1
|
Nahas G, Chen Y, Ningundi A, Tercyak S, Preciado D. Middle Ear microRNAs Drive Mucin Gene Response. Laryngoscope 2025; 135:1815-1820. [PMID: 39569770 DOI: 10.1002/lary.31912] [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: 07/24/2024] [Revised: 09/29/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE(S) To investigate the role of microRNA-378 (miR-378) in the regulation of mucin gene expression and inflammatory response in human middle ear epithelial cells (HMEEC) during bacterial infection by non-typeable Haemophilus influenzae (NTHi). METHODS Human middle ear epithelial cells (HMEEC) were cultured and transfected with miR-378 or control miRNA. Post-transfection, cells were exposed to NTHi lysates. mRNA levels of MUC5B, MUC5AC, and IL-8 were quantified using RT-qPCR, and promoter activity was measured via luciferase assays. The effects of miR-378 on mucin and cytokine gene expression were analyzed. RESULTS Transfection with miR-378 significantly increased the expression of MUC5B (3.6 fold, p < 0.01), MUC5AC (19.1 fold, p < 0.01), and IL-8 (2.01 fold, p < 0.05) in HMEEC. NTHi exposure reduced MUC5B (1.385 fold, p < 0.05) and MUC5AC (1.61 fold, p < 0.05) gene expression in miR-378 transfected cells but significantly increased IL-8 levels (1.32 fold, p < 0.05). Luciferase assays showed that miR-378 upregulated the promoter activity of MUC5B (1.4 fold, p < 0.01) and MUC5AC (1.6 fold, p < 0.01) genes, indicating its role in transcriptional regulation. CONCLUSION miR-378 plays a crucial role in promoting mucin overproduction and an inflammatory response in the middle ear epithelium during OM. Targeting miR-378 could offer a novel therapeutic strategy for preventing the progression from AOM to COM. LEVEL OF EVIDENCE NA Laryngoscope, 135:1815-1820, 2025.
Collapse
Affiliation(s)
- Gabriel Nahas
- Division of Otolaryngology, Sheikh Zayed Center for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, USA
| | - Yajun Chen
- Division of Otolaryngology, Sheikh Zayed Center for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, USA
| | - Apurva Ningundi
- Division of Otolaryngology, Sheikh Zayed Center for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, USA
| | - Samuel Tercyak
- Division of Otolaryngology, Sheikh Zayed Center for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, USA
| | - Diego Preciado
- Division of Otolaryngology, Sheikh Zayed Center for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, USA
- Division of Pediatric Otolaryngology, Children's National Hospital, Washington, DC, USA
| |
Collapse
|
2
|
Zimbru RI, Zimbru EL, Bojin FM, Haidar L, Andor M, Harich OO, Tănasie G, Tatu C, Mailat DE, Zbîrcea IM, Hirtie B, Uța C, Bănărescu CF, Panaitescu C. Connecting the Dots: How MicroRNAs Link Asthma and Atherosclerosis. Int J Mol Sci 2025; 26:3570. [PMID: 40332077 DOI: 10.3390/ijms26083570] [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: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Asthma and atherosclerosis are chronic conditions with distinct pathophysiologies, but overlapping inflammatory mechanisms that suggest a potential common regulatory framework. MicroRNAs (miRNAs), small non-coding RNA molecules that modulate gene expression post-transcriptionally, could be key players in linking these disorders. This review outlines how miRNAs contribute to the complex interplay between asthma and atherosclerosis, focusing on key miRNAs involved in inflammatory pathways, immune cell regulation and vascular remodeling. We discuss specific miRNAs, such as miR-155, miR-21 and miR-146a, which have been shown to modulate inflammatory cytokine production and T cell differentiation, impacting respiratory and cardiovascular health. The common miRNAs found in both asthma and atherosclerosis emphasize their role as potential biomarkers, but also as therapeutic targets. Understanding these molecular connections may unlock novel approaches for innovative, integrated treatment strategies that address both conditions and may significantly improve patient outcomes. Further research is needed to explore mechanistic pathways and validate the translational potential of miRNA-based interventions in preclinical and clinical settings.
Collapse
Affiliation(s)
- Răzvan-Ionuț Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer-OncoGen, Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Elena-Larisa Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer-OncoGen, Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Multidisciplinary Heart Research Center, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Florina-Maria Bojin
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer-OncoGen, Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Laura Haidar
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Minodora Andor
- Multidisciplinary Heart Research Center, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Cardiology Clinic, Timisoara Municipal Clinical Emergency Hospital, 12 Revoluției din 1989 Bd., 300040 Timisoara, Romania
| | - Octavia Oana Harich
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Gabriela Tănasie
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer-OncoGen, Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Carmen Tatu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer-OncoGen, Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Diana-Evelyne Mailat
- Multidisciplinary Heart Research Center, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Cardiology Clinic, Timisoara Municipal Clinical Emergency Hospital, 12 Revoluției din 1989 Bd., 300040 Timisoara, Romania
| | - Iulia-Maria Zbîrcea
- Department of Automation and Applied Informatics, "Politehnica" University of Timisoara, 300006 Timișoara, Romania
| | - Bogdan Hirtie
- ENT Department, "Victor Babes" University of Medicine and Pharmacy, 300042 Timișoara, Romania
| | - Cristina Uța
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Camelia-Felicia Bănărescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Carmen Panaitescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer-OncoGen, Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital "Pius Brinzeu", 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| |
Collapse
|
3
|
Chen Y, Mao R, Chang Q, Yuan Y, Zhang H, Li F. A causal effects of neutrophil extracellular traps and its biomarkers on acute respiratory distress syndrome: a two-sample Mendelian randomization study. Sci Rep 2025; 15:11995. [PMID: 40199908 PMCID: PMC11978891 DOI: 10.1038/s41598-025-95676-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
Previous studies have indicated an association between neutrophil extracellular traps (NETs) and acute respiratory distress syndrome (ARDS). This study aimed to investigate the potential causal effects of NETs and NETs-related biomarkers on ARDS or vice-versa. A two-sample Mendelian randomization (MR) utilizing genome-wide association studies (GWAS) data was employed to analyze the causality. The primary analysis was conducted using inverse-variance weighted (IVW) methods; weighted median, MR-Egger, and weighted model methods were used to validate the results. Horizontal pleiotropy and outlier detection were assessed via MR-Egger and MR pleiotropy residual sum and outlier (MR-PRESSO), respectively; Cochran's Q test evaluated heterogeneity, while Leave-one-out analyses were used to evaluate the presence of predominant instrumental variables (IVs). IVW method suggested causal associations between genetically predicted IL-13 and a higher risk of ARDS [OR (95%CI) = 1.52 (1.03-2.23), P = 0.047], while there was no causal effect of other factors on ARDS (all P > 0.05). Also, ARDS had no effect on NETs and NETs-related biomarkers (all P > 0.05). Cochran's Q confirmed no significant heterogeneity. MR-Egger regression ruled out horizontal pleiotropy's influence, and MR-PRESSO analysis identified no outliers, reinforcing the study's findings. This MR study established a causal relationship between IL-13 and ARDS, suggesting its potential role as a therapeutic target and biomarker of ARDS. Future work should delve into the underlying mechanisms and clinical applications.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Ruolin Mao
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qing Chang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yueyang Yuan
- School of Mechanical and Electrical Engineering, Hu Nan City University, Yiyang, 413099, China
| | - Hai Zhang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Feng Li
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|
4
|
Yan X, Liu Q, Adams T, Schupp J, Li S, Huang S, Grant N, Wilson G, Gomez J, Cohn L, Kaminski N, Weiss ST, Tantisira K, Chupp GL. Single-cell RNA Sequencing Analysis of Sputum Cell Transcriptomes Reveals Pathways and Communication Networks That Contribute to the Pathogenesis of Asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646405. [PMID: 40236103 PMCID: PMC11996526 DOI: 10.1101/2025.03.31.646405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Asthma is driven by complex interactions amongst structural airway cells, cells of the immune system, and the environmental. While sputum cell characterization has been instrumental in studying asthma pathogenesis and refining treatment strategies, the nuances of cellular transcriptomes and intercellular communication in asthmatic sputum remain poorly understood. Methods We employed single-cell RNA sequencing to analyze cells isolated form the sputum from 16 asthma patients and 8 non-asthmatic controls. Cell identities were established using curated marker genes and SingleR annotation. We compared cell-specific gene expression and communication networks between asthmatic and control groups, correlating findings with distinct pathways that were dysregulated in asthma. Findings 37,565 cellular transcriptomes were captured and analyzed. 15 distinct cell populations were identified, including various macrophages, monocytes, dendritic cells, and lymphocytes, along with rare cell types such as mast cells, innate lymphoid cells, bronchial epithelial cells, and eosinophils. Intercellular communication analysis indicated heightened signaling activity in asthma compared to controls, particularly in CD4+ T cells and dendritic cells which exhibited the most significant increases in RNA expression of outgoing signaling molecules. Notably, the ADAM12-SDC4 and CCL22-CCR4 ligand-receptor pathways demonstrated the strongest shifts between asthma and control subjects, particularly between dendritic cells and CD4 lymphocytes. Interpretation SC RNA seq profiling the asthma cellular transcriptome analysis of sputum highlights both innate and adaptive immune mechanisms that are significantly amplified in asthma. The elevated expression of ADAM12-SCD4 and CCL22-CC4 point to their critical role in asthma pathogenesis, suggesting potential avenues for targeted therapies and improved management of this chronic condition. Research in context Evidence Before This Study: Asthma is a chronic inflammatory disease of the airways driven by intricate interactions between airway structural and immune cells. Previous transcriptomic studies have focused on bulk RNA samples from the airway, leaving significant gaps in our understanding of the cellular dynamics that characterize the disease.Added Value of This Study: This study pioneers the use of single-cell RNA sequencing on sputum samples from patients with asthma, revealing a detailed landscape of cell phenotypes and dynamic communication patterns that distinguish asthmatic individuals from those without the disease. Notably, heightened intercellular communication was observed in asthma, particularly between CD4+ T cells and dendritic cells, confirming that there is a robust network of interactions between immune and structural cells. The notable increase of ADAM12-CCR4 communication from dendritic cells to other cell populations further emphasizes the dysregulation present in asthma.Implications of All Available Evidence: Our transcriptomic profiling illuminates distinct and amplified communication pathways involving CD4+ T cells and dendritic cells, aligning with established paradigms of both adaptive and innate immune responses in asthma pathogenesis. The identification of ADAM12 and CCR4 pathway dysregulation adds a critical layer to our understanding of the molecular mechanisms underpinning asthma, paving the way for potential therapeutic targets and personalized treatment strategies. Single cell profiling of the sputum has the capacity to characterize the breadth of cellular phenotypes, their functional status, and the communication in the airway at a level not previously attainable.
Collapse
|
5
|
Dorscheid D, Gauvreau GM, Georas SN, Hiemstra PS, Varricchi G, Lambrecht BN, Marone G. Airway epithelial cells as drivers of severe asthma pathogenesis. Mucosal Immunol 2025:S1933-0219(25)00029-7. [PMID: 40154790 DOI: 10.1016/j.mucimm.2025.03.003] [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: 05/20/2024] [Revised: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Our understanding of the airway epithelium's role in driving asthma pathogenesis has evolved over time. From being regarded primarily as a physical barrier that could be damaged via inflammation, the epithelium is now known to actively contribute to asthma development through interactions with the immune system. The airway epithelium contains multiple cell types with specialized functions spanning barrier action, mucociliary clearance, immune cell recruitment, and maintenance of tissue homeostasis. Environmental insults may cause direct or indirect injury to the epithelium leading to impaired barrier function, epithelial remodelling, and increased release of inflammatory mediators. In severe asthma, the epithelial barrier repair process is inhibited and the response to insults is exaggerated, driving downstream inflammation. Genetic and epigenetic mechanisms also maintain dysregulation of the epithelial barrier, adding to disease chronicity. Here, we review the role of the airway epithelium in severe asthma and how targeting the epithelium can contribute to asthma treatment.
