1
|
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
|
2
|
Zhou J, Feng D, Chen Y, Li X, Cen J, Wu W, Zheng W, Gan W, Zhang T. Effect of leucine on mitochondria and oxidative stress to reduce virulence and pathogenicity of Acinetobacter baumannii. Microbiol Res 2025; 290:127932. [PMID: 39454348 DOI: 10.1016/j.micres.2024.127932] [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: 06/19/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Elucidating the virulence mechanisms of A. baumannii is essential for developing strategies to mitigate pathogenicity. Although high-virulent strains are associated with increased mortality rate in severely infected patients, the underlying mechanisms remains not well understood. Our analysis revealed leucine as a pivotal biomarker, with the 11dP and paaK being significant contributors to virulence. The ATP-dependent activity and antioxidant activity were identified as the most important pathways in distinguishing the virulence of A. baumannii. Exogenous leucine was found to modulate mitochondria dysfunction and oxidative stress, thereby diminishing the pathogenicity of A. baumannii towards Beas 2B cells. Moreover, leucine reduced the virulence of A. baumannii to Galleria mellonella (G. mellonella) and alleviated pathological damage to lung tissues in mice. Our study offers a novel treatment strategy based on metabolomics, which may assist in the exploration and management of infections caused by highly virulent pathogens. It sets a new course for reducing the impact of highly virulent A. baumannii infections and has significant implications for the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Jianxia Zhou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Dingyun Feng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Yuetao Chen
- The State Key Laboratory of Bio-Control, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Xia Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Jiemei Cen
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Wenbin Wu
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Wenzheng Zheng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Wenlei Gan
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China
| | - Tiantuo Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
3
|
Guo X, Wang P, Wei H, Yan J, Zhang D, Qian Y, Guo B. Interleukin(IL)-37 attenuates isoproterenol (ISO)-induced cardiac hypertrophy by suppressing JAK2/STAT3-signaling associated inflammation and oxidative stress. Int Immunopharmacol 2024; 142:113134. [PMID: 39293311 DOI: 10.1016/j.intimp.2024.113134] [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: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Inflammation and oxidative stress have drawn more and more interest in the realm of cardiovascular disease. In many different disorders, IL-37 acts as an anti-inflammatory and suppressor of inflammation. This study aimed to investigate whether IL-37 could alleviate cardiac hypertrophy by reducing inflammation and oxidative stress. METHODS In vivo, a cardiac hypertrophy model was induced by 14 d of daily isoproterenol (ISO, 30 mg/kg/d) injection, followed by weeks of treatment with recombinant human IL-37 (1000 ng/animal), administered three times weekly. Assessments concentrated on markers of inflammation and oxidative stress, apoptosis, myocardial disease, and cardiac shape and function. In vitro, neonatal rat cardiomyocytes (NRCMs) were subjected to ISO (10 µM) to establish a cardiomyocytes hypertrophy model. Subsequent IL-37 treatment (100 ng/ml) was applied to determine its cardioprotective efficacy and to elucidate further the underlying mechanisms involved. RESULTS Significant cardioprotective benefits of IL-37 were seen (in vitro as well as in vivo), primarily through the reduction of oxidative stress, inflammation, apoptosis, and heart hypertrophy markers. Furthermore, IL-37 treatment was associated with a decrease in JAK2 and STAT3 phosphorylation. It is interesting to note that WP1066, a JAK2/STAT3 inhibitor, exhibited antioxidant and anti-inflammatory properties comparable to IL-37, as well as synergistic effects when mixed with the latter. CONCLUSION ISO-induced cardiac hypertrophy is lessened by IL-37 through the reduction of oxidative stress and inflammation. Additionally, the effects of IL-37 are closely related to inactivation of the JAK2/STAT3 signaling pathway. It is anticipated that IL-37 will one day be used to treat cardiovascular illnesses such as heart hypertrophy.
Collapse
Affiliation(s)
- Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Pengfei Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Huiqing Wei
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Jie Yan
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Donglei Zhang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Yuxing Qian
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China; Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
4
|
Marshall CL, Mostafa D, Hemshehkar M, Lao Y, Balshaw R, Spicer V, Mookherjee N. Biological Sex Is an Effect Modifier of Allergen-Mediated Alteration of the Lung Proteome. J Proteome Res 2024; 23:4203-4215. [PMID: 39214566 DOI: 10.1021/acs.jproteome.4c00025] [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] [Indexed: 09/04/2024]
Abstract
Asthma exhibits a distinct sex bias in the disease prevalence, severity, and response to therapy. However, sex-related differences in alterations of the lung proteome mediated by aeroallergens critical in asthma, such as house dust mites (HDM), remain unknown. In this study, we define sex-related differences in the lung proteome using an HDM-challenged mouse model by 1D LC-MS/MS. Sex-disaggregated data analysis showed that 406 proteins were uniquely altered in females, 273 proteins were uniquely altered in males, and 414 proteins were altered in both females and males in response to HDM. In a linear mixed model analysis, sex modified the HDM exposure effect for 163 proteins, i.e., a significant sex:exposure interaction was identified in 84 proteins in females and 35 proteins in males. Of these, 12 proteins showed a significant sex effect in both female and male lungs. We further selected 3 proteins Tjp1, Lamtor1, and G3BP2 for independent confirmation studies. Our findings detail the sex-specific lung proteome in response to an aeroallergen critical in asthma and demonstrate that sex is a significant effect modifier of HDM response. These results will serve as a valuable resource for delineating sex-specific mechanisms in aeroallergen-driven responses in asthma research.
Collapse
Affiliation(s)
- Courtney Lynn Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Dina Mostafa
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Mahadevappa Hemshehkar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Ying Lao
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Robert Balshaw
- George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T6, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| | - Neeloffer Mookherjee
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E0T5, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Mercedes-Benz, Winnipeg R3E3P4, Canada
| |
Collapse
|
5
|
Chen JH, Li JJ, Yuan Y, Tian Q, Feng DD, Zhuang LL, Cao Q, Zhou GP, Jin R. ETS1 and RBPJ transcriptionally regulate METTL14 to suppress TGF-β1-induced epithelial-mesenchymal transition in human bronchial epithelial cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167349. [PMID: 39002703 DOI: 10.1016/j.bbadis.2024.167349] [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: 02/27/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling. Epithelial-mesenchymal transition (EMT) of bronchial epithelial cells is considered to be a crucial player in asthma. Methyltransferase-like 14 (METTL14), an RNA methyltransferase, is implicated in multiple pathological processes, including EMT, cell proliferation and migration. However, the role of METTL14 in asthma remains uncertain. This research aimed to explore the biological functions of METTL14 in asthma and its underlying upstream mechanisms. METTL14 expression was down-regulated in asthmatic from three GEO datasets (GSE104468, GSE165934, and GSE74986). Consistent with this trend, METTL14 was decreased in the lung tissues of OVA-induced asthmatic mice and transforming growth factor-β1 (TGF-β1)-stimulated human bronchial epithelial cells (Beas-2B) in this study. Overexpression of METTL14 caused reduction in mesenchymal markers (FN1, N-cad, Col-1 and α-SMA) in TGF-β1-treated cells, but caused increase in epithelial markers (E-cad), thus inhibiting EMT. Also, METTL14 suppressed the proliferation and migration ability of TGF-β1-treated Beas-2B cells. Two transcription factors, ETS1 and RBPJ, could both bind to the promoter region of METTL14 and drive its expression. Elevating METTL14 expression could reversed EMT, cell proliferation and migration promoted by ETS1 or RBPJ deficiency. These results indicate that the ETS1/METTL14 and RBPJ/METTL14 transcription axes exhibit anti-EMT, anti-proliferation and anti-migration functions in TGF-β1-induced bronchial epithelial cells, implying that METTL14 may be considered an alternative candidate target for the treatment of asthma.
