1
|
Luo L, Yang X, Zhao H, Wang L, Li W, Zhang Y. High expression of ITGB3 ameliorates asthma by inhibiting epithelial-mesenchymal transformation through suppressing the activation of NF-kB pathway. Sci Rep 2025; 15:13837. [PMID: 40263524 DOI: 10.1038/s41598-025-98842-y] [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: 12/16/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Integrin β3 (ITGB3) has been identified as an asthma-associated gene; however, its molecular mechanisms remain poorly understood. Epithelial-mesenchymal transition (EMT) is a critical driver of airway remodeling in asthma, which underpins disease progression. This study aimed to elucidate the role of ITGB3 in asthma pathogenesis by investigating its regulation of EMT. Asthma models were established in vivo using C57BL/6 mice and in vitro with A549 cells, both exposed to house dust mite (HDM) extract. The effects of HDM and ITGB3 modulation on cellular viability, apoptosis, and inflammatory cytokines (IL-4, IL-5, IL-13) were assessed in cultured cells and murine lungs. EMT was evaluated via western blot analysis of E-cadherin, N-cadherin, and vimentin expression. The NF-κB pathway was examined by quantifying phosphorylated p65 and IkBa levels. Lung tissue pathology and ITGB3 expression were assessed using hematoxylin and eosin (H&E) staining and immunohistochemistry. Results demonstrated that HDM exposure reduced A549 cell viability, increased cytotoxicity, apoptosis, and pro-inflammatory cytokine production, while promoting EMT. ITGB3 knockdown exacerbated these effects, whereas ITGB3 overexpression mitigated them. Furthermore, HDM activated the NF-κB pathway, an effect reversed by ITGB3 overexpression. In HDM-challenged cells, NF-κB activation via an agonist counteracted the protective effects of ITGB3 overexpression on apoptosis, inflammation, and EMT. Notably, ITGB3 overexpression suppressed inflammation, EMT, and pathological remodeling in asthmatic mice. Collectively, our findings reveal that ITGB3 exerts protective effects in asthma by inhibiting EMT through suppression of the NF-κB signaling pathway, thereby identifying ITGB3 as a potential therapeutic target for asthma management.
Collapse
Affiliation(s)
- Lu Luo
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Xiaoshan Yang
- Department of Rheumatology And Immunology, Binzhou People's Hospital, Binzhou, China
| | - Haitao Zhao
- Department of Hematology, Binzhou People's Hospital, Binzhou, China
| | - Lingling Wang
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Wengang Li
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Yan Zhang
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China.
| |
Collapse
|
2
|
Sahnoon L, Bajbouj K, Mahboub B, Hamoudi R, Hamid Q. Targeting IL-13 and IL-4 in Asthma: Therapeutic Implications on Airway Remodeling in Severe Asthma. Clin Rev Allergy Immunol 2025; 68:44. [PMID: 40257546 PMCID: PMC12011922 DOI: 10.1007/s12016-025-09045-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/22/2025]
Abstract
Asthma is a chronic respiratory disorder affecting individuals across all age groups. It is characterized by airway inflammation and remodeling and leads to progressive airflow restriction. While corticosteroids remain a mainstay therapy, their efficacy is limited in severe asthma due to genetic and epigenetic alterations, as well as elevated pro-inflammatory cytokines interleukin-4 (IL-4), interleukin-13 (IL-13), and interleukin-5 (IL-5), which drive structural airway changes including subepithelial fibrosis, smooth muscle hypertrophy, and goblet cell hyperplasia. This underscores the critical need for biologically targeted therapies. This review systematically examines the roles of IL-4 and IL-13, key drivers of type-2 inflammation, in airway remodeling and their potential as therapeutic targets. IL-4 orchestrates eosinophil recruitment, immunoglobulin class switching, and Th2 differentiation, whereas IL-13 directly modulates structural cells, including fibroblasts and epithelial cells, to promote mucus hypersecretion and extracellular matrix (ECM) deposition. Despite shared signaling pathways, IL-13 emerges as the dominant cytokine in remodeling processes including mucus hypersecretion, fibrosis and smooth muscle hypertrophy. While IL-4 primarily amplifies inflammatory cascades by driving IgE switching, promoting Th2 cell polarization that sustain cytokine release, and inducing chemokines to recruit eosinophils. In steroid-resistant severe asthma, biologics targeting IL-4/IL-13 show promise in reducing exacerbations and eosinophilic inflammation. However, their capacity to reverse established remodeling remains inconsistent, as clinical trials prioritize inflammatory biomarkers over long-term structural outcomes. This synthesis highlights critical gaps in understanding the durability of IL-4/IL-13 inhibition on airway structure and advocates for therapies combining biologics with remodeling-specific strategies. Through the integration of mechanistic insights and clinical evidence, this review emphasizes the need for long-term studies utilizing advanced imaging, histopathological techniques, and patient-reported outcomes to evaluate how IL-4/IL-13-targeted therapies alter airway remodeling and symptom burden, thereby informing more effective treatment approaches for severe, steroid-resistant asthma.
Collapse
Affiliation(s)
- Lina Sahnoon
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Bassam Mahboub
- Rashid Hospital, Dubai Health, 4545, Dubai, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Division of Surgery and Interventional Science, University College London, London, UK.
- Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah, United Arab Emirates.
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Meakins-Christie Laboratories, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
3
|
Liu C, Huang X, Li S, Ji W, Luo T, Liang J, Lv Y. M2 macrophage-derived exosomes reverse TGF-β1-induced epithelial mesenchymal transformation in BEAS-2B cells via the TGF-βRI/Smad2/3 signaling pathway. Eur J Med Res 2025; 30:271. [PMID: 40211426 PMCID: PMC11987241 DOI: 10.1186/s40001-025-02516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/26/2025] [Indexed: 04/13/2025] Open
Abstract
INTRODUCTION Airway remodeling in bronchial asthma can be inhibited by disrupting the epithelial mesenchymal transition (EMT) of activated airway epithelial cells. Exosomes, as key mediators of intercellular communication, have been implicated in the pathophysiology of asthma-related airway inflammation, remodeling, and hyperresponsiveness. This study aimed to investigate the role of M2 macrophage-derived exosomes (M2φ-exos) in modulating TGF-β1-induced EMT in airway epithelial (BEAS-2B) cells and elucidate the underlying molecular mechanism, if any. METHODS THP-1 cells were induced to differentiate into M2 macrophages via phorbol 12-myristate 13-acetate (PMA) and IL-4. Exosomes were subsequently isolated and purified via ultracentrifugation. M2φ-exos expression was characterized by protein marker levels, transmission electron microscopy imaging, and nanoparticle tracking analysis. TGF-β1-induced BEAS-2B cells were exposed to M2φ-exos to determine the latter's effects. RESULTS THP-1 cells were successfully differentiated into M2 macrophages, as confirmed by in vitro flow cytometry. The isolated exosomes presented typical cup-shaped structures and expressed CD81 and TSG101. TGF-β1 induction altered the morphological characteristics of BEAS-2B cells and activated the TGF-βRI/Smad2/3 signaling pathway, leading to increased expression of Snail, Vimentin and Collagen 1 and decreased expression of E-cadherin. After exosome or SB431542 induction, TGF-β1-induced EMT was reversed. GW4869, an exosome release inhibitor, exhibited the ability to block the beneficial effects of exosomes. CONCLUSION M2Φ-exos inhibited EMT in BEAS-2B cells through the TGF-βRI/Smad2/3 signaling pathway. This novel insight into the role of M2Φ-exos in modulating EMT may have important implications for the beneficial effects of asthma, particularly in addressing airway remodeling.
Collapse
Affiliation(s)
- Chao Liu
- Department of Respiratory and Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Xiaolin Huang
- Dental Implant and Restoration Centre, Zhongshan Stomatological Hospital, Zhongshan, Guangdong, China
| | - Siqi Li
- Department of Respiratory and Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Wentao Ji
- Department of Respiratory and Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Tian Luo
- Department of Respiratory and Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Jianping Liang
- Department of Respiratory and Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong, China.
| | - Yanhua Lv
- Department of Respiratory and Critical Care Medicine, Shunde Hospital, Southern Medical University (the First People'S Hospital of Shunde), Foshan, Guangdong, China.
| |
Collapse
|
4
|
Fazlıoğlu N, Sasani H, Fazlıoğlu M, Çiftçi EP, Mutlu LC. Association of tracheal diameter with respiratory function and fibrosis severity in idiopathic pulmonary fibrosis patients. BMC Pulm Med 2025; 25:157. [PMID: 40188355 PMCID: PMC11972493 DOI: 10.1186/s12890-025-03624-x] [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: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND In this research project, we examined the relationship between tracheal size and respiratory function in individuals with Idiopathic Pulmonary Fibrosis (IPF). IPF is a long-term condition that affects the functioning of the lungs. METHODS This retrospective study included 86 patients diagnosed with IPF. Tracheal and bronchial diameters were measured using high-resolution computed tomography (HRCT) and pulmonary function tests (PFTs); Force vital capacity (FVC), diffusion capacity for carbon monoxide (DLCO) and the gender, age, physiology (GAP) index was calculated. Patients were grouped according to demographic characteristics such as age, gender and smoking. RESULTS There was a significant positive correlation between the anteroposterior (AP) and transverse diameters of the trachea in the subcricoid region and the GAP index (r = 0.318, p = 0.003 and r = 0.312, p = 0.004, respectively). Similarly, subcricoid and carina areas were significantly correlated with both GAP index (r = 0.307, p = 0.006 and r = 0.334, p = 0.003, respectively) and FVC/DLCO ratio (r = 0.218, p = 0.049 and r = 0.245, p = 0.027, respectively). The main bronchial areas were also positively correlated with the GAP index, but no significant correlation was found between FVC and DLCO values and airway measurements. Each unit increase in GAP index was associated with a 1.69-fold increase in mortality risk (p = 0.0016, 95% confidence interval: 1.22-2.34). CONCLUSION Tracheal and main bronchial areas can be used as potential biomarkers in the assessment of disease severity and prognosis in IPF patients. In particular, the significant correlation of subcricoid and carina areas with both GAP index and FVC/DLCO ratio suggests that these measurements may be useful in the evaluation of disease progression.