Collapse
Affiliation(s)
- Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Bart N Lambrecht
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium.
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy.
| |
Collapse
|
6
|
Wu Y, Wang Q, Zhu W, Wang D, Yong Y, Li W, Sun J. NOTCH pathway was involved in Kaempferol 3-O-gentiobioside attenuated airway inflammation and mucus hypersecretion. Sci Rep 2025; 15:10383. [PMID: 40140655 PMCID: PMC11947102 DOI: 10.1038/s41598-025-95280-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
Allergic asthma is an inflammatory condition characterized by the release of pro-inflammatory cytokines and the expansion of mucus-producing cells. This research aimed to evaluate the impact of Kaempferol 3-O-gentiobioside (K3G), extracted from Sauropus spatulifolius Beille leaves, on inflammatory cytokine secretion and mucus overproduction in IL-13-stimulated airway epithelial cells (16HBE cells) and ovalbumin (OVA)-induced allergic asthma mouse models. Studies have found that K3G significantly reduces the release of pro-inflammatory cytokines (IgE, TNF-α, histamine, IL-1β, IL-6, and IL-8) induced by IL-13. It also mitigating the expression of the mucin5AC (MUC5AC). Additionally, K3G also downregulated the expression of NLRP3, TLR4, p-IκBα, and p-P65 proteins. In an OVA-induced mouse asthma model, K3G treatment reduced the secretion of pro-inflammatory cytokines, and inhibited the increase in mucus-secreting cells in a dose-dependent manner. Furthermore, exposure to IL-13 and OVA increased the expression of NOTCH signaling receptors in both 16HBE cells and lung tissues of the mice. K3G treatment effectively targeted NOTCH1 and inhibits activation of the NOTCH pathway. In conclusion, K3G alleviates asthma-related airway alterations by suppressing inflammatory cytokines and excessive mucus secretion via the NOTCH signaling pathway. These results indicate the therapeutic promise of K3G in treating allergic asthma.
Collapse
Affiliation(s)
- Yumiao Wu
- Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Qinqin Wang
- Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Wanqing Zhu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China
| | - Danyi Wang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China
| | - Yayun Yong
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China
| | - Weiwei Li
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530023, China.
| | - Jichao Sun
- Guangxi University of Chinese Medicine, Nanning, 530200, China.
| |
Collapse
|
7
|
Kumar S, Corkran M, Cheema Y, Scull MA, Duncan GA. AAV-mediated MUC5AC siRNA delivery to prevent mucociliary dysfunction in asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642720. [PMID: 40161599 PMCID: PMC11952410 DOI: 10.1101/2025.03.12.642720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The main structural components of mucus produced in the lung are mucin 5B (MUC5B) and mucin 5AC (MUC5AC) where a relatively higher expression of MUC5B is typical in health. In the lungs of individuals with asthma, there is a shift from MUC5B to MUC5AC as the predominantly secreted mucin which has been shown to impair mucociliary clearance (MCC) and increase mucus plug formation in the airways. Given its role in asthmatic lung disease, MUC5AC represents a potential therapeutic target where a gene delivery approach could be leveraged to modulate its expression. For these purposes, we explored adeno-associated virus serotype 6 (AAV6), as a lung-tropic viral gene vector to target airway epithelial cells and reduce MUC5AC expression via siRNA delivery. We confirmed that AAV6 was able to transduce epithelial cells in the airways of healthy mice with high transgene expression in mucus-secreting goblet cells. Using multiple particle tracking analysis, we observed that AAV6 was capable of penetrating both normal and MUC5AC-enriched mucus barriers. Successful transduction with AAV6 was also achieved in IL-13 stimulated human airway epithelial (HAE) cells differentiated at air-liquid interface (ALI). AAV6 expressing MUC5AC-targeting siRNA was evaluated as a prophylactic treatment in HAE cell cultures before IL-13 challenge. IL-13 stimulated HAE cultures treated with AAV6-MUC5AC siRNA had significantly reduced MUC5AC mRNA and protein expression compared to untreated controls. Mucociliary transport in IL-13 stimulated HAE cultures was also maintained and comparable to healthy controls following AAV6-MUC5AC siRNA treatment. Together, these findings support that AAV6 may be used as an inhaled gene therapy to suppress MUC5AC overexpression and restore normal airway clearance function in asthma.
Collapse
Affiliation(s)
- Sahana Kumar
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
| | - Maria Corkran
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
| | - Yahya Cheema
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Margaret A Scull
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Gregg A Duncan
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| |
Collapse
|
8
|
Yuan X, Li C, Yang L, Gao J, Wang B, Li Z. Unraveling asthma through single-cell RNA sequencing in understanding disease mechanisms. J Asthma 2025:1-9. [PMID: 40014380 DOI: 10.1080/02770903.2025.2472358] [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: 01/10/2025] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE To elucidate the fundamental principles of single-cell RNA sequencing (scRNA-seq) and summarize its application in asthma research, aiming to enhance understanding of asthma pathophysiology and guide future research directions. DATASOURCE Recent advances and emerging research in scRNA-seq and its role in the pathogenesis of asthma. STUDY SELECTIONS This review incorporates studies that analyzed the heterogeneity of asthma cell types and their functional states using scRNA-seq, with particular emphasis on immune cells and airway remodeling. The selection of specific cell types and markers was based on their relevance to asthma pathogenesis, and we discuss the rationale for favoring certain scRNA-seq technologies in these investigations. RESULTS ScRNA-seq technology has provided insights into the key mechanisms underlying inflammation and airway remodeling in asthma. It has uncovered the diversity of immune cell subtypes and their specific roles in asthma pathogenesis, revealing critical pathways that contribute to disease progression. These findings offer a theoretical foundation for the development of targeted therapeutic strategies, paving the way for personalized medicine and improved patient outcomes. CONCLUSION ScRNA-seq reveals the complex heterogeneity and functional roles of immune cells in asthma, offering key insights into disease mechanisms and the potential for targeted therapies. However, challenges remain, such as the need for further refinement of data integration methods and addressing the limited clinical applicability of current findings. Future research should focus on overcoming these limitations, improving cell type annotation, and expanding studies to include longitudinal and clinical data to better understand disease dynamics and therapy responses.
Collapse
Affiliation(s)
- Xingxing Yuan
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Chaofan Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Liuxin Yang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiawei Gao
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bingyu Wang
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Zhuying Li
- Department of Respiratory, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
9
|
Alladina J, Medoff BD, Cho JL. Innate Immunity and Asthma Exacerbations: Insights From Human Models. Immunol Rev 2025; 330:e70016. [PMID: 40087882 PMCID: PMC11922041 DOI: 10.1111/imr.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Asthma is a common chronic respiratory disease characterized by the presence of airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. Repeated asthma exacerbations can lead to progressive airway remodeling and irreversible airflow obstruction. Thus, understanding and preventing asthma exacerbations are of paramount importance. Although multiple endotypes exist, asthma is most often driven by type 2 airway inflammation. New therapies that target specific type 2 mediators have been shown to reduce the frequency of asthma exacerbations but are incompletely effective in a significant number of asthmatics. Furthermore, it remains unknown whether current treatments lead to sustained changes in the airway or if targeting additional pathways may be necessary to achieve asthma remission. Activation of innate immunity is the initial event in the inflammatory sequence that occurs during an asthma exacerbation. However, there continue to be critical gaps in our understanding of the innate immune response to asthma exacerbating factors. In this review, we summarize the current understanding of the role of innate immunity in asthma exacerbations and the methods used to study them. We also identify potential novel therapeutic targets for asthma and future areas for investigation.
Collapse
Affiliation(s)
- Jehan Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Josalyn L. Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
10
|
Zhao Y, Patel J, Fan J, Wang X, Chen L, Li Y, Luo Z. Integrated analysis reveals that EGR1 promotes epithelial IL33 production in T2 asthma. J Transl Med 2025; 23:203. [PMID: 39966984 PMCID: PMC11837401 DOI: 10.1186/s12967-025-06116-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Airway epithelial cells constitute the first line of defense against external noxious stimuli and play crucial roles in the release of epithelial inflammatory cytokines (IL33, IL25 and TSLP), initiating airway allergic inflammatory diseases such as asthma. IL33 plays critical physiological processes in T2-endotype asthma. However, the mechanisms by which allergen exposure triggers IL33 release from airway epithelial cells remain unclear. METHODS Integrated bioinformatic analysis and transcriptional analysis of bulk RNA-seq and single cell RNA-seq (scRNA-seq) data were used to identify core genes and determine the internal gene network associated with IL33. The expression of EGR1 was subsequently analyzed in vitro in the BEAS-2B cell line and in vivo in a house dust mite (HDM)-induced mouse asthma model. The functional experiments of EGR1 were investigated in vitro via siRNA knockdown and over-expressed plasmid. Chromatin immunoprecipitation (ChIP)-PCR and dual-luciferase reporter assay validation were subsequently performed to investigate the mechanisms by which EGR1 regulates IL33 secretion. RESULTS Bulk RNA-seq and scRNA-seq data identified EGR1 as an epithelial cell-derived gene implicated in IL33 expressions in asthma. The comprehensive analysis of multiple datasets indicated that the high EGR1 expression in epithelial cells may suggest a mechanistic basis of T2-endotype childhood asthma. Moreover, we verified that the expressions of EGR1 in airway epithelial cells were elevated both in vitro and in vivo asthma models. EGR1 regulated the production of IL33. Ultimately, ChIP and luciferase reporter assays confirmed that transcription factor EGR1 directly regulate the transcription of IL33 mRNA. CONCLUSIONS Our integrated bioinformatic analysis elucidated that EGR1 directly regulates the production of IL33 in T2-asthma and provide insights underlying the progression of asthma.
Collapse
Affiliation(s)
- Yan Zhao
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, China
| | - Jenil Patel
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston (UTHealth Houston) School of Public Health, Dallas, TX, USA
| | - Jinhua Fan
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyang Wang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Chen
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Li
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Zhengxiu Luo
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
- China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, China.
- Department of Respiratory Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
11
|
Paplińska-Goryca M, Misiukiewicz-Stępień P, Wróbel M, Mycroft-Rzeszotarska K, Adamska D, Rachowka J, Królikowska M, Goryca K, Krenke R. The impaired response of nasal epithelial cells to microplastic stimulation in asthma and COPD. Sci Rep 2025; 15:4242. [PMID: 39905077 PMCID: PMC11794662 DOI: 10.1038/s41598-025-87242-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
Microplastic particles from the air are inhaled and accumulate in the lungs, potentially causing immunological reactions and airway tissue injury. This study aimed to evaluate the biological effects of polyamide fibres on nasal epithelium co-cultivated with macrophages in control, asthma, and COPD groups. Nasal epithelial cells alone or in co-culture with monocyte-derived macrophages were exposed to polyamide fibres for 48 h. We identified 8 differentially expressed genes (DEGs) in controls, 309 DEGs in asthma (including ANKRD36C, BCL2L15, FCGBP, and IL-19), and 22 DEGs in COPD (e.g., BCL2L15, IL-19, CAPN14, PGBD5, PTPRH), particularly in epithelial/moMφ co-cultures. Microplastic exposure induced inflammatory cytokine secretion only for IL-8 production in controls (epithelial/ moMφs co-culture) and asthmatic (monoculture) epithelial cells in contrast to PM2.5, which was a strong inflammatory inducer. Gene Ontology analysis revealed that microplastic exposure affected sterol and cholesterol biosynthesis, secondary alcohol metabolism, and acetyl-CoA metabolism in asthma, and cell motility, chemokine signaling, leukocyte migration, and chemotaxis in COPD. Microplastic stimulation altered the response of airway epithelial cells in obstructive lung diseases differently than in controls, linking to Th2 inflammation, stress response modulation, and carcinogenesis. Asthmatic and COPD epithelial cells are more susceptible to damage from microplastic fibre exposure.