Collapse
Affiliation(s)
- Jia-He Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Jiao-Jiao Li
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Yue Yuan
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Dan-Dan Feng
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Li-Li Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China; Clinical Allergy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China; Clinical Allergy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
6
|
Geng X, Yuan J, Xu W, Zou D, Sun Y, Li J. YWHAB is regulated by IRX5 and inhibits the migration and invasion of breast cancer cells. Oncol Lett 2024; 28:469. [PMID: 39119237 PMCID: PMC11306988 DOI: 10.3892/ol.2024.14602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/06/2024] [Indexed: 08/10/2024] Open
Abstract
Highly metastatic and heterogeneous breast cancer affects the health of women worldwide. Abnormal expression of tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein β (YWHAB), also known as 14-3-3β, is associated with the tumorigenesis and progression of bladder cancer, lung cancer and hepatocellular carcinoma; however, to the best of our knowledge, the role of YWHAB in breast cancer remains unknown. In the present study, a dual luciferase assay demonstrated that the transcription factor iroquois homeobox 5 may regulate YWHAB expression by affecting the promoter sequence upstream of its transcription start site. Subsequently, it was demonstrated that overexpression of YWHAB did not affect proliferation, but did reduce the migration and invasion of MDA-MB-231 cells. Furthermore, knockdown of YWHAB promoted the migration and invasion of MCF7 cells. Transcriptomics analysis demonstrated that when YWHAB was overexpressed, 61 genes were differentially expressed, of which 43 genes were upregulated and 18 genes were downregulated. These differentially expressed genes (DEGs) were enriched in cancer-related pathways, such as 'TNF signaling pathway' [Kyoto Encyclopedia of Genes and Genomes (KEGG): map04688]. The pathway with the largest number of DEGs was 'Rheumatoid arthritis' (KEGG: map05323). Notably, YWHAB downregulated vimentin, which is a mesenchymal marker, thus suggesting that it may weaken the mesenchymal properties of cells. These findings indicate that YWHAB may be a potential therapeutic target in breast cancer and further work should be performed to assess its actions as a potential tumor suppressor.
Collapse
Affiliation(s)
- Xuexia Geng
- School of Life Science, Huaibei Normal University, Huaibei, Anhui 235000, P.R. China
| | - Jun Yuan
- School of Life Science, Huaibei Normal University, Huaibei, Anhui 235000, P.R. China
| | - Wenjie Xu
- School of Life Science, Huaibei Normal University, Huaibei, Anhui 235000, P.R. China
| | - Deng Zou
- School of Life Science, Huaibei Normal University, Huaibei, Anhui 235000, P.R. China
| | - Yuxuan Sun
- School of Life Science, Huaibei Normal University, Huaibei, Anhui 235000, P.R. China
| | - Jun Li
- School of Life Science, Huaibei Normal University, Huaibei, Anhui 235000, P.R. China
| |
Collapse
|
7
|
Tang L, Chen B, Wang B, Xu J, Yan H, Shan Y, Zhao X. Mediation of FOXA2/IL-6/IL-6R/STAT3 signaling pathway mediates benzo[a]pyrene-induced airway epithelial mesenchymal transformation in asthma. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 357:124384. [PMID: 38901818 DOI: 10.1016/j.envpol.2024.124384] [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: 04/05/2024] [Revised: 05/30/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Benzo [a]pyrene (BaP), a toxic pollutant, increases the incidence and severity of asthma. However, the molecular mechanisms underlying the effects of BaP in asthma remain unclear. In terms of research methods, we used BaP to intervene in the animal model of asthma and the human bronchial epithelial (16HBE) cells, and the involved mechanisms were found from the injury, inflammation, and airway epithelial to mesenchymal transition (EMT) in asthma. We also constructed small interfering RNAs and overexpression plasmids to knockdown/overexpress IL-6R and FOXA2 in 16HBE cells and a serotype 9 adeno-associated viral vector for lung tissue overexpression of FOXA2 in mice to determine the mechanism of action of BaP-exacerbated asthma airway EMT. We observed that BaP aggravated inflammatory cell infiltration into the lungs, reduced the Penh value, increased collagen fibres in the lung tissue, and increased serum IgE levels in asthmatic mice. After BaP intervention, the expression of FOXA2 in the lung tissue of asthmatic mice decreased, the production and secretion of IL-6 were stimulated, and STAT3 phosphorylation and nuclear translocation increased, leading to changes in EMT markers. However, EMT decreased after increasing FOXA2 expression and decreasing that of IL-6R and was further enhanced after low FOXA2 expression. Our results revealed that BaP exacerbated airway epithelial cell injury and interfered with FOXA2, activating the IL-6/IL-6R/STAT3 signaling pathway to promote airway EMT in asthma. These findings provide toxicological evidence for the mechanism underlying the contribution of BaP to the increased incidence of asthma and its exacerbations.
