1
|
Tao P, Su B, Mao X, Lin Y, Zheng L, Zou X, Yang H, Liu J, Li H. Interleukin-35 inhibits NETs to ameliorate Th17/Treg immune imbalance during the exacerbation of cigarette smoke exposed-asthma via gp130/STAT3/ferroptosis axis. Redox Biol 2025; 82:103594. [PMID: 40101533 PMCID: PMC11964675 DOI: 10.1016/j.redox.2025.103594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
Cigarette smoke (CS) exposure amplifies neutrophil accumulation. IL-35, a novel cytokine with anti-inflammatory properties, is involved in protection against asthma. However, the biological roles of neutrophils and the precise molecular mechanisms of IL-35 in CS exposed-asthma remain unclear. We showed that the exacerbation of CS exposed-asthma leads to dramatically increased neutrophil counts and an imbalance in DC-Th17/Treg immune responses. RNA sequencing revealed that NETs, part of a key biological process in neutrophils, were significantly upregulated in the context of CS exposed-asthma exacerbation and that IL-35 treatment downregulated NET-associated gene expression. Targeted degradation of NETs, rather than neutrophil depletion, alleviated the CS exposed-asthma. Mechanistically, STAT3 phosphorylation promoted ferroptosis, exacerbating NET release, which in turn enhanced dendritic cell (DC) antigen presentation, activated T cells, and specifically promoted Th17 cell differentiation while inhibiting Treg cells. IL-35 acting on the gp130 receptor alleviated STAT3-mediated ferroptosis-associated NET formation. In summary, our study revealed a novel mechanism by which IL-35 inhibited NET formation, subsequently alleviating neutrophilic inflammation and restoring the DC-Th17/Treg imbalance in CS exposed-asthma, highlighting the potential of IL-35 as a targeted therapeutic strategy.
Collapse
Affiliation(s)
- Peizhi Tao
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Beiting Su
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xueyan Mao
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yusen Lin
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Zheng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hailing Yang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Liu
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Hongtao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
2
|
Guixing X, Yilin L, Huaying F, Fanrong L, Dehua L. Effect of the Antibody-mediated Immune Responses on COPD, Asthma, and Lung Function: A Mendelian Randomization Study. Arch Bronconeumol 2025; 61:212-219. [PMID: 39489629 DOI: 10.1016/j.arbres.2024.10.003] [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: 07/18/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION The precise cause of antibody-mediated immune responses on chronic obstructive pulmonary disease (COPD), asthma, and lung function remains unclear. We characterized the relationship between antibody-mediated immune responses to COPD, asthma, and lung function, ultimately achieve the prevention or treatment. METHODS We obtained summary data from published genome-wide association studies, including antibody-mediated immune responses, COPD, asthma, forced expiratory volume in the first second (FEV1), forced expiratory volume (FVC), and FEV1/FVC. Bidirectional two-sample mendelian randomization (MR) analysis was used to assess causal relationships of antibody-mediated immune responses, COPD, asthma, FEV1, FVC, and FEV1/FVC. RESULTS A total of 20 antibody-mediated immune responses were identified have a significant causal effect on COPD, asthma, FEV1, and FVC, with six exhibiting reverse causality. Importantly, the results of the five MR analyses were almost identical with respect to the causal effect of anti-polyomavirus 2 IgG seropositivity and varicella zoster virus glycoprotein E and I antibody levels on the risk of COPD, asthma, FEV1, and FVC. CONCLUSIONS This study contributes to existing knowledge by investigating the causal relationship between antibody-mediated immune responses and respiratory conditions, including COPD, asthma, and lung function, using a two-sample MR design. The key findings can aid in identifying individuals at risk of these conditions and facilitate early prevention and diagnosis.
Collapse
Affiliation(s)
- Xu Guixing
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Liu Yilin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine Chinese Medicine, Chengdu, Sichuan, China
| | - Fan Huaying
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Liang Fanrong
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine Chinese Medicine, Chengdu, Sichuan, China
| | - Li Dehua
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Toumpanakis D, Bartziokas K, Bakakos A, Fouka E, Bakakos P, Loukides S, Steiropoulos P, Papaioannou AI. Monoclonal Antibodies for the Treatment of Chronic Obstructive Pulmonary Disease. Pulm Ther 2025:10.1007/s41030-025-00291-5. [PMID: 40123030 DOI: 10.1007/s41030-025-00291-5] [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: 12/27/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common and complex disease characterized by persistent airflow limitation and the presence of exacerbations, resulting in significant morbidity and mortality. Although the pathogenesis of COPD is multifactorial, airway inflammation plays a significant role in disease progression. Despite the advantages of non-pharmaceutical and pharmaceutical interventions that have significantly improved the symptom burden and exacerbation frequency in COPD, there is a lack of disease-modifying therapies that target the underlying disease mechanisms. Monoclonal antibodies (mAbs), a drug class that has improved treatment in severe asthma by blocking mediators of the type 2 (Th2) and allergic inflammatory cascades, are currently under investigation for their efficacy in COPD. Our review summarizes the evidence for the use of monoclonal antibodies in COPD and discusses current limitations and promising advances. Although targeting Th1 inflammation has failed to improve COPD outcomes, recent clinical trials have shown beneficial effects of monoclonal antibodies targeting Th2 inflammation, providing evidence for a personalized approach in COPD treatment.
Collapse
Affiliation(s)
- Dimitrios Toumpanakis
- 2Nd Department of Critical Care, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Konstantinos Bartziokas
- 2Nd Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Agamemnon Bakakos
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Evangelia Fouka
- Respiratory Medicine Department, Aristotle University of Thessaloniki, G Papanikolaou Hospital, Thessaloniki, Greece
| | - Petros Bakakos
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Stelios Loukides
- 2Nd Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece.
| | - Andriana I Papaioannou
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| |
Collapse
|
4
|
Cao S, Gu Y, Lu G, Zhu L, Feng S, Bian T. Causal Correlations Between Plasma Metabolites, Inflammatory Proteins, and Chronic Obstructive Pulmonary Disease: A Mendelian Randomization and Bioinformatics-Based Investigation. J Inflamm Res 2025; 18:4057-4073. [PMID: 40125086 PMCID: PMC11929513 DOI: 10.2147/jir.s513526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Background An increasing number of studies have demonstrated a strong correlation between metabolism, inflammation, and chronic obstructive pulmonary disease (COPD). However, it remains unclear if there is a causal relationship between these factors. This study employed the Mendelian randomization (MR) approach to investigate the associations between these factors and explore the mediating roles of key inflammatory proteins. Methods MR was used to assess the causal associations between plasma metabolites, inflammatory proteins, and COPD. Sensitivity analyses were performed to verify the robustness of the findings. Mediation analysis was conducted to explore the roles of inflammatory proteins in the metabolism-COPD pathway. We constructed protein-protein interaction (PPI) network and explored the potential mechanism through gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Single-cell sequencing and transcriptome datasets were used for auxiliary validation. Finally, experimental validation was performed using human lung tissue. Results This study identified 63 metabolites, 10 metabolite ratios, and 48 inflammatory proteins that were associated with COPD, all of which exhibited potential causal relationships. Furthermore, three proteins were identified as mediators in the metabolite-to-COPD pathway. PPI network, GO and KEGG enrichment analysis revealed the biological pathways in which they were involved. Validation of the expression of these three intermediary proteins in lung tissue demonstrated that NRXN3 was expressed in pulmonary endothelial cells and exerted a protective effect against COPD development. Conclusion The MR analysis revealed causal associations among metabolism, inflammation, and COPD. These findings offer novel insights into metabolism-inflammation-COPD mechanisms, suggesting that interventions targeting metabolic processes may represent a promising strategy for preventing the onset or progression of COPD.