Collapse
Affiliation(s)
- Magdalena Paplińska-Goryca
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland.
| | - Paulina Misiukiewicz-Stępień
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Monika Wróbel
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Katarzyna Mycroft-Rzeszotarska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Dorota Adamska
- Genomic Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Julia Rachowka
- Genomic Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | | | - Krzysztof Goryca
- Genomic Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Rafał Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| |
Collapse
|
12
|
Yang Z, Zheng Y, Zhao J, Zhong Y, Zhang L, Wu H, Xie T, Ding Y. Impact of IL13 genetic polymorphisms on COPD susceptibility in the Chinese Han population. Respir Med 2025; 237:107923. [PMID: 39733814 DOI: 10.1016/j.rmed.2024.107923] [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: 08/08/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized by persistent respiratory symptoms and airflow limitation. Interleukin-13 (IL13), associated with T-helper type 2 cells, plays a crucial role in COPD pathophysiology. This study aimed to investigate the relationship of single nucleotide polymorphisms (SNPs) in IL13 to COPD risk. METHODS Five candidate SNPs of IL13 were genotyped using MassARRAY iPLEX platform in a cohort of472 COPD patients and 472 healthy controls. Logistic regression analysis was used to calculate odds ratios (ORs) and 95 % confidence intervals (CIs). Additionally, Multifactor dimensionality reduction (MDR) software was utilized to assess the combined impact of SNP-SNP interactions on COPD risk. RESULTS IL13 rs20541 (OR: 1.24, p: 0.028), rs1295685 (OR: 1.31, p: 0.006), rs848 (OR: 1.27, p: 0.016), and rs847 (OR: 1.30, p: 0.007) were associated with COPD risk. Moreover, IL13 variants were related to the increased COPD risk in females, individuals aged ≥68 year, non-smokers or non-drinkers. The optimal multi-locus model was identified as the combination of rs20541 and rs1295685. CONCLUSION Our findings indicated the association between IL13 variants and an elevated risk of developing COPD, especially rs1295685 and rs847. These findings could have implications for understanding the role of IL13 variants in COPD predisposition.
Collapse
Affiliation(s)
- Zehua Yang
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Yamei Zheng
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Jie Zhao
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Yi Zhong
- Department of General Practice, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Haihong Wu
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Tian Xie
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China.
| | - Yipeng Ding
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China.
| |
Collapse
|
13
|
Zhu X, Cheng F, Duan H, Fu S, Zhao C. Novel insights into the study of goblet cell hypersecretion in allergic rhinitis. Front Immunol 2025; 16:1525928. [PMID: 39958344 PMCID: PMC11825788 DOI: 10.3389/fimmu.2025.1525928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/06/2025] [Indexed: 02/18/2025] Open
Abstract
Goblet cell hypersecretion is a hallmark of airway inflammation and is driven by complex neuroimmune regulation involving submucosal glands and goblet cells. Although studies have focused on mast cell degranulation as a critical driver of nasal secretion, the role of goblet cells in this process is relatively under-researched. In allergic airway inflammation, goblet cells exhibit metaplasia and hypersecretion. However, allergen exposure does not directly trigger goblet cell degranulation, raising questions regarding the underlying mechanisms of these reactions. The activation of enteric neurons promotes goblet cell degranulation by stimulating the calcitonin gene-related peptide (CGRP)-receptor active modification protein-1 (RAMP1) axis. Meanwhile, airway goblet cells express various neuropeptide receptors, and their activation by neuropeptides such as substance P and CGRP induces mucus secretion, exacerbating allergic rhinitis-associated hypersecretion. Thus, although previously less recognised, the neuron-goblet cell signalling axis plays a critical role in allergic rhinitis mucus secretion. This review highlights current research on the neuroimmune mechanisms underlying goblet cell metaplasia and degranulation, focusing on allergic rhinitis, so as to guide clinical treatment strategies.
Collapse
Affiliation(s)
- Xiaojia Zhu
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Fengli Cheng
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Hongying Duan
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Sirui Fu
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Changqing Zhao
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Liu Y, Li Y, Wu R, Wang Y, Li P, Jiang T, Wang K, Liu Y, Cheng Z. Epithelial and immune transcriptomic characteristics and possible regulatory mechanisms in asthma exacerbation: insights from integrated studies. Front Immunol 2025; 16:1512053. [PMID: 39917297 PMCID: PMC11798785 DOI: 10.3389/fimmu.2025.1512053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Background Asthma exacerbation significantly contribute to disease mortality and result in heightened health care expenditures. This study was aimed at gaining important new insights into the heterogeneity of epithelial and immune cells and elucidating key regulatory genes involved in the pathogenesis of asthma exacerbation. Methods Functional enrichment, pseudotime, metabolism and cell-cell communication analyses of epithelial cells and immune cells in single-cell RNA sequencing (scRNA-seq) dataset were applied. Immune infiltration analysis was performed in bulk RNA sequencing (bulk RNA-seq) dataset. Key regulatory genes were obtained by taking the intersection of the differentially expressed genes (DEGs) between control and asthma group in epithelial cells, immune cells and bulk RNA-seq data. Asthma animal and in vitro cell line models were established to verify the key regulatory genes expression by employing quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results ScRNA-seq analysis identified 7 epithelial subpopulations and 14 distinct immune cell types based on gene expression profiles. Further analysis demonstrated that these cells manifested high heterogeneity at the levels of functional variations, dynamics, communication patterns and metabolic changes. Notably, TMPRSS11A, TUBA1A, SCEL, ICAM4, TMPRSS11B, IGFBP2, CLC, NFAM1 and F13A1 were identified as key regulatory genes of asthma. The results of the qRT-PCR demonstrated that the 9 key regulatory genes were involved in asthma. Conclusions We systematically explored epithelial and immune characteristics in asthma exacerbation and identified 9 key regulatory genes underlying asthma occurrence and progression, which may be valuable for providing new insights into the cellular and molecular mechanisms driving asthma exacerbations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhe Cheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, He’nan, China
| |
Collapse
|
15
|
Xu J, Yao Y, Zhuang Q, Li Z, Zhang M, Wang S, Hu H, Ye J. Characterization of a chitinase from Trichinella spiralis and its immunomodulatory effects on allergic airway inflammation in mice. Parasit Vectors 2025; 18:6. [PMID: 39806495 PMCID: PMC11730484 DOI: 10.1186/s13071-024-06656-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND A fundamental tenet of the hygiene theory is the inverse association between helminth infections and the emergence of immune-mediated diseases. Research has been done to clarify the processes by which helminth-derived molecules can inhibit immunological disorders. This study aimed to evaluate the ability of Trichinella spiralis chitinase (Ts-chit) to ameliorate the symptoms of allergic airway inflammation. METHODS Recombinant Trichinella spiralis chitinase (rTs-chit) was expressed in Escherichia coli BL21, and its structural homology to murine acidic mammalian chitinase (AMCase) was comprehensively analyzed. The expression of Ts-chit was examined across all T. spiralis life stages. To explore its immunomodulatory potential, a murine model of allergen-induced airway inflammation was established. The effects of rTs-chit were evaluated by assessing airway hyperresponsiveness and cytokine profiles in bronchoalveolar lavage fluid and performing detailed histopathological and immunohistochemical analyses. RESULTS Recombinant Ts-chit (rTs-chit) was successfully expressed in E. coli BL21, showing strong structural similarity to murine acidic mammalian chitinase (AMCase). Expression profiling revealed that Ts-chit is present throughout all stages of the T. spiralis life cycle. In an allergic airway inflammation model, rTs-chit reduced weight loss and lung inflammation, lowering inflammatory cell infiltration and Th2 cytokines (IL-4, IL-5, IL-13) while increasing the immunosuppressive cytokine IL-10. Additionally, rTs-chit treatment decreased the expression of GATA3, arginase-1, MCP-1, CCL-11, and AMCase, along with reducing OVA-specific IgE, IgG, and IgG1 levels, suggesting its potential as an immunomodulatory agent. CONCLUSIONS This study highlights rTs-chit's potential as a therapeutic agent for allergic airway diseases, leveraging its structural similarity to host chitinases to regulate Th2 responses and inflammatory pathways. The findings provide new insights into helminth-derived proteins as promising candidates for immune-based therapies.
Collapse
Affiliation(s)
- Jia Xu
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China
| | - Ye Yao
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China
| | - Qisheng Zhuang
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China
| | - Zixuan Li
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China
| | - Min Zhang
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China
| | - Shouan Wang
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China
| | - Hongxin Hu
- The Affiliated Hospital of Putian University, Putian City, 351100, Fujian Province, China.
| | - Jianbin Ye
- School of Basic Medicine Science, Fujian Province, Putian University, Key Laboratory of Translational Tumor Medicine in , Putian City, 351100, Fujian Province, China.
- School of Pharmacy, Fujian Medical University, Fuzhou City, 350004, Fujian Province, China.
- School of Pharmacy, Putian University, Putian City, 351100, Fujian Province, China.
| |
Collapse
|
16
|
Wang W, Chen Z, Cui K, Chen N, Gao Q. Transforming growth factor-β3/Smad2/Smad3 signaling pathway inhibition and autophagy by the Yunpi-Xiefei-Huatan decoction ameliorated airway inflammation and mucus hypersecretion in asthmatic rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119125. [PMID: 39581284 DOI: 10.1016/j.jep.2024.119125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Yunpi-Xiefei-Huatan decoction (YXHD) is a traditional Chinese medicine that can improve asthma-related symptoms, including cough, phlegm in the throat, and shortness of breath. However, the YXHD mechanism on asthma has not yet been elucidated. STUDY AIM The aim of this study is to investigate the effect of YXHD on airway inflammation, mucus hypersecretion, and autophagy modulation in asthma. MATERIALS AND METHODS The YXHD chemical constituents were observed and analyzed using high-performance liquid chromatography-mass spectrometry. Ovalbumin sensitization and stimulation were used to establish an asthma rat model. A total of 80 Sprague-Dawley (SD) rats were segmented into eight groups at random: a Normal (NC) group, a Model (Mod) group, a YXHD low-dose group (10 g/kg/d), a YXHD moderate-dose group (20 g/kg/d), a YXHD high-dose group (40 g/kg/d), a Rapamycin group (4 mg/kg/d), a 3-methyladenine (3-MA) group (15 mg/kg/d), and a Dexamethasone (DEX) group (0.5 mg/kg/d). Whole-body plethysmography (WBP) detection was used to evaluate airway hyperresponsiveness. An enzyme-linked immunosorbent assay (ELISA) was used to detect inflammatory factors in the peripheral blood. Inflammatory cells in the bronchoalveolar lavage fluid (BALF) were also counted. Pathological changes in the lung tissues were marked using hematoxylin and eosin (H&E) staining and periodic acid-Schiff (PAS) staining. The localization of MUC5AC and the co-localization of LC3B + MUC5AC were observed using immunofluorescence. The expressions of autophagy and the TGF-β3/Smad2/Smad3 pathway in the lung tissues were detected using a Western blot assay (WB) and qPCR, and the autophagosomes in the lung tissues were detected using a transmission electron microscope (TEM). RESULTS Twenty signal peaks were identified using ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) technology. The TGF-β3/Smad2/Smad3 signal pathway activation was induced using ovalbumin (OVA) exposure in the rats. The upregulated expression of autophagy, enhanced MUC5AC fluorescence and LC3B fluorescence, and their co-localized expression in the airway epithelium indicated inflammatory cell infiltration and excessive mucus secretion in the lungs. This resulted in airway hyper-responsiveness. The YXHD inhibited the activation of the TGF-β3/Smad2/Smad3 signaling pathway, and autophagy expression reduced inflammatory factors, abnormal mucus secretion, and airway hyperresponsiveness. CONCLUSION The YXHD improved lung function, relieved lung inflammation, and inhibited airway mucus secretions in asthmatic rat models. Its mechanism may have been related to the blockage of the TGF-β3/Smad2/Smad3 signaling pathway and autophagy downregulation.