Collapse
Affiliation(s)
- Lingling Tang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Bailei Chen
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Bohan Wang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210004, China
| | - Jing Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Hua Yan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yiwen Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xia Zhao
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China; Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210004, China.
| |
Collapse
|
8
|
Feng K, Cen J, Zou X, Zhang T. Novel insight into MDA-7/IL-24: A potent therapeutic target for autoimmune and inflammatory diseases. Clin Immunol 2024; 266:110322. [PMID: 39033900 DOI: 10.1016/j.clim.2024.110322] [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: 05/29/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Melanoma differentiation-associated gene-7 (MDA-7)/interleukin-24 (IL-24) is a pleiotropic member of the IL-10 family of cytokines, and is involved in multiple biological processes, including cell proliferation, cell differentiation, tissue fibrosis, the inflammatory response, and antitumor activity. MDA-7/IL-24 can regulate epithelial integrity, homeostasis, mucosal immunity and host resistance to various pathogens by enhancing immune and inflammatory responses. Our recent study revealed the mechanism of MDA-7/IL-24 in promoting airway inflammation and airway remodeling through activating the JAK/STAT3 and ERK signaling pathways in bronchial epithelial cells. Herein, we summarize the cellular sources, inducers, target cells, signaling pathways, and biological effects of MDA-7/IL-24 in several allergic and autoimmune diseases. This review also synopsizes recent advances in clinical research targeting MDA-7/IL-24 or its receptors. Based on these advancements, we emphasize its potential as a target for immunotherapy and discuss the challenges of developing immunotherapeutic drugs targeting MDA-7/IL-24 or its receptors in autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Kangni Feng
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Jiemei Cen
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Xiaoling Zou
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Tiantuo Zhang
- Department of Pulmonary and Critical Care Medicine, the Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
9
|
Goleij P, Rahimi M, Pourshahroudi M, Tabari MAK, Muhammad S, Suteja RC, Daglia M, Majma Sanaye P, Hadipour M, Khan H, Sadeghi P. The role of IL-2 cytokine family in asthma. Cytokine 2024; 180:156638. [PMID: 38761716 DOI: 10.1016/j.cyto.2024.156638] [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: 03/09/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND The interleukin-2 (IL-2) family of cytokines, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, are pivotal regulators of the immune response, impacting both innate and adaptive immunity. Understanding their molecular characteristics, receptor interactions, and signalling pathways is essential for elucidating their roles in health and disease. OBJECTIVES This review provides a comprehensive overview of the IL-2 family of cytokines, highlighting their molecular biology, receptor interactions, and signalling mechanisms. Furthermore, it explores the involvement of IL-2 family cytokines in the pathogenesis of chronic respiratory diseases, with a specific focus on chronic obstructive pulmonary disease (COPD) and asthma. METHODS A thorough literature review was conducted to gather insights into the molecular biology, receptor interactions, and signalling pathways of IL-2 family cytokines. Additionally, studies investigating the roles of these cytokines in chronic respiratory diseases, particularly COPD and asthma, were analysed to discern their implications in wider pathophysiology of disease. RESULTS IL-2 family cytokines exert pleiotropic effects on immune cells, modulating cellular proliferation, differentiation, and survival. Dysregulation of IL-2 family cytokines has been implicated in the pathogenesis of chronic respiratory illnesses, including COPD and asthma. Elevated levels of IL-2 and IL-9 have been associated with disease severity in COPD, while IL-4 and IL-9 play crucial roles in asthma pathogenesis by promoting airway inflammation and remodelling. CONCLUSION Understanding the intricate roles of IL-2 family cytokines in chronic respiratory diseases provides valuable insights into potential therapeutic targets for these conditions. Targeting specific cytokines or their receptors may offer novel treatment modalities to attenuate disease progression and improve clinical outcomes in patients with COPD and asthma.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mohammad Rahimi
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran.
| | - Motahareh Pourshahroudi
- Department of Public Health, Faculty of Health, Education and Life Sciences, Birmingham City University, Birmingham, United Kingdom.
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran; Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Syed Muhammad
- Farooqia College of Pharmacy, Mysuru, Karnataka, India.
| | | | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | | | - Mahboube Hadipour
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| | - Parniyan Sadeghi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Cheng WH, Chang PL, Wu YC, Wang SA, Chen CL, Hsu FL, Neoh MM, Lin LY, Yuliani FS, Lin CH, Chen BC. Neutralization of CX3CL1 Attenuates TGF-β-Induced Fibroblast Differentiation Through NF-κB Activation and Mitochondrial Dysfunction in Airway Fibrosis. Lung 2024; 202:343-356. [PMID: 38678499 DOI: 10.1007/s00408-024-00701-6] [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: 01/25/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Severe asthma, characterized by inflammation and airway remodeling, involves fibroblast differentiation into myofibroblasts expressing α-SMA. This process leads to the production of fibronectin and connective tissue growth factor (CTGF), driven by factors such as transforming growth factor (TGF)-β. Furthermore, the persistent presence of myofibroblasts is associated with resistance to apoptosis and mitochondrial dysfunction. The chemokine (C-X3-C motif) ligand 1 (CX3CL1) plays a role in tissue fibrosis. However, it is currently unknown whether neutralization of CX3CL1 decreases TGF-β-induced fibroblast differentiation and mitochondrial dysfunction in normal human lung fibroblasts (NHLFs). METHODS CX3CL1/C-X3-C motif chemokine receptor 1 (CX3CR1), CX3CL1 was analyzed by immunofluorescence (IF) or immunohistochemical (IHC) staining of ovalbumin-challenged mice. CX3CL1 release was detected by ELISA. TGF-β-induced CTGF, fibronectin, and α-SMA expression were evaluated in NHLFs following neutralization of CX3CL1 (TP213) treatment for the indicated times by Western blotting or IF staining. Mitochondrion function was detected by a JC-1 assay and seahorse assay. Cell apoptosis was observed by a terminal uridine nick-end labeling (TUNEL) assay. RESULTS An increase in CX3CL1 expression was observed in lung tissues from mice with ovalbumin-induced asthma by IF staining. CX3CR1 was increased in the subepithelial layer of the airway by IHC staining. Moreover, CX3CR1 small interfering (si)RNA downregulated TGF-β-induced CTGF and fibronectin expression in NHLFs. CX3CL1 induced CTGF and fibronectin expression in NHLFs. TGF-β-induced CX3CL1 secretion from NHLFs. Furthermore, TP213 decreased TGF-β-induced CTGF, fibronectin, and α-SMA expression in NHLFs. Mitochondrion-related differentially expressed genes (DEGs) were examined after CX3CL1 neutralization in TGF-β-treated NHLFs. TP213 alleviated TGF-β-induced mitochondrial dysfunction and apoptosis resistance in NHLFs. CX3CL1 induced p65, IκBα, and IKKα phosphorylation in a time-dependent manner. Furthermore, CX3CL1-induced fibronectin expression and JC-1 monomer were decreased by p65 siRNA. TP213 reduced TGF-β-induced p65 and α-SMA expression in NHLFs. CONCLUSIONS These findings suggest that neutralizing CX3CL1 attenuates lung fibroblast activation and mitochondrial dysfunction. Understanding the impacts of CX3CL1 neutralization on fibroblast mitochondrial function could contribute to the development of therapeutic strategies for managing airway remodeling in severe asthma.
