1
|
Li N, Qiu G, Xu X, Shen Y, Chen Y. TRIM11 Prevents Ferroptosis in model of asthma by UBE2N-TAX1BP1 signaling. BMC Pulm Med 2024; 24:542. [PMID: 39472837 PMCID: PMC11523820 DOI: 10.1186/s12890-024-03351-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Asthma is a complex chronic respiratory inflammatory disease affected by both genetic and environmental factors. Therefore, our study explored the influence of TRIM11 on asthma and its underlying mechanisms. Our research involved patients diagnosed with asthma and healthy volunteers recruited from our hospital. We observed a reduction in serum TRIM11 expression in asthma patients, which positively correlated with the levels of anti-IgE or IgE. Additionally, both TRIM11 mRNA and protein expression in lung tissue were diminished. The introduction of the TRIM11 gene resulted in a reduction in inflammation in an in vitro asthma model and prevented the development of asthma in a mouse model. Moreover, the TRIM11 gene exhibited a suppressive effect on Ferroptosis and mitigated ROS-induced mitochondrial damage in the asthma model. TRIM11 was found to stimulate UBE2N-TAX1BP1 signaling in the asthma model, with UBE2N being identified as the specific target for TRIM11's effects on Ferroptosis. Furthermore, TRIM11 protein interacted with UBE2N protein and facilitated the dissociation of UBE2N-UBE2N in the asthma model. In conclusion, TRIM11 plays a vital role in preventing Ferroptosis in the asthma model through UBE2N-TAX1BP1 signaling. This indicates that targeting the TRIM11 mechanism could serve as a promising strategy for anti-Ferroptosis immunotherapy in asthma treatment.
Collapse
Affiliation(s)
- Na Li
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China.
| | - Guoqing Qiu
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| | - Xiangqin Xu
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| | - Yan Shen
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| | - Yuming Chen
- Department of Respiratory and Critical Care Medicine, Longgang Central Hospital, 6082 Longgang Avenue, Shenzhen, 518116, China
| |
Collapse
|
2
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [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: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
3
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
Xu C, Huang H, Zou H, Zhao Y, Liu L, Chai R, Zhang J. The miR-9-5p/KLF5/IL-1β Axis Regulates Airway Smooth Muscle Cell Proliferation and Apoptosis to Aggravate Airway Remodeling and Inflammation in Asthma. Biochem Genet 2024; 62:3996-4010. [PMID: 38267617 DOI: 10.1007/s10528-023-10640-1] [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: 10/08/2023] [Accepted: 12/12/2023] [Indexed: 01/26/2024]
Abstract
The aim of this study was to investigate the underlying mechanism of miR-9-5p in airway smooth muscle cells (ASMCs) of asthmatic mice. An asthmatic mouse model was established through the intraperitoneal injection of ovalbumin. Histopathological changes in lung tissues of asthmatic mice were observed using HE staining. ASMCs was identified using immunofluorescence staining and cell morphology. The mRNA expressions of miR-9-5p, KLF5, and IL-1β were measured using RT-qPCR. Additionally, CCK8 assay and flow cytometry were applied for ASMC proliferation and apoptosis, respectively. The protein levels of OPN, KLF5, and IL-1β were assessed using western blotting. The results showed that miR-9-5p was abnormally downregulated in lung tissues and ASMCs of asthmatic mice. Dual-Luciferase Reporter Assay and Chromatin immunoprecipitation confirmed that miR-9-5p targeted KLF5 that bounds to IL-1β promoter. Besides, miR-9-5p negatively regulated IL-1β mRNA and protein level via KLF5. Moreover, miR-9-5p was found to positively regulate ASMC apoptosis, negatively regulate ASMC proliferation and OPN protein expression, albeit with partial reversal by KLF5. Mechanistically, the regulation of ASMC proliferation and apoptosis by miR-9-5p is achieved by targeting KLF5/IL-1β axis.