Collapse
Affiliation(s)
- Shurui Cao
- Department of Respiratory Medicine, Wuxi People’s Hospital, Wuxi Medical Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Yongqi Gu
- Department of Respiratory Medicine, Wuxi People’s Hospital, Wuxi Medical Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Guye Lu
- Department of Respiratory Medicine, Wuxi People’s Hospital, Wuxi Medical Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Lizhen Zhu
- Department of Respiratory Medicine, Wuxi People’s Hospital, Wuxi Medical Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Shumin Feng
- Department of Respiratory Medicine, Wuxi People’s Hospital, Wuxi Medical Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Tao Bian
- Department of Respiratory Medicine, Wuxi People’s Hospital, Wuxi Medical Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| |
Collapse
|
5
|
Zeng Z, Liu X, Xiang F, He X, Li J, Liu H, Xie L. MicroRNA-21 plays a role in exacerbating chronic obstructive pulmonary disease by regulating necroptosis and apoptosis in bronchial epithelial cells. Tob Induc Dis 2025; 23:TID-23-32. [PMID: 40104397 PMCID: PMC11915094 DOI: 10.18332/tid/202182] [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: 03/27/2024] [Revised: 10/31/2024] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
INTRODUCTION Bronchial epithelial cell damage is an important determinant of the severity of chronic obstructive pulmonary (COPD). However, the exact molecular mechanisms underlying this cell death in COPD development are not well understood. This study investigates the involvement of microRNA-21 (miR-21/miRNA-21) in COPD and its underlying molecular mechanism. METHODS A mouse model of COPD was created by exposing the mice to cigarette smoke (CS) and injecting them with cigarette smoke extract (CSE). Both wild-type mice and miR-21 knockout (miR-21-/-) mice were used to investigate the role of microRNA-21 (miR-21) in exacerbating COPD. Various assays and analyses were performed, including HE staining, tunel staining, enzyme-linked immunosorbent assay (ELISA), flow cytometry, quantitative real-time polymerase chain reaction (RT-qPCR), and western blotting (WB) to measure outcomes such as the pathological morphological changes, necroptosis, apoptosis, and levels of inflammatory factors. RESULTS Our results revealed an upregulation of miR-21 in the lung tissue of COPD model mice. Additionally, knockout of miR-21 resulted in decreased levels of bronchial epithelial cell necroptosis and apoptosis, as evidenced by the downregulation of tumor necrosis factor receptor 1 (TNFR1), phosphoryl-mixed lineage kinase domain-like protein (p-MLKL) and caspase-3. This downregulation of necroptosis and apoptosis ultimately led to a reduction of inflammatory factors and damage-associated molecular patterns (DAMPs), such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL- 1β), and interleukin-6 (IL-6) and high mobility group protein B1(HMGB1) in the lungs, thereby ameliorating COPD. CONCLUSIONS Our findings suggest that miR-21 contributes to the worsening of chronic obstructive pulmonary disease by modulating necroptosis and apoptosis in bronchial epithelial cells, providing a new theoretical basis for the pathogenesis of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Zhengpeng Zeng
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xuelian Liu
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Fei Xiang
- Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xue He
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiahui Li
- Changsha Kexin Cancer Hospital, Changsha, China
| | - Hanying Liu
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Xie
- Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Pham VD, Lee JH, Shin D, Vu HM, Jung J, Kashyap MK, Lee SH, Kim MS. On the Ocean of Biomarkers for the Precise Diagnosis and Prognosis of Lung Diseases. Proteomics Clin Appl 2025:e70003. [PMID: 40098318 DOI: 10.1002/prca.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/19/2025]
Abstract
Bronchoalveolar lavage fluid (BALF) has long been used for diagnosing various lung diseases through its cellular components. However, the clinical utility of biomolecules in the BALF remains largely unexplored. Recently, mass spectrometry-based proteomics has been applied to profile the BALF proteomes to identify novel biomarkers for lung diseases. This review discusses the current progress in the field of BALF proteomics and highlights its potential as a valuable source of biomarkers for different lung diseases. Additionally, we explored the latest advancements and findings from BALF studies. Finally, we address the current limitations and propose future directions and research opportunities to advance the study of BALF.
Collapse
Affiliation(s)
- Van Duc Pham
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Jung-Hyung Lee
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Doyun Shin
- Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Hung M Vu
- Bertis R&D Division, Bertis Inc., Gwacheon-si, Gyeonggi-do, Republic of Korea
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Junyang Jung
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Manoj K Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India
| | - Seung Hyeun Lee
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Min-Sik Kim
- Department of New Biology, DGIST, Daegu, Republic of Korea
- New Biology Research Center, DGIST, Daegu, Republic of Korea
| |
Collapse
|
7
|
Tian M, Huang W, Chen J, Liu X, Wang H, Pan X, Wang L, Li Q, Gao L, Ye Y. The extract from Quzhou Aurantii Fructus attenuates cough variant asthma through inhibiting the TRPV1/Ca 2+/NFAT/TSLP pathway and ferroptosis via TRPV1 mediation in ovalbumin-induced mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119038. [PMID: 39510426 DOI: 10.1016/j.jep.2024.119038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/20/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cough variant asthma (CVA), a prevalent chronic inflammatory disease, is the most common cause of chronic cough. Over the years, the aqueous extract of Quzhou Aurantii Fructus (QAFA) has been widely used to treat respiratory diseases, particularly cough. AIM OF THE STUDY This study aimed to elucidate the therapeutic effect of QAFA on allergen-induced CVA, providing deep insights into the underlying mechanisms. MATERIALS AND METHODS Ultra-high performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) was employed to characterize the compositions, while UPLC was used to quantify the contents of its major components in QAFA. CVA model was established via sensitization and atomization with ovalbumin (OVA), and received 600 and 1200 mg/kg of QAFA via intragastric gavage. Cough response was assessed by stimulation with capsaicin (CAP). Then, airway hyperresponsiveness (AHR), ELISA, western blotting, RT-qPCR, and histological analyses, were applied to assess pulmonary function, pathological changes, and investigate mechanisms in CVA mice following QAFA treatment through the TRPV1/Ca2+-dependent NFAT-induced expression of TSLP and ferroptosis. Additionally, the effects and mechanisms of QAFA were validated using IL-4, CAP for stimulation, capsazepine (CPZ) for inhibition, and TRPV1 siRNA transfection in cells. RESULTS Chemical analysis revealed that QAFA primarily contained sixteen compounds, with four main components including narirutin, naringin, hesperidin, and neohesperidin. In vivo, QAFA treatment alleviated cough and AHR, while concurrently reducing airway inflammation and mucus secretion in CVA mice. These effects were achieved by suppressing the TRPV1/NFAT/TSLP pathway and modulating the expression of ferroptosis-related proteins. In vitro, siTRPV1-transfected BEAS-2B cells demonstrated the involvement of the TRPV1 channel in IL-4-mediated Ca2+ influxes, ferroptosis, and regulation of TSLP production. QAFA and CPZ suppressed IL-4-induced TSLP production via the TRPV1/NFAT pathway and regulated the levels of ferroptosis-related proteins, while CAP counteracted the effect of QAFA on TSLP production in BEAS-2B cells. Furthermore, QAFA reduced IL-4 or CAP induced Ca2+ influx and IL-4 induced ferroptosis through TRPV1 mediation. CONCLUSIONS This study demonstrated that QAFA improved pulmonary function and alleviated asthmatic inflammatory response in treating CVA probably through suppressing the TRPV1/Ca2+/NFAT/TSLP pathway and ferroptosis via TRPV1 mediation.