Collapse
Affiliation(s)
- Wun-Hao Cheng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Respiratory Therapy, Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pao-Lung Chang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chih Wu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shao-An Wang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Feng-Lin Hsu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Mei-May Neoh
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Lee-Yuan Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Fara Silvia Yuliani
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Chien-Huang Lin
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
11
|
Mao Z, Ding Z, Liu Z, Shi Y, Zhang Q. miR-21-5p Modulates Airway Inflammation and Epithelial-Mesenchymal Transition Processes in a Mouse Model of Combined Allergic Rhinitis and Asthma Syndrome. Int Arch Allergy Immunol 2024; 185:775-785. [PMID: 38588656 PMCID: PMC11309074 DOI: 10.1159/000538252] [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: 12/22/2023] [Accepted: 03/05/2024] [Indexed: 04/10/2024] Open
Abstract
INTRODUCTION Combined allergic rhinitis and asthma syndrome (CARAS) is a concurrent allergic symptom of diseases of allergic rhinitis and asthma. However, the mechanism of CARAS remains unclear. The study aimed to investigate the impact of microRNA-21 (miR-21) on CARAS via targeting poly (ADP-ribose) polymerase-1 (PARP-1) and phosphoinositide 3-kinase (PI3K)/AKT pathways. METHODS The levels of miR-21-5p and PARP-1 in CARAS patients were detected by quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA). An ovalbumin-sensitized mouse model of CARAS was established. And knock down of miR-21-5p was constructed by intranasally administering with miR-21-5p shRNA-encoding adeno-associated virus vector. Airway resistance and airway inflammatory response were detected. ELISA was used to evaluate IL-4/IL-5/IL-13 levels in bronchoalveolar lavage fluid (BALF). Expression levels of E-cadherin, fibronectin, and α-SMA were determined using Western blotting. The levels of PARP-1 and the activation of PI3K/AKT were assayed. RESULTS Downregulation of miR-21-5p relieved pathophysiological symptoms of asthma including airway hyperreactivity and inflammatory cell infiltration. Downregulation of miR-21-5p significantly reduced the levels of IL4, IL-5, and IL-13 in BALF. Additionally, downregulation of miR-21-5p inhibited the epithelial-mesenchymal transition (EMT) process in CARAS mice. Furthermore, miR-21-5p regulated PARP-1 and was involved in PI3K/AKT activation in CARAS mice. CONCLUSION Downregulation of miR-21-5p ameliorated CARAS-associated lung injury by alleviating airway inflammation, inhibiting the EMT process, and regulating PARP-1/PI3K/AKT in a mouse model of CARAS.
Collapse
Affiliation(s)
- Zhengdao Mao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Ziqi Ding
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Zhiguang Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
12
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
13
|
Qin Y, Shao B, Ren SH, Ye K, Qin H, Wang HD, Sun C, Zhu Y, Wang Z, Zhang J, Li X, Wang H. Interleukin-37 contributes to endometrial regenerative cell-mediated immunotherapeutic effect on chronic allograft vasculopathy. Cytotherapy 2024; 26:299-310. [PMID: 38159090 DOI: 10.1016/j.jcyt.2023.12.004] [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: 05/19/2023] [Revised: 11/26/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AIMS Chronic allograft vasculopathy (CAV) remains a predominant contributor to late allograft failure after organ transplantation. Several factors have already been shown to facilitate the progression of CAV, and there is still an urgent need for effective and specific therapeutic approaches to inhibit CAV. Human mesenchymal-like endometrial regenerative cells (ERCs) are free from the deficiencies of traditional invasive acquisition methods and possess many advantages. Nevertheless, the exact immunomodulation mechanism of ERCs remains to be elucidated. METHODS C57BL/6 (B6) mouse recipients receiving BALB/c mouse donor abdominal aorta transplantation were treated with ERCs, negative control (NC)-ERCs and interleukin (IL)-37-/-ERCs (ERCs with IL-37 ablation), respectively. Pathologic lesions and inflammatory cell infiltration in the grafts, splenic immune cell populations, circulating donor-specific antibody levels and cytokine profiles were analyzed on postoperative day (POD) 40. The proliferative capacities of Th1, Th17 and Treg subpopulations were assessed in vitro. RESULTS Allografts from untreated recipients developed typical pathology features of CAV, namely endothelial thickening, on POD 40. Compared with untreated and IL-37-/-ERC-treated groups, IL-37-secreting ERCs (ERCs and NC-ERCs) significantly reduced vascular stenosis, the intimal hyperplasia and collagen deposition. IL-37-secreting ERCs significantly inhibited the proliferation of CD4+T cells, reduced the proportions of Th1 and Th17 cells, but increased the proportion of Tregs in vitro. Furthermore, in vitro results also showed that IL-37-secreting ERCs significantly inhibited Th1 and Th17 cell responses, abolished B-cell activation, diminished donor-specific antibody production and increased Treg proportions. Notably, IL-37-secreting ERCs remarkably downregulated the levels of pro-inflammatory cytokines (interferon-γ, tumor necrosis factor-α, IL-1β, IL-6 and IL-17A) and increased IL-10 levels in transplant recipients. CONCLUSIONS The knockdown of IL-37 dramatically abrogates the therapeutic ability of ERCs for CAV. Thus, this study highlights that IL-37 is indispensable for ERC-mediated immunomodulation for CAV and improves the long-term allograft acceptance.
Collapse
Affiliation(s)
- Yafei Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Department of Vascular Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, PR China.
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Shao-Hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Kui Ye
- Department of Vascular Surgery, Tianjin Fourth Central Hospital, The Fourth Central Clinical College, Tianjin Medical University, Tianjin, PR China.
| | - Hong Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Chenglu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yanglin Zhu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Zhaobo Wang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, PR China.
| | - Jingyi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, PR China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, PR China.
| |
Collapse
|
14
|
Yoshie S, Murono S, Hazama A. Approach for Elucidating the Molecular Mechanism of Epithelial to Mesenchymal Transition in Fibrosis of Asthmatic Airway Remodeling Focusing on Cl - Channels. Int J Mol Sci 2023; 25:289. [PMID: 38203460 PMCID: PMC10779031 DOI: 10.3390/ijms25010289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Airway remodeling caused by asthma is characterized by structural changes of subepithelial fibrosis, goblet cell metaplasia, submucosal gland hyperplasia, smooth muscle cell hyperplasia, and angiogenesis, leading to symptoms such as dyspnea, which cause marked quality of life deterioration. In particular, fibrosis exacerbated by asthma progression is reportedly mediated by epithelial-mesenchymal transition (EMT). It is well known that the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling is closely associated with several signaling pathways, including the TGF-β1/Smad, TGF-β1/non-Smad, and Wnt/β-catenin signaling pathways. However, the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling has not yet been fully clarified. Given that Cl- transport through Cl- channels causes passive water flow and consequent changes in cell volume, these channels may be considered to play a key role in EMT, which is characterized by significant morphological changes. In the present article, we highlight how EMT, which causes fibrosis and carcinogenesis in various tissues, is strongly associated with activation or inactivation of Cl- channels and discuss whether Cl- channels can lead to elucidation of the molecular mechanism of EMT in fibrosis of asthmatic airway remodeling.