Collapse
Affiliation(s)
- Chong Xu
- Department of Respiration, General Hospital of Northern Theater Command, No 83 Wenhua Road, Shenyang, 110016, China
| | - Hehua Huang
- Department of Respiration, General Hospital of Northern Theater Command, No 83 Wenhua Road, Shenyang, 110016, China
| | - Hongmei Zou
- Department of Respiration, General Hospital of Northern Theater Command, No 83 Wenhua Road, Shenyang, 110016, China
| | - Yumeng Zhao
- Department of Respiration, General Hospital of Northern Theater Command, No 83 Wenhua Road, Shenyang, 110016, China
| | - Lu Liu
- Department of Respiration, Xinmi Traditional Chinese Medicine Hospital, Xinmi, 452370, China
| | - Ruonan Chai
- Department of Respiration, General Hospital of Northern Theater Command, No 83 Wenhua Road, Shenyang, 110016, China.
| | - Junli Zhang
- Department of Respiration, General Hospital of Northern Theater Command, No 83 Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
5
|
Djeddi S, Fernandez-Salinas D, Huang GX, Aguiar VRC, Mohanty C, Kendziorski C, Gazal S, Boyce JA, Ober C, Gern JE, Barrett NA, Gutierrez-Arcelus M. Rhinovirus infection of airway epithelial cells uncovers the non-ciliated subset as a likely driver of genetic risk to childhood-onset asthma. CELL GENOMICS 2024; 4:100636. [PMID: 39197446 PMCID: PMC11480861 DOI: 10.1016/j.xgen.2024.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/11/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024]
Abstract
Asthma is a complex disease caused by genetic and environmental factors. Studies show that wheezing during rhinovirus infection correlates with childhood asthma development. Over 150 non-coding risk variants for asthma have been identified, many affecting gene regulation in T cells, but the effects of most risk variants remain unknown. We hypothesized that airway epithelial cells could also mediate genetic susceptibility to asthma given they are the first line of defense against respiratory viruses and allergens. We integrated genetic data with transcriptomics of airway epithelial cells subject to different stimuli. We demonstrate that rhinovirus infection significantly upregulates childhood-onset asthma-associated genes, particularly in non-ciliated cells. This enrichment is also observed with influenza infection but not with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or cytokine activation. Overall, our results suggest that rhinovirus infection is an environmental factor that interacts with genetic risk factors through non-ciliated airway epithelial cells to drive childhood-onset asthma.
Collapse
Affiliation(s)
- Sarah Djeddi
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniela Fernandez-Salinas
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Licenciatura en Ciencias Genómicas, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62210, México
| | - George X Huang
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vitor R C Aguiar
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chitrasen Mohanty
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Steven Gazal
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90007, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Joshua A Boyce
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - James E Gern
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53726, USA; Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA
| | - Nora A Barrett
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
6
|
Li M, Li Z. Research progress on the relationship between phenotype and signaling pathways of pulmonary macrophages and asthma. J Asthma 2024:1-8. [PMID: 39072611 DOI: 10.1080/02770903.2024.2386634] [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: 04/18/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE The purpose of this study is to systematically investigate the different phenotypes and functional analyses of macrophages in lung tissue. DATA SOURCES A search was performed using three databases (Web of Science, Science Direct, and MEDLINE) for all relevant studies published from January 1, 2019, to December 31, 2023. STUDY SELECTIONS This systematic review complied with the PRlSMA document's requirements, including studies related to the signaling pathway relationship between pulmonary macrophages and asthma phenotype. The search includedstudies published in English or French lanquage, and was based on title, abstract, and complete textDocuments not meeting inclusion requirements were excluded. RESULTS We have identified studies published within the past five years that meet the criteria for inclusion in this review. We found that asthma is a heterogeneous chronic inflammatory lung disease, and lung tissue macrophages are important immune cells in the respiratory tract. Pulmonary macrophages are also heterogeneous, as they have different subgroups with varying effector functions depending on the environment. They have different phenotypes and biological functions in different disease environments. The phenotypic changes of pulmonary macrophages occur during asthma, and the study of the different phenotypes and functions of macrophages in lung tissue is of great significance for treatment. CONCLUSIONS This review summarizes current literature and provides a detailed introduction to the role of macrophages as key inflammatory mediators in the pathogenesis of asthma, as well as existing knowledge gaps. In addition, we propose that regulatory macrophages may prevent the development of asthma by producing IL-10, and regulating the polarization of pulmonary macrophages may be a new direction for asthma treatment.