Collapse
Affiliation(s)
- Meizi Tian
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Wenkang Huang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Jiahui Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Xiaotong Liu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Haiou Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Xiaoya Pan
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Lixia Wang
- Changshan Characteristic Industry Development Center, Quzhou, Zhejiang, 324000, China
| | - Qin Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
| | - Lijuan Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China.
| | - Yiping Ye
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China.
| |
Collapse
|
8
|
Tanabe N, Matsumoto H, Morimoto C, Hirai T. Sputum short-chain fatty acids, microbiome, inflammation, and mucus plugging in obstructive airway disease. J Allergy Clin Immunol 2025:S0091-6749(25)00120-4. [PMID: 39914553 DOI: 10.1016/j.jaci.2025.01.031] [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: 11/28/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Short-chain fatty acids (SCFAs), produced by anaerobic bacteria through fermentation in the gut, may suppress eosinophilic inflammation while potentially promoting neutrophilic inflammation. However, the role of local SCFAs in the airway microbiome, inflammation, and mucus plugging in type 2-dominant obstructive airway diseases remains unclear. OBJECTIVE Our aim was to investigate associations between sputum SCFAs and the relative abundance of anaerobic bacteria, neutrophil and eosinophil counts in sputum, and mucus plug scores on computed tomography images in patients with obstructive airway diseases. METHODS Sputum samples and chest computed tomography images were prospectively collected in stable patients with asthma with fixed airflow limitation, chronic obstructive pulmonary disease, and asthma-chronic obstructive pulmonary disease overlap (ACO). Sputum samples were analyzed for concentrations of SCFA, including n-butyrate, acetate, and propionate; microbiome composition using 16S rRNA sequencing; and inflammatory cell differentials. RESULTS In 46 patients, enriched for ACO with relatively high levels of type 2 markers, higher SCFA levels were associated with higher relative abundance of bacteria of the phylum Bacteroidetes and lower relative abundance of bacteria of the phylum Proteobacteria. Hierarchic clustering identified a severe eosinophil-dominant inflammation cluster characterized by lower SCFAs levels and higher mucus plug scores. In the 2 neutrophilic clusters, one characterized by higher SCFAs levels and the other by lower SCFAs levels, lower butyrate levels were significantly associated with higher mucus plug scores. CONCLUSION Local SCFA concentrations may be closely associated with the airway microbiome and influence mucus plugging in ACO-enriched populations. Understanding these interactions could inform therapeutic strategies targeting SCFAs or the microbiome to manage type 2-dominant obstructive airway diseases.
Collapse
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Chie Morimoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
9
|
Liang Q, Wang Y, Li Z. Comprehensive bioinformatics analysis identifies metabolic and immune-related diagnostic biomarkers shared between diabetes and COPD using multi-omics and machine learning. Front Endocrinol (Lausanne) 2025; 15:1475958. [PMID: 39845878 PMCID: PMC11750655 DOI: 10.3389/fendo.2024.1475958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background Diabetes and chronic obstructive pulmonary disease (COPD) are prominent global health challenges, each imposing significant burdens on affected individuals, healthcare systems, and society. However, the specific molecular mechanisms supporting their interrelationship have not been fully defined. Methods We identified the differentially expressed genes (DEGs) of COPD and diabetes from multi-center patient cohorts, respectively. Through cross-analysis, we identified the shared DEGs of COPD and diabetes, and investigated alterations of signaling pathways using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA). By using weighted gene correlation network analysis (WGCNA), key gene modules for COPD and diabetes were identified, and various machine learning algorithms were employed to identify shared biomarkers. Using xCell, we investigated the relationship between shared biomarkers and immune infiltration in diabetes and COPD. Single-cell sequencing, clinical samples, and animal models were used to confirm the robustness of shared biomarkers. Results Cross-analysis identified 186 shared DEGs between diabetes and COPD patients. Functional enrichment results demonstrate that metabolic and immune-related pathways are common features altered in both diabetes and COPD patients. WGCNA identified 526 genes from key gene modules in COPD and diabetes. Multiple machine learning algorithms identified 4 shared biomarkers for COPD and diabetes, including CADPS, EDNRB, THBS4 and TMEM27. Finally, the 4 shared biomarkers were validated in single-cell sequencing data, clinical samples, and animal models, and their expression changes were consistent with the results of bioinformatic analysis. Conclusions Through comprehensive bioinformatics analysis, we revealed the potential connection between diabetes and COPD, providing a theoretical basis for exploring the common regulatory genes.
Collapse
Affiliation(s)
- Qianqian Liang
- Department of Integrated Pulmonology, Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yide Wang
- Department of Integrated Pulmonology, Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zheng Li
- Department of Integrated Pulmonology, Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
- Xinjiang National Clinical Research Base of Traditional Chinese Medicine, The Affiliated Hospital of Xinjiang University of Traditional Chinese Medicine, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Respiratory Disease Research, The Affiliated Hospital of Xinjiang University of Traditional Chinese Medicine, Urumqi, Xinjiang, China
- Xinjiang Clinical Medical Research Center of Respiratory Obstructive Diseases, The Affiliated Hospital of Xinjiang University of Traditional Chinese Medicine, Urumqi, Xinjiang, China
| |
Collapse
|
10
|
Gyawali B, Georas SN, Khurana S. Biologics in severe asthma: a state-of-the-art review. Eur Respir Rev 2025; 34:240088. [PMID: 39778920 PMCID: PMC11707604 DOI: 10.1183/16000617.0088-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025] Open
Abstract
Asthma is considered severe if it remains uncontrolled despite optimal conventional therapy, characterised by poor symptom control, frequent exacerbations and increased exposure to systemic corticosteroids. This has a significant impact on morbidity, mortality and healthcare resource utilisation. Recent advances in the understanding of asthma heterogeneity and immunopathogenesis have helped delineate precise disease pathways. The discovery of these pivotal pathways has led to the development of highly effective biologic therapies. Currently available asthma biologics target immunoglobulin E, interleukin (IL)-5/IL-5Rα, IL-4Rα and thymic stromal lymphopoietin. Identification of specific asthma phenotypes, utilising easily measurable biomarkers, has paved the way towards personalised and precision asthma management. Biologic therapies play a significant role in reducing exacerbations, hospitalisations and the need for maintenance systemic steroids, while also improving the quality of life in patients with severe asthma. The evidence for their clinical efficacy comes from randomised controlled trials (RCTs), extension studies, metanalyses and real-world data. This review synthesises findings from early, pivotal RCTs and subsequent studies following the approval of biologics for severe asthma. The safety and efficacy data from these studies, completed in a variety of settings, provide practical perspectives on their application and enhance their generalisability.
Collapse
Affiliation(s)
- Bishal Gyawali
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Mary Parkes Center for Asthma, Allergy and Pulmonary Care, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Sandhya Khurana
- Division of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Mary Parkes Center for Asthma, Allergy and Pulmonary Care, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
11
|
Shen TC. Molecular Mechanisms, Diagnoses, and Treatments of Respiratory Diseases. Biomedicines 2024; 13:4. [PMID: 39857587 PMCID: PMC11760475 DOI: 10.3390/biomedicines13010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
The Special Issue "Molecular Mechanisms, Diagnoses, and Treatments of Respiratory Diseases" in the journal Biomedicines compiles critical advancements in the understanding of respiratory diseases, focusing on their molecular mechanisms, diagnostic approaches, and therapeutic strategies [...].