Collapse
Affiliation(s)
- Wenzhou Wang
- Guizhou University of Traditional Chinese Medicine, Guizhou, 550000, China.
| | - Zhu Chen
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guizhou, 550000, China.
| | - Kainan Cui
- Guizhou University of Traditional Chinese Medicine, Guizhou, 550000, China.
| | - Na Chen
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guizhou, 550000, China
| | - Qianqian Gao
- Guizhou University of Traditional Chinese Medicine, Guizhou, 550000, China.
| |
Collapse
|
17
|
Shan B, Huo Y, Guo Z, Li Q, Pan Z, Li Q, Du X. miR-184, a downregulated ovary-elevated miRNA transcriptionally activated by SREBF2, exerts anti-apoptotic properties in ovarian granulosa cells through inducing SMAD3 expression. Cell Death Dis 2024; 15:892. [PMID: 39695116 DOI: 10.1038/s41419-024-07286-1] [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: 07/13/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
Follicular atresia is the primary threat to female fertility. miRNAs are dysregulated in granulosa cells (GCs) during follicular atresia, and have emerged as crucial regulators of the initiation and progression of follicular atresia. However, the downregulated ovary-elevated (OE) miRNAs and their biological functions in ovary remain elusive. Here, 13 downregulated OE miRNAs were systematically identified by integrating tissue expression high-throughput data and comparative transcriptome analyses, among which miR-184 was specifically highly expressed in ovary but dramatically downregulated during follicular atresia. Low miR-184 levels were also positively correlated with follicular atresia. Based on the in vitro GC and follicle culture system, we found that miR-184 suppressed GC apoptosis and follicular atresia. Mechanistically, miR-184 induced SMAD3 transcription by acting as a saRNA, and also stabilized SMAD3 mRNA by directly binding to its 5'-UTR, which promoted TGF-β pathway activity and its anti-apoptotic effect. In addition, miR-184 was transcribed independently of host gene, which was activated by SREBF2 in an H3K4me3-dependent manner. Comparative analysis revealed that SREBF2 expression and H3K4me3 enrichment on miR-184 promoter in GCs from atretic follicles were dramatically reduced, which leads to the downregulation of miR-184 during follicular atresia. Moreover, the expression pattern, function, target, and regulatory mechanism of miR-184 among mammals are highly conserved and universal. Taken together, our findings demonstrate that miR-184, transcriptionally activated by SREBF2 in an H3K4me3-dependent manner, exerts anti-atretic effects by inducing SMAD3 expression, highlighting that it is a promising regulator for improving follicular development, ovarian health and female fertility.
Collapse
Affiliation(s)
- Baosen Shan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yangan Huo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhennan Guo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qiqi Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- College of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Zhenjiang, 212400, China
| | - Zengxiang Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
18
|
Ortiz-Melo MT, Campos JE, Sánchez-Guzmán E, Herrera-Aguirre ME, Castro-Muñozledo F. Regulation of corneal epithelial differentiation: miR-141-3p promotes the arrest of cell proliferation and enhances the expression of terminal phenotype. PLoS One 2024; 19:e0315296. [PMID: 39642122 PMCID: PMC11623785 DOI: 10.1371/journal.pone.0315296] [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: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 12/08/2024] Open
Abstract
In recent years, different laboratories have provided evidence on the role of miRNAs in regulation of corneal epithelial metabolism, permeability and wound healing, as well as their alteration after surgery and in some ocular pathologies. We searched the available databases reporting miRNA expression in the human eye, looking for miRNAs highly expressed in central cornea, which could be crucial for maintenance of the epithelial phenotype. Using the rabbit RCE1(5T5) cell line as a model of corneal epithelial differentiation, we describe the participation of miR-141-3p as a possible negative regulator of the proliferative/migratory phenotype in corneal epithelial cells. The expression of miR-141-3p followed a time course similar to the differentiation-linked KRT3 cytokeratin, being delayed 24-48 hours relative to PAX6 expression; such result suggested that miR-141-3p only regulates the expression of terminal phenotype. Inhibition of miR-141-3p led to increased cell proliferation and motility, and induced the expression of molecular makers characteristic of an Epithelial Mesenchymal Transition (EMT). Comparison between the transcriptional profile of cells in which miR-141-3p was knocked down, and the transcriptomes from proliferative non-differentiated and differentiated stratified epithelia suggest that miR-141-3p is involved in the expression of terminal differentiation mediating the arrest of cell proliferation and inhibiting the EMT in highly motile early differentiating cells.
Collapse
Affiliation(s)
- María Teresa Ortiz-Melo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
- Unidad de Investigación en Biomedicina (UBIMED), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Jorge E. Campos
- Unidad de Biotecnología y Prototipos (UBIPRO), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Erika Sánchez-Guzmán
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - María Esther Herrera-Aguirre
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - Federico Castro-Muñozledo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| |
Collapse
|
19
|
Mach N. The forecasting power of the mucin-microbiome interplay in livestock respiratory diseases. Vet Q 2024; 44:1-18. [PMID: 38606662 PMCID: PMC11018052 DOI: 10.1080/01652176.2024.2340003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 03/31/2024] [Indexed: 04/13/2024] Open
Abstract
Complex respiratory diseases are a significant challenge for the livestock industry worldwide. These diseases considerably impact animal health and welfare and cause severe economic losses. One of the first lines of pathogen defense combines the respiratory tract mucus, a highly viscous material primarily composed of mucins, and a thriving multi-kingdom microbial ecosystem. The microbiome-mucin interplay protects from unwanted substances and organisms, but its dysfunction may enable pathogenic infections and the onset of respiratory disease. Emerging evidence also shows that noncoding regulatory RNAs might modulate the structure and function of the microbiome-mucin relationship. This opinion paper unearths the current understanding of the triangular relationship between mucins, the microbiome, and noncoding RNAs in the context of respiratory infections in animals of veterinary interest. There is a need to look at these molecular underpinnings that dictate distinct health and disease outcomes to implement effective prevention, surveillance, and timely intervention strategies tailored to the different epidemiological contexts.
Collapse
Affiliation(s)
- Núria Mach
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| |
Collapse
|
20
|
Wang T, Yang Y, Yang M, Wang J, Hai M, Zhang W, Ma R, Ma G, Wang N, Qin Y, Wang P, Wan Q. Mechanisms of Mt.b Ag85B-Fc fusion protein against allergic asthma in mice by intranasal immunization. Int J Biol Macromol 2024; 283:137535. [PMID: 39549815 DOI: 10.1016/j.ijbiomac.2024.137535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/03/2024] [Accepted: 11/09/2024] [Indexed: 11/18/2024]
Abstract
Ag85B, the primary component of the Ag85 complex and an early secreted protein by Mycobacterium tuberculosis, has shown potential for the treatment of allergic asthma (AA) when used as a Fc-fusion protein. Administered via nasal immunization, Ag85B-Fc fusion protein significantly alleviated airway inflammation and reduced the proportions of some anaphylaxis related cells in lungs, with no significant histopathological injury to major organs in ovalbumin (OVA)-induced AA model mice. To investigate the underlying immune regulatory mechanisms of Ag85B protein, integrated proteomics and transcriptomics analyses were conducted, identifying the complement and coagulation cascades, and phagosomes as the two significantly enriched pathways at both gene and protein levels. Moreover, C3ar1 (C3aR1), Itgam (CD11b), Itgb2 (CD18), fgg (FGG), Cybb (CYBB), and Ncf4 (NCF4) were identified as core target factors that play a central role in allergic and asthmatic responses. Among them, C3aR1 and CR3 consisting of CD11b and CD18, are main complement receptors, indicating that Ag85B alleviated AA by regulating C3aR1- and CR3-mediated signal transduction. The validation results were consistent with the aforementioned findings. Overall, these results provide valuable insight into the application of mucosal immunotherapy in treatment of AA, positioning Ag85B-Fc fusion protein as a safe mucosal immunotherapeutic agent for AA.
Collapse
Affiliation(s)
- Tiansong Wang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Yuxin Yang
- Clinical College of Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Maosheng Yang
- School of Geographic Science and Planning, Ningxia University, Yinchuan, Ningxia, 750021, PR China
| | - Jing Wang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Maiyan Hai
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Wei Zhang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Rui Ma
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Gang Ma
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750003, PR China
| | - Na Wang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Yanyan Qin
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China
| | - Peng Wang
- Ningxia Key Laboratory of Craniocerebral Diseases, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China.
| | - Qiaofeng Wan
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China.
| |
Collapse
|
21
|
Romano Ibarra GS, Lei L, Yu W, Thurman AL, Gansemer ND, Meyerholz DK, Pezzulo AA, McCray PB, Thornell IM, Stoltz DA. IL-13 induces loss of CFTR in ionocytes and reduces airway epithelial fluid absorption. J Clin Invest 2024; 134:e181995. [PMID: 39255033 PMCID: PMC11527443 DOI: 10.1172/jci181995] [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: 04/22/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024] Open
Abstract
The airway surface liquid (ASL) plays a crucial role in lung defense mechanisms, and its composition and volume are regulated by the airway epithelium. The cystic fibrosis transmembrane conductance regulator (CFTR) is abundantly expressed in a rare airway epithelial cell type called an ionocyte. Recently, we demonstrated that ionocytes can increase liquid absorption through apical CFTR and basolateral barttin/chloride channels, while airway secretory cells mediate liquid secretion through apical CFTR channels and basolateral NKCC1 transporters. Th2-driven (IL-4/IL-13) airway diseases, such as asthma, cause goblet cell metaplasia, accompanied by increased mucus production and airway secretions. In this study, we investigate the effect of IL-13 on chloride and liquid transport performed by ionocytes. IL-13 treatment of human airway epithelia was associated with reduced epithelial liquid absorption rates and increased ASL volume. Additionally, IL-13 treatment reduced the abundance of CFTR-positive ionocytes and increased the abundance of CFTR-positive secretory cells. Increasing ionocyte abundance attenuated liquid secretion caused by IL-13. Finally, CFTR-positive ionocytes were less common in asthma and chronic obstructive pulmonary disease and were associated with airflow obstruction. Our findings suggest that loss of CFTR in ionocytes contributes to the liquid secretion observed in IL-13-mediated airway diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paul B. McCray
- Department of Internal Medicine
- Department of Pediatrics
- Pappajohn Biomedical Institute, and
| | - Ian M. Thornell
- Department of Internal Medicine
- Pappajohn Biomedical Institute, and
| | - David A. Stoltz
- Department of Internal Medicine
- Pappajohn Biomedical Institute, and
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
22
|
Zhu L, Jin Z. Exploring the causal relationship between the immune cell-inflammatory factor axis and lung cancer: a Mendelian randomization study. Front Oncol 2024; 14:1345765. [PMID: 39267832 PMCID: PMC11390355 DOI: 10.3389/fonc.2024.1345765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
Background Lung cancer is a major health burden globally and smoking is a well-known risk factor. It has been observed that chronic inflammation contributes to lung cancer progression, with immune cells and inflammatory cytokines implicated in tumor development. Clarifying the causal links between these immune components and lung cancer could enhance prevention and therapy. Methods We performed Mendelian randomization (MR) to explore causal connections between immune cells, inflammatory markers, and lung cancer risk, using genetic variants as instruments. Data from GWAS on these variables underpinned our MR analyses. Results Our findings indicated an inverse association between some immune cells and lung cancer risk, implying that more immune cells might be protective. NK T cells (CD16-CD56) and myeloid cells (HLA DR+ on CD33dim HLA DR+ CD11b+) had an inverse correlation with lung cancer risk. Furthermore, a direct relationship was observed between inflammatory cytokines and these immune cells. In contrast, IL-18 was inversely associated with lung cancer, while IL-13 showed a direct correlation. Conclusion The study underscores the role of immune and inflammatory factors in lung cancer. These insights could lead to new therapeutic strategies for combating lung cancer.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Zhi Jin
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
23
|
Wang G, Liu Z, Zhan J, Li R, Ye Y, Qi Y, Wei X, Zheng J. Serum exosomal miR-141-3p and miR-3679-5p levels associated with endotype and postoperative recurrence in chronic rhinosinusitis with nasal polyps. World Allergy Organ J 2024; 17:100938. [PMID: 39156601 PMCID: PMC11327455 DOI: 10.1016/j.waojou.2024.100938] [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: 03/18/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024] Open
Abstract
Background Chronic rhinosinusitis with nasal polyps (CRSwNP) is a chronic inflammatory disease. Exosomes were involved in different inflammatory diseases, but their roles in CRSwNP were poorly explored. Method We collected serum samples from 8 CRSwNP patients and 8 healthy controls (HC) and isolated their exosomes. MiRNA sequencing was performed for the exosome samples and differentially expressed miRNAs were identified. The top 3 differentially expressed exosomal miRNAs were confirmed in 2 validation cohorts, and their diagnostic values, predictive values for eosinophilic endotype, and recurrence were evaluated. Results Distinctive serum exosomal miRNA profiles were observed between CRSwNP and HC groups. Reverse transcription-polymerase chain reaction results in the first validation cohort revealed that serum exosomal miR-141-3p levels were increased, and miR-18a-5p and miR-3679-5p levels were decreased in the CRSwNP group compared to the HC group. These 3 miRNAs were further validated in the second validation cohort, and the results showed that miR-141-3p levels were elevated and miR-3679-5p levels were reduced in the serum exosomes in the eosinophilic CRSwNP group in comparison with the non-eosinophilic CRSwNP group. Receiver operating characteristic (ROC) curves highlighted that exosomal miR-141-3p and miR-3679-5p exhibited promising values for predicting the eosinophilic endotype. The patients in the second cohort were followed up for 2 years, and categorized into recurrence and non-recurrence groups. The serum exosomal miR-141-3p levels were increased and miR-3679-5p levels were reduced in the recurrence group in comparison with the non-recurrence group. ROC curves and Kaplan-Meier survival analysis revealed significant associations between the levels of exosomal miR-141-3p and miR-3679-5p and the risk of postoperative recurrence. Conclusions This study identified unique miRNA expression patterns in serum exosomes of CRSwNP patients. Circulating exosomal miR-141-3p and miR-3679-5p emerged as novel biomarkers for diagnosing CRSwNP, predicting the eosinophilic endotype, and forecasting postoperative recurrence.