Collapse
Affiliation(s)
- Nevin Fazlıoğlu
- Department of Pulmonology, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, 59030, Turkey.
| | - Hadi Sasani
- Department of Radiology, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, Turkey
| | - Mithat Fazlıoğlu
- Department of Thoracic Surgery, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, Turkey
| | - Ezgi Pınar Çiftçi
- Department of Pulmonology, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, 59030, Turkey
| | - Levent Cem Mutlu
- Department of Pulmonology, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, 59030, Turkey
| |
Collapse
|
5
|
Mo J, Zuo J, Yu L, Zhang H, Weng S, Ye L. New insights into the effects of PFOS exposure on rat lung development: morphological, functional, and single-cell sequencing analysis. Arch Toxicol 2025:10.1007/s00204-025-04014-2. [PMID: 40128328 DOI: 10.1007/s00204-025-04014-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Perfluorooctane sulfonate (PFOS), a widely persistent environmental pollutant, has been demonstrated to disrupt lung development in animal models. However, its cellular and molecular mechanisms remain insufficiently understood. This study examines the effects of prenatal PFOS exposure on lung development and function in offspring rats. Pregnant rats were exposed to PFOS at concentrations relevant to both environmental and occupational exposures, with doses of 0, 0.01, 0.1, and 1 mg/kg/day from gestational day 11-20. We primarily evaluated morphological changes, pulmonary function, bronchoalveolar lavage fluid composition, and alterations in trace element and fatty acid metabolism at postnatal days 0, 4, 14, 21, and 60. Single-cell RNA sequencing was employed to profile cellular and molecular responses in the lungs. Our results show that PFOS exposure leads to dose-dependent reductions in alveolar development, increased pulmonary injury, fibrosis, and impaired lung function. PFOS also changes lung cell composition, particularly affecting structural and immune cells, and shifts immune responses from innate to adaptive immunity. Differential gene expression analyses revealed the upregulation of Fam111a and downregulation of Stk35, implicating these genes in PFOS-induced lung injury and repair processes. In addition, pathway analyses demonstrated suppression of immune-related signaling pathways and disruption of cell adhesion and phagocytosis, which may exacerbate lung tissue injury. These findings provide novel insights into the developmental toxicity of PFOS and highlight its potential long-term health risks.
Collapse
Affiliation(s)
- Jiali Mo
- Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, Beijing, 102627, China
| | - Jingye Zuo
- Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, Beijing, 102627, China
| | - Lin Yu
- Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, Beijing, 102627, China
| | - Huishan Zhang
- Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, Beijing, 102627, China
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shuting Weng
- Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, Beijing, 102627, China
| | - Leping Ye
- Department of Pediatric Pulmonology, Children's Medical Center, Peking University First Hospital, Beijing, 102627, China.
| |
Collapse
|
6
|
Huang Z, Li L, Zhang B, Yao D, Xiao B, Mo B. Investigation of the mechanistic impact of CBL0137 on airway remodeling in asthma. BMC Pulm Med 2025; 25:129. [PMID: 40114084 PMCID: PMC11927260 DOI: 10.1186/s12890-025-03596-y] [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: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Bronchial asthma, a chronic inflammatory airway disease, is characterized by airway remodeling, including thickening of the airway smooth muscle layer, primarily due to abnormal proliferation of airway smooth muscle cells (ASMCs). CBL0137 (Curaxin-137 hydrochloride), a histone chaperone facilitate chromatin transcription (FACT) inhibitor, has demonstrated anti-tumor properties, including inhibition of proliferation, promotion of apoptosis, and increased autophagy. However, its effects on ASMCs and airway remodeling remain unexplored. METHODS Asthma models were established using ovalbumin (OVA) in female C57BL/6 J mice, with therapeutic interventions using CBL0137 and budesonide. Lung tissues were analyzed using Hematoxylin and eosin (H&E), PAS, Masson's trichrome, and α-SMA immunofluorescence staining. ASMCs extracted from Sprague-Dawley rats were cultured in vitro experiments, with phenotypic changes assessed via flow cytometry. Gene and protein expressions were analyzed using RT-PCR and Western blotting. RESULTS CBL0137 significantly reduced airway resistance, goblet cell proliferation, alveolar collagen deposition, and airway smooth muscle layer thickening in asthmatic mice. In vitro, CBL0137 inhibited ASMC proliferation and induced apoptosis, downregulating cyclin-B1, Cdc2, and Bcl-2 while upregulating caspase-3. CONCLUSIONS CBL0137 mitigates airway remodeling of asthmatic mice by modulating ASMC proliferation and apoptosis, presenting a potential therapeutic strategy for asthma treatment.
Collapse
Affiliation(s)
- Zhiheng Huang
- Department of Respiratory and Critical Care Medicine, Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Chest Hospital of Guangxi Zhuang Autonomous Region, Liuzhou, 545005, China
| | - Liangxian Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541000, China
| | - Bingxi Zhang
- Department of Pulmonary and Critical Care Medicine, The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Dong Yao
- Department of Respiratory and Critical Care Medicine, Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China.
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, the Key Laboratory of Respiratory Diseases,Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541001, China.
| | - Bo Xiao
- Department of Pulmonary and Critical Care Medicine, The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
- Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China.
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, the Key Laboratory of Respiratory Diseases,Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
7
|
Liu M, Gao C, Li J, Zhang Y, Gao R, Yang C, Zhang J. The association between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and pulmonary function: evidence from NHANES 2007-2012. Front Nutr 2025; 12:1534958. [PMID: 40177182 PMCID: PMC11961415 DOI: 10.3389/fnut.2025.1534958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Background This research aims to explore the potential association between lung function and the ratio of non-high-density lipoprotein cholesterol (NHL) to high-density lipoprotein cholesterol (NHHR). Previous research has shown that lipid metabolism imbalance is closely linked to cardiovascular disease, however, there is a lack of information regarding its impact on lung function. Methods This research used information from the National Health and Nutrition Examination Survey (NHANES) spanning the years 2007 to 2012, including a large-scale sample of 9,498 adults aged 20 years and older. A cross-sectional study employing multivariable regression models was aimed at examining the relevance between NHHR and indicators of lung function (FEV1, FVC, and FEV1/FVC). Adjustments were made for a wide range of confounding factors, encompassing race, gender, age, BMI, smoking status, physical activity, diabetes, alcohol consumption, and education level. Data analysis included categorizing NHHR into quartiles and using trend tests to evaluate dose-response relationships between NHHR quartiles and lung function. Sensitivity analyses were conducted by excluding participants with asthma and COPD to ensure the reliability of the results. Results The results manifested a significant correlation between decreased FEV1 and FVC values and elevated NHHR, most notably within the highest quartile of NHHR (Q4), where the association was most pronounced. Additionally, trend test results indicated a significant linear negative correlation between NHHR and both FEV1 and FVC. However, the correlation between FEV1/FVC and NHHR showed a nonlinear U-shaped pattern. Suggesting differential impacts of NHHR on various lung function indicators. The findings' robustness was shown by sensitivity analysis, which revealed that even after omitting people with asthma and COPD, the negative correlation between NHHR and FEV1 and FVC remained significant. Conclusion This research emphasizes the significance of tracking lipid levels in evaluating respiratory health and offers early evidence in favor of NHHR as a probable biomarker for respiratory function. Further longitudinal research has occasion to prove the causal relationship between NHHR and lung function and to explore its underlying biological mechanisms.
Collapse
Affiliation(s)
- Miaoyan Liu
- Department of Respiratory Medicine, Chest Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Chaofeng Gao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinggeng Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yibo Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Rui Gao
- Graduate Work Department, Xi'an Medical University, Xi'an, China
| | - Chaoting Yang
- Graduate Work Department, Xi'an Medical University, Xi'an, China
| | - Jian Zhang
- Department of Respiratory Medicine, Chest Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| |
Collapse
|
8
|
Zhou J, Yang X, Zhou J, Xiong M, Wen L. Efficacy of medical education combined with extended care on adherence to inhaled glucocorticoids and clinical effects in patients with bronchial asthma. J Asthma 2025; 62:492-499. [PMID: 39352707 DOI: 10.1080/02770903.2024.2410423] [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: 07/31/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
OBJECTIVE To observe the clinical efficacy of medical education combined with extended care in patients with bronchial asthma and its effect on adherence to inhaled glucocorticoids. METHODS Ninety-eight patients with bronchial asthma were divided into the control group and the experimental group, n = 49, by utilizing the random number table method. The control group was given routine education and care as well as routine out-of-hospital instructions, and the experimental group was given medical education and extended care based on the control group. Asthma disease knowledge mastery, asthma control, quality of life, medication adherence and lung function were compared between both groups, and the number of asthma attacks and re-hospitalizations were recorded. RESULTS The experimental group performed higher scores of health knowledge, asthma control test and quality of life, rate of complete adherence, forced expiratory volume in one second (FEV1), peak expiratory flow rate, and FEV1/forced vital capacity. The number of asthma attacks and the times of re-hospitalizations were lower in the experimental group (all p < 0.05). CONCLUSION Medical education combined with extended care can improve bronchial asthma patients' mastery of asthma disease knowledge, effectively control patients' conditions, enhance patients' quality of life and lung function, increase patients' adherence to inhaled glucocorticoids, and reduce the recurrence of bronchial asthma patients.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Toxicosis/Nephrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuhao Yang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jiaqing Zhou
- Department of Pneumoconiology and Respiratory and Critical Care Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Maoyu Xiong
- Department of Pneumoconiology and Respiratory and Critical Care Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Wen
- Department of Pneumoconiology and Respiratory and Critical Care Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Alladina J, Medoff BD, Cho JL. Innate Immunity and Asthma Exacerbations: Insights From Human Models. Immunol Rev 2025; 330:e70016. [PMID: 40087882 PMCID: PMC11922041 DOI: 10.1111/imr.70016] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Asthma is a common chronic respiratory disease characterized by the presence of airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. Repeated asthma exacerbations can lead to progressive airway remodeling and irreversible airflow obstruction. Thus, understanding and preventing asthma exacerbations are of paramount importance. Although multiple endotypes exist, asthma is most often driven by type 2 airway inflammation. New therapies that target specific type 2 mediators have been shown to reduce the frequency of asthma exacerbations but are incompletely effective in a significant number of asthmatics. Furthermore, it remains unknown whether current treatments lead to sustained changes in the airway or if targeting additional pathways may be necessary to achieve asthma remission. Activation of innate immunity is the initial event in the inflammatory sequence that occurs during an asthma exacerbation. However, there continue to be critical gaps in our understanding of the innate immune response to asthma exacerbating factors. In this review, we summarize the current understanding of the role of innate immunity in asthma exacerbations and the methods used to study them. We also identify potential novel therapeutic targets for asthma and future areas for investigation.