Collapse
Affiliation(s)
- Te-Chun Shen
- School of Medicine, China Medical University, Taichung 404, Taiwan; ; Tel.: +886-4-22052121
- Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
- Department of Internal Medicine, Chu Shang Show Chwan Hospital, Nantou 557, Taiwan
| |
Collapse
|
12
|
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
|
13
|
Elawad S, Saad-Omer SI, Elawad SOM, Elbadawi MH, Ahmed SA, Bashir SB, Abdelmutaleb SA, Mohamed SO, Babiker SB, Osman SM, Fadul MA, Abdalkareem EO, M. Ahmed AB, Galal A. Knowledge, attitude, and practice of asthma among the adults in Shendi locality, Sudan: A cross-sectional study. Medicine (Baltimore) 2024; 103:e40395. [PMID: 39496011 PMCID: PMC11537662 DOI: 10.1097/md.0000000000040395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Asthma is a chronic inflammatory condition leading to intermittent airway narrowing and obstruction. Although millions suffer from asthma globally, there is limited data from African countries, particularly Sudan. This study addresses the gap in research on asthma knowledge, attitudes, and practices among the adults in Shendi locality, Northern Sudan. This cross-sectional, community-based study was conducted in Kabushiya village, Shendi, Sudan, involving 148 participants selected through multistage random sampling. Data were collected via face-to-face interviews using a validated, structured questionnaire. Statistical analyses included Mann-Whitney, Kruskal-Wallis, Chi-square, Fisher exact tests, and multiple linear regression. Participants demonstrated high levels of knowledge, positive attitudes, and good practices regarding asthma. Knowledge scores were significantly associated with marital status, education, occupation, and income (P values: .040, .003, .000, and .006, respectively), with the highest medians among divorced individuals, those with higher education, professional workers, and people earning <50,000 SDGs (medians: 17, 16, 17, and 16, respectively). Attitude scores varied significantly by marital status, education, occupation, and family asthma history (P values: .001, .000, .003, and .016, respectively), with highest scores among divorced individuals, those with high school or higher education, professionals, and those with a first-degree relative with asthma (medians: 6, 5, 5, and 5, respectively). Patient practice scores differed by marital status and income (P values: .026 and .006, respectively), with higher scores among singles and those earning <50,000 SDGs. Co-patient practice scores were significantly influenced by occupation, personal or familial asthma experience, and family asthma status (P values: .026, .003, and .000, respectively). No significant differences were observed in co-patient practice scores between groups. The study highlights a generally good level of asthma knowledge and positive attitudes among participants, with some variability in practices based on socio-demographic factors. These findings emphasize the need for targeted education and intervention programs to improve asthma management in rural Sudan.
Collapse
Affiliation(s)
- Sara Elawad
- University of Khartoum, Faculty of Medicine, Khartoum, Sudan
| | | | | | | | | | - Sara B. Bashir
- University of Khartoum, Faculty of Medicine, Khartoum, Sudan
| | | | | | - Suad B. Babiker
- University of Khartoum, Faculty of Medicine, Khartoum, Sudan
| | - Sara M.S. Osman
- University of Khartoum, Faculty of Medicine, Khartoum, Sudan
| | | | | | | | - Ali Galal
- University of Khartoum, Faculty of Medicine, Khartoum, Sudan
| |
Collapse
|
14
|
Desai B, Adrish M, Mohan A, Lugogo NL. Biologics in Asthma: Emerging Biologics. Immunol Allergy Clin North Am 2024; 44:751-763. [PMID: 39389722 DOI: 10.1016/j.iac.2024.07.008] [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] [Indexed: 10/12/2024]
Abstract
Advances in our understanding of asthma pathophysiology have led to the advent of multiple targeted asthma therapies such as biologics. However, partial response to biologics occurs, indicating residual disease activity in some patients. Hence, there exists a need for new therapies that focus on novel pathways, alongside perhaps evaluation of combination biologic therapies and modulators of downstream cytokine activation. Therefore, although our current focus is on biologics; it is critical to take a more holistic approach including consideration for nonbiologic therapies that have the potential to significantly advance asthma care.
Collapse
Affiliation(s)
- Brinda Desai
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Muhammad Adrish
- Department of Pulmonary & Critical Care, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Arjun Mohan
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2d21, Ann Arbor, MI 48109, USA
| | - Njira L Lugogo
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2c40, Ann Arbor, MI 48109, USA.
| |
Collapse
|
15
|
Zhang Y, Chen L, Ouyang H. Shikonin alleviates asthma phenotypes in mice via an airway epithelial STAT3-dependent mechanism. Open Med (Wars) 2024; 19:20241016. [PMID: 39444792 PMCID: PMC11497215 DOI: 10.1515/med-2024-1016] [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: 02/19/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 10/25/2024] Open
Abstract
Background Asthma is an inflammatory disease where the balance between Th1/Th2 and Th17/Treg plays a crucial role in its pathogenesis. Shikonin is used to treat a variety of autoimmune diseases due to its good anti-inflammatory activity. However, the effect and mechanism of shikonin on asthma remain unknown. Method Mice were sensitized with ovalbumin (OVA)/house dust mite (HDM) and treated with shikonin. Lung inflammation was assessed histologically and via flow cytometry. Bronchoalveolar lavage fluid (BALF) was analyzed for cell counts and cytokines. Shikonin's impact on p-STAT3 was studied in vivo and in vitro. Results Shikonin inhibited OVA or HDM-induced inflammation and airway hyperresponsiveness. Upon treatment, a restoration of the Th1/Th2 and Th17/Treg balance was observed, evidenced by a reduction in IL-4 and IL-17A levels in BALF, alongside an elevation in interferon-gamma and IL-10. Furthermore, shikonin impeded the infiltration of eosinophils, neutrophils, macrophages, and lymphocytes into lung tissue. The observed decrease in STAT3 phosphorylation and diminished nuclear translocation of p-STAT3 confirmed that shikonin promotes the balance of Th1/Th2 and Th17/Treg by regulating airway epithelial STAT3. Conclusion Shikonin mitigates asthma symptoms through a STAT3-dependent mechanism, indicating its potential as an anti-asthmatic therapeutic agent.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Respiratory Medicine, Xi’an International Medical Center Hospital, Xi’an, 710101, China
| | - Lizhan Chen
- Department of Respiratory Medicine, Xi’an International Medical Center Hospital, Xi’an, 710101, China
| | - Haifeng Ouyang
- Department of Respiratory Medicine, Xi’an International Medical Center Hospital, No. 777 Xitai Road, Xi’an, 710101, China
| |
Collapse
|
16
|
Yu Q, Cai Z, Liu X, Lin S, Li P, Ruan Y, Liang J, He X, Lin W. Research Progress on Treating Spinal Cord Injury by Modulating the Phenotype of Microglia. J Integr Neurosci 2024; 23:171. [PMID: 39344231 DOI: 10.31083/j.jin2309171] [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: 03/30/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 10/01/2024] Open
Abstract
Spinal cord injury (SCI) is a severe central nervous system disorder with no currently available effective treatment. Microglia are immune cells in the central nervous system that play crucial roles in the SCI occurrence, development, and recovery stages. They exhibit dynamic polarization over time and can switch between classical activation (M1) and alternative activation (M2) phenotypes to respond to environmental stimuli. The M1 phenotype is involved in initiating and sustaining inflammatory responses, while the M2 phenotype exerts anti-inflammatory effects and promotes tissue repair in damaged areas. Inhibiting M1 polarization and promoting M2 polarization have become hotspots in regulating neuroinflammation and treating SCI. This article provides a comprehensive review centered on modulating microglial polarization phenotypes for SCI treatment.