Collapse
Affiliation(s)
- Susumu Yoshie
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Shigeyuki Murono
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akihiro Hazama
- Department of Cellular and Integrative Physiology, Graduate School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
15
|
Zhang M, Zhou JX, Huang CQ, Feng KN, Zou XL, Cen JM, Meng P, Li HT, Zhang TT. IL-38 alleviates airway remodeling in chronic asthma via blocking the profibrotic effect of IL-36γ. Clin Exp Immunol 2023; 214:260-274. [PMID: 37586814 PMCID: PMC10719219 DOI: 10.1093/cei/uxad099] [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: 02/18/2023] [Revised: 06/30/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023] Open
Abstract
Airway remodeling is a major feature of asthma. Interleukin (IL)-36γ is significantly upregulated and promotes airway hyper-responsiveness (AHR) in asthma, but its role in airway remodeling is unknown. Here, we aimed to investigate the role of IL-36γ in airway remodeling, and whether IL-38 can alleviate airway remodeling in chronic asthma by blocking the effects of IL-36γ. IL-36γ was quantified in mice inhaled with house dust mite (HDM). Extracellular matrix (ECM) deposition in lung tissues and AHR were assessed following IL-36γ administration to mice. Airway inflammation, AHR, and remodeling were evaluated after IL-38 or blocking IL-36 receptor (IL-36R) treatment in asthmatic mice. The effects of lung fibroblasts stimulated with IL-36γ and IL-38 were quantified in vitro. Increased expression of IL-36γ was detected in lung tissues of HDM-induced asthmatic mice. The intratracheal instillation of IL-36γ to mice significantly enhanced the ECM deposition, AHR, and the number of activated lung fibroblasts around the airways. IL-38 or blocking IL-36R treated asthmatic mice showed a significant alleviation in the airway inflammation, AHR, airway remodeling, and number of activated fibroblasts around airways as compared with the HDM group. In vitro, IL-36γ promoted the activation and migration of human lung fibroblasts (HFL-1). The administration of IL-38 can counteract these biological processes induced by IL-36γ in HFL-1cells. The results indicated that IL-38 can mitigate airway remodeling by blocking the profibrotic effects of IL-36γ in chronic asthma. IL-36γ may be a new therapeutic target, and IL-38 is a potential candidate agent for inhibiting airway remodeling in asthma.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Xia Zhou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chu-Qin Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kang-Ni Feng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao-Ling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jie-Mei Cen
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ping Meng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hong-Tao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tian-Tuo Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Institute of Respiratory Disease of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
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
|
17
|
Gai Y, Bai C, Zhang W, Xiao H, Xu J, Hou J, Ge X. Nootkatone attenuates airway inflammation in asthmatic mice through repressing ROS-induced NLRP3 inflammasome activation. Biochem Cell Biol 2023; 101:513-522. [PMID: 37466343 DOI: 10.1139/bcb-2023-0009] [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] [Indexed: 07/20/2023] Open
Abstract
Nootkatone (NKT) exhibits potential pharmacological activities including anti-oxidation and anti-inflammation. Nevertheless, little is known about the roles of NKT in asthmatic airway inflammation. In the study, mice were sensitized and challenged with ovalbumin (OVA) to establish experimental allergic asthma model. After treatment with NKT, lung tissues, peripheral blood, and bronchoalveolar lavage fluid (BALF) were collected to assess inflammatory cytokines, oxidative stress, and pathological alternations. The effects of NKT on regulating reactive oxygen species (ROS)-induced NLR family pyrin domain containing 3 (NLRP3) inflammasome activation was assessed in IL-13-treated BEAS-2B cell model. We found that NKT treatment decreased the production of Th2 inflammatory cytokines (IL-4, IL-5, and IL-13) in BALF and IgE levels in serum, and alleviated inflammatory cell penetration, goblet cell proliferation, collagen accumulation, and mucus hypersecretion in lung tissues. NKT treatment mitigated oxidative stress and NLRP3 inflammasome activation in asthmatic mice. IL-13 treatment induced oxidative stress and NLRP3-mediated pyroptosis in BEAS-2B bronchial epithelial cells, whereas these effects were blocked by NKT. NKT protected against airway remodeling, as indicated by decreased epithelial-mesenchymal transition. Taken together, these results demonstrate that NKT mitigates asthmatic airway inflammation by inhibiting ROS-triggered NLRP3 activation and may be a potential agent for treating asthma.
Collapse
Affiliation(s)
- Yun Gai
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, Datong Road, Shanghai 200137, People's Republic of China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University, Changhai Road, Shanghai 200433, People's Republic of China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Shu Guang Hospital of Shanghai University of TCM, Shanghai 201203, People's Republic of China
| | - Hua Xiao
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, Datong Road, Shanghai 200137, People's Republic of China
| | - Jing Xu
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, Datong Road, Shanghai 200137, People's Republic of China
| | - Jia Hou
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia, People's Republic of China
| | - Xiahui Ge
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, Datong Road, Shanghai 200137, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
He H, Ji X, Cao L, Wang Z, Wang X, Li XM, Miao M. Medicine Targeting Epithelial-Mesenchymal Transition to Treat Airway Remodeling and Pulmonary Fibrosis Progression. Can Respir J 2023; 2023:3291957. [PMID: 38074219 PMCID: PMC10701063 DOI: 10.1155/2023/3291957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 09/18/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023] Open
Abstract
Objective. Dysregulation of epithelial-mesenchymal transition (EMT) in the airway epithelium is associated with airway remodeling and the progression of pulmonary fibrosis. Many treatments have been shown to inhibit airway remodeling and pulmonary fibrosis progression in asthma and chronic obstructive pulmonary disease (COPD) by regulating EMT and have few side effects. This review aimed to describe the development of airway remodeling through the EMT pathway, as well as the potential therapeutic targets in these pathways. Furthermore, this study aimed to review the current research on drugs to treat airway remodeling and their effects on the EMT pathway. Findings. The dysregulation of EMT was associated with airway remodeling in various respiratory diseases. The cytokines released during inflammation may induce EMT and subsequent airway remodeling. Various drugs, including herbal formulations, specific herbal compounds, cytokines, amino acid or protein inhibitors, microRNAs, and vitamins, may suppress airway remodeling by inhibiting EMT-related pathways.