Collapse
Affiliation(s)
- Jehan Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Josalyn L Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
10
|
Schmidt C, Hatziklitiu W, Trinkmann F, Cattaneo G, Port J. Investigation of inert gas washout methods in a new numerical model based on an electrical analogy. Med Biol Eng Comput 2025; 63:447-466. [PMID: 39373835 PMCID: PMC11750920 DOI: 10.1007/s11517-024-03200-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
Inert gas washout methods have been shown to detect pathological changes in the small airways that occur in the early stages of obstructive lung diseases such as asthma and COPD. Numerical lung models support the analysis of characteristic washout curves, but are limited in their ability to simulate the complexity of lung anatomy over an appropriate time period. Therefore, the interpretation of patient-specific washout data remains a challenge. A new numerical lung model is presented in which electrical components describe the anatomical and physiological characteristics of the lung as well as gas-specific properties. To verify that the model is able to reproduce characteristic washout curves, the phase 3 slopes (S3) of helium washouts are simulated using simple asymmetric lung anatomies consisting of two parallel connected lung units with volume ratios of1.25 0.75 ,1.50 0.50 , and1.75 0.25 and a total volume flow of 250 ml/s which are evaluated for asymmetries in both the convection- and diffusion-dominated zone of the lung. The results show that the model is able to reproduce the S3 for helium and thus the processes underlying the washout methods, so that electrical components can be used to model these methods. This approach could form the basis of a hardware-based real-time simulator.
Collapse
Affiliation(s)
- Christoph Schmidt
- Institute of Biomedical Engineering, University of Stuttgart, Seidenstraße 36, 70174, Stuttgart, Germany.
| | - Wasilios Hatziklitiu
- Institute of Biomedical Engineering, University of Stuttgart, Seidenstraße 36, 70174, Stuttgart, Germany
| | - Frederik Trinkmann
- Pneumology and Critical Care Medicine, Thoraxklinik at University Hospital Heidelberg, Translational Lung Research Center Heidelberg (TLRC), Member of German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Biomedical Informatics, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medical Center Mannheim, Heidelberg University, Heidelberg, Germany
| | - Giorgio Cattaneo
- Institute of Biomedical Engineering, University of Stuttgart, Seidenstraße 36, 70174, Stuttgart, Germany
| | - Johannes Port
- Institute of Biomedical Engineering, University of Stuttgart, Seidenstraße 36, 70174, Stuttgart, Germany
| |
Collapse
|
11
|
Lei J, Shu Z, Zhu H, Zhao L. AMPK Regulates M1 Macrophage Polarization through the JAK2/STAT3 Signaling Pathway to Attenuate Airway Inflammation in Obesity-Related Asthma. Inflammation 2025; 48:372-392. [PMID: 38886294 DOI: 10.1007/s10753-024-02070-x] [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: 04/07/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Abstract-Obesity-related asthma is primarily characterized by nonallergic inflammation, with pathogenesis involving oxidative stress, metabolic imbalance, and immunoinflammatory mechanisms. M1 macrophages, which predominantly secrete pro-inflammatory factors, mediate insulin resistance and systemic metabolic inflammation in obese individuals. Concurrently, adenosine monophosphate-activated protein kinase (AMPK) serves as a critical regulator of intracellular energy metabolism and is closely associated with macrophage activation. However, their specific roles and associated mechanisms in obesity-related asthma remain to be explored. In this study, we investigated the macrophage polarization status and potential interventional mechanisms through obesity-related asthmatic models and lipopolysaccharide (LPS) -treated RAW264.7 cell with a comprehensive series of evaluations, including HE, PAS and Masson staining of lung histopathology, immunohistochemical staining, immunofluorescence technology, qRT-PCR, Western Blot, and ELISA inflammatory factor analysis. The results revealed M1 macrophage polarization in obesity-related asthmatic lung tissue alongside downregulation of AMPK expression. Under LPS stimulation, exogenous AMPK activation attenuated M1 macrophage polarization via the Janus kinase 2/ signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway. Additionally, in obesity-related asthmatic mice, AMPK activation was found to alleviate airway inflammation by regulating M1 macrophage polarization, the mechanism closely associated with the JAK2/STAT3 pathway. These findings not only advance our understanding of macrophage polarization in obesity-related asthma, but also provide new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jiahui Lei
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Zhenhui Shu
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, 450003, China
| | - He Zhu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Limin Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, China.
- To whom correspondence should be addressed at Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China.
| |
Collapse
|
12
|
Luo T, Ji W, Gong Y, Chen L, Liu C, Zhang D, Li X, Lv Y. REDD1 mediates HDM-induced nuclear-cytoplasmic translocation and release of IL-33 in airway epithelial cells by downregulating Nrf2. Respir Res 2025; 26:47. [PMID: 39893427 PMCID: PMC11786574 DOI: 10.1186/s12931-025-03119-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
OBJECTIVE This study aims to investigate whether REDD1 (Regulated in Development and DNA Damage Responses 1) mediates the nuclear-to-cytoplasmic translocation and release of IL-33 in airway epithelial cells induced by house dust mites (HDM). METHODS REDD1 expression levels in bronchial asthma patients were validated using public databases, followed by immunohistochemical analysis of REDD1 protein in airway epithelial cells from these patients. An asthma model was then established using HDM-induced 16HBE cell lines, with REDD1 gene knockout performed. The relationship between varying levels of REDD1 expression, Nrf2, and related inflammatory factors was assessed using Western blot and qPCR. To further investigate the role of the REDD1-Nrf2-IL-33 axis in the development of asthma, we employed Nrf2 activators and inhibitors to reassess the impact of REDD1 on IL-33. RESULTS At both mRNA and protein levels, we found that REDD1 was significantly overexpressed in samples from asthma patients (P < 0.05). In vitro, 24-hour exposure to HDM induced a notable nuclear-to-cytoplasmic translocation of IL-33 and increased its levels in the culture medium of 16HBE cells. In addition, HDM treatment significantly upregulated the expression of both REDD1 and Nrf2. Knockdown of REDD1 markedly suppressed HDM-induced IL-33 release and the expression of TNF-α, IL-6, and IL-1β, while enhancing Nrf2 expression. Moreover, treatment with the Nrf2 agonist curcumin inhibited HDM-induced nuclear-to-cytoplasmic translocation and extracellular secretion of IL-33, whereas the opposite effect was observed when using the Nrf2 antagonist ML385. CONCLUSION This study reveals the crucial regulatory role of the REDD1-Nrf2-IL-33 axis in the pathological process of bronchial asthma. REDD1 modulates the expression of IL-33 and other inflammatory factors through the Nrf2 signaling pathway, thereby influencing the onset and progression of asthma. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Tian Luo
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Wentao Ji
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Yuxin Gong
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Foshan, Guangdong, 510280, China
| | - Lichang Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Foshan, Guangdong, 510280, China
| | - Chao Liu
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Dandan Zhang
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Xi Li
- Department of Respiratory and Critical Care Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528300, China.
| | - Yanhua Lv
- Department of Respiratory and Critical Care Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528300, China.
| |
Collapse
|
13
|
Abbasi Moajani F, Soozangar N, Amani M, Jeddi F, Salimnejad R, Aslani MR. The suppressive effects of crocin from saffron on allergic airway inflammation through Drp1/Nfr1/Mfn2/Pgc1-alpha signaling pathway in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118862. [PMID: 39326816 DOI: 10.1016/j.jep.2024.118862] [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: 06/15/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saffron is derived from the dried stigmas of Crocus sativus L., which was considered by ancient nations for food and medicinal purposes. In traditional medicine, the therapeutic use of Crocus sativus includes antispasmodic, antitussive and expectorant. AIM OF THE STUDY Mitochondrial fusion, fission, biogenesis, and mitophagy are essential processes for maintaining mitochondrial dynamics in response to cellular stress. The primary objective of this research was to examine how crocin affected the levels of important mitochondrial regulators, including Drp1, Pgc1α, Nrf1, and Mfn2, in the lung tissue of ovalbumin-sensitized mice. MATERIALS AND METHODS A total of fifty male BALB/C mice were randomly assigned to five unique groups (n = 10 for each group), including the control group, ovalbumin-sensitized group (OVA), OVA group treated with 30 mg/kg of crocin, OVA group treated with 60 mg/kg of crocin, and OVA group treated with 1 mg/kg of dexamethasone. Post-sensitization and ovalbumin challenge, mice lung tissues were evaluated for the expression of Drp1, Pgc1α, Nrf1, and Mfn2 mRNA levels using real-time PCR as well as histopathological assessments. RESULTS In the OVA group, there was a significant elevated in inflammatory cells such as eosinophils, neutrophils, macrophages, and lymphocytes; however, crocin (both concentrations) and dexamethasone intervention showed significant inhibitory effects (P < 0.01 to P < 0.001). Moreover, an increase in the expression of Drp1, Pgc1α, and Nrf1 levels was seen in the OVA group, while crocin and dexamethasone showed protective benefits (P < 0.05 to P < 0.001). Furthermore, the levels of Mfn2 were reduced in the lung tissue of mice exposed to ovalbumin, but this decrease was reversed by crocin 60 (P < 0.05) and dexamethasone treatment (P < 0.001). CONCLUSION In mice with OVA sensitization, the balance of mitochondrial dynamics in lung tissue was disrupted, but intervention of crocin identified to have a protective effect.