Collapse
Affiliation(s)
- Gang Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zizhen Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jiabin Zhan
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Rui Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yi Ye
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yanyan Qi
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xin Wei
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jing Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
24
|
Miao Y, Zhong C, Bao S, Wei K, Wang W, Li N, Bai C, Chen W, Tang H. Impaired tryptophan metabolism by type 2 inflammation in epithelium worsening asthma. iScience 2024; 27:109923. [PMID: 38799558 PMCID: PMC11126962 DOI: 10.1016/j.isci.2024.109923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/16/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Previous researches indicate that tryptophan metabolism is critical to allergic inflammation and that indoleamine 2,3-dioxygenase 1 (IDO1), as a key enzyme, is known for its immunosuppressive properties. Therefore, we are aimed to explore whether tryptophan metabolism, especially IDO1, influences allergic asthma and clarify specific mechanism. With the analysis of clinical data, exploration in cell experiments, and verifying in HDM-induced asthma mice models, we finally found that in allergic asthma, low level of T1 cytokines along with high level of T2 cytokines inhibited the expression of IDO1 in airway epithelium, hampering the kynurenine pathway in tryptophan metabolism and decreasing the level of intracellular kynurenine (Kyn). As an endogenous ligand of aryl hydrocarbon receptor, Kyn regulated the expression of cystathionine-γ-lyase (CTH). Notably, in asthma models, enhancing either IDO1 or H2S relieved asthma, while inhibiting the activity of CTH exacerbated it. IDO1-Kyn-CTH pathway could be a potential target for treatment for allergic asthma.
Collapse
Affiliation(s)
- Yushan Miao
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Caiming Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shujun Bao
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Kunchen Wei
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Na Li
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Wei Chen
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Hao Tang
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
25
|
Zhang Y, Zhan L, Jiang X, Tang X. Comprehensive review for non-coding RNAs: From mechanisms to therapeutic applications. Biochem Pharmacol 2024; 224:116218. [PMID: 38643906 DOI: 10.1016/j.bcp.2024.116218] [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: 02/01/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Abstract
Non-coding RNAs (ncRNAs) are an assorted collection of transcripts that are not translated into proteins. Since their discovery, ncRNAs have gained prominence as crucial regulators of various biological functions across diverse cell types and tissues, and their abnormal functioning has been implicated in disease. Notably, extensive research has focused on the relationship between microRNAs (miRNAs) and human cancers, although other types of ncRNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are also emerging as significant contributors to human disease. In this review, we provide a comprehensive summary of our current knowledge regarding the roles of miRNAs, lncRNAs, and circRNAs in cancer and other major human diseases, particularly cancer, cardiovascular, neurological, and infectious diseases. Moreover, we discuss the potential utilization of ncRNAs as disease biomarkers and as targets for therapeutic interventions.
Collapse
Affiliation(s)
- YanJun Zhang
- College of Pharmacy and Traditional Chinese Medicine, Jiangsu College of Nursing, Huaian, Jiangsu, 223005, China
| | - Lijuan Zhan
- College of Pharmacy and Traditional Chinese Medicine, Jiangsu College of Nursing, Huaian, Jiangsu, 223005, China
| | - Xue Jiang
- College of Pharmacy and Traditional Chinese Medicine, Jiangsu College of Nursing, Huaian, Jiangsu, 223005, China.
| | - Xiaozhu Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
26
|
Kratchmarov R, Djeddi S, Dunlap G, He W, Jia X, Burk CM, Ryan T, McGill A, Allegretti JR, Kataru RP, Mehrara BJ, Taylor EM, Agarwal S, Bhattacharyya N, Bergmark RW, Maxfield AZ, Lee S, Roditi R, Dwyer DF, Boyce JA, Buchheit KM, Laidlaw TM, Shreffler WG, Rao DA, Gutierrez-Arcelus M, Brennan PJ. TCF1-LEF1 co-expression identifies a multipotent progenitor cell (T H2-MPP) across human allergic diseases. Nat Immunol 2024; 25:902-915. [PMID: 38589618 PMCID: PMC11849131 DOI: 10.1038/s41590-024-01803-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/06/2024] [Indexed: 04/10/2024]
Abstract
Repetitive exposure to antigen in chronic infection and cancer drives T cell exhaustion, limiting adaptive immunity. In contrast, aberrant, sustained T cell responses can persist over decades in human allergic disease. To understand these divergent outcomes, we employed bioinformatic, immunophenotyping and functional approaches with human diseased tissues, identifying an abundant population of type 2 helper T (TH2) cells with co-expression of TCF7 and LEF1, and features of chronic activation. These cells, which we termed TH2-multipotent progenitors (TH2-MPP) could self-renew and differentiate into cytokine-producing effector cells, regulatory T (Treg) cells and follicular helper T (TFH) cells. Single-cell T-cell-receptor lineage tracing confirmed lineage relationships between TH2-MPP, TH2 effectors, Treg cells and TFH cells. TH2-MPP persisted despite in vivo IL-4 receptor blockade, while thymic stromal lymphopoietin (TSLP) drove selective expansion of progenitor cells and rendered them insensitive to glucocorticoid-induced apoptosis in vitro. Together, our data identify TH2-MPP as an aberrant T cell population with the potential to sustain type 2 inflammation and support the paradigm that chronic T cell responses can be coordinated over time by progenitor cells.
Collapse
Affiliation(s)
- Radomir Kratchmarov
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Djeddi
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Garrett Dunlap
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenqin He
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaojiong Jia
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caitlin M Burk
- Center for Immunology and Inflammatory Diseases and Food Allergy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tessa Ryan
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alanna McGill
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica R Allegretti
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raghu P Kataru
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Babak J Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin M Taylor
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Shailesh Agarwal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Neil Bhattacharyya
- Massachusetts Eye & Ear Institute, Harvard Medical School, Boston, MA, USA
| | - Regan W Bergmark
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Alice Z Maxfield
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Lee
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel Roditi
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel F Dwyer
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua A Boyce
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen M Buchheit
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanya M Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wayne G Shreffler
- Center for Immunology and Inflammatory Diseases and Food Allergy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patrick J Brennan
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Xu X, Yin J, Yang Y, Liu H, Yu J, Luo X, Zhang Y, Song X. Advances in co-pathogenesis of the united airway diseases. Respir Med 2024; 225:107580. [PMID: 38484897 DOI: 10.1016/j.rmed.2024.107580] [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: 11/15/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/19/2024]
Abstract
According to the concept of "united airway diseases", the airway is a single organ in which upper and lower airway diseases are commonly comorbid. A range of inflammatory factors have been found to play an important role in the chain reaction of upper and lower airway diseases. However, the amount of research on this concept remains limited. The underlying mechanism of the relationship between typical diseases of the united airway, such as asthma, allergic rhinitis, and chronic sinusitis, also needs to be further explored. This review highlights the interaction between upper and lower respiratory diseases gathered from epidemiological, histoembryology, neural mechanistic, microbiological, and clinical studies, revealing the relationship between the upper and lower respiratory tracts.
Collapse
Affiliation(s)
- Xinjun Xu
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Jiali Yin
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Yujuan Yang
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Huifang Liu
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China; The 2nd School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong, China
| | - Jingyi Yu
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China
| | - Xianghuang Luo
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China; School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
| | - Yu Zhang
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China.
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery. Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, China.
| |
Collapse
|
28
|
Li J, Hong X, Jiang M, Kho AT, Tiwari A, Wang AL, Chase RP, Celedón JC, Weiss ST, McGeachie MJ, Tantisira KG. A novel piwi-interacting RNA associates with type 2-high asthma phenotypes. J Allergy Clin Immunol 2024; 153:695-704. [PMID: 38056635 DOI: 10.1016/j.jaci.2023.10.032] [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: 02/23/2023] [Revised: 10/14/2023] [Accepted: 10/25/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Piwi-interacting RNAs (piRNAs), comprising the largest noncoding RNA group, regulate transcriptional processes. Whether piRNAs are associated with type 2 (T2)-high asthma is unknown. OBJECTIVE We sought to investigate the association between piRNAs and T2-high asthma in childhood asthma. METHODS We sequenced plasma samples from 462 subjects in the Childhood Asthma Management Program (CAMP) as the discovery cohort and 1165 subjects in the Genetics of Asthma in Costa Rica Study (GACRS) as a replication cohort. Sequencing reads were filtered first, and piRNA reads were annotated and normalized. Linear regression was used for the association analysis of piRNAs and peripheral blood eosinophil count, total serum IgE level, and long-term asthma exacerbation in children with asthma. Mediation analysis was performed to investigate the effect direction. We then ascertained if the circulating piRNAs were present in asthmatic airway epithelial cells in a Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo) public data set. RESULTS Fifteen piRNAs were significantly associated with eosinophil count in CAMP (P ≤ .05), and 3 were successfully replicated in GACRS. Eleven piRNAs were associated with total IgE in CAMP, and one of these was replicated in GACRS. All 22 significant piRNAs were identified in epithelial cells in vitro, and 6 of these were differentially expressed between subjects with asthma and healthy controls. Fourteen piRNAs were associated with long-term asthma exacerbation, and effect of piRNAs on long-term asthma exacerbation are mediated through eosinophil count and serum IgE level. CONCLUSION piRNAs are associated with peripheral blood eosinophils and total serum IgE in childhood asthma and may play important roles in T2-high asthma.