Collapse
Affiliation(s)
- Qinghe Yu
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| | - Ziming Cai
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| | - Xiaofeng Liu
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| | - Shuhui Lin
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| | - Pian Li
- Department of The Third School of Clinical Medicine, Guangzhou University of Chinese Medicine, 510006 Guangzhou, Guangdong, China
| | - Ye Ruan
- Department of The Third School of Clinical Medicine, Guangzhou University of Chinese Medicine, 510006 Guangzhou, Guangdong, China
| | - Jinzhu Liang
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| | - Xu He
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| | - Wenping Lin
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, 518118 Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Pelaia C. Interleukin 33: a suitable target for biological therapies of COPD? ERJ Open Res 2024; 10:00433-2024. [PMID: 39319048 PMCID: PMC11417601 DOI: 10.1183/23120541.00433-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 09/26/2024] Open
Abstract
Because of the relevant importance of the IL-33/ST2 axis in COPD pathobiology, the future results of AERIFY-1 and AERIFY-2 trials could disclose new information about subgroups of responder patients to biologic therapies for this highly prevalent disease https://bit.ly/3USJCUP.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Medical and Surgical Sciences, University “Magna Graecia”, Catanzaro, Italy
| |
Collapse
|
18
|
Schlecht-Louf G, Federici S, Ryffel B, Russo RC. Editorial: The chemokine system in epithelial immunity. Front Immunol 2024; 15:1481686. [PMID: 39267756 PMCID: PMC11390664 DOI: 10.3389/fimmu.2024.1481686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Affiliation(s)
| | - Sara Federici
- Systems Immunology Department, Weizmann Institute of Science Rehovot, Rehovot, Israel
| | - Bernhard Ryffel
- Immuno-Neuro Modulation-INEM National Centre for Scientific Research (CNRS-UMR7355) and University of Orleans, Orleans, France
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
19
|
Zhang Y, Wu K, Mao D, Iberg CA, Yin-Declue H, Sun K, Wikfors HA, Keeler SP, Li M, Young D, Yantis J, Crouch EC, Chartock JR, Han Z, Byers DE, Brody SL, Romero AG, Holtzman MJ. A first-in-kind MAPK13 inhibitor that can correct stem cell reprogramming and post-injury disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608990. [PMID: 39229202 PMCID: PMC11370402 DOI: 10.1101/2024.08.21.608990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The stress kinase MAPK13 (aka p38δ-MAPK) is an attractive entry point for therapeutic intervention because it regulates the structural remodeling that can develop after epithelial barrier injury in the lung and likely other tissue sites. However, a selective, safe, and effective MAPK13 inhibitor is not yet available for experimental or clinical application. Here we identify a first-in-kind MAPK13 inhibitor using structure-based drug design combined with a screening funnel for cell safety and molecular specificity. This inhibitor (designated NuP-4) down-regulates basal-epithelial stem cell reprogramming, structural remodeling, and pathophysiology equivalently to Mapk13 gene-knockout in mouse and mouse organoid models of post-viral lung disease. This therapeutic benefit persists after stopping treatment as a sign of disease modification and attenuates key aspects of inflammation and remodeling as an indication of disease reversal. Similarly, NuP-4 treatment can directly control cytokine-stimulated growth, immune activation, and mucinous differentiation in human basal-cell organoids. The data thereby provide a new tool and potential fix for long-term stem cell reprogramming after viral injury and related conditions that require MAPK13 induction-activation.
Collapse
Affiliation(s)
- Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Ming Li
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joshua R Chartock
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenfu Han
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
20
|
Sweef O, Mahfouz R, Taşcıoğlu T, Albowaidey A, Abdelmonem M, Asfar M, Zaabout E, Corcino YL, Thomas V, Choi ES, Furuta S. Decoding LncRNA in COPD: Unveiling Prognostic and Diagnostic Power and Their Driving Role in Lung Cancer Progression. Int J Mol Sci 2024; 25:9001. [PMID: 39201688 PMCID: PMC11354875 DOI: 10.3390/ijms25169001] [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: 07/01/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer represent formidable challenges in global health, characterized by intricate pathophysiological mechanisms and multifaceted disease progression. This comprehensive review integrates insights from diverse perspectives to elucidate the intricate roles of long non-coding RNAs (lncRNAs) in the pathogenesis of COPD and lung cancer, focusing on their diagnostic, prognostic, and therapeutic implications. In the context of COPD, dysregulated lncRNAs, such as NEAT1, TUG1, MALAT1, HOTAIR, and GAS5, emerge as pivotal regulators of genes involved in the disease pathogenesis and progression. Their identification, profiling, and correlation with the disease severity present promising avenues for prognostic and diagnostic applications, thereby shaping personalized disease interventions. These lncRNAs are also implicated in lung cancer, underscoring their multifaceted roles and therapeutic potential across both diseases. In the domain of lung cancer, lncRNAs play intricate modulatory roles in disease progression, offering avenues for innovative therapeutic approaches and prognostic indicators. LncRNA-mediated immune responses have been shown to drive lung cancer progression by modulating the tumor microenvironment, influencing immune cell infiltration, and altering cytokine production. Their dysregulation significantly contributes to tumor growth, metastasis, and chemo-resistance, thereby emphasizing their significance as therapeutic targets and prognostic markers. This review summarizes the transformative potential of lncRNA-based diagnostics and therapeutics for COPD and lung cancer, offering valuable insights into future research directions for clinical translation and therapeutic development.
Collapse
Affiliation(s)
- Osama Sweef
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Reda Mahfouz
- Core Laboratory, University Hospital Cleveland Medical Center, Department of Pathology, School of Medicine, Case Western Reserve University, 1100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Clinical Pathology, Faculty of Medicine, Menofia University, Shebin-Elkom 32511, Egypt
| | - Tülin Taşcıoğlu
- Department of Molecular Biology and Genetics, Demiroglu Bilim University, Esentepe Central Campus, Besiktas, 34394 Istanbul, Turkey
| | - Ali Albowaidey
- The Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Mohamed Abdelmonem
- Department of Pathology, Transfusion Medicine Service, Stanford Healthcare, Stanford, CA 94305, USA
| | - Malek Asfar
- Department of Pathology, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Elsayed Zaabout
- Department of Therapeutics & Pharmacology, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
| | - Yalitza Lopez Corcino
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Venetia Thomas
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Eun-Seok Choi
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Saori Furuta
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| |
Collapse
|
21
|
Varricchi G, Poto R. Towards precision medicine in COPD: Targeting type 2 cytokines and alarmins. Eur J Intern Med 2024; 125:28-31. [PMID: 38762432 DOI: 10.1016/j.ejim.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a main global epidemic increasing as population age and affecting approximately 10% of subjects over 45 years. COPD is a heterogeneous inflammatory disease with several endo-phenotypes and clinical presentations. Although neutrophilic inflammation is canonically considered a hallmark of COPD, eosinophilic inflammation can also be present in a subgroup of patients. Several other immune cells and cytokines play a key role in orchestrating and perpetuating the inflammatory pathways in COPD, making them attractive targets for treating this disorder. Recent studies have started to evaluate the possible role of type 2 (T2) inflammation and epithelial-derived alarmins (TSLP and IL-33) in COPD. Two phase III randomized clinical trials (RCTs) showed a modest reduction in exacerbations in COPD patients with eosinophilic phenotype treated with mepolizumab (anti-IL-5) or benralizumab (anti-IL-5Rα). A phase III RCT showed a 30% reduction in exacerbations in COPD patients with ≥ 300 eosinophils/μL treated with dupilumab (anti-IL-4Rα). These results suggest that blocking a single cytokine (e.g., IL-5) or its main target (i.e., IL-5Rα) is less promising than blocking a wider spectrum of cytokines (i.e., IL-4 and IL-13) in COPD. TSLP and IL-33 are upstream regulators of T2-high and T2-low immune responses in airway inflammation. Several ongoing RCTs are evaluating the efficacy and safety of anti-TSLP (tezepelumab), anti-IL-33 (itepekimab, tozorakimab), and anti-ST2 (astegolimab) in patients with COPD, who experience exacerbations. In conclusion, targeting T2 inflammation or epithelial-derived alarmins might represent a step forward in precision medicine for the treatment of a subset of COPD.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy
| |
Collapse
|
22
|
Inoue D, Inoue R. Mechanisms of osteoporosis associated with chronic obstructive pulmonary disease. J Bone Miner Metab 2024; 42:428-437. [PMID: 38977438 DOI: 10.1007/s00774-024-01527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease characterized by chronic respiratory symptoms due to inflammatory and destructive changes of the lung leading to progressive airflow obstruction. Fragility fractures associated with osteoporosis are among major comorbidities and have significant impacts on quality of life and prognosis of patients with COPD. Evidence suggests that both decreased bone mineral density (BMD) and impaired bone quality contribute to bone fragility and resultant fractures in COPD. Although various clinical risk factors of osteoporosis have been described, mechanisms of COPD-associated osteoporosis are still largely unknown. In addition, its specific treatment has not been established, either. Previous studies have suggested involvement of low BMI and sarcopenia in the pathogenesis of COPD-associated osteoporosis. In this narrative review, we will propose critical roles of vitamin D deficiency and inflammation, both of which are often present in COPD and may underlie the development of osteosarcopenia and impaired bone quality, ultimately causing fractures in COPD patients.