Collapse
Affiliation(s)
- Fatima Abbasi Moajani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Narges Soozangar
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mojtaba Amani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ramin Salimnejad
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
14
|
Cheng SC, Liou CJ, Wu SJ, Lin CF, Huang TH, Huang WC. Neochlorogenic acid ameliorates allergic airway inflammation by suppressing type 2 immunity and upregulating HO-1 expression. Int Immunopharmacol 2025; 146:113867. [PMID: 39689596 DOI: 10.1016/j.intimp.2024.113867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Neochlorogenic acid is a natural compound isolated from various fruits and vegetables that has anti-inflammation and anti-oxidative effects in macrophages. Inflammatory immune cells and tracheal epithelial cells can stimulate airway hyperresponsiveness, inflammation, and reactive oxygen species. In this study, we investigated the effect of neochlorogenic acid in ameliorating inflammatory and oxidative responses in asthmatic mice. We used an ovalbumin (OVA)-induced mouse model, treating mice with neochlorogenic acid by intraperitoneal injection. We also treated inflammatory human tracheal epithelial (BEAS-2B) cells with neochlorogenic acid to evaluate inflammatory cytokine levels and oxidative responses. The results demonstrate that neochlorogenic acid attenuated airway hyperresponsiveness, eosinophil infiltration, and goblet cell hyperplasia in the lungs of asthmatic mice. Neochlorogenic acid also reduced type 2 cytokine expression in bronchoalveolar lavage fluid and improved oxidative stress in the lung. Neochlorogenic acid effectively blocked monocyte attachment to adherent BEAS-2B cells, and reduced pro-inflammatory cytokine and reactive oxygen species production in inflammatory BEAS-2B cells. These findings suggest that neochlorogenic acid is a potential immunomodulator that can ameliorate airway hyperresponsiveness and airway inflammation in asthmatic mice.
Collapse
Affiliation(s)
- Shu-Chen Cheng
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33303, Taiwan
| | - Chian-Jiun Liou
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City 33303, Taiwan
| | - Shu-Ju Wu
- Department of Nutrition and Health Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33303, Taiwan
| | - Chwan-Fwu Lin
- Department of Cosmetic Science, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan City 33303, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan City 33303, Taiwan
| | - Tse-Hung Huang
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33303, Taiwan.
| | - Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City 33303, Taiwan; Department of Pediatrics, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei 23656, Taiwan.
| |
Collapse
|
15
|
Kim YH, Park CH, Kim JM, Yoon YC. Chitooligosaccharides suppress airway inflammation, fibrosis, and mucus hypersecretion in a house dust mite-induced allergy model. FRONTIERS IN ALLERGY 2025; 6:1533928. [PMID: 39927112 PMCID: PMC11799285 DOI: 10.3389/falgy.2025.1533928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
Background Respiratory allergy is a serious respiratory disorder characterized by inflammation, mucus hypersecretion, and airway tissue sclerosis. Disruption of the T helper 1 (Th1) and T helper 2 (Th2) immune systems by stimuli induced by house dust mites (HDM) and fine particulate matter leads to the secretion of various inflammatory cytokines, resulting in immune respiratory diseases characterized by airway inflammation. Chitooligosaccharides (COS) are known for their antioxidant and anti-inflammatory properties. Methods Human airway epithelial cells (BEAS-2B) were cultured in DMEM/F12 medium containing COS at concentrations of 25-100 µg/ml for 24 h. No intracellular toxicity was observed up to 1,000 µg/ml. Cell experiments were conducted at COS concentrations below 100 µg/ml, while animal experiments were performed at concentrations below 100 mg/kg body weight for 4 weeks. Samples of right lung tissue obtained from the experimental animals were used for gene and protein expression analysis, whereas samples of contralateral lung tissue were used for immunohistochemical analysis. Results COS regulated Th1 immunity by inhibiting major cytokines, including inflammatory tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), in BEAS-2B cells. In the HDM-induced allergic respiratory model, COS suppressed the infiltration of inflammatory cells around the airways and inhibited the mRNA expression of Th1 immune cytokines in lung tissues, while also reducing the expression of nuclear factor kappa B (NF-κB)-related proteins. Furthermore, the results confirmed the suppression of the levels of immunoglobulin E (IgE) in the blood secreted by mast cells activated by HDM, which led to a reduction in allergic mucus hypersecretion and airway sclerosis. Conclusion In summary, COS are thought to improve airway resistance by alleviating inflammatory allergic respiratory diseases caused by HDM and are regarded as substances that regulate the balance of the Th1 and Th2 immune systems in epithelial cells affected by mucus hypersecretion.
Collapse
Affiliation(s)
| | | | | | - Yeo Cho Yoon
- Healthcare & Nutrition Laboratory, Amicogen, Inc., Seongnam, Republic of Korea
| |
Collapse
|
16
|
Wang H, Chen Y, Zhang J, Wang N, Tian T. Deletion of BRCC3 ameliorates airway inflammation in asthma by inhibiting the activation of NLRP3 inflammasome. Int Immunopharmacol 2025; 145:113720. [PMID: 39642564 DOI: 10.1016/j.intimp.2024.113720] [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: 07/10/2024] [Revised: 11/14/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
BRCA1/BRCA2-containing complex subunit 3 (BRCC3) serves as a deubiquitinating enzyme contributing to multiple inflammation-related disorders. However, the role of BRCC3 in modulating airway inflammation in asthma has not been investigated. This study aimed to examine the role of BRCC3 in airway inflammation using a mouse model of asthma induced by ovalbumin (OVA). BRCC3 levels were found to be elevated in mice with asthma. BRCC3 knockout (KO) mice demonstrated a notable improvement in pathological changes, accompanied by reduced levels of inflammatory cell infiltration and inflammatory cytokines, compared to wild-type (WT) mice following OVA challenge. The NLRP3 inflammasome was high activated in asthmatic mice, which was restrained by BRCC3 KO, as companied by a decrease in NLRP3, ASC, cleaved Caspase-1, cleaved Gasdermin D (GSDMD), IL-1β, and IL-18. In vitro studies demonstrated BRCC3 levels increased in airway epithelial cells in response to house dust mite (HDM) stimulation, depending on the dose and duration of exposure. Silencing BRCC3 in airway epithelial cells protected against HDM-induced cell injury and inflammation, along with inhibiting the NLRP3 inflammasome and pyroptosis. Conversely, the overexpression of BRCC3 in airway epithelial cells worsened DM-induced cell injury and inflammation while also enhancing the NLRP3 inflammasome and pyroptosis. Further investigations revealed that silencing BRCC3 increased the ubiquitination of NLRP3, whereas overexpressing BRCC3 decreased it. Pharmacological inhibition of the NLRP3 inflammasome diminished the effects of BRCC3 overexpression on the inflammation and pyroptosis induced by HDM in airway epithelial cells. Overall, these findings underscore the importance of BRCC3 in the pathogenesis of asthma. Deletion of BRCC3 alleviates airway inflammation in asthma by impeding the activation of the NLRP3 inflammasome, thus indicating that BRCC3 could serve as a potential target for asthma therapy.
Collapse
Affiliation(s)
- Hao Wang
- The First Department of Pediatrics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi Province 710100, China
| | - Yao Chen
- Department of Pediatrics, Xi'an Zhongda International Hospital, Xi'an, Shaanxi Province 710000, China
| | - Jin Zhang
- The First Department of Pediatrics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi Province 710100, China
| | - Ning Wang
- The First Department of Pediatrics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi Province 710100, China
| | - Tian Tian
- The First Department of Pediatrics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi Province 710100, China.
| |
Collapse
|
17
|
Zhang H, Zheng W, Peng R, Wu D, Hu Y, Sun T, Gao L, Liu Y, Guo L, Ding Y, Liu L. First-in-human study on tolerability, pharmacokinetics and pharmacodynamics of single and multiple escalating doses of XKH001, a recombinant humanized monoclonal antibody against IL-25 in healthy Chinese volunteers. Expert Opin Investig Drugs 2025; 34:81-87. [PMID: 39815604 DOI: 10.1080/13543784.2025.2453162] [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: 10/31/2024] [Revised: 12/04/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND XKH001 is a recombinant humanized IgG1 monoclonal antibody against IL-25 for the treatment of type 2 inflammatory diseases. This study aimed to evaluate the tolerability, pharmacokinetics, and pharmacodynamics of XKH001 in humans for the first time. RESEARCH DESIGN AND METHODS This clinical investigation adopted a randomized, double-blind, and placebo-controlled single ascending dose (SAD) and multiple ascending dose (MAD) design. RESULTS XKH001 was well tolerated in healthy Chinese subjects. Following repeated administration, XKH001 showed a slow absorption with a median Tmax of 4-7 days and a mean half-life (t1/2) of 22-25 days. The accumulation ratio ranged from 1.34 to 1.99. The exposure was mostly dose proportional, with a mean slope of 0.85-1.06. All subjects tested negative for ADA (except three subjects tested positive). The subjects who received 600 mg XKH001 in the MAD study showed a 78.2 ng/mL decrease in the total immunoglobulin E (IgE) level 85 days after the first administration, while the subjects who received matched placebo exhibited only an 8.6 ng/mL decrease. CONCLUSIONS XKH001 showed favorable safety and pharmacokinetics profiles and a low immunogenicity in its first-in-human study. The data support its further clinical evaluation in patients with type 2 inflammatory diseases. TRIAL REGISTRATION The study was registered in ClinicalTrials.gov (NCT05991661).