Collapse
Affiliation(s)
- Minghui Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin City, China
| | - Zhuying Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin City, China
| |
Collapse
|
7
|
Liu C, He L, Zheng X. UNVEILING THE CAUSAL ASSOCIATION BETWEEN NONINFECTIOUS RESPIRATORY DISORDERS AND SEPSIS THROUGH MENDELIAN RANDOMIZATION ANALYSIS. Shock 2024; 62:179-185. [PMID: 38526151 DOI: 10.1097/shk.0000000000002358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
ABSTRACT Background: The association between sepsis and noninfectious respiratory diseases is well-documented, yet the specific causal link between the two remains unclear. In order to explore this relationship further, we employed a Mendelian randomization (MR) analysis utilizing data from the UK Biobank and FinnGen Biobank. Methods: We analyzed the summary statistics of a genome-wide association study summary statistics for chronic obstructive pulmonary disease (COPD), asthma, pulmonary embolism (PE), idiopathic pulmonary fibrosis (IPF), obstructive sleep apnea (OSA), lung cancer, sepsis, and sepsis-related mortality. We employed the inverse-variance weighted (IVW) method and four additional MR methods. Heterogeneity and horizontal pleiotropy were assessed using the Cochrane's Q test, MR-Egger intercept, and MR-PRESSO test. A sensitivity analysis was also performed. Results: MR analysis showed associations between COPD and lung cancer with increased sepsis risk (odds ratio (OR)IVW 1.138, P = 0.006; (OR)IVW 1.123, P = 0.031; respectively) and sepsis mortality ((OR)IVW 1.350, P = 0.022; (OR)IVW 1.312, P = 0.022; respectively). Asthma exhibited a potential protective effect against sepsis mortality ((OR)IVW = 0.300, P = 0.039), while PE demonstrated a risk effect ((OR)IVW = 1.148, P = 0.032). No causal association was observed between asthma, PE, and sepsis ( P > 0.05). IPF and OSA were not significantly associated with sepsis or sepsis-related mortality ( P > 0.05). Heterogeneity and horizontal pleiotropy were not evident for asthma or lung cancer ( P > 0.05). However, horizontal pleiotropy was suggested for COPD by the MR-Egger regression ( P < 0.05), but not by the MR-PRESSO test ( P > 0.05). IPF and OSA were not significantly associated with sepsis or sepsis-related mortality ( P > 0.05). Conclusion: Our MR analysis offers new insights into potential links between noninfectious respiratory diseases and the risk of sepsis. However, additional investigation into the underlying mechanisms and clinical studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Critical Care Medicine, Dazhou Central Hospital, Dazhou, People's Republic of China
| | | | | |
Collapse
|
8
|
Gu W, Huang C, Chen G, Kong W, Zhao L, Jie H, Zhen G. The role of extracellular traps released by neutrophils, eosinophils, and macrophages in asthma. Respir Res 2024; 25:290. [PMID: 39080638 PMCID: PMC11290210 DOI: 10.1186/s12931-024-02923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Extracellular traps (ETs) are a specialized form of innate immune defense in which leukocytes release ETs composed of chromatin and active proteins to eliminate pathogenic microorganisms. In addition to the anti-infection effect of ETs, researchers have also discovered their involvement in the pathogenesis of inflammatory disease, tumors, autoimmune disease, and allergic disease. Asthma is a chronic airway inflammatory disease involving multiple immune cells. The increased level of ETs in asthma patients suggests that ETs play an important role in the pathogenesis of asthma. Here we review the research work on the formation mechanism, roles, and therapeutic strategies of ETs released by neutrophils, eosinophils, and macrophages in asthma.