Collapse
Affiliation(s)
- Daisuke Inoue
- Division of Endocrinology and Metabolism, Third Department of Medicine, Teikyo University Chiba Medical Center, 3426-3 Anesaki, Ichihara-city, Chiba, 299-0111, Japan.
| | - Reiko Inoue
- Division of Endocrinology and Metabolism, Third Department of Medicine, Teikyo University Chiba Medical Center, 3426-3 Anesaki, Ichihara-city, Chiba, 299-0111, Japan
| |
Collapse
|
23
|
Sun B, Shen K, Zhao R, Li Y, Xiang M, Lin J. Precision medicine for severe asthma - Biological targeted therapy. Int Immunopharmacol 2024; 134:112189. [PMID: 38759375 DOI: 10.1016/j.intimp.2024.112189] [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] [Accepted: 04/29/2024] [Indexed: 05/19/2024]
Abstract
Severe asthma is a complex and heterogeneous chronic airway inflammatory disease. Current treatment strategies are increasingly focused on disease classification, facilitating the transition towards personalized medicine by integrating biomarkers and monoclonal antibodies for tailored therapeutic approaches. Several approved biological agents, including anti-immunoglobulin E (IgE), anti-interleukin (IL)-4, anti-IL-5, and anti-thymic stromal lymphopoietin (TSLP) monoclonal antibodies, have demonstrated significant efficacy in reducing asthma exacerbations, eosinophil counts, improving lung function, minimizing oral corticosteroid usage, and enhancing patients' quality of life. The utilization of these biological agents has brought about profound transformations in the management of severe asthma. This article provides a comprehensive review on biomarkers and biological agents for severe asthma while emphasizing the increasing importance of further research into its pathogenesis and novel treatment modalities.
Collapse
Affiliation(s)
- Bingqing Sun
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kunlu Shen
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruiheng Zhao
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Yun Li
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Min Xiang
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Jiangtao Lin
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Li Y, Wang Y, Wu R, Li P, Cheng Z. HTR2B as a novel biomarker of chronic obstructive pulmonary disease with lung squamous cell carcinoma. Sci Rep 2024; 14:13206. [PMID: 38851806 PMCID: PMC11162446 DOI: 10.1038/s41598-024-63896-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: 01/30/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is often associated with lung squamous cell carcinoma (LUSC), which has the same etiology (smoking, inflammation, oxidative stress, microenvironmental changes, and genetics). Smoking, inflammation, and airway remodeling are the most important and classical mechanisms of COPD comorbidity in LUSC patients. Cancer can occur during repeated airway damage and repair (airway remodeling). Changes in the inflammatory and immune microenvironments, which can cause malignant transformation of some cells, are currently being revealed in both LUSC and COPD patients. We obtained the GSE76925 dataset from the Gene Expression Omnibus database. Screening for possible COPD biomarkers was performed using the LASSO regression model and a random forest classifier. The compositional patterns of the immune cell fraction in COPD patients were determined using CIBERSORT. HTR2B expression was analyzed using validation datasets (GSE47460, GSE106986, and GSE1650). HTR2B expression in COPD cell models was determined via real-time quantitative PCR. Epithelial-mesenchymal transition (EMT) marker expression levels were determined after knocking down or overexpressing HTR2B. HTR2B function and mechanism in LUSC were analyzed with the Kaplan‒Meier plotter database. HTR2B expression was inhibited to detect changes in LUSC cell proliferation. A total of 1082 differentially expressed genes (DEGs) were identified in the GSE76925 dataset (371 genes were significantly upregulated, and 711 genes were significantly downregulated). Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that the DEGs were mainly enriched in the p53 signaling and β-alanine metabolism pathways. Gene Ontology enrichment analysis indicated that the DEGs were largely related to transcription initiation from the RNA polymerase I promoter and to the regulation of mononuclear cell proliferation. The LASSO regression model and random forest classifier results revealed that HTR2B, DPYS, FRY, and CD19 were key COPD genes. Immune cell infiltration analysis indicated that these genes were closely associated with immune cells. Analysis of the validation sets suggested that HTR2B was upregulated in COPD patients. HTR2B was significantly upregulated in COPD cell models, and its upregulation was associated with increased EMT marker expression. Compared with that in bronchial epithelial cells, HTR2B expression was upregulated in LUSC cells, and inhibiting HTR2B expression led to the inhibition of LUSC cell proliferation. In conclusions, HTR2B might be a new biomarker and therapeutic target in COPD patients with LUSC.
Collapse
MESH Headings
- Humans
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/metabolism
- Epithelial-Mesenchymal Transition/genetics
- Receptor, Serotonin, 5-HT2B/genetics
- Receptor, Serotonin, 5-HT2B/metabolism
- Gene Expression Regulation, Neoplastic
- Cell Proliferation/genetics
- Cell Line, Tumor
Collapse
Affiliation(s)
- Yue Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yu Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ruhao Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Pengfei Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zhe Cheng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
25
|
Szczesny B, Boorgula MP, Chavan S, Campbell M, Johnson RK, Kammers K, Thompson EE, Cox MS, Shankar G, Cox C, Morin A, Lorizio W, Daya M, Kelada SNP, Beaty TH, Doumatey AP, Cruz AA, Watson H, Naureckas ET, Giles BL, Arinola GA, Sogaolu O, Falade AG, Hansel NN, Yang IV, Olopade CO, Rotimi CN, Landis RC, Figueiredo CA, Altman MC, Kenny E, Ruczinski I, Liu AH, Ober C, Taub MA, Barnes KC, Mathias RA. Multi-omics in nasal epithelium reveals three axes of dysregulation for asthma risk in the African Diaspora populations. Nat Commun 2024; 15:4546. [PMID: 38806494 PMCID: PMC11133339 DOI: 10.1038/s41467-024-48507-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Asthma has striking disparities across ancestral groups, but the molecular underpinning of these differences is poorly understood and minimally studied. A goal of the Consortium on Asthma among African-ancestry Populations in the Americas (CAAPA) is to understand multi-omic signatures of asthma focusing on populations of African ancestry. RNASeq and DNA methylation data are generated from nasal epithelium including cases (current asthma, N = 253) and controls (never-asthma, N = 283) from 7 different geographic sites to identify differentially expressed genes (DEGs) and gene networks. We identify 389 DEGs; the top DEG, FN1, was downregulated in cases (q = 3.26 × 10-9) and encodes fibronectin which plays a role in wound healing. The top three gene expression modules implicate networks related to immune response (CEACAM5; p = 9.62 × 10-16 and CPA3; p = 2.39 × 10-14) and wound healing (FN1; p = 7.63 × 10-9). Multi-omic analysis identifies FKBP5, a co-chaperone of glucocorticoid receptor signaling known to be involved in drug response in asthma, where the association between nasal epithelium gene expression is likely regulated by methylation and is associated with increased use of inhaled corticosteroids. This work reveals molecular dysregulation on three axes - increased Th2 inflammation, decreased capacity for wound healing, and impaired drug response - that may play a critical role in asthma within the African Diaspora.