Collapse
Affiliation(s)
- Hong Zhang
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Wenbo Zheng
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Ran Peng
- Beijing Kanova Biopharmaceutical Co. Ltd, Beijing, China
| | - Dandan Wu
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Yue Hu
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Tiantian Sun
- Beijing Kanova Biopharmaceutical Co. Ltd, Beijing, China
| | - Lei Gao
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Yusi Liu
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Li Guo
- Beijing Kanova Biopharmaceutical Co. Ltd, Beijing, China
| | - Yanhua Ding
- Phase I Clinical Research Center, The First Hospital of Jilin University, Jilin, China
| | - Li Liu
- Department of Pediatric Respiratory, Children's Medical Center, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
18
|
Perfilyeva YV, Aquino AD, Borodin MA, Kali A, Abdolla N, Ostapchuk YO, Tleulieva R, Perfilyeva AV, Jainakbayev NT, Sharipov KO, Belyaev NN. Can interventions targeting MDSCs improve the outcome of vaccination in vulnerable populations? Int Rev Immunol 2024:1-17. [PMID: 39707917 DOI: 10.1080/08830185.2024.2443423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/26/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Preventive vaccination is a crucial strategy for controlling and preventing infectious diseases, offering both effectiveness and cost-efficiency. However, despite the widespread success of vaccination programs, there are still certain population groups who struggle to mount adequate responses to immunization. These at-risk groups include but are not restricted to the elderly, overweight individuals, individuals with chronic infections and cancer patients. All of these groups are characterized by persistent chronic inflammation. Recent studies have demonstrated that one of the key players in immune regulation and the promotion of chronic inflammation are myeloid-derived suppressor cells (MDSCs). These cells possess a wide range of immunosuppressive mechanisms and are able to dampen immune responses in both antigen-specific and antigen-nonspecific manner, thus contributing to the establishment and maintenance of an inflammatory environment. Given their pivotal role in immune modulation, there is growing interest in understanding how MDSCs may influence the efficacy of vaccines, particularly in vulnerable populations. In this narrative review, we discuss whether MDSCs are able to regulate vaccine-induced immunity and whether their suppression can potentially enhance vaccine efficacy in vulnerable populations.
Collapse
Affiliation(s)
- Yuliya V Perfilyeva
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Arthur D Aquino
- Almazov National Medical Research Center, St. Petersburg, Russia
| | - Maxim A Borodin
- Almazov National Medical Research Center, St. Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Aikyn Kali
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Nurshat Abdolla
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Al-Farabi, Kazakh National University, Almaty, Kazakhstan
| | | | - Raikhan Tleulieva
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | | | | - Kamalidin O Sharipov
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | |
Collapse
|
19
|
Ignatova GL, Avdeev SN, Antonov VN, Blinova EV. [The effectiveness of vaccination against pneumococcal infection in patients with severe bronchial asthma]. TERAPEVT ARKH 2024; 96:1057-1062. [PMID: 39731766 DOI: 10.26442/00403660.2024.11.203038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Indexed: 12/30/2024]
Abstract
INTRODUCTION The article discusses topical issues of the use of conjugated 13-valent pneumococcal vaccine Prevenar®13 (PCV13) in patients with severe bronchial asthma (SBA), including those receiving targeted therapy with genetically engineered biological drugs (GEBD). AIM To study the effectiveness of vaccination against pneumococcal infection (PI) in patients with SBA. MATERIALS AND METHODS The study included 381 patients with SBA. The average age in the study groups was 45.5 (42.0; 52.5) years. All patients underwent clinical and instrumental studies, including spirography with bronchodilation test. After confirming the diagnosis of BA, the patients were divided into 2 observation groups. Group 1 (n=191) consisted of patients undergoing treatment with GEBD. Group 2 included patients with asthma receiving standard therapy, according to the 4th-5th stage according to the criteria of the Global Initiative for Asthma 2022 (Global Initiative for Asthma - GINA). The observation group consisted of 190 patients. In each group, there are subgroups of patients who have been vaccinated against PI and have not been vaccinated for various reasons. The following criteria were used as the main endpoints of observation for 12 months to assess the effectiveness: the number of pneumonia during the observation period, the number of exacerbations of asthma (severe, non-severe), the number of hospitalizations, the duration of exacerbations, the level of control according to the Asthma Control Questionnaire (ACQ5), functional indicators. RESULTS The coverage of PI vaccination in patients with BA remains quite low, further organizational and methodological work is required to increase their involvement in vaccination. Immunization of PCV13 in patients with SBA at the 4th-5th stage of therapy reduces the risk of community-acquired pneumonia by at least 28.5%. PCV13 vaccination may be an additional effective tool for controlling the symptoms of SBA, including in patients undergoing treatment with GEBD. Vaccination allows to normalize the functional parameters of respiratory function in patients with SBA. PCV13 is well tolerated and does not cause any significant allergic reactions in patients with asthma. CONCLUSION PCV13 vaccination is an effective tool for reducing the risk of community-acquired pneumonia in patients with severe bronchial asthma, including those on targeted therapy with genetically engineered biological drugs.
Collapse
Affiliation(s)
| | - S N Avdeev
- Sechenov First Moscow State Medical University (Sechenov University)
- Research Institute of Pulmonology
| | | | | |
Collapse
|
20
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
21
|
Quan J, Xie D, Li Z, Yu X, Liang Z, Chen Y, Wu L, Huang D, Lin L, Fan L. Luteolin alleviates airway remodeling in asthma by inhibiting the epithelial-mesenchymal transition via β-catenin regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156090. [PMID: 39393303 DOI: 10.1016/j.phymed.2024.156090] [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: 04/19/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Asthma is a prevalent long-term inflammatory condition that causes airway inflammation and remodeling. Increasing evidence indicates that epithelial-mesenchymal transition (EMT) holds a prominent implication in airway reconstruction in patients with asthma. Flavonoids obtained from Chinese Materia Medica (CMM), such as Luteolin (Lut), exhibit various beneficial effects in various asthma models. Lut has been shown to mitigate various asthma symptoms, including airway inflammation, hyperresponsiveness, bronchoconstriction, excessive mucus production, pulmonary autophagy, and neutrophilic asthma. However, whether flavonoids can suppress EMT-associated airway remodeling in asthma and the fundamental mechanisms involved remain unclear, with no studies specifically addressing Lut in this context. PURPOSE To evaluate the inhibition of airway remodeling in asthma by Lut and its potential mechanisms, while examining the significance of β-catenin in this process through cellular and animal studies. METHODS A BEAS-2B cell model stimulated by lipopolysaccharide (LPS) was established in vitro. Wound closure and Transwell assays were utilized to assess the cellular migratory ability. EMT- and fibrosis-related markers in LPS-stimulated cells were evaluated using RT-qPCR and western blotting. The status of the β-catenin/E-cadherin and β-catenin destruction complexes was evaluated using western blotting, immunofluorescence (IF) staining, and co-immunoprecipitation (Co-IP) analysis. The regulatory function of Lut in β-catenin-dependent EMT was further validated by β-catenin overexpression with adenovirus transduction and siRNA-mediated knockdown of β-catenin. Moreover, the counts of different types of bronchoalveolar lavage fluid (BALF) inflammatory cells from mice with asthma induced by ovalbumin (OVA) were evaluated in vivo using Congo red staining. Hematoxylin and eosin (H&E), Masson's trichrome, and periodic acid-Schiff (PAS) staining were used to evaluate collagen deposition, mucus production, and inflammation in murine lung tissues. Western blotting and immunohistochemistry (IHC) assays were used to assess EMT- and fibrosis-related markers in the lung tissues in vivo. RESULT Six naturally derived flavonoids, including Lut, attenuated cell migration and prevented EMT in LPS-treated BEAS-2B cells. Moreover, Lut suppressed TGF-β1, MMP-9, fibronectin (FN), and α-smooth muscle actin (α-SMA) levels in LPS-stimulated BEAS-2B cells. Additionally, Lut downregulated the levels of β-catenin by modulating the β-catenin/E-cadherin and β-catenin destruction complexes, highlighting the pivotal role of β-catenin in EMT inhibition by Lut in LPS-stimulated BEAS-2B cells. Furthermore, Lut suppressed airway inflammation and attenuated EMT-associated airway remodeling through β-catenin blockade in OVA-induced asthmatic mice. The bronchial wall thickness notably reduced from 37.24 ± 4.00 μm in the asthmatic model group to 30.06 ± 4.40 μm in the Lut low-dose group and 24.69 ± 2.87 μm in the Lut high-dose group. CONCLUSION According to our current understanding, this research is the first to reveal that Lut diminishes airway remodeling in asthma by inhibiting EMT via β-catenin regulation, thereby filling a research gap concerning Lut and flavonoids. These results provide a theoretical basis for treating asthma with anti-asthmatic CMM, as well as a candidate and complementary therapeutic approach to treat asthma.