Collapse
Affiliation(s)
- Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Mingye Jiang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Alvin T Kho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Computational Health Informatics Program, Boston Children's Hospital, Boston, Mass
| | - Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Alberta L Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Robert P Chase
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Juan C Celedón
- Division of Pulmonary Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pa
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Partners Personalized Medicine, Partners Healthcare, Boston, Mass
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Kelan G Tantisira
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Department of Pediatrics, Division of Respiratory Medicine, University of California-San Diego, La Jolla, Calif.
| |
Collapse
|
29
|
Zhai Z, Shao L, Lu Z, Yang Y, Wang J, Liu Z, Wang H, Zheng Y, Lu H, Song X, Zhang Y. Characteristics of mucin hypersecretion in different inflammatory patterns based on endotypes of chronic rhinosinusitis. Clin Transl Allergy 2024; 14:e12334. [PMID: 38282195 PMCID: PMC10802810 DOI: 10.1002/clt2.12334] [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: 09/17/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is usually accompanied by mucin hypersecretion that can lead to mucus accumulation and impair nasal mucociliary clearance, thus exacerbating airway inflammation. Abnormal mucin hypersecretion is regulated by different T helper (Th) cytokines, which are associated with different endotype-driven inflammatory responses. Therefore, it is of great significance to understand how these factors regulate mucin hypersecretion to provide precise treatment strategies for different endotypes of CRS. BODY: Thus far, the most common endotypes of CRS are classified as type 1, type 2, or type 3 immune responses based on innate and adaptive cell-mediated effector immunity, and the representative Th cytokines in these immune responses, such as IFN-γ, TNF-α, IL-4, IL-5, IL-13, IL-10, IL-17, and IL-22, play an important regulatory role in mucin secretion. We reviewed all the related literature in the PubMed database to determine the expression of these Th cytokines in CRS and the role they play in the regulation of mucin secretion. CONCLUSION We believe that the main Th cytokines involved in specific endotypes of CRS play a key role in regulating abnormal mucin secretion, which contributes to better understanding of the pathogenesis of CRS and provides therapeutic targets for airway inflammatory diseases associated with mucin hypersecretion.
Collapse
Affiliation(s)
- Zhaoxue Zhai
- Second Clinical Medicine CollegeBinzhou Medical UniversityYantaiChina
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Liting Shao
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Zhaoyang Lu
- Second Clinical Medicine CollegeBinzhou Medical UniversityYantaiChina
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Yujuan Yang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| | - Jianwei Wang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| | - Zhen Liu
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Huikang Wang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Yang Zheng
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Haoran Lu
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Xicheng Song
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| | - Yu Zhang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| |
Collapse
|
30
|
Bolger GB. Therapeutic Targets and Precision Medicine in COPD: Inflammation, Ion Channels, Both, or Neither? Int J Mol Sci 2023; 24:17363. [PMID: 38139192 PMCID: PMC10744217 DOI: 10.3390/ijms242417363] [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: 11/13/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The development of a wider range of therapeutic options is a key objective in drug discovery for chronic obstructive pulmonary disease (COPD). Fundamental advances in lung biology have the potential to greatly expand the number of therapeutic targets in COPD. The recently reported successful Phase 3 clinical trial of the first biologic agent for COPD, the monoclonal antibody dupilumab, adds additional support to the importance of targeting inflammatory pathways in COPD. However, numerous other cellular mechanisms are important targets in COPD therapeutics, including airway remodeling, the CFTR ion channel, and mucociliary function. Some of these emerging targets can be exploited by the expanded use of existing COPD drugs, such as roflumilast, while targeting others will require the development of novel molecular entities. The identification of additional therapeutic targets and agents has the potential to greatly expand the value of using clinical and biomarker data to classify COPD into specific subsets, each of which can be predictive of an enhanced response to specific subset(s) of targeted therapies. The author reviews established and emerging drug targets in COPD and uses this as a framework to define a novel classification of COPD based on therapeutic targets. This novel classification has the potential to enhance precision medicine in COPD patient care and to accelerate clinical trials and pre-clinical drug discovery efforts.
Collapse
Affiliation(s)
- Graeme B Bolger
- BZI Pharma LLC, 1500 1st Ave N., Unit 36, Birmingham, AL 35203-1872, USA
| |
Collapse
|
31
|
Yang R, Wu X, Gounni AS, Xie J. Mucus hypersecretion in chronic obstructive pulmonary disease: From molecular mechanisms to treatment. J Transl Int Med 2023; 11:312-315. [PMID: 38130649 PMCID: PMC10732574 DOI: 10.2478/jtim-2023-0094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Affiliation(s)
- Ruonan Yang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, Hubei Province, China
| | - Xiaojie Wu
- Department of Respiratory and Critical Care Medicine, Wuhan NO. 1 Hospital, Wuhan Hospital of traditional Chinese and Western Medicine, Wuhan430022, Hubei Province, China
| | - Abdelilah Soussi Gounni
- Department of Immunology, Faculty of Medicine, University of Manitoba, ManitobaR3E 0W3, Canada
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, Hubei Province, China
| |
Collapse
|
32
|
Wang J, Li J, He Y, Huang X, Feng J, Liu L, Liu Y, Jiang X, Jia J. The SIRT3 activator ganoderic acid D regulates airway mucin MUC5AC expression via the NRF2/GPX4 pathway. Pulm Pharmacol Ther 2023; 83:102262. [PMID: 37879430 DOI: 10.1016/j.pupt.2023.102262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
PURPOSE The expression of MUC5AC, a highly prevalent airway mucin, is regulated by stimulatory factors such as oxidative stress. Ganoderic acid D (GAD) activates mitochondrial deacetylase SIRT3. SIRT3 regulates mitochondrial function through deacetylation of mitochondrial proteins, thereby playing a significant role in alleviating oxidative stress-related diseases. Therefore, this study aimed to investigate the mechanisms and rationale underlying the regulation of MUC5AC expression by GAD. METHODS Human airway epithelial cells (NCI-H292) were exposed to pyocyanin (PCN) to establish an in vitro cell model of airway mucus hypersecretion. The expression of SIRT3, MUC5AC, and NRF2 pathway proteins in cells was assessed. Cellular mitochondrial morphology and oxidative stress markers were analyzed. C57BL/6 mice were induced with Pseudomonas aeruginosa (PA) to establish an in vivo mouse model of airway mucus hypersecretion. The expression of SIRT3 and MUC5AC in the airways was examined. In addition, the differential expression of target genes in the airway epithelial tissues of patients with chronic obstructive pulmonary disease (COPD) was analyzed using publicly available databases. RESULTS The results revealed a significant upregulation of MUC5AC expression and a significant downregulation of SIRT3 expression in relation to airway mucus hypersecretion. GAD inhibited the overexpression of MUC5AC in PCN-induced NCI-H292 cells and PA-induced mouse airways by upregulating SIRT3. GAD activated the NRF2/GPX4 pathway and inhibited PCN-induced oxidative stress and mitochondrial morphological changes in NCI-H292 cells. However, ML385 inhibited the regulatory effects of GAD on MUC5AC expression. CONCLUSION The SIRT3 activator GAD downregulated MUC5AC expression, potentially through activation of the NRF2/GPX4 pathway. Accordingly, GAD may be a potential treatment approach for airway mucus hypersecretions.
Collapse
Affiliation(s)
- Jiancheng Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiayao Li
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yingying He
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaochun Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yulin Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, Sichuan Province, China.
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
33
|
Park JM, Park JW, Lee J, Kim SH, Seo DY, Ahn KS, Han SB, Lee JW. Aromadendrin inhibits PMA-induced cytokine formation/NF-κB activation in A549 cells and ovalbumin-induced bronchial inflammation in mice. Heliyon 2023; 9:e22932. [PMID: 38125474 PMCID: PMC10730751 DOI: 10.1016/j.heliyon.2023.e22932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Hyperproduction of immune cell-derived inflammatory molecules and recruitment of immune cells promote the development of allergic asthma (AA). Aromadendrin (ARO) has various biological properties including anti-inflammatory effects. In this study, we evaluated the ameliorative effects of ARO on the development of AA in vitro and in vivo. Phorbol 12-myristate 13-acetate (PMA, 100 nM) was used to induce inflammation in A549 airway epithelial cells. The cohesion of A549 and eosinophil EOL-1 cells was studied. Ovalbumin (30 or 60 μg)/Alum (3 mg) mixture was adapted for AA induction in mice. ARO (5 or 10 mg/kg, p. o.) was administered to mice to investigate its ameliorative effect on AA development. Enzyme-linked immunosorbent assay, western blotting, and hematoxylin and eosin/periodic acid Schiff staining were performed to study the ameliorative effect of ARO on bronchial inflammation. In PMA-stimulated A549 cells, the upregulation of cytokines (interleukin [IL]-1β/IL-6/tumor necrosis factor alpha [TNF-α]/monocyte chemoattractant protein [MCP]-1]) and nuclear factor kappa B (NF-κB) activation was effectively reduced by ARO pretreatment. ARO suppressed the adhesion of A549 cells and eosinophils. In ovalbumin-induced AA mice, the levels of cells, such as eosinophils, Th2 cytokines, MCP-1 in bronchoalveolar lavage fluid, IgE in serum, and inducible nitric oxide synthase/cyclooxygenase-2 expression in the lung tissue were upregulated, which were all suppressed by ARO. In addition, the increase in cell inflow and mucus formation in the lungs of AA mice was reversed by ARO as per histological analysis. ARO also modulated NF-κB activation in the lungs of AA mice. Overall, the anti-inflammatory properties of ARO in vitro/in vivo studies of AA were notable. Thus, ARO has a modulatory effect on bronchial inflammation and may be a potential adjuvant for AA treatment.
Collapse
Affiliation(s)
- Jin-Mi Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Ji-Won Park
- Practical Research Division, Honam National Institute of Biological Resources (HNIBR), 99, Gohadoan-gil, Mokpo-si, Jeollanam-do, 58762, Republic of Korea
| | - Juhyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Seung-Ho Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Da-Yun Seo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| |
Collapse
|
34
|
Yang M, Sun L, Zhu D, Meng C, Sha J. Recent advances in understanding the effects of T lymphocytes on mucosal barrier function in allergic rhinitis. Front Immunol 2023; 14:1224129. [PMID: 37771581 PMCID: PMC10523012 DOI: 10.3389/fimmu.2023.1224129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/27/2023] [Indexed: 09/30/2023] Open
Abstract
Allergic rhinitis is a non-infectious chronic inflammatory disease of the nasal mucosa that affects T cells and their cytokines. T cells play significant roles in the development of allergic inflammatory diseases by orchestrating mechanisms underlying innate and adaptive immunity. Although many studies on allergic rhinitis have focused on helper T cells, molecular makeup, and pathogenesis-related transduction pathways, pathological mechanisms have not yet been completely explored. Recent studies have suggested that T cell status may play an important role in the interaction between T cells and the nasal mucosal barrier in allergic rhinitis. This study aimed to explore the interactions between T cells and nasal mucosal barriers in allergic rhinitis and to review the therapeutic modalities of pertinent biological agents involving T cells.
Collapse
Affiliation(s)
- Maolin Yang
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Liwei Sun
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases (20190901003JC), Changchun, China
| | - Dongdong Zhu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases (20190901003JC), Changchun, China
| | - Cuida Meng
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases (20190901003JC), Changchun, China
| | - Jichao Sha
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic Diseases (20190901003JC), Changchun, China
| |
Collapse
|
35
|
Guz M, Jeleniewicz W, Cybulski M. Interactions between circRNAs and miR-141 in Cancer: From Pathogenesis to Diagnosis and Therapy. Int J Mol Sci 2023; 24:11861. [PMID: 37511619 PMCID: PMC10380543 DOI: 10.3390/ijms241411861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
The function of non-coding RNAs (ncRNAs) in the pathogenesis and development of cancer is indisputable. Molecular mechanisms underlying carcinogenesis involve the aberrant expression of ncRNAs, including circular RNAs (circRNAs), and microRNAs (miRNAs). CircRNAs are a class of single-stranded, covalently closed RNAs responsible for maintaining cellular homeostasis through their diverse functions. As a part of the competing endogenous RNA (ceRNAs) network, they play a central role in the regulation of accessibility of miRNAs to their mRNA targets. The interplay between these molecular players is based on the primary role of circRNAs that act as miRNAs sponges, and the circRNA/miRNA imbalance plays a central role in different pathologies including cancer. Herein, we present the latest state of knowledge about interactions between circRNAs and miR-141, a well-known member of the miR-200 family, in malignant transformation, with emphasis on the biological role of circRNA/miR-141/mRNA networks as a future target for novel anti-cancer therapies.