Collapse
Affiliation(s)
- Brooke Szczesny
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Meher Preethi Boorgula
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Sameer Chavan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Monica Campbell
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Randi K Johnson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
- Quantitative Sciences Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kai Kammers
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Emma E Thompson
- Division of Allergy and Infectious Diseases, Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Madison S Cox
- Division of Allergy and Infectious Diseases, Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Gautam Shankar
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Corey Cox
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Andréanne Morin
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Wendy Lorizio
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michelle Daya
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Samir N P Kelada
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alvaro A Cruz
- Fundacao ProAR and Federal University of Bahia, Salvador, Bahia, Brazil
| | - Harold Watson
- Faculty of Medical Sciences, The University of the West Indies, Queen Elizabeth Hospital, St. Michael, Bridgetown, Barbados
| | | | - B Louise Giles
- Departments of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Ganiyu A Arinola
- Department of Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumide Sogaolu
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adegoke G Falade
- Department of Pediatrics, University of Ibadan, and University College Hospital, Ibadan, Nigeria
| | - Nadia N Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ivana V Yang
- Departments of Biomedical Informatics and Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | | | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - R Clive Landis
- Edmund Cohen Laboratory for Vascular Research, George Alleyne Chronic Disease Research Centre, Caribbean Institute for Health Research, The University of the West Indies, Cave Hill Campus, Wanstead, Barbados
| | - Camila A Figueiredo
- Federal University of Bahia and Funda. Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil
- Instituto de Ciências de Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Matthew C Altman
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Eimear Kenny
- Center for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew H Liu
- Department of Pediatrics, Childrens Hospital Colorado and University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Carole Ober
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
26
|
Jürgens DC, Müller JT, Nguyen A, Merkel OM. Tailoring lipid nanoparticles for T-cell targeting in allergic asthma: Insights into efficacy and specificity. Eur J Pharm Biopharm 2024; 198:114242. [PMID: 38442794 PMCID: PMC7616735 DOI: 10.1016/j.ejpb.2024.114242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Asthma impacts over 300 million patients globally, with significant health implications, especially in cases of its allergic subtype. The disease is characterized by a complex interplay of airway inflammation and immune responses, often mediated by Th2 cell-related cytokines. In this study, we engineered lipid nanoparticles (LNPs) to specifically deliver therapeutic siRNA via the transferrin receptor to T cells. Strain-promoted azide-alkyne cycloaddition (SPAAC) was employed for the conjugation of transferrin ligands to PEGylated lipids in the LNPs, with the goal of enhancing cellular uptake and gene knockdown. The obtained LNPs exhibited characteristics that make them suitable for pulmonary delivery. Using methods such as nanoparticle tracking analysis (NTA) and enzyme-linked immunosorbent assay (ELISA), we determined the average number of transferrin molecules bound to individual LNPs. Additionally, we found that cellular uptake was ligand-dependent, achieving a GATA3 knockdown of more than 50% in relevant in vitro and ex vivo models. Notably, our findings highlight the limitations inherent to modifying the surface of LNPs, particularly with regard to their targeting capabilities. This work paves the way for future research aimed at optimizing targeted LNPs for the treatment of immunologic diseases such as allergic asthma.
Collapse
Affiliation(s)
- David C Jürgens
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany
| | - Joschka T Müller
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany
| | - Anny Nguyen
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-University Munich, 80799 Munich, Germany; Ludwig-Maximilians-University Munich, Member of the German Center for Lung Research (DZL), Germany
| |
Collapse
|
27
|
Mariotti EB, Corrà A, Aimo C, Ruffo di Calabria V, Quintarelli L, Verdelli A, Caproni M. Exacerbation of clinical manifestations of bullous pemphigoid after treatment with dupilumab. Clin Exp Dermatol 2024; 49:399-400. [PMID: 38011325 DOI: 10.1093/ced/llad412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023]
Abstract
The article presents an adverse event that occurred after the induction phase of dupilumab, administered in another country to a patient affected by bullous pemphigoid.
Collapse
Affiliation(s)
| | - Alberto Corrà
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Cristina Aimo
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Lavinia Quintarelli
- Rare Diseases Unit, Section of Dermatology, Department of Health Sciences, USL Toscana Centre, European Reference Network-Skin Member, University of Florence, Florence, Italy
| | - Alice Verdelli
- Rare Diseases Unit, Section of Dermatology, Department of Health Sciences, USL Toscana Centre, European Reference Network-Skin Member, University of Florence, Florence, Italy
| | - Marzia Caproni
- Rare Diseases Unit, Section of Dermatology, Department of Health Sciences, USL Toscana Centre, European Reference Network-Skin Member, University of Florence, Florence, Italy
| |
Collapse
|
28
|
Baglivo I, Colantuono S, Lumaca A, Papa A, Gasbarrini A, Caruso C. The last step to achieve barrier damage control. Front Immunol 2024; 15:1354556. [PMID: 38415254 PMCID: PMC10897052 DOI: 10.3389/fimmu.2024.1354556] [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: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Heterogeneity characterises inflammatory diseases and different phenotypes and endotypes have been identified. Both innate and adaptive immunity contribute to the immunopathological mechanism of these diseases and barrier damage plays a prominent role triggering type 2 inflammation through the alarmins system, such as anti-Thymic Stromal Lymphopoietin (TSLP). Treatment with anti-TSLP monoclonal antibodies showed efficacy in severe asthma and clinical trials for other eosinophilic diseases are ongoing. The aim of this perspective review is to analyse current advances and future applications of TSLP inhibition to control barrier damage.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’ApparatoDigerente, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Arianna Lumaca
- Unità Operativa Semplice Dipartimentale (UOSD) di Allergologia, Ospedale Maria Santissima (SS) Dello Splendore, Teramo, Italy
| | - Alfredo Papa
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’ApparatoDigerente, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Antonio Gasbarrini
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’ApparatoDigerente, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
29
|
Chen CY, Wu KH, Guo BC, Lin WY, Chang YJ, Wei CW, Lin MJ, Wu HP. Personalized Medicine in Severe Asthma: From Biomarkers to Biologics. Int J Mol Sci 2023; 25:182. [PMID: 38203353 PMCID: PMC10778979 DOI: 10.3390/ijms25010182] [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: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Severe asthma is a complex and heterogeneous clinical condition presented as chronic inflammation of the airways. Conventional treatments are mainly focused on symptom control; however, there has been a shift towards personalized medicine. Identification of different phenotypes driven by complex pathobiological mechanisms (endotypes), especially those driven by type-2 (T2) inflammation, has led to improved treatment outcomes. Combining biomarkers with T2-targeting monoclonal antibodies is crucial for developing personalized treatment strategies. Several biological agents, including anti-immunoglobulin E, anti-interleukin-5, and anti-thymic stromal lymphopoietin/interleukin-4, have been approved for the treatment of severe asthma. These biological therapies have demonstrated efficacy in reducing asthma exacerbations, lowering eosinophil count, improving lung function, diminishing oral corticosteroid use, and improving the quality of life in selected patients. Severe asthma management is undergoing a profound transformation with the introduction of ongoing and future biological therapies. The availability of novel treatment options has facilitated the adoption of phenotype/endotype-specific approaches and disappearance of generic interventions. The transition towards precision medicine plays a crucial role in meticulously addressing the individual traits of asthma pathobiology. An era of tailored strategies has emerged, allowing for the successful targeting of immune-inflammatory responses that underlie uncontrolled T2-high asthma. These personalized approaches hold great promise for improving the overall efficacy and outcomes in the management of severe asthma. This article comprehensively reviews currently available biological agents and biomarkers for treating severe asthma. With the expanding repertoire of therapeutic options, it is becoming increasingly crucial to comprehend the influencing factors, understand the pathogenesis, and track treatment progress in severe asthma.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Department of Emergency Medicine, Tungs’ Taichung Metro Harbor Hospital, Taichung 435403, Taiwan; (C.-Y.C.); (C.-W.W.)
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Bei-Cyuan Guo
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Wen-Ya Lin
- Division of Pediatric Emergency Medicine, Department of Pediatrics, Taichung Veteran General Hospital, Taichung 43503, Taiwan;
| | - Yu-Jun Chang
- Laboratory of Epidemiology and Biostastics, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Chih-Wei Wei
- Department of Emergency Medicine, Tungs’ Taichung Metro Harbor Hospital, Taichung 435403, Taiwan; (C.-Y.C.); (C.-W.W.)