Collapse
Affiliation(s)
- Jingyu Quan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Dan Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Zihong Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xuhua Yu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Ziyao Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Yuanbin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Lei Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Donghui Huang
- Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, China.
| | - Lin Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| | - Long Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| |
Collapse
|
22
|
Lin Z, Zhuang J, He L, Zhu S, Kong W, Lu W, Zhang Z. Exploring Smad5: a review to pave the way for a deeper understanding of the pathobiology of common respiratory diseases. Mol Med 2024; 30:225. [PMID: 39578779 PMCID: PMC11585160 DOI: 10.1186/s10020-024-00961-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024] Open
Abstract
Smad5 (small mothers against decapentaplegic 5) protein is a receptor-regulated member of the Smad family proteins, mainly participating in the bone morphogenetic protein (BMP) signaling pathway in its phosphorylated form. This article will provide a detailed review of Smad5, focusing on its gene characteristics, protein structure, and subcellular localization properties. We will also explore the related signaling pathways and the mechanisms of Smad5 in respiratory diseases, including chronic obstructive pulmonary disease (COPD), bronchial asthma, pulmonary arterial hypertension(PAH), lung cancer, and idiopathic pulmonary fibrosis (IPF). Additionally, the review will cover aspects such as proliferation, differentiation, apoptosis, anti-fibrosis, and mitochondrial function metabolism. In addition, the review will cover aspects of proliferation, differentiation, apoptosis, anti-fibrosis and functional mitochondrial metabolism related to the above topics. Numerous studies suggest that Smad5 may play a unique and important role in the pathogenesis of respiratory system diseases. However, in previous research, Smad5 was mainly used to broadly determine the activation of the BMP signaling pathway, and its own function has not been given much attention. It is worth noting that Smad5 has distinct nuclear-cytoplasmic distribution characteristics different from Smad1 and Smad8. It can undergo significant nuclear-cytoplasmic shuttling when intracellular pH (pHi) changes, playing important roles in both the classical BMP signaling pathway and non-BMP signaling pathways. Given that Smad5 can move intracellularly in response to changes in physicochemical properties, its cellular localization may play a crucial role in the development of respiratory diseases. This article will explore the possibility that its distribution characteristics may be an important factor that is easily overlooked and not adequately considered in disease research.
Collapse
Affiliation(s)
- Zeqiang Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiayu Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixia He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Siyuan Zhu
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiguo Kong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Zili Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Miralles-López JC, Alvarez-Gutiérrez FJ, Delgado-Romero J, Quirce S, Soto-Campos JG, Andújar-Espinosa R, Cabrejos-Perotti S, Castilla-Martínez M, Flores-Martín I, Pajarón-Fernández MJ, Valverde-Molina J. Disease Modification in Asthma: Are We on the Right Way? A Multidisciplinary Expert Delphi Consensus (MODIASTHMA Consensus). J Asthma Allergy 2024; 17:1163-1171. [PMID: 39558970 PMCID: PMC11571985 DOI: 10.2147/jaa.s488764] [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: 08/27/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose With the advent of biological therapies, emerging concepts regarding establishing new targets in asthma management, such as disease modification, have entered the debate among the scientific community. The definitions that form the conceptual basis of this goal need to be agreed upon. Methods A multidisciplinary expert group was assembled as the steering committee. A systematic literature review was conducted to identify the scientific background for constructing appropriate definitions. Based on the literature review and the clinical experience of the experts, the committee built a list of statements that could be applied to establish the definition of disease modification in asthma. After that, a Delphi validation was performed to assess the appropriateness of the list of statements. The questionnaire included a total of 22 statements, divided into "Essential criteria for disease modification in asthma" (5 statements) and "Disease modification indicators and other considerations" (17 statements). Panelists used a 9-point Likert scale to measure agreement on each statement. The cut-off point for high consensus was defined as a minimum score of 7 and had to be reached by at least two-thirds of the experts. Results A total of 192 asthma experts voted on statements anonymously. Of those, 104 (54%) were Pneumologists, 65 (34%) were allergologists, and 23 (12%) were Pediatricians. An interim analysis of round 1 data was performed. All statements reached consensus on the first round, with a median score above 7 in all cases. Conclusion In conclusion, in this Delphi study, a large number of experts in the management of severe asthma from different specialties agreed on the clinical-functional and pathophysiological aspects to be considered in order to try to achieve disease modification.
Collapse
Affiliation(s)
- Juan Carlos Miralles-López
- Allergy Department, University General Hospital Reina Sofia, Murcia, Spain
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
| | | | | | - Santiago Quirce
- Department of Allergy, La Paz University Hospital, IdiPAZ, Madrid, Spain
- CIBER of Respiratory Diseases (CIBERES), Madrid, Spain
| | | | - Ruben Andújar-Espinosa
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
- Pulmonology Department. H. U. Virgen de la Arrixaca, Murcia, Spain
| | - Sheila Cabrejos-Perotti
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
- Allergy Department, University General Hospital Santa Lucia, Cartagena, Spain
| | - Manuel Castilla-Martínez
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
- Pulmonology Department, University General Hospital Los Arcos, San Javier, Spain
| | - Isabel Flores-Martín
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
- Allergy Department, Hospital Comarcal del Noroeste, Caravaca, Murcia, Spain
| | - Manuel José Pajarón-Fernández
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
- Allergy Department, Hospital de la Vega Lorenzo Guirao, Cieza, Spain
| | - José Valverde-Molina
- Severe Asthma Association of the Region of Murcia. ASGRAMUR, Murcia, Spain
- Paediatrics Department, University General Hospital Santa Lucia (Cartagena University Hospital Complex), Cartagena, Spain
- IMIB Biomedical Research Institute, Murcia, Spain
| |
Collapse
|
24
|
Trusculescu AA, Ancusa VM, Pescaru CC, Wellmann N, Fira-Mladinescu C, Oancea CI, Fira-Mladinescu O. A Multifaceted Exploration of Status Asthmaticus: A Retrospective Analysis in a Romanian Hospital. J Clin Med 2024; 13:6615. [PMID: 39518753 PMCID: PMC11546779 DOI: 10.3390/jcm13216615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Status asthmaticus is a severe, life-threatening asthma exacerbation requiring urgent medical intervention. This study aims to examine its epidemiology in Timis County, Romania, over 11 years. Methods: A retrospective analysis was conducted using hospital records from 2013 to 2023, focusing on demographic, geospatial, and temporal distributions. Network analysis of the recorded comorbidities was used to identify phenotypic clusters among patients. Results: Females and older adults were disproportionately affected. Several triggers and geospatial patterns were identified. Five phenotypic clusters were determined: two in the T2-high endotype, two in T2-low, and a mixed one. Conclusions: The findings highlight the need for personalized asthma management strategies and public healthcare interventions in Timiș County, addressing specific demographic and geospatial factors. This study also provides a valuable reference for similar regions.
Collapse
Affiliation(s)
- Adriana Ana Trusculescu
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| | - Versavia Maria Ancusa
- Department of Computer and Information Technology, Automation and Computers Faculty, “Politehnica” University of Timis, Vasile Pârvan Blvd, no. 2, 300223 Timisoara, Romania
| | - Camelia Corina Pescaru
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| | - Norbert Wellmann
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Corneluta Fira-Mladinescu
- Hygiene Division, Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy Timisoara, Victor Babes Street, no. 16, 300226 Timisoara, Romania;
- Center for Study in Preventive Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania
| | - Cristian Iulian Oancea
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| | - Ovidiu Fira-Mladinescu
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| |
Collapse
|
25
|
Du L, Chen Z, Tao Q, Yang J, Chen N, Wang Q. Yunpi Xiefei Huatan Tang decoction reduces airway inflammation and airway remodeling in asthmatic mice through Wnt/β-catenin signaling pathway. Allergol Immunopathol (Madr) 2024; 52:110-116. [PMID: 39515805 DOI: 10.15586/aei.v52i6.1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Asthma is one of the most prevalent chronic respiratory diseases among children, markedly impairing patient's health and imposing an increasing burden on the healthcare system. Several traditional Chinese medicines have demonstrated efficacy in alleviating asthma symptoms through studies conducted on animal models. Recent studies have shown that the Yunpi Xiefei Huatan Tang decoction (YPD) exhibits significant therapeutic outcomes in treating phlegm-obstructed pulmonary asthma. However, the precise regulatory effects of YPD on the progression of asthma require additional investigation. OBJECTIVE To explore the functions of YPD in asthma progression. MATERIAL AND METHODS The asthma rat model triggered by ovalbumin (OVA) was established successfully. The pathological changes of lung tissues were examined through Hematoxylin and Eosin (H&E) staining. The levels of Interleukin 6 (IL-6) and IL-1β were tested through Enzyme-Linked-Immunosorbent Serologic Assay (ELISA). The number of total cells or eosinophils in bronchoalveolar lavage fluid was confirmed through cell counter. The collagen deposition in bronchi was assessed through Masson staining. The protein expressions were measured through western blot. RESULTS This study demonstrated that YPD could mitigate airway inflammation in an OVA-triggered asthma rat model. Furthermore, YPD was found to decrease the production of inflammatory cytokines in the lungs and suppress the infiltration of inflammatory cells into bronchoalveolar lavage fluid. Additionally, the airway remodeling stimulated by OVA could be suppressed following YPD treatment. Finally, it was disclosed that YPD inhibited the wingless-related integration site-beta-catenin (Wnt/β-catenin) signaling pathway in the OVA-stimulated asthma rat model. CONCLUSION YPD alleviated airway inflammation and remodeling in asthmatic mice via the Wnt/β-catenin signaling pathway. This research offers significant insights into the potential application of YPD in the treatment of asthma.