Collapse
Affiliation(s)
- Wei Gu
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Chunli Huang
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Gongqi Chen
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Weiqiang Kong
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Lu Zhao
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Huiru Jie
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China
| | - Guohua Zhen
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, Wuhan, China.
| |
Collapse
|
9
|
Ray JL, Walum J, Jelic D, Barnes R, Bentley ID, Britt RD, Englert JA, Ballinger MN. scRNA-seq identifies unique macrophage population in murine model of ozone induced asthma exacerbation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604740. [PMID: 39211080 PMCID: PMC11361036 DOI: 10.1101/2024.07.23.604740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ozone (O 3 ) inhalation triggers asthmatic airway hyperresponsiveness (AHR), but the mechanisms by which this occurs are unknown. Previously, we developed a murine model of dust mite, ragweed, and aspergillus (DRA)-induced allergic lung inflammation followed by O 3 exposure for mechanistic investigation. The present study used single cell RNA-sequencing for unbiased profiling of immune cells within the lungs of mice exposed to DRA, O 3 , or DRA+O 3 , to identify the components of the immune cell niche that contribute to AHR. Alveolar macrophages (AMs) had the greatest number of differentially expressed genes following DRA+O 3 , most of which were unique to the 2-hit exposure. Following DRA+O 3 , AMs activated transcriptional pathways related to cholesterol biosynthesis, degradation of the extracellular matrix, endosomal TLR processing, and various cytokine signals. We also identified AM and monocyte subset populations that were unique to the DRA+O 3 group. These unique AMs activated gene pathways related to inflammation, sphingolipid metabolism, and bronchial constriction. The unique monocyte population had a gene signature that suggested phospholipase activation and increased degradation of the extracellular matrix. Flow cytometry analysis of BAL immune cells showed recruited monocyte-derived AMs after DRA and DRA+O 3 , but not after O 3 exposure alone. O 3 alone increased BAL neutrophils but this response was attenuated in DRA+O 3 mice. DRA-induced changes in the airspace immune cell profile were reflected in elevated BAL cytokine/chemokine levels following DRA+O 3 compared to O 3 alone. The present work highlights the role of monocytes and AMs in the response to O 3 and suggests that the presence of distinct subpopulations following allergic inflammation may contribute to O 3 -induced AHR.
Collapse
|
10
|
Hernandez-Lara MA, Richard J, Deshpande DA. Diacylglycerol kinase is a keystone regulator of signaling relevant to the pathophysiology of asthma. Am J Physiol Lung Cell Mol Physiol 2024; 327:L3-L18. [PMID: 38742284 PMCID: PMC11380957 DOI: 10.1152/ajplung.00091.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
Signal transduction by G protein-coupled receptors (GPCRs), receptor tyrosine kinases (RTKs) and immunoreceptors converge at the activation of phospholipase C (PLC) for the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). This is a point for second-messenger bifurcation where DAG via protein kinase C (PKC) and IP3 via calcium activate distinct protein targets and regulate cellular functions. IP3 signaling is regulated by multiple calcium influx and efflux proteins involved in calcium homeostasis. A family of lipid kinases belonging to DAG kinases (DGKs) converts DAG to phosphatidic acid (PA), negatively regulating DAG signaling and pathophysiological functions. PA, through a series of biochemical reactions, is recycled to produce new molecules of PIP2. Therefore, DGKs act as a central switch in terminating DAG signaling and resynthesis of membrane phospholipids precursor. Interestingly, calcium and PKC regulate the activation of α and ζ isoforms of DGK that are predominantly expressed in airway and immune cells. Thus, DGK forms a feedback and feedforward control point and plays a crucial role in fine-tuning phospholipid stoichiometry, signaling, and functions. In this review, we discuss the previously underappreciated complex and intriguing DAG/DGK-driven mechanisms in regulating cellular functions associated with asthma, such as contraction and proliferation of airway smooth muscle (ASM) cells and inflammatory activation of immune cells. We highlight the benefits of manipulating DGK activity in mitigating salient features of asthma pathophysiology and shed light on DGK as a molecule of interest for heterogeneous diseases such as asthma.