| | - Mao-Jen Lin
- Division of Cardiology, Department of Medicine, Taichung Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan
- Department of Medicine, College of Medicine, Tzu Chi University, Hualien 97002, Taiwan
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| |
Collapse
|
30
|
Schleich F, Moermans C, Seidel L, Kempeneers C, Louis G, Rogister F, Tombu S, Pottier L, Poirrier AL, Ziant S, Henket M, Sanchez C, Paulus V, Guissard F, Donneau AF, Louis R. Benralizumab in severe eosinophilic asthma in real life: confirmed effectiveness and contrasted effect on sputum eosinophilia versus exhaled nitric oxide fraction - PROMISE. ERJ Open Res 2023; 9:00383-2023. [PMID: 38020567 PMCID: PMC10680030 DOI: 10.1183/23120541.00383-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Randomised controlled trials have shown that benralizumab, an anti-interleukin-5 receptor monoclonal antibody, reduces exacerbations and oral corticosteroid dose and improves asthma control and lung function in severe eosinophilic asthma. The aim of this study was to confirm results of randomised controlled trials in real life in a population of 73 patients with severe eosinophilic asthma treated with benralizumab for at least 12 months. Methods Patients underwent careful monitoring of asthma exacerbations, exhaled nitric oxide fraction, lung function, asthma control and quality of life questionnaire responses and sputum induction, and gave a blood sample at baseline, after 6 months and then every year. Results We found significant reductions in exacerbations (by 92%, p<0.0001) and oral corticosteroid dose (by 83%, p<0.001) after 6 months that were maintained over time, with 78% of patients able to stop oral corticosteroid therapy. Patients improved their Asthma Control Test (ACT) score (from 11.7±5.1 to 16.9±5.35, p<0.0001), Asthma Control Questionnaire (ACQ) score (from 2.88±1.26 to 1.77±1.32, p<0.0001) and Asthma Quality of Life Questionnaire score (+1.04, p<0.0001) at 6 months and this was maintained during follow-up. Only 35% and 43% of patients reached asthma control according to an ACT score ≥20 and ACQ score <1.5, respectively. We observed stable post-bronchodilation lung function over time and a significant reduction in sputum eosinophil count, with 85% of patients exhibiting sputum eosinophil counts <3% after 6 months (p<0.01) with no effect on exhaled nitric oxide fraction. Conclusion In our real-life study, we confirm the results published in randomised controlled trials showing a sharp reduction in exacerbations and oral corticosteroid therapy, an improvement in asthma control and quality of life, and a dramatic reduction in sputum eosinophil count.
Collapse
Affiliation(s)
- Florence Schleich
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Catherine Moermans
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | | | - Céline Kempeneers
- Biostatistics and Research Method Center (B-STAT), University of Liege and CHU Liege, Liege, Belgium
| | - Gilles Louis
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Florence Rogister
- Division of Respirology, Department of Pediatrics, CHU Liege, Liege, Belgium
| | - Sophie Tombu
- Department of Ear, Nose and Throat, CHU Sart-Tilman, Liege, Belgium
| | - Laurence Pottier
- Department of Ear, Nose and Throat, CHU Sart-Tilman, Liege, Belgium
| | | | - Stéphanie Ziant
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Monique Henket
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Carole Sanchez
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Virginie Paulus
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Françoise Guissard
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| | - Anne-Françoise Donneau
- Biostatistics and Research Method Center (B-STAT), University of Liege and CHU Liege, Liege, Belgium
| | - Renaud Louis
- Department of Pulmonary Medicine, CHU Sart-Tilman, Liege, Belgium
- IGIGA Research Group, University of Liege, Liege, Belgium
| |
Collapse
|
31
|
Sardon-Prado O, Diaz-Garcia C, Corcuera-Elosegui P, Korta-Murua J, Valverde-Molina J, Sanchez-Solis M. Severe Asthma and Biological Therapies: Now and the Future. J Clin Med 2023; 12:5846. [PMID: 37762787 PMCID: PMC10532431 DOI: 10.3390/jcm12185846] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Recognition of phenotypic variability in pediatric asthma allows for a more personalized therapeutic approach. Knowledge of the underlying pathophysiological and molecular mechanisms (endotypes) of corresponding biomarkers and new treatments enables this strategy to progress. Biologic therapies for children with severe asthma are becoming more relevant in this sense. The T2 phenotype is the most prevalent in childhood and adolescence, and non-T2 phenotypes are usually rare. This document aims to review the mechanism of action, efficacy, and potential predictive and monitoring biomarkers of biological drugs, focusing on the pediatric population. The drugs currently available are omalizumab, mepolizumab, benralizumab, dupilumab, and 1ezepelumab, with some differences in administrative approval prescription criteria between the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA). Previously, we described the characteristics of severe asthma in children and its diagnostic and therapeutic management.
Collapse
Affiliation(s)
- Olaia Sardon-Prado
- Division of Paediatric Respiratory Medicine, Donostia University Hospital, 20014 San Sebastián, Spain; (O.S.-P.); (P.C.-E.); (J.K.-M.)
- Department of Pediatrics, University of the Basque Country (UPV/EHU), 20014 Leioa, Spain
| | - Carolina Diaz-Garcia
- Paediatric Pulmonology and Allergy Unit, Santa Lucia General University Hospital, 30202 Cartagena, Spain;
| | - Paula Corcuera-Elosegui
- Division of Paediatric Respiratory Medicine, Donostia University Hospital, 20014 San Sebastián, Spain; (O.S.-P.); (P.C.-E.); (J.K.-M.)
| | - Javier Korta-Murua
- Division of Paediatric Respiratory Medicine, Donostia University Hospital, 20014 San Sebastián, Spain; (O.S.-P.); (P.C.-E.); (J.K.-M.)
| | - Jose Valverde-Molina
- Department of Paediatrics, Santa Lucía General University Hospital, 30202 Cartagena, Spain
- IMIB Biomedical Research Institute, 20120 Murcia, Spain;
| | - Manuel Sanchez-Solis
- IMIB Biomedical Research Institute, 20120 Murcia, Spain;
- Department of Pediatrics, University of Murcia, 20120 Murcia, Spain
- Paediatric Allergy and Pulmonology Units, Virgen de la Arrixaca University Children’s Hospital, 20120 Murcia, Spain
| |
Collapse
|
32
|
Sekaran K, Varghese RP, Karthik A, Sasikumar K, Shree Devi MS, Sathiyarajeswaran P, George Priya Doss C. In silico network pharmacology analysis and molecular docking validation of Swasa Kudori tablet for screening druggable phytoconstituents of asthma. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:257-274. [PMID: 38220427 DOI: 10.1016/bs.apcsb.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Traditional medicines are impactful in treating a cluster of respiratory-related illnesses. This paper demonstrates screening active, druggable phytoconstituents from a classical Siddha-based poly-herbal formulation called Swasa Kudori Tablet to treat asthma. The phytoconstituents of Swasa Kudori are identified as Calotropis gigantea, Piper nigrum, and (Co-drug) Abies webbiana. Active chemical compounds are extracted with the Chemical Entities of Biological Interest (ChEBI) database. The gene targets of each compound are identified based on the pharmacological activity using the DIGEP-Pred database. Thirty-two genes showing Pa> 0.7 is screened, and the target markers are selected after performing gene overlap evaluation with the asthma genes reported in GeneCards and DisGeNET database. Ten markers are identified, such as ADIPOQ, CASP8, CAT, CCL2, CD86, FKBP5, HMOX1, NFE2L2, TIMP1, VDR, in common, listed as molecular targets. Pharmacokinetic assessment (ADME) revealed five natural drug compounds 2-5-7-trihydroxy-2-(4-hydroxyphenyl)-2,3-dihydro-4H-chromen-4-one, (+)-catechin-3'-methyl ether, futoenone, 5-hydroxy-4',7-dimethoxyflavanone, and pinocembrin showing better druggability. Further screening delineates the target (HMOX1) and drug (pinocembrin) for molecular docking evaluation. When docked with HO-1, Pinocembrin showed a binding affinity of -8.0 kcal/mol. MD simulation studies substantiate the docking studies as HO-1 in complex with pinocembrin remains stable in the simulated trajectory. The current findings exhibit the significance of traditional medicines as potential drug candidates against asthma.
Collapse
Affiliation(s)
- Karthik Sekaran
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | | | - Ashwini Karthik
- Department of Biology, Mount Carmel College Autonomous, Bengaluru, India
| | - K Sasikumar
- School of Electronics Engineering, Vellore Institute of Technology, Vellore, India
| | - M S Shree Devi
- Siddha Central Research Institute (CCRS), Chennai, India
| | | | - C George Priya Doss
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India.
| |
Collapse
|