Collapse
Affiliation(s)
- Li Du
- Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Zhu Chen
- Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China;
| | - Qiong Tao
- Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Jianhui Yang
- Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Na Chen
- Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Qiao Wang
- Department of Pediatrics, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| |
Collapse
|
26
|
Fan W, Gui B, Zhou X, Li L, Chen H. A narrative review on lung injury: mechanisms, biomarkers, and monitoring. Crit Care 2024; 28:352. [PMID: 39482752 PMCID: PMC11526606 DOI: 10.1186/s13054-024-05149-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: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 11/03/2024] Open
Abstract
Lung injury is closely associated with the heterogeneity, severity, mortality, and prognosis of various respiratory diseases. Effective monitoring of lung injury is crucial for the optimal management and improved outcomes of patients with lung diseases. This review describes acute and chronic respiratory diseases characterized by significant lung injury and current clinical tools for assessing lung health. Furthermore, we summarized the mechanisms of lung cell death observed in these diseases and highlighted recently identified biomarkers in the plasma indicative of injury to specific cell types and scaffold structure in the lung. Last, we propose an artificial intelligence-driven lung injury monitoring model to assess disease severity, and predict mortality and prognosis, aiming to achieve precision and personalized medicine.
Collapse
Affiliation(s)
- Wenping Fan
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China
| | - Biyu Gui
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China
| | - Xiaolei Zhou
- Department of Pulmonary Medicine, Chest Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Li Li
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
| | - Huaiyong Chen
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, 300350, China.
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China.
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
| |
Collapse
|
27
|
Liu Y, Yin Q, Liu B, Lu Z, Liu M, Meng L, He C, Chang J. Fisetin reduces ovalbumin-triggered airway remodeling by preventing phenotypic switching of airway smooth muscle cells. Respir Res 2024; 25:370. [PMID: 39402516 PMCID: PMC11479573 DOI: 10.1186/s12931-024-03005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The transformation of airway smooth muscle cells (ASMCs) from a quiescent phenotype to a hypersecretory and hypercontractile phenotype is a defining feature of asthmatic airway remodeling. Fisetin, a flavonoid compound, possesses anti-inflammatory characteristics in asthma; yet, its impact on airway remodeling and ASMCs phenotype transition has not been investigated. OBJECTIVES This research seeked to assess the impact of fisetin on ovalbumin (OVA) induced asthmatic airway remodeling and ASMCs phenotype transition, and clarify the mechanisms through network pharmacology predictions as well as in vivo and in vitro validation. METHODS First, a fisetin-asthma-ASMCs network was constructed to identify potential targets. Subsequently, cellular and animal studies were carried out to examine the inhibitory effects of fisetin on airway remodeling in asthmatic mice, and to detemine how fisetin impacts the phenotypic transition of ASMCs. RESULTS Network analysis indicated that fisetin might affect asthma via mediating the phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (AKT) pathway. Intraperitoneal administration of fisetin in vivo reduced airway inflammation and remodeling, as shown by reduced inflammatory cells, decreased T helper type 2 (Th2) cytokine release, diminished collagen accumulation, mitigated airway smooth muscle thickening, and decreased expression of osteopontin (OPN), collagen-I and α-smooth muscle actin (α-SMA). Moreover, fisetin suppressed the PI3K/AKT pathway in asthmatic lung tissue. According to the in vitro data, fisetin downregulated the expression of the synthetic phenotypic proteins OPN and collagen-I, contractile protein α-SMA, and inhibited cellular migration, potentially through the PI3K/AKT pathway. CONCLUSION These results suggest that fisetin inhibits airway remodeling in asthma by regulating ASMCs phenotypic shift, emphasizing that fisetin is a promising candidate for the treatment of airway smooth muscle remodeling.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Qiling Yin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Bin Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Zheng Lu
- Tai'an Tumour Prevention and Treatment Hospital, Tai'an, Shandong, 271000, China
| | - Meijun Liu
- Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Ling Meng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| | - Chao He
- Department of Gastrointestinal Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, 271000, China.
| | - Jin Chang
- Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| |
Collapse
|
28
|
Bakalenko N, Kuznetsova E, Malashicheva A. The Complex Interplay of TGF-β and Notch Signaling in the Pathogenesis of Fibrosis. Int J Mol Sci 2024; 25:10803. [PMID: 39409132 PMCID: PMC11477142 DOI: 10.3390/ijms251910803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Fibrosis is a major medical challenge, as it leads to irreversible tissue remodeling and organ dysfunction. Its progression contributes significantly to morbidity and mortality worldwide, with limited therapeutic options available. Extensive research on the molecular mechanisms of fibrosis has revealed numerous factors and signaling pathways involved. However, the interactions between these pathways remain unclear. A comprehensive understanding of the entire signaling network that drives fibrosis is still missing. The TGF-β and Notch signaling pathways play a key role in fibrogenesis, and this review focuses on their functional interplay and molecular mechanisms. Studies have shown synergy between TGF-β and Notch cascades in fibrosis, but antagonistic interactions can also occur, especially in cardiac fibrosis. The molecular mechanisms of these interactions vary depending on the cell context. Understanding these complex and context-dependent interactions is crucial for developing effective strategies for treating fibrosis.
Collapse
Affiliation(s)
| | | | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia; (N.B.); (E.K.)
| |
Collapse
|
29
|
Bae J. Asymptomatic Pulmonary Fibrosis Associated With Ipsilateral Proximal Interruption of a Pulmonary Artery (PIPA). Cureus 2024; 16:e72572. [PMID: 39606523 PMCID: PMC11602208 DOI: 10.7759/cureus.72572] [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] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Pulmonary fibrosis is a chronic condition typically affecting both lungs; however, cases of unilateral pulmonary fibrosis are exceedingly rare and often result from specific unilateral inflammatory conditions like radiation pneumonitis or infection. An even rarer occurrence is the unilateral proximal interruption of a pulmonary artery (PIPA), a developmental anomaly resulting from the failed connection of the sixth aortic arch to the pulmonary trunk. This condition can manifest alone or alongside other cardiac abnormalities. There are limited reports of pulmonary fibrosis associated with PIPA. In this case, a 33-year-old male with chronic mild asthma presented with bilateral shoulder pain. Initial radiographs showed reticular opacities and volume loss in the right lung with a rightward mediastinal shift, suggesting possible fibrosis. Further investigations with chest Computed Tomography (CT) and CT angiogram confirmed right-sided pulmonary fibrosis and the absence of the right pulmonary artery, with no other significant cardiopulmonary symptoms reported. This case highlights the complexity of diagnosing and managing rare unilateral pulmonary conditions.
Collapse
Affiliation(s)
- Jinyong Bae
- Radiology, Brooke Army Medical Center, San Antonio, USA
| |
Collapse
|
30
|
Jung JH, Kang SA, Park JH, Kim SD, Yu HS, Mun SJ, Cho KS. Immunomodulatory Effect of Adipose Stem Cell-Derived Extra-Cellular Vesicles on Cytokine Expression and Regulatory T Cells in Patients with Asthma. Int J Mol Sci 2024; 25:10524. [PMID: 39408853 PMCID: PMC11477288 DOI: 10.3390/ijms251910524] [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: 08/16/2024] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Although mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are as effective as MSCs in the suppression of allergic airway inflammation, few studies have evaluated the immunomodulatory capacity of MSC-derived EVs in patients with asthma. Thus, we assessed the effects of adipose stem cell (ASC)-derived EVs on cytokine expression and regulatory T cells (Tregs) in peripheral blood mononuclear cells (PBMCs) of asthmatic patients. PBMCs (1 × 106 cells/mL) were isolated from asthmatic patient and healthy controls and co-cultured with 1 μg/mL of ASC-derived EVs. Th (T helper) 1-, Th2-, and Treg-related cytokine expression, fluorescence-activated cell sorting analysis of CD4+CD25+FOXP3+ T cells, and co-stimulatory molecules were analyzed before and after ASC-derived EV treatment. The expression levels of IL-4 and costimulatory molecules such as CD83 and CD86 were significantly higher in PBMCs of asthmatic patients than in control PBMCs. However, ASC-derived EV treatment significantly decreased the levels of interleukin (IL)-4 and co-stimulatory molecules such as CD83 and CD86 in the phytohemagglutinin (PHA)-stimulated PBMC of asthmatic patients. Furthermore, ASC-derived EVs remarkably increased the transforming growth factor-β (TGF-β) levels and expression of Tregs in the PBMC of asthmatic patients. ASC-derived EVs induce Treg expansion and have immunomodulatory effects by downregulating IL-4 and upregulating TGF-β in PBMCs of asthmatic patients.