Collapse
Affiliation(s)
- Miguel A Hernandez-Lara
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Joshua Richard
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Deepak A Deshpande
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
11
|
Fang Q, Wu W, Xiao Z, Zeng D, Liang R, Wang J, Yuan J, Su W, Xu X, Zheng Y, Lai T, Sun J, Fu Q, Zheng SG. Gingival-derived mesenchymal stem cells alleviate allergic asthma inflammation via HGF in animal models. iScience 2024; 27:109818. [PMID: 38766356 PMCID: PMC11099335 DOI: 10.1016/j.isci.2024.109818] [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: 02/21/2024] [Revised: 03/25/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
Allergic asthma is a chronic non-communicable disease characterized by lung tissue inflammation. Current treatments can alleviate the clinical symptoms to some extent, but there is still no cure. Recently, the transplantation of mesenchymal stem cells (MSCs) has emerged as a potential approach for treating allergic asthma. Gingival-derived mesenchymal stem cells (GMSCs), a type of MSC recently studied, have shown significant therapeutic effects in various experimental models of autoimmune diseases. However, their application in allergic diseases has yet to be fully elucidated. In this study, using an OVA-induced allergic asthma model, we demonstrated that GMSCs decrease CD11b+CD11c+ proinflammatory dendritic cells (DCs), reduce Th2 cells differentiation, and thus effectively diminish eosinophils infiltration. We also identified that the core functional factor, hepatocyte growth factor (HGF) secreted by GMSCs, mediated its effects in relieving airway inflammation. Taken together, our findings indicate GMSCs as a potential therapy for allergic asthma and other related diseases.
Collapse
Affiliation(s)
- Qiannan Fang
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Jiaotong University School of Medicine Affiliated Songjiang Hospital, Shanghai, China
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Wenbin Wu
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zexiu Xiao
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Donglan Zeng
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rongzhen Liang
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Jiaotong University School of Medicine Affiliated Songjiang Hospital, Shanghai, China
| | - Julie Wang
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Jiaotong University School of Medicine Affiliated Songjiang Hospital, Shanghai, China
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Jia Yuan
- Division of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Zheng
- Department of Dermatology Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tianwen Lai
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jianbo Sun
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Qingling Fu
- Otorhinolaryngology Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Jiaotong University School of Medicine Affiliated Songjiang Hospital, Shanghai, China
| |
Collapse
|
12
|
Yang S, Guo R, Meng X, Zhang Y. AIM2 participates in house dust mite (HDM)-induced epithelial dysfunctions and ovalbumin (OVA)-induced allergic asthma in infant mice. J Asthma 2024; 61:479-490. [PMID: 38078661 DOI: 10.1080/02770903.2023.2289157] [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: 07/28/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 01/13/2024]
Abstract
Objective: Allergen sensitization and high rates of concomitant allergic diseases are characteristic of severe pediatric asthma. The present study was aimed to explore the mechanism of allergic asthma via bioinformatics and experiment investigation. Methods: The GSE27011 dataset contained the expression profiles of normal and pediatric asthma white blood cells was downloaded for analyzing the different expression genes and function enrichment. The allergic asthma model in infant mice was established by ovalbumin (OVA) stimulation. The cellular model was established by house dust mite (HDM)-stimulation in human bronchial epithelial cells. The absent in melanoma 2 (AIM2) knockdown was achieved by intranasal lentivirus injection or cell infection. The bronchoalveolar lavage fluid (BALF) was collected for cell counting and ELISA assessment of cytokines. Lung tissues were collected for HE staining and immunohistochemical (IHC) staining. Real-time PCR and immunoblotting were used for the determination of key gene expressions in mouse and cell models. Results: upregulation of AIM2 gene expression was observed in pediatric asthma patients based on GSE27011 and OVA-induced infant mouse allergic asthma model. AIM2 knockdown ameliorated OVA caused elevation in airway hyper-responsiveness (AHR), elevation in cell quantities (eosinophils, neutrophils, lymphocytes), and levels of cytokines (IL-4, IL-13, TNF-α, and OVA-specific IgE) in BALF. Moreover, AIM2 knockdown relieved OVA-caused histopathological alterations in mouse lungs, up-regulation of AIM2 levels, and NOD1 and receptor-interacting protein 2 (RIP2) protein levels, as well as p65 phosphorylation. In the cell model, AIM2 knockdown partially ameliorated HDM-induced epithelial dysfunctions by promoting cell viability, down-regulating inflammatory cytokines levels, and decreasing the protein levels of AIM2, NOD1, RIP2, and phosphorylated p65. Conclusion: AIM2 participates in HDM-induced epithelial dysfunctions and OVA-induced allergic asthma progression. AIM2 could be a promising target for pediatric allergic asthma treatment regimens, which warrants further in vivo investigations.
