining and Masson's trichrome staining, respectively. Th1/Th2 cytokines in serum or broncho alveolar lavage fluid (BALF) were measured by ELISA kits. Rh1 significantly alleviated the lung resistance and airway resistance, and reduced the number of total inflammation cells, eosinophils, neutrophils, and lymphocytes in BALF of the asthmatic mice. The morphological changes and collagen deposition of airway were also reduced by Rh1 in asthmatic mice. The increase of Eotaxin, IL-4, IL-5, IL-13, and IL-33 and the decrease of IL-12 and IFN-γ in both BALF and serum of OVA exposed mice were reversed by Rh1. Rh1 attenuates OVA-induced asthma in the mice model by regulating Th1/Th2 cytokines balance.
Collapse
Affiliation(s)
- Qiang Li
- Department of Geriatrics, Hangzhou Hospital of Traditional Chinese Medicine, Xihu District, Hangzhou City, Zhejiang Province, China
| | - Chunmiao Zhai
- Department of Rehabilitation Medicine, Hangzhou Dingqiao Hospital, Jianggan District, Hangzhou City, Zhejiang Province, China
| | - Guodong Wang
- Department of Geriatrics, Hangzhou Hospital of Traditional Chinese Medicine, Xihu District, Hangzhou City, Zhejiang Province, China
| | - Jia Zhou
- Department of Geriatrics, Hangzhou Hospital of Traditional Chinese Medicine, Xihu District, Hangzhou City, Zhejiang Province, China
| | - Weiguang Li
- Department of Geriatrics, Hangzhou Hospital of Traditional Chinese Medicine, Xihu District, Hangzhou City, Zhejiang Province, China
| | - Liquan Xie
- Department of Geriatrics, Hangzhou Hospital of Traditional Chinese Medicine, Xihu District, Hangzhou City, Zhejiang Province, China
| | - Zhanli Shi
- Department of Geriatrics, Hangzhou Red Cross Hospital, Xiacheng District, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
50
|
Hur J, Rhee CK, Lee SY, Kim YK, Kang JY. MicroRNA-21 inhibition attenuates airway inflammation and remodelling by modulating the transforming growth factor β-Smad7 pathway. Korean J Intern Med 2021; 36:706-720. [PMID: 33601867 PMCID: PMC8137415 DOI: 10.3904/kjim.2020.132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/05/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND/AIMS Current asthma therapies remain unsatisfactory for controlling airway remodelling in asthma. MicroRNA-21 is a key player in asthma pathogenesis, but the molecular mechanisms underlying its effects on airway remodelling are not completely understood. We investigated the effects of inhibition of microRNA-21 on allergic airway inflammation and remodelling. METHODS Female BALB/c mice were divided into four groups: control, ovalbumin-sensitized and -challenged for 3 months, microRNA-negative control-treated ovalbumin-treated, and microRNA-21 inhibitor-treated ovalbumin-treated groups. Parameters related to airway remodelling, cytokine production, airway inflammation, and airway hyperresponsiveness were compared between groups. Human bronchial smooth muscle cells were used in a mechanism study. RESULTS In this asthma model, ovalbumin-sensitized and -challenged mice exhibited allergic airway inf lammation and airway remodelling. MicroRNA-21 inhibitor-treated mice had fewer inflammatory cells, lower TH2 cytokine production, and suppressed parameters related to remodelling such as goblet cell hyperplasia, collagen deposition, hydroxyproline content, and expression of smooth muscle actin. Inhibition of microRNA-21 decreased transforming growth factor β1 expression and induced Smad7 expression in lung tissue. In human bronchial smooth muscle cells stimulated with transforming growth factor β1, microRNA-21 inhibition upregulated Smad7 expression and decreased markers of airway remodelling. CONCLUSION Inhibition of microRNA-21 had both anti-inflammatory and anti-remodelling effects in this model of ovalbumin-induced chronic asthma. Our data suggest that the microRNA-21-transforming growth factor β1-Smad7 axis modulates the pathogenesis of ovalbumin-induced chronic asthma and in human bronchial smooth muscle cells. MicroRNA-21 inhibitors may be a novel therapeutic target in patients with allergic asthma, especially those with airway remodelling.
Collapse
Affiliation(s)
- Jung Hur
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chin Kook Rhee
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sook Young Lee
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young Kyoon Kim
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Young Kang
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|