Collapse
Affiliation(s)
- Jae Hoon Jung
- Department of Otorhinolaryngology, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Shin Ae Kang
- Department of Environmental Medical Biology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea;
| | - Ji-Hwan Park
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| | - Sung-Dong Kim
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| | - Hak Sun Yu
- Department of Parasitology and Tropical medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Sue Jean Mun
- Department of Otorhinolaryngology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| |
Collapse
|
31
|
Ghobadi H, Aslani F, Boskabady MH, Saadat S, Aslani MR. Saffron ( Crocus sativus) and its constituents in ovalbumin-induced asthma model: a preclinical systematic review and meta-analysis. Front Pharmacol 2024; 15:1436295. [PMID: 39295946 PMCID: PMC11408349 DOI: 10.3389/fphar.2024.1436295] [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: 05/22/2024] [Accepted: 08/01/2024] [Indexed: 09/21/2024] Open
Abstract
Background Animal and human studies have demonstrated that the saffron and the active components of saffron, including crocin, crocetin, and safranal, possess anti-inflammatory, antioxidant, and immunomodulatory properties. In this meta-analysis, the preclinical evidence and potential mechanism of saffron were explored in an animal model of ovalbumin-induced asthma. Methods Studies related to saffron and its constituents in an animal model of ovalbumin-induced asthma from the beginning to March 2024 were searched from Scopus, PubMed, and Web of Science databases. The methodological quality of the studies was evaluated using the 15-item CAMARADES checklist. Data analysis was performed using STATA software version 17. Results Thirteen studies with 536 animals (268 animals in the intervention group and 268 animals in the ovalbumin-induced group) were analyzed. The meta-analysis findings demonstrated that saffron and its constituents played a significant role in reducing total WBC, eosinophil, lymphocyte, and monocyte counts. Moreover, saffron showed a significant decrease in the levels of IL-4, IL-5, IL-13, IgE, histamine, endothelin, nitric oxide, and nitrite. Moreover, saffron was found to elevate EC50 thresholds and lower maximum response rates in experimental animals. The analysis revealed a significant identification of modulation in endoplasmic reticulum (ER) stress markers and miRNAs pathways. Conclusion Saffron and its components may impact ovalbumin-induced asthma model in animals through anti-inflammatory, antioxidant, and immunomodulatory pathways, as well as improving pulmonary function and modulating ER stress markers and miRNAs pathways. As a result, saffron should be considered for further clinical trials in individuals suffering from asthma.
Collapse
Affiliation(s)
- Hassan Ghobadi
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farzin Aslani
- Department of Orthopedics, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saeideh Saadat
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
32
|
Huang CY, Wu YK, Yang MC, Huang KL, Su WL, Huang YC, Chih-Wei W, Tzeng IS, Lan CC. Assessing post-COVID-19 respiratory dynamics: a comprehensive analysis of pulmonary function, bronchial hyperresponsiveness and bronchodilator response. ERJ Open Res 2024; 10:00149-2024. [PMID: 39377091 PMCID: PMC11456966 DOI: 10.1183/23120541.00149-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/01/2024] [Indexed: 10/09/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) has a considerable impact on the global healthcare system. Individuals who have recovered from COVID often experience chronic respiratory symptoms that affect their daily lives. This study aimed to assess respiratory dynamics such as airway hyperresponsiveness (AHR) and bronchodilator response in post-COVID patients. Methods This study included 282 adults with respiratory symptoms who underwent provocation tests. The demographic details, clinical symptoms and medical histories were recorded. Baseline spirometry, methacholine challenge tests (MCT) and post-bronchodilator spirometry were performed. Patients were divided into the following four groups: Group 1: non-COVID-19 and negative MCT; Group 2: post-COVID-19 and negative MCT; Group 3: non-COVID-19 and positive MCT; and Group 4: post-COVID-19 and positive MCT. Results Most post-COVID-19 patients (43.7%) experienced AHR, and wheezing was more common. Patients in Group 4 exhibited increased intensities of dyspnoea, cough and wheezing with the lowest pulmonary function test (PFT) parameters at baseline. Moreover, significant decreases in PFT parameters after the MCT were observed in these patients. Although the prevalence of a low forced expiratory volume in 1 s to forced vital capacity ratio (<70%) was initially 2% in Group 4, it increased to 29% after MCT. No significant differences in allergic history or underlying diseases were observed between the groups. Conclusions These findings provide comprehensive insights into the AHR and respiratory symptoms of post-COVID-19 individuals, highlighting the characteristics and potential exacerbations in patients with positive MCT results. This emphasises the need of MCT to address respiratory dynamics in post-COVID-19 individuals.
Collapse
Affiliation(s)
- Chun-Yao Huang
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yao-Kuang Wu
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Mei-Chen Yang
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kuo-Liang Huang
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Lin Su
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yi-Chih Huang
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wu Chih-Wei
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Chou-Chin Lan
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
33
|
Harrell CR, Djonov V, Volarevic A, Arsenijevic A, Volarevic V. Molecular Mechanisms Responsible for the Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes in the Treatment of Lung Fibrosis. Int J Mol Sci 2024; 25:4378. [PMID: 38673961 PMCID: PMC11050301 DOI: 10.3390/ijms25084378] [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: 02/03/2024] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) are nano-sized extracellular vesicles which contain various MSC-sourced anti-fibrotic, immunoregulatory and angio-modulatory proteins (growth factors, immunoregulatory cytokines, chemokines), lipids, and nucleic acids (messenger RNA and microRNAs). Due to their lipid envelope, MSC-Exos easily by-pass all barriers in the body and deliver their cargo directly in target cells, modulating their viability, proliferation, phenotype and function. The results obtained in recently published experimental studies demonstrated beneficial effects of MSC-Exos in the treatment of lung fibrosis. MSC-Exos reduced activation of fibroblasts and prevented their differentiation in myofibroblasts. By delivering MSC-sourced immunoregulatory factors in lung-infiltrated monocytes and T cells, MSC-Exos modulate their function, alleviating on-going inflammation and fibrosis. MSC-Exos may also serve as vehicles for the target delivery of anti-fibrotic and immunomodulatory agents, enabling enhanced attenuation of lung fibrosis. Although numerous pre-clinical studies have demonstrated the therapeutic potential of MSC-Exos in the treatment of pulmonary fibrosis, there are several challenges that currently hinder their clinical implementation. Therefore, in this review article, we summarized current knowledge and we discussed future perspectives regarding molecular and cellular mechanisms which were responsible for the anti-fibrotic, anti-inflammatory and immunoregulatory properties of MSC-Exos, paving the way for their clinical use in the treatment of lung fibrosis.
Collapse
Affiliation(s)
- Carl Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N, Palm Harbor, FL 34684, USA;
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland;
| | - Ana Volarevic
- Department of Psychology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
| | - Aleksandar Arsenijevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia
| | - Vladislav Volarevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia
- Faculty of Pharmacy Novi Sad, Trg Mladenaca 5, 21000 Novi Sad, Serbia
| |
Collapse
|
34
|
Zhuo Z, Nie J, Xie B, Wang F, Shi M, Jiang Y, Zhu W. A comprehensive study of Ephedra sinica Stapf-Schisandra chinensis (Turcz.) Baill herb pair on airway protection in asthma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117614. [PMID: 38113990 DOI: 10.1016/j.jep.2023.117614] [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: 11/04/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ephedra sinica Stapf (Mahuang) and Schisandra chinensis (Turcz.) Baill (Wuweizi) are commonly utilized in traditional Chinese medicine for the treatment of cough and asthma. The synergistic effect of Mahuang-Wuweizi herb pair enhances their efficacy in alleviating respiratory symptoms, making them extensively employed in the management of respiratory disorders. Although previous studies have demonstrated the therapeutic potential of Mahuang-Wuweizi in pulmonary fibrosis, the precise mechanism underlying their effectiveness against asthma remains elusive. AIM OF THE STUDY The objective of this study is to investigate the mechanism underlying the preventive and therapeutic effects of Mahuang-Wuweizi herb pair on asthma progression, focusing on airway inflammation and airway remodeling. MATERIALS AND METHODS The active constituents and potential mechanisms of Mahuang-Wuweizi in the management of asthma were elucidated through network pharmacology analysis. Liquid chromatography tandem mass spectrometry (LC-MS/MS) was used to detect the main components of Mahuang-Wuweizi decoction. A rat model of bronchial asthma was established, and the effects of Mahuang-Wuweizi were investigated using hematoxylin-eosin (HE) staining, immunohistochemistry (IHC) staining, enzyme-linked immunosorbent assay (ELISA), Western blotting (WB), and real-time reverse transcription polymerase chain reaction (RT-qPCR). RESULTS The results of network pharmacological prediction showed that Mahuang had 22 active components and Wuweizi had 8 active components, with 225 potential targets. 1159 targets associated with asthma and 115 targets that overlap between drugs and diseases were identified. These include interleukin-6 (IL-6), tumor necrosis factor (TNF), Tumor Protein 53, interleukin-1β (IL-1β), as well as other essential targets. Additionally, there is a potential correlation between asthma and Phosphatidylinositol 3 kinase (PI3K)/Protein Kinase B (AKT) signaling pathway, calcium ion channels, nuclear factor-kappa B (NF-κB) signaling pathway, and other signaling pathways. The animal experiment results demonstrated that treatment with Mahuang and Wuweizi, in comparison to the model group, exhibited improvements in lung tissue pathological injury, reduction in collagen fiber accumulation around the airway and proliferation of airway smooth muscle, decrease in concentration levels of IL-6, TNF-α and IL-1β in lung tissue, as well as alleviation of airway inflammation. Furthermore, Mahuang and Wuweizi suppressed the expression of phospholipase C (PLC), transient receptor potential channel 1 (TRPC1), myosin light chain kinase (MLCK), NF-κB P65 protein in ovalbumin (OVA)-sensitized rat lung tissue and downregulated the mRNA expression of PLC, TRPC1, PI3K, AKT, NF-κB P65 in asthmatic rats. These findings were consistent with network pharmacological analysis. CONCLUSION The results show that the synergistic interaction between Mahuang and Wuweizi occur, and they can effectively reduce airway remodeling and airway inflammation induced by inhaling OVA in bronchial asthma rats by inhibiting the expression of PLC/TRPC1/PI3K/AKT/NF-κB signaling pathway. Therefore, Mahuang and Wuweizi may be potential drugs to treat asthma.
Collapse
Affiliation(s)
- Zushun Zhuo
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Jianhua Nie
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Bin Xie
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Fei Wang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Min Shi
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Yini Jiang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Weifeng Zhu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| |
Collapse
|