Collapse
Affiliation(s)
- Hongjuan He
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Henan, Zhengzhou 450046, China
| | - Xiaoyan Ji
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Henan, Zhengzhou 450046, China
| | - Lihua Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Henan, Zhengzhou 450046, China
| | - Zhenzhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Henan, Zhengzhou 450046, China
| | - Xiaoyu Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Henan, Zhengzhou 450046, China
| | - Xiu-Min Li
- Department of Otolaryngology, Microbiology and Immunology, New York Medical College, New York, NY 10595, USA
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Henan, Zhengzhou 450046, China
| |
Collapse
|
19
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
20
|
Mottais A, Riberi L, Falco A, Soccal S, Gohy S, De Rose V. Epithelial-Mesenchymal Transition Mechanisms in Chronic Airway Diseases: A Common Process to Target? Int J Mol Sci 2023; 24:12412. [PMID: 37569787 PMCID: PMC10418908 DOI: 10.3390/ijms241512412] [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: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a reversible process, in which epithelial cells lose their epithelial traits and acquire a mesenchymal phenotype. This transformation has been described in different lung diseases, such as lung cancer, interstitial lung diseases, asthma, chronic obstructive pulmonary disease and other muco-obstructive lung diseases, such as cystic fibrosis and non-cystic fibrosis bronchiectasis. The exaggerated chronic inflammation typical of these pulmonary diseases can induce molecular reprogramming with subsequent self-sustaining aberrant and excessive profibrotic tissue repair. Over time this process leads to structural changes with progressive organ dysfunction and lung function impairment. Although having common signalling pathways, specific triggers and regulation mechanisms might be present in each disease. This review aims to describe the various mechanisms associated with fibrotic changes and airway remodelling involved in chronic airway diseases. Having better knowledge of the mechanisms underlying the EMT process may help us to identify specific targets and thus lead to the development of novel therapeutic strategies to prevent or limit the onset of irreversible structural changes.
Collapse
Affiliation(s)
- Angélique Mottais
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
| | - Luca Riberi
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Andrea Falco
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Simone Soccal
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Virginia De Rose
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
21
|
Yuan J, Wang M, Wang C, Zhang L. Epithelial cell dysfunction in chronic rhinosinusitis: the epithelial-mesenchymal transition. Expert Rev Clin Immunol 2023; 19:959-968. [PMID: 37386882 DOI: 10.1080/1744666x.2023.2232113] [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: 05/23/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/01/2023]
Abstract
INTRODUCTION Epithelial-mesenchymal transition (EMT) is a type of epithelial cell dysfunction, which is widely present in the nasal mucosa of patients with chronic rhinosinusitis (CRS), especially CRS with nasal polyps, and contributes to pathogenesis of the disease. EMT is mediated via complex mechanisms associated with multiple signaling pathways. AREAS COVERED We have summarized the underlying mechanisms and signaling pathways promoting EMT in CRS. Strategies or drugs/agents targeting the genes and pathways related to the regulation of EMT are also discussed for their potential use in the treatment of CRS and asthma. A literature search of studies published in English from 2000 to 2023 was conducted using the PubMed database, employing CRS, EMT, signaling, mechanisms, targeting agents/drugs, as individual or combinations of search terms. EXPERT OPINION EMT in nasal epithelium not only leads to epithelial cell dysfunction but also plays an important role in nasal tissue remodeling in CRS. A comprehensive understanding of the mechanisms underlying EMT and the development of drugs/agents targeting these mechanisms may provide new treatment strategies for CRS.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Nasal Diseases, Beijing Laboratory of Allergic Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Ming Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Nasal Diseases, Beijing Laboratory of Allergic Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Chengshuo Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Nasal Diseases, Beijing Laboratory of Allergic Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Nasal Diseases, Beijing Laboratory of Allergic Diseases, Beijing Institute of Otolaryngology, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Chu F, Liang L, Chen F. Effect of budesonide combined with montelukast sodium on pulmonary function parameters and immunoglobulin levels in children with bronchial asthma. Allergol Immunopathol (Madr) 2023; 51:151-157. [PMID: 37422792 DOI: 10.15586/aei.v51i4.897] [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: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND AND AIM Bronchial asthma is a prevalent type of respiratory disease that affects a large proportion of pediatric patients. The purpose of this study is to further investigate the clinical effects of budesonide combined with montelukast sodium in treating bronchial asthma. METHODS Eighty six children with bronchial asthma were equally divided into study and control groups via randomized double-blind controlled trial. The control group was treated with aerosol inhalation of budesonide combined with placebo, while the study group was treated with budesonide combined with montelukast sodium. Pulmonary function parameters, immunoglobulin, and recovery of related symptoms, along with the adverse reaction rate, were observed and compared between both groups. RESULTS Before treatment, there was no marked difference in pulmonary function parameters and immunoglobulin indexes between both groups (P > 0.05). All pulmonary function indicators and immunoglobulin indexes in both groups improved following therapy, with the study group outperforming the control group (P < 0.05). The recovery time of related symptoms in the study group was shorter than that in the control group (P < 0.05). The incidence of adverse reactions in both groups was compared, with notable differences (P < 0.05). CONCLUSION Budesonide combined with montelukast sodium in the treatment of bronchial asthma has the value of clinical application and promotion.
Collapse
Affiliation(s)
- Fangfang Chu
- Department of Pediatric Respiratory, Anhui Provincial Children's Hospital, Hefei City, Anhui Province, 230051, China
| | - Lei Liang
- Department of Pediatric Respiratory, Anhui Provincial Children's Hospital, Hefei City, Anhui Province, 230051, China;
| | - Fuzhe Chen
- Department of Pediatric Respiratory, Anhui Provincial Children's Hospital, Hefei City, Anhui Province, 230051, China
| |
Collapse
|
23
|
Ji T, Li H. T-helper cells and their cytokines in pathogenesis and treatment of asthma. Front Immunol 2023; 14:1149203. [PMID: 37377958 PMCID: PMC10291091 DOI: 10.3389/fimmu.2023.1149203] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Prosperous advances in understanding the cellular and molecular mechanisms of chronic inflammation and airway remodeling in asthma have been made over the past several decades. Asthma is a chronic inflammatory disease of the airways characterized by reversible airway obstruction that is self-resolving or remits with treatment. Around half of asthma patients are "Type-2-high" asthma with overexpression of type 2 inflammatory pathways and elevated type 2 cytokines. When stimulated by allergens, airway epithelial cells secrete IL-25, IL-33, and TSLP to derive a Th2 immune response. First ILC2 followed by Th2 cells produces a series of cytokines such as IL-4, IL-5, and IL-13. TFH cells control IgE synthesis by secreting IL-4 to allergen-specific B cells. IL-5 promotes eosinophil inflammation, while IL-13 and IL-4 are involved in goblet cell metaplasia and bronchial hyperresponsiveness. Currently, "Type-2 low" asthma is defined as asthma with low levels of T2 biomarkers due to the lack of reliable biomarkers, which is associated with other Th cells. Th1 and Th17 are capable of producing cytokines that recruit neutrophils, such as IFN-γ and IL-17, to participate in the development of "Type-2-low" asthma. Precision medicine targeting Th cells and related cytokines is essential in the management of asthma aiming at the more appropriate patient selection and better treatment response. In this review, we sort out the pathogenesis of Th cells in asthma and summarize the therapeutic approaches involved as well as potential research directions.