Collapse
Affiliation(s)
- Shengzhi Yang
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Ru Guo
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Xianmei Meng
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Yunhong Zhang
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| |
Collapse
|
13
|
Camp B, Jorde I, Sittel F, Pausder A, Jeron A, Bruder D, Schreiber J, Stegemann-Koniszewski S. Comprehensive analysis of lung macrophages and dendritic cells in two murine models of allergic airway inflammation reveals model- and subset-specific accumulation and phenotypic alterations. Front Immunol 2024; 15:1374670. [PMID: 38529288 PMCID: PMC10961404 DOI: 10.3389/fimmu.2024.1374670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Allergic asthma has been mainly attributed to T helper type 2 (Th2) and proinflammatory responses but many cellular processes remain elusive. There is increasing evidence for distinct roles for macrophage and dendritic cell (DC) subsets in allergic airway inflammation (AAI). At the same time, there are various mouse models for allergic asthma that have been of utmost importance in identifying key inflammatory pathways in AAI but that differ in the allergen and/or route of sensitization. It is unclear whether and how the accumulation and activation of specialized macrophage and DC subsets depend on the experimental model chosen for analyses. Methods In our study, we employed high-parameter spectral flow cytometry to comprehensively assess the accumulation and phenotypic alterations of different macrophage- and DC-subsets in the lung in an OVA- and an HDM-mediated mouse model of AAI. Results We observed subset-specific as well as model-specific characteristics with respect to cell numbers and functional marker expression. Generally, alveolar as opposed to interstitial macrophages showed increased MHCII surface expression in AAI. Between the models, we observed significantly increased numbers of alveolar macrophages, CD103+ DC and CD11b+ DC in HDM-mediated AAI, concurrent with significantly increased airway interleukin-4 but decreased total serum IgE levels. Further, increased expression of CD80 and CD86 on DC was exclusively detected in HDM-mediated AAI. Discussion Our study demonstrates a model-specific involvement of macrophage and DC subsets in AAI. It further highlights spectral flow cytometry as a valuable tool for their comprehensive analysis under inflammatory conditions in the lung.
Collapse
Affiliation(s)
- Belinda Camp
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Ilka Jorde
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Franka Sittel
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Alexander Pausder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andreas Jeron
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
14
|
Liu P, Wang Y, Chen C, Liu H, Ye J, Zhang X, Ma C, Zhao D. Research trends on airway remodeling: A bibliometrics analysis. Heliyon 2024; 10:e24824. [PMID: 38333835 PMCID: PMC10850909 DOI: 10.1016/j.heliyon.2024.e24824] [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: 08/09/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Background Airway remodeling is an essential pathological basis of respiratory diseases such as asthma and COPD, which is significantly related to pulmonary function and clinical symptoms. And pulmonary disease can be improved by regulating airway remodeling. This study aimed to establish a knowledge map of airway remodeling to clarify current research hotspots and future research trends. Methods A comprehensive search was performed to analyze all relevant articles on airway remodeling using the Web of Science Core Collection Database from January 01, 2004 to June 03, 2023.2 reviewers screened the retrieved literature. Besides, the CiteSpace (6.2. R3) and VOSviewer (1.6.19) were utilized to visualize the research focus and trend regarding the effect of airway remodeling. Results A total of 4077 articles about airway remodeling were retrieved. The United States is the country with the most published literature, underscoring the country's role in airway remodeling. In recent years, China has been the country with the fastest growth in the number of published literature, suggesting that China will play a more critical role in airway remodeling in the future. From the perspective of co-operation among countries, European co-operation was closer than Asian co-operation. The co-citation analysis showed that 98,313 citations were recorded in 3594 articles, and 25 clusters could be realized. In recent years, Burst detection shows that oxidative stress and epithelial-mesenchymal transition are hot words. Conclusions Based on the bibliometric analysis of airway remodeling studies in the past 20 years, a multi-level knowledge structure map was drawn, it mainly includes countries, institutions, research fields, authors, journals, keywords and so on. The research directions represented by obstructive airway disease, PDGF-BB treatment of airway smooth muscle, allergen-induced airway remodeling, extracellular matrix, and non-coding RNA are the research hotspots in the field of airway remodeling. While the risk factors for airway remodeling, the application of new noninvasively assessing tools, biomarkers as well as The molecular mechanism represented by EMT and autophagy had been frontiers in recent years.