Collapse
Affiliation(s)
- Małgorzata Guz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Witold Jeleniewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Marek Cybulski
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
36
|
Kierbiedź-Guzik N, Sozańska B. miRNAs as Modern Biomarkers in Asthma Therapy. Int J Mol Sci 2023; 24:11499. [PMID: 37511254 PMCID: PMC10380449 DOI: 10.3390/ijms241411499] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Asthma is a chronic inflammatory disease of the airways characterized by shortness of breath, chest tightness, coughing, and wheezing. For several decades (approximately 30 years), miRNAs and their role in asthma have been of constant interest among scientists. These small, non-coding RNA fragments, 18-25 nucleotides long, regulate gene expression at the post-transcriptional level by binding to the target mRNA. In this way, they affect several biological processes, e.g., shaping airway structures, producing cytokines and immune mediators, and controlling defense mechanisms. Publications confirm their potential role in the diagnosis and monitoring of the disease, but only some articles address the use of miRNAs in the treatment of asthma. The following paper reviews the latest available studies and presents miRNAs as a useful tool for predicting the effectiveness of the included treatment, early diagnosis of exacerbations, and in assessing patient compliance for different groups of drugs used in asthma. The latest known pathways underlying the pathogenesis of the disease, which are associated with a change in miRNA expression, may be precise targets of therapeutic activity in the future.
Collapse
Affiliation(s)
- Natalia Kierbiedź-Guzik
- 14th Paediatric Ward-Pulmonology and Allergology, J. Gromkowski Provincial Specialist Hospital, ul. Koszarowa 5, 51-149 Wrocław, Poland
| | - Barbara Sozańska
- 1st Department and Clinic of Paediatrics, Allergology and Cardiology Wrocław Medical University, ul. Chałubińskiego 2a, 50-368 Wrocław, Poland
| |
Collapse
|
37
|
Lee RE, Reidel B, Nelson MR, Macdonald JK, Kesimer M, Randell SH. Air-Liquid interface cultures to model drug delivery through the mucociliary epithelial barrier. Adv Drug Deliv Rev 2023; 198:114866. [PMID: 37196698 PMCID: PMC10336980 DOI: 10.1016/j.addr.2023.114866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/23/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
Epithelial cells from mucociliary portions of the airways can be readily grown and expanded in vitro. When grown on a porous membrane at an air-liquid interface (ALI) the cells form a confluent, electrically resistive barrier separating the apical and basolateral compartments. ALI cultures replicate key morphological, molecular and functional features of the in vivo epithelium, including mucus secretion and mucociliary transport. Apical secretions contain secreted gel-forming mucins, shed cell-associated tethered mucins, and hundreds of additional molecules involved in host defense and homeostasis. The respiratory epithelial cell ALI model is a time-proven workhorse that has been employed in various studies elucidating the structure and function of the mucociliary apparatus and disease pathogenesis. It serves as a critical milestone test for small molecule and genetic therapies targeting airway diseases. To fully exploit the potential of this important tool, numerous technical variables must be thoughtfully considered and carefully executed.
Collapse
Affiliation(s)
- Rhianna E Lee
- Marsico Lung Institute and Cystic Fibrosis Research Center, United States; Department of Cell Biology and Physiology, United States
| | - Boris Reidel
- Marsico Lung Institute and Cystic Fibrosis Research Center, United States; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Mark R Nelson
- Marsico Lung Institute and Cystic Fibrosis Research Center, United States
| | - Jade K Macdonald
- Marsico Lung Institute and Cystic Fibrosis Research Center, United States
| | - Mehmet Kesimer
- Marsico Lung Institute and Cystic Fibrosis Research Center, United States; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Scott H Randell
- Marsico Lung Institute and Cystic Fibrosis Research Center, United States; Department of Cell Biology and Physiology, United States.
| |
Collapse
|
38
|
Boboltz A, Kumar S, Duncan GA. Inhaled drug delivery for the targeted treatment of asthma. Adv Drug Deliv Rev 2023; 198:114858. [PMID: 37178928 PMCID: PMC10330872 DOI: 10.1016/j.addr.2023.114858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Asthma is a chronic lung disease affecting millions worldwide. While classically acknowledged to result from allergen-driven type 2 inflammatory responses leading to IgE and cytokine production and the influx of immune cells such as mast cells and eosinophils, the wide range in asthmatic pathobiological subtypes lead to highly variable responses to anti-inflammatory therapies. Thus, there is a need to develop patient-specific therapies capable of addressing the full spectrum of asthmatic lung disease. Moreover, delivery of targeted treatments for asthma directly to the lung may help to maximize therapeutic benefit, but challenges remain in design of effective formulations for the inhaled route. In this review, we discuss the current understanding of asthmatic disease progression as well as genetic and epigenetic disease modifiers associated with asthma severity and exacerbation of disease. We also overview the limitations of clinically available treatments for asthma and discuss pre-clinical models of asthma used to evaluate new therapies. Based on the shortcomings of existing treatments, we highlight recent advances and new approaches to treat asthma via inhalation for monoclonal antibody delivery, mucolytic therapy to target airway mucus hypersecretion and gene therapies to address underlying drivers of disease. Finally, we conclude with discussion on the prospects for an inhaled vaccine to prevent asthma.
Collapse
Affiliation(s)
- Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - Sahana Kumar
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States; Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW The study of microRNA in asthma has revealed a vibrant new level of gene regulation underlying asthma pathology. Several miRNAs have been shown to be important in asthma, influencing various biological mechanisms which lead to asthma pathology and symptoms. In addition, miRNAs have been proposed as biomarkers of asthma affection status, asthma severity, and asthma treatment response. We review all recent asthma-miRNA work, while also presenting comprehensive tables of all miRNA results related to asthma. RECENT FINDINGS We here reviewed 63 recent studies published reporting asthma and miRNA research, and an additional 14 reviews of the same. We summarized the information for both adult and childhood asthma, as well as research on miRNAs in asthma-COPD overlap syndrome (ACOs), and virus-induced asthma exacerbations. We attempted to present a comprehensive collection of recently published asthma-associated miRNAs as well as tables of all published asthma-related miRNA results.
Collapse
Affiliation(s)
- Rinku Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Tang W, Li M, Teng F, Cui J, Dong J, Wang W. Single-cell RNA-sequencing in asthma research. Front Immunol 2022; 13:988573. [PMID: 36524132 PMCID: PMC9744750 DOI: 10.3389/fimmu.2022.988573] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022] Open
Abstract
Asthma is a complex and heterogeneous disease with multicellular involvement, and knowledge gaps remain in our understanding of the pathogenesis of asthma. Efforts are still being made to investigate the immune pathogenesis of asthma in order to identify possible targets for prevention. Single cell RNA sequencing (scRNA-seq) technology is a useful tool for exploring heterogeneous diseases, identifying rare cell types and distinct cell subsets, enabling elucidation of key processes of cell differentiation, and understanding regulatory gene networks that predict immune function. In this article, we provide an overview of the importance of scRNA-seq for asthma research, followed by an in-depth discussion of the results in recent years, in order to provide new ideas for the pathogenesis, drug development and treatment of asthma.
Collapse
Affiliation(s)
- Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China,The Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Mihui Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China,The Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Fangzhou Teng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China,The Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jie Cui
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China,The Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China,The Institutes of Integrative Medicine, Fudan University, Shanghai, China,*Correspondence: Wenqian Wang, ; Jingcheng Dong,
| | - Wenqian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China,The Institutes of Integrative Medicine, Fudan University, Shanghai, China,*Correspondence: Wenqian Wang, ; Jingcheng Dong,
| |
Collapse
|
41
|
Boateng E, Kovacevic D, Oldenburg V, Rådinger M, Krauss-Etschmann S. Role of airway epithelial cell miRNAs in asthma. FRONTIERS IN ALLERGY 2022; 3:962693. [PMID: 36203653 PMCID: PMC9530201 DOI: 10.3389/falgy.2022.962693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/01/2022] [Indexed: 12/07/2022] Open
Abstract
The airway epithelial cells and overlying layer of mucus are the first point of contact for particles entering the lung. The severity of environmental contributions to pulmonary disease initiation, progression, and exacerbation is largely determined by engagement with the airway epithelium. Despite the cellular cross-talk and cargo exchange in the microenvironment, epithelial cells produce miRNAs associated with the regulation of airway features in asthma. In line with this, there is evidence indicating miRNA alterations related to their multifunctional regulation of asthma features in the conducting airways. In this review, we discuss the cellular components and functions of the airway epithelium in asthma, miRNAs derived from epithelial cells in disease pathogenesis, and the cellular exchange of miRNA-bearing cargo in the airways.
Collapse
Affiliation(s)
- Eistine Boateng
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Correspondence: Eistine Boateng
| | - Draginja Kovacevic
- DZL Laboratory for Experimental Microbiome Research, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Vladimira Oldenburg
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Madeleine Rådinger
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- DZL Laboratory for Experimental Microbiome Research, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
42
|
Jakwerth CA, Kitzberger H, Pogorelov D, Müller A, Blank S, Schmidt-Weber CB, Zissler UM. Role of microRNAs in type 2 diseases and allergen-specific immunotherapy. FRONTIERS IN ALLERGY 2022; 3:993937. [PMID: 36172292 PMCID: PMC9512106 DOI: 10.3389/falgy.2022.993937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 12/07/2022] Open
Abstract
MicroRNAs (miRs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases as well as their potential as biomarkers in allergen-specific treatment options. Their function as post-transcriptional regulators, controlling various cellular processes, is of high importance since any single miR can target multiple mRNAs, often within the same signalling pathway. MiRs can alter dysregulated expression of certain cellular responses and contribute to or cause, but in some cases prevent or repress, the development of various diseases. In this review article, we describe current research on the role of specific miRs in regulating immune responses in epithelial cells and specialized immune cells in response to various stimuli, in allergic diseases, and regulation in the therapeutic approach of allergen-specific immunotherapy (AIT). Despite the fact that AIT has been used successfully as a causative treatment option since more than a century, very little is known about the mechanisms of regulation and its connections with microRNAs. In order to fill this gap, this review aims to provide an overview of the current knowledge.
Collapse
|
43
|
Zhi W, Jiang S, Xu Z, An Y, Chen J, Li Y, Liu Y, Zhang H. Oxysophocarpine inhibits airway inflammation and mucus hypersecretion through JNK/AP-1 pathway in vivo and in vitro. Fitoterapia 2022; 162:105278. [PMID: 35970410 DOI: 10.1016/j.fitote.2022.105278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 02/07/2023]
Abstract
Asthma is a high-incidence disease in the world. Oxysophocarpine (OSC), a quinolizidine alkaloid displays various pharmacological functions including anti-inflammation, neuroprotective, anti-virus and antioxidant. Here, we established mice and cell asthmatic model to explore the effects of OSC for asthma treatment. Mice were sensitized and challenged with ovalbumin (OVA) and treated with OSC before challenge. Enzyme-linked immuno sorbent assay (ELISA), hematoxylin and eosin (H&E), periodic acid-schiff (PAS), tolonium chloride staining and immunohistochemical assay were performed. OSC treatment inhibited inflammatory cell infiltration and mucus secretion in the airway, reduced IgE level in mouse serum and decreased IL-4, IL-5 production in bronchoalveolar lavage fluid (BALF). OSC also reduced the spleen index to regulate immune function. Meanwhile, NCI-H292 cells were induced by lipopolysaccharide (LPS) to simulate airway epithelial injury. OSC pretreatment decreased the IL-6 and IL-8 cytokine levels, mucin 5 AC expression, and mucin 5 AC mRNA level in the cell model. Further, OSC suppressed the phosphorylation of c-Jun N-terminal kinase (JNK), and activator protein 1 (AP-1, Fos and Jun). These findings revealed that OSC alleviated bronchial asthma associated with JNK/AP-1 signaling pathway.