Collapse
Affiliation(s)
| | - Hequan Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Huang AS, Tong BCK, Hung HCH, Wu AJ, Ho OKY, Kong AHY, Leung MMK, Bai J, Fu X, Yu Z, Li M, Leung TF, Mak JCW, Leung GPH, Cheung KH. Targeting calcium signaling by inositol trisphosphate receptors: A novel mechanism for the anti-asthmatic effects of Houttuynia cordata. Biomed Pharmacother 2023; 164:114935. [PMID: 37245337 DOI: 10.1016/j.biopha.2023.114935] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023] Open
Abstract
Asthma is a chronic inflammatory disease characterized by airway hypersensitivity and remodeling. The current treatments provide only short-term benefits and may have undesirable side effects; thus, alternative or supplementary therapy is needed. Because intracellular calcium (Ca2+) signaling plays an essential role in regulating the contractility and remodeling of airway smooth muscle cells, the targeting of Ca2+ signaling is a potential therapeutic strategy for asthma. Houttuynia cordata is a traditional Chinese herb that is used to treat asthma due to its anti-allergic and anti-inflammatory properties. We hypothesized that H. cordata might modulate intracellular Ca2+ signaling and could help relieve asthmatic airway remodeling. We found that the mRNA and protein levels of inositol trisphosphate receptors (IP3Rs) were elevated in interleukin-stimulated primary human bronchial smooth muscle cells and a house dust mite-sensitized model of asthma. The upregulation of IP3R expression enhanced intracellular Ca2+ release upon stimulation and contributed to airway remodeling in asthma. Intriguingly, pretreatment with H. cordata essential oil rectified the disruption of Ca2+ signaling, mitigated asthma development, and prevented airway narrowing. Furthermore, our analysis suggested that houttuynin/2-undecanone could be the bioactive component in H. cordata essential oil because we found similar IP3R suppression in response to the commercially available derivative sodium houttuyfonate. An in silico analysis showed that houttuynin, which downregulates IP3R expression, binds to the IP3 binding domain of IP3R and may mediate a direct inhibitory effect. In summary, our findings suggest that H. cordata is a potential alternative treatment choice that may reduce asthma severity by targeting the dysregulation of Ca2+ signaling.
Collapse
Affiliation(s)
- Alexis Shiying Huang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Benjamin Chun-Kit Tong
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Harry Chun-Hin Hung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Aston Jiaxi Wu
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Olivia Ka-Yi Ho
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Anna Hau-Yee Kong
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Maggie Ming-Ki Leung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Jingxuan Bai
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Xiuqiong Fu
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Zhiling Yu
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Min Li
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China
| | - Ting Fan Leung
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Judith Choi-Wo Mak
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - King-Ho Cheung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region of China.
| |
Collapse
|
25
|
Wang L, Liu X. Long noncoding RNA antisense noncoding RNA in the INK4 locus inhibition alleviates airway remodeling in asthma through the regulation of the microRNA-7-5p/early growth response factor 3 axis. Immun Inflamm Dis 2023; 11:e823. [PMID: 37102654 PMCID: PMC10091379 DOI: 10.1002/iid3.823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/07/2023] [Accepted: 03/10/2023] [Indexed: 04/28/2023] Open
Abstract
Asthma, a chronic inflammatory disease of the airways, clinically manifests as airway remodeling. The purpose of this study was to probe the potential role of long noncoding RNA (lncRNA) antisense noncoding RNA in the INK4 locus (lncRNA ANRIL) in the proliferation and migration of airway smooth muscle cell (ASMC) and to explore its potential mechanisms in asthma. Serum samples were obtained from 30 healthy volunteers and 30 patients with asthma. Additionally, platelet-derived growth factor-BB (PDGF-BB) was used to induce airway remodeling in ASMCs. The level of lncRNA ANRIL and microRNA (miR)-7-5p in serum samples were measured by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). TargetScan predicted the binding site of miR-7-5p to early growth response factor 3 (EGR3) and validated the results using a dual-luciferase reporter assay. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) and Transwell assays were used to detect cellular proliferation and migration, respectively. Subsequently, changes in proliferation- and migration-related genes were verified using western blot analysis and qRT-PCR. These results indicate that lncRNA ANRIL was upregulated in the serum and PDGF-BB-induced ASMCs of patients with asthma, whereas miR-7-5p expression was reduced. EGR3 was a direct target of miR-7-5p. LncRNA ANRIL silencing inhibited the proliferation or migration of ASMCs induced by PDGF-BB through miR-7-5p upregulation. Mechanistic studies indicated that miR-7-5p inhibits the proliferation or migration of PDGF-BB-induced ASMCs by decreasing EGR3 expression. EGR3 upregulation reverses the role of miR-7-5p in airway remodeling. Thus, downregulation of lncRNA ANRIL inhibits airway remodeling through inhibiting the proliferation and migration of PDGF-BB-induced ASMCs by regulating miR-7-5p/EGR3 signaling.
Collapse
Affiliation(s)
- Liyan Wang
- Department of PediatricsWuhan Third HospitalWuhanChina
| | - Xueru Liu
- Department of Respiratory Medicine, Wuhan Children's Hospital, Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanChina
| |
Collapse
|
26
|
Zhou Y, Duan Q, Yang D. In vitro human cell-based models to study airway remodeling in asthma. Biomed Pharmacother 2023; 159:114218. [PMID: 36638596 DOI: 10.1016/j.biopha.2023.114218] [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: 07/20/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Airway remodeling, as a predominant characteristic of asthma, refers to the structural changes that occurred both in the large and small airways. These pathological changes not only contribute to airway hyperresponsiveness and airway obstruction, but also predict poor outcomes of patients. In vitro models are the alternatives to animal models that facilitate airway remodeling research. Current approaches to mimic airway remodeling in vitro include mono cultures of cell lines and primary cells that are derived from the respiratory tract, and co-culture systems that consist of different cell subpopulations. Moreover, recent advances in microfluid chips and organoids show promise in simulating the complex architecture and functionality of native organs. According, they enable highly physiological-relevant investigations of human diseases in vitro. Here we aim to detail the current human cell-based models regarding their key pros and cons, and to discuss how they may be used to facilitate our understanding of airway remodeling in asthma.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China
| | - Qirui Duan
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China
| | - Dong Yang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China.