Collapse
Affiliation(s)
- Pengcheng Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Yu Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Chen Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Hui Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Jing Ye
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Xiaoming Zhang
- School of Basic Medicine, Anhui Medical University, Hefei, 230000, China
| | - Changxiu Ma
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| | - Dahai Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230000, China
| |
Collapse
|
15
|
Tian C, Gao J, Yang L, Yuan X. Non-coding RNA regulation of macrophage function in asthma. Cell Signal 2023; 112:110926. [PMID: 37848099 DOI: 10.1016/j.cellsig.2023.110926] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
As a chronic respiratory disease, asthma is related to airway inflammation and remodeling. Macrophages are regarded as main innate immune cells in the airway that exert various functions like antigen recognition and presentation, phagocytosis, and pathogen clearance, playing a crucial role in the pathogeneses of asthma. Non-coding RNAs (ncRNAs), mainly include microRNA, long non-coding RNA and circular RNA, have been extensively investigated on the regulation of pathological process in asthma. Recent studies have indicated that ncRNA-regulated macrophages affect macrophage polarization, airway inflammation, immune regulation and airway remodeling, which suggests that modulating macrophages by ncRNAs may be a promising strategy for the treatment of asthma. This review summarizes the effect of macrophages in asthma and the regulatory mechanisms of ncRNAs, as well as focuses on the role of ncRNAs-regulated macrophages in asthma, for the development of novel therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Chunyan Tian
- Heilongjiang University of Chinese Medicine, Harbin 150040, China; Department of Respiratory Medicine, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Jiawei Gao
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Liuxin Yang
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xingxing Yuan
- Heilongjiang University of Chinese Medicine, Harbin 150040, China; Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150006, China.
| |
Collapse
|
16
|
Savin IA, Zenkova MA, Sen’kova AV. Bronchial Asthma, Airway Remodeling and Lung Fibrosis as Successive Steps of One Process. Int J Mol Sci 2023; 24:16042. [PMID: 38003234 PMCID: PMC10671561 DOI: 10.3390/ijms242216042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Bronchial asthma is a heterogeneous disease characterized by persistent respiratory system inflammation, airway hyperreactivity, and airflow obstruction. Airway remodeling, defined as changes in airway wall structure such as extensive epithelial damage, airway smooth muscle hypertrophy, collagen deposition, and subepithelial fibrosis, is a key feature of asthma. Lung fibrosis is a common occurrence in the pathogenesis of fatal and long-term asthma, and it is associated with disease severity and resistance to therapy. It can thus be regarded as an irreversible consequence of asthma-induced airway inflammation and remodeling. Asthma heterogeneity presents several diagnostic challenges, particularly in distinguishing between chronic asthma and other pulmonary diseases characterized by disruption of normal lung architecture and functions, such as chronic obstructive pulmonary disease. The search for instruments that can predict the development of irreversible structural changes in the lungs, such as chronic components of airway remodeling and fibrosis, is particularly difficult. To overcome these challenges, significant efforts are being directed toward the discovery and investigation of molecular characteristics and biomarkers capable of distinguishing between different types of asthma as well as between asthma and other pulmonary disorders with similar structural characteristics. The main features of bronchial asthma etiology, pathogenesis, and morphological characteristics as well as asthma-associated airway remodeling and lung fibrosis as successive stages of one process will be discussed in this review. The most common murine models and biomarkers of asthma progression and post-asthmatic fibrosis will also be covered. The molecular mechanisms and key cellular players of the asthmatic process described and systematized in this review are intended to help in the search for new molecular markers and promising therapeutic targets for asthma prediction and therapy.
Collapse
Affiliation(s)
| | | | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrent’ev Ave 8, 630090 Novosibirsk, Russia; (I.A.S.); (M.A.Z.)
| |
Collapse
|