| |
Collapse
|
27
|
Agarwood Oil Nanoemulsion Attenuates Cigarette Smoke-Induced Inflammation and Oxidative Stress Markers in BCi-NS1.1 Airway Epithelial Cells. Nutrients 2023; 15:nu15041019. [PMID: 36839377 PMCID: PMC9959783 DOI: 10.3390/nu15041019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an irreversible inflammatory respiratory disease characterized by frequent exacerbations and symptoms such as cough and wheezing that lead to irreversible airway damage and hyperresponsiveness. The primary risk factor for COPD is chronic cigarette smoke exposure, which promotes oxidative stress and a general pro-inflammatory condition by stimulating pro-oxidant and pro-inflammatory pathways and, simultaneously, inactivating anti-inflammatory and antioxidant detoxification pathways. These events cause progressive damage resulting in impaired cell function and disease progression. Treatments available for COPD are generally aimed at reducing the symptoms of exacerbation. Failure to regulate oxidative stress and inflammation results in lung damage. In the quest for innovative treatment strategies, phytochemicals, and complex plant extracts such as agarwood essential oil are promising sources of molecules with antioxidant and anti-inflammatory activity. However, their clinical use is limited by issues such as low solubility and poor pharmacokinetic properties. These can be overcome by encapsulating the therapeutic molecules using advanced drug delivery systems such as polymeric nanosystems and nanoemulsions. In this study, agarwood oil nanoemulsion (agarwood-NE) was formulated and tested for its antioxidant and anti-inflammatory potential in cigarette smoke extract (CSE)-treated BCi-NS1.1 airway basal epithelial cells. The findings suggest successful counteractivity of agarwood-NE against CSE-mediated pro-inflammatory effects by reducing the expression of the pro-inflammatory cytokines IL-1α, IL-1β, IL-8, and GDF-15. In addition, agarwood-NE induced the expression of the anti-inflammatory mediators IL-10, IL-18BP, TFF3, GH, VDBP, relaxin-2, IFN-γ, and PDGF. Furthermore, agarwood-NE also induced the expression of antioxidant genes such as GCLC and GSTP1, simultaneously activating the PI3K pro-survival signalling pathway. This study provides proof of the dual anti-inflammatory and antioxidant activity of agarwood-NE, highlighting its enormous potential for COPD treatment.
Collapse
|
28
|
Chen G, Li J, Liu H, Zhou H, Liu M, Liang D, Meng Z, Gan H, Wu Z, Zhu X, Han P, Liu T, Gu R, Liu S, Dou G. Cepharanthine Ameliorates Pulmonary Fibrosis by Inhibiting the NF-κB/NLRP3 Pathway, Fibroblast-to-Myofibroblast Transition and Inflammation. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020753. [PMID: 36677811 PMCID: PMC9864377 DOI: 10.3390/molecules28020753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/27/2022] [Accepted: 01/06/2023] [Indexed: 01/15/2023]
Abstract
Pulmonary fibrosis (PF) is one of the sequelae of Corona Virus Disease 2019 (COVID-19), and currently, lung transplantation is the only viable treatment option. Hence, other effective treatments are urgently required. We investigated the therapeutic effects of an approved botanical drug, cepharanthine (CEP), in a cell culture model of transforming growth factor-β1 (TGF-β1) and bleomycin (BLM)-induced pulmonary fibrosis rat models both in vitro and in vivo. In this study, CEP and pirfenidone (PFD) suppressed BLM-induced lung tissue inflammation, proliferation of blue collagen fibers, and damage to lung structures in vivo. Furthermore, we also found increased collagen deposition marked by α-smooth muscle actin (α-SMA) and Collagen Type I Alpha 1 (COL1A1), which was significantly alleviated by the addition of PFD and CEP. Moreover, we elucidated the underlying mechanism of CEP against PF in vitro. Various assays confirmed that CEP reduced the viability and migration and promoted apoptosis of myofibroblasts. The expression levels of myofibroblast markers, including COL1A1, vimentin, α-SMA, and Matrix Metallopeptidase 2 (MMP2), were also suppressed by CEP. Simultaneously, CEP significantly suppressed the elevated Phospho-NF-κB p65 (p-p65)/NF-κB p65 (p65) ratio, NOD-like receptor thermal protein domain associated protein 3 (NLRP3) levels, and elevated inhibitor of NF-κB Alpha (IκBα) degradation and reversed the progression of PF. Hence, our study demonstrated that CEP prevented myofibroblast activation and treated BLM-induced pulmonary fibrosis in a dose-dependent manner by regulating nuclear factor kappa-B (NF-κB)/ NLRP3 signaling, thereby suggesting that CEP has potential clinical application in pulmonary fibrosis in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Ruolan Gu
- Correspondence: (R.G.); (S.L.); (G.D.)
| | | | | |
Collapse
|
29
|
Alnuqaydan AM, Almutary AG, Azam M, Manandhar B, De Rubis G, Madheswaran T, Paudel KR, Hansbro PM, Chellappan DK, Dua K. Phytantriol-Based Berberine-Loaded Liquid Crystalline Nanoparticles Attenuate Inflammation and Oxidative Stress in Lipopolysaccharide-Induced RAW264.7 Macrophages. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4312. [PMID: 36500935 PMCID: PMC9737637 DOI: 10.3390/nano12234312] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 06/17/2023]
Abstract
Inflammation and oxidative stress are interrelated processes that represent the underlying causes of several chronic inflammatory diseases that include asthma, cystic fibrosis, chronic obstructive pulmonary disease (COPD), allergies, diabetes, and cardiovascular diseases. Macrophages are key initiators of inflammatory processes in the body. When triggered by a stimulus such as bacterial lipopolysaccharides (LPS), these cells secrete inflammatory cytokines namely TNF-α that orchestrate the cellular inflammatory process. Simultaneously, pro-inflammatory stimuli induce the upregulation of inducible nitric oxide synthase (iNOS) which catalyzes the generation of high levels of nitric oxide (NO). This, together with high concentrations of reactive oxygen species (ROS) produced by macrophages, mediate oxidative stress which, in turn, exacerbates inflammation in a feedback loop, resulting in the pathogenesis of several chronic inflammatory diseases. Berberine is a phytochemical embedded with potent in vitro anti-inflammatory and antioxidant properties, whose therapeutic application is hindered by poor solubility and bioavailability. For this reason, large doses of berberine need to be administered to achieve the desired pharmacological effect, which may result in toxicity. Encapsulation of such a drug in liquid crystalline nanoparticles (LCNs) represents a viable strategy to overcome these limitations. We encapsulated berberine in phytantriol-based LCNs (BP-LCNs) and tested the antioxidant and anti-inflammatory activities of BP-LCNs in vitro on LPS-induced mouse RAW264.7 macrophages. BP-LCNs showed potent anti-inflammatory and antioxidant activities, with significant reduction in the gene expressions of TNF-α and iNOS, followed by concomitant reduction of ROS and NO production at a concentration of 2.5 µM, which is lower than the concentration of free berberine concentration required to achieve similar effects as reported elsewhere. Furthermore, we provide evidence for the suitability for BP-LCNs both as an antioxidant and as an anti-inflammatory agent with potential application in the therapy of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Abdullah M. Alnuqaydan
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| | - Abdulmajeed G. Almutary
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| | - Mohd Azam
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| | - Bikash Manandhar
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Philip M. Hansbro
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|