Collapse
Affiliation(s)
- Wenbing Zhi
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an 710003, PR China
| | - Shengnan Jiang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an 710003, PR China
| | - Zongren Xu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an 710003, PR China
| | - Yuye An
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Jing Chen
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Ye Li
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an 710003, PR China; Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an 710003, PR China.
| | - Hong Zhang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an 710003, PR China; Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China.
| |
Collapse
|
44
|
Wu L, Zhao W, Tang S, Chen R, Ji M, Yang X. Role of ILC2s in Solid Tumors: Facilitate or Inhibit? Front Immunol 2022; 13:886045. [PMID: 35720302 PMCID: PMC9203687 DOI: 10.3389/fimmu.2022.886045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are important mediators of type 2 immunity and play an important role in allergic diseases, helminth infections, and tissue fibrosis. However, the role of ILC2s in tumor immunity requires further elucidation. Studies over the past decade have reported that ILC2s play a promoting or suppressing role in different tumors. Here we reviewed the role of ILC2s in solid tumors demonstrating that ILC2s act as a crucial regulator in tumor immunity. We proposed that ILC2s could be an important predictor for tumor prognosis and a new therapeutic target after immunotherapy resistance. In conclusion, our study shed new light on modifying and targeting ILC2s for anti-tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Mei Ji
- *Correspondence: Mei Ji, ; Xin Yang,
| | - Xin Yang
- *Correspondence: Mei Ji, ; Xin Yang,
| |
Collapse
|
45
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
46
|
Tai J, Han M, Kim TH. Therapeutic Strategies of Biologics in Chronic Rhinosinusitis: Current Options and Future Targets. Int J Mol Sci 2022; 23:ijms23105523. [PMID: 35628333 PMCID: PMC9141505 DOI: 10.3390/ijms23105523] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/11/2022] [Accepted: 05/14/2022] [Indexed: 12/25/2022] Open
Abstract
Chronic rhinosinusitis is a chronic inflammatory disease of the upper airways, for which treatment options include medical or surgical therapy. However, there are limitations to conservative treatment strategies, such as the relapse of nasal polyps. In this review, we discuss the rising role of biomolecular mechanisms associated with various biologics that have been approved or are undergoing clinical trials to treat chronic rhinosinusitis. We also highlight the potential molecular therapeutic targets for managing and treating chronic rhinosinusitis.
Collapse
|
47
|
Gao X, Leung TF, Wong GWK, Ko WH, Cai M, He EJ, Chu IMT, Tsang MSM, Chan BCL, Ling J, Fan X, Lu L, Lam CWK, Wong CK. Meteorin-β/Meteorin like/IL-41 attenuates airway inflammation in house dust mite-induced allergic asthma. Cell Mol Immunol 2022; 19:245-259. [PMID: 34848868 PMCID: PMC8803866 DOI: 10.1038/s41423-021-00803-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/27/2021] [Indexed: 02/03/2023] Open
Abstract
We sought to examine the regulatory effect of Meteorin-β (Metrnβ)/Meteorin like (Metrnl)/IL-41 on lung inflammation in allergic asthma. We found that Metrnβ was elevated significantly in asthmatic patients and in mice with allergic asthma induced by house dust mite (HDM) extract. Upon exposure to HDM, Metrnβ was secreted predominantly by airway epithelial cells and inflammatory cells, including macrophages and eosinophils. The increased Metrnβ effectively blocked the development of airway hyperreactivity (AHR) and decreased inflammatory cell airway infiltration and type 2 cytokine production, which was associated with downregulated DC-mediated adaptive immune responses. Moreover, Metrnβ impaired the maturation and function of bone marrow-derived dendritic cells in vitro. Asthmatic mice adoptively transferred with dendritic cells isolated from Metrnβ-treated allergic mice displayed decreased AHR, airway inflammation, and lung injury. Metrnβ also displayed anti-inflammatory properties in immunodeficient SCID mice with allergic asthma and in in vitro 3D ALI airway models. Moreover, blockade of Metrnβ by anti-Metrnβ antibody treatment promoted the development of allergic asthma. These results revealed the unappreciated protective roles of Metrnβ in alleviating DC-mediated Th2 inflammation in allergic asthma, providing the novel treatment strategy of therapeutic targeting of Metrnβ in allergic asthma.
Collapse
Affiliation(s)
- Xun Gao
- grid.10784.3a0000 0004 1937 0482Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ting-Fan Leung
- grid.10784.3a0000 0004 1937 0482Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary Wing-Kin Wong
- grid.10784.3a0000 0004 1937 0482Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing-Hung Ko
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Mengyun Cai
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ellie Jiayi He
- grid.10784.3a0000 0004 1937 0482Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China ,grid.189967.80000 0001 0941 6502Emory College of Arts and Sciences, Emory University, Atlanta, GA USA
| | - Ida Miu-Ting Chu
- grid.10784.3a0000 0004 1937 0482Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- grid.10784.3a0000 0004 1937 0482Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China ,grid.10784.3a0000 0004 1937 0482Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- grid.10784.3a0000 0004 1937 0482Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiawei Ling
- grid.10784.3a0000 0004 1937 0482Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Fan
- grid.194645.b0000000121742757Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Liwei Lu
- grid.194645.b0000000121742757Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Christopher Wai-Kei Lam
- grid.259384.10000 0000 8945 4455Faculty of Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China. .,Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China. .,Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
48
|
Wu W, Gao J, Chen D, Chen G, Feng Y, Chang C, Chen S, Yi L, Zhen G. Epithelial microRNA-30a-3p targets RUNX2/HMGB1 axis to suppress airway eosinophilic inflammation in asthma. Respir Res 2022; 23:17. [PMID: 35093061 PMCID: PMC8800331 DOI: 10.1186/s12931-022-01933-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Background Type 2-high asthma is a prominent endotype of asthma which is characterized by airway eosinophilic inflammation. Airway epithelial cells play a critical role in the pathogenesis of asthma. Our previous miRNA profiling data showed that miR-30a-3p was downregulated in bronchial epithelial cells from asthma patients. We hypothesize that epithelial miR-30a-3p plays a role in asthma airway inflammation. Methods We measured miR‐30a-3p expression in bronchial brushings of asthma patients (n = 51) and healthy controls (n = 16), and analyzed the correlations between miR‐30a-3p expression and airway eosinophilia. We examined whether Runt-related transcription factor 2 (RUNX2) was a target of miR‐30a-3p and whether RUNX2 bound to the promoter of high mobility group box 1 (HMGB1) by using luciferase reporter assay and chromatin immunoprecipitation (ChIP)-PCR. The role of miR‐30a-3p was also investigated in a murine model of allergic airway inflammation. Results We found that miR-30a-3p expression were significantly decreased in bronchial brushings of asthma patients compared to control subjects. Epithelial miR-30a-3p expression was negatively correlated with parameters reflecting airway eosinophilia including eosinophils in induced sputum and bronchial biopsies, and fraction of exhaled nitric oxide in asthma patients. We verified that RUNX2 is a target of miR-30a-3p. Furthermore, RUNX2 bound to the promoter of HMGB1 and upregulated HMGB1 expression. RUNX2 and HMGB1 expression was both enhanced in airway epithelium and was correlated with each other in asthma patients. Inhibition of miR-30a-3p enhanced RUNX2 and HMGB1 expression, and RUNX2 overexpression upregulated HMGB1 in BEAS-2B cells. Intriguingly, airway overexpression of mmu-miR-30a-3p suppressed Runx2 and Hmgb1 expression, and alleviated airway eosinophilia in a mouse model of allergic airway inflammation. Conclusions Epithelial miR-30a-3p could possibly target RUNX2/HMGB1 axis to suppress airway eosinophilia in asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01933-x.
Collapse
|
49
|
Wang X, Chen H, Liu J, Gai L, Yan X, Guo Z, Liu F. Emerging Advances of Non-coding RNAs and Competitive Endogenous RNA Regulatory Networks in Asthma. Bioengineered 2021; 12:7820-7836. [PMID: 34635022 PMCID: PMC8806435 DOI: 10.1080/21655979.2021.1981796] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 12/31/2022] Open
Abstract
Asthma is a chronic inflammatory disease characterized by airway remodeling and bronchial hyperresponsiveness. A variety of effector cells and cytokines jointly stimulate the occurrence of inflammatory response in asthma. Although the pathogenesis of asthma is not entirely clear, the possible roles of non-coding RNAs (ncRNAs) have been recently demonstrated. NcRNAs are non-protein-coding RNA molecules, such as circular RNAs (circRNAs), long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), which are involved in the regulation of a variety of biological processes. Mounting studies have shown that ncRNAs play pivotal roles in the occurrence and progression of asthma via competing endogenous RNA (ceRNA) regulatory networks. However, the specific mechanism and clinical application of ncRNAs and ceRNA regulatory networks in asthma have not been fully elucidated, which are worthy of further investigation. This paper comprehensively summarized the current progress on the roles of miRNAs, lncRNAs, circRNAs, and ceRNA regulatory networks in asthma, which can provide a better understanding for the disease pathogenesis and is helpful for identifying novel biomarkers for asthma.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Clinical Medicine College, Weifang Medical University, WeifangChina
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| | - Hui Chen
- Clinical Medicine College, Weifang Medical University, WeifangChina
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| | - Jingjing Liu
- Clinical Medicine College, Weifang Medical University, WeifangChina
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| | - Linlin Gai
- Department of Central Laboratory, The First Affiliated Hospital of Weifang Medical University/Weifang People’s Hospital, WeifangChina
| | - Xinyi Yan
- Department of Central Laboratory, The First Affiliated Hospital of Weifang Medical University/Weifang People’s Hospital, WeifangChina
| | - Zhiliang Guo
- Department of Spine Surgery, The 80th Group Army Hospital of Chinese PLA, WeifangChina
| | - Fengxia Liu
- Department of Allergy, The First Affiliated Hospital of Weifang Medical University/ Weifang People’s Hospital, WeifangChina
| |
Collapse
|
50
|
Hu X, Shen Y, Zhao Y, Wang J, Zhang X, Tu W, Kaufman W, Feng J, Gao P. Epithelial Aryl Hydrocarbon Receptor Protects From Mucus Production by Inhibiting ROS-Triggered NLRP3 Inflammasome in Asthma. Front Immunol 2021; 12:767508. [PMID: 34868022 PMCID: PMC8634667 DOI: 10.3389/fimmu.2021.767508] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/28/2021] [Indexed: 02/05/2023] Open
Abstract
Background Despite long-standing recognition in the significance of mucus overproduction in asthma, its etiology remains poorly understood. Muc5ac is a secretory mucin that has been associated with reduced pulmonary function and asthma exacerbations. Objectives We sought to investigate the immunological pathway that controls Muc5ac expression and allergic airway inflammation in asthma. Methods Cockroach allergen-induced Muc5ac expression and aryl hydrocarbon receptor (AhR) signaling activation was examined in the human bronchial epithelial cells (HBECs) and mouse model of asthma. AhR regulation of Muc5ac expression, mitochondrial ROS (Mito-ROS) generation, and NLRP3 inflammasome was determined by AhR knockdown, the antagonist CH223191, and AhR-/- mice. The role of NLRP3 inflammasome in Muc5ac expression and airway inflammation was also investigated. Results Cockroach allergen induced Muc5ac overexpression in HBECs and airways of asthma mouse model. Increased expression of AhR and its downstream genes CYP1A1 and CYP1B1 was also observed. Mice with AhR deletion showed increased allergic airway inflammation and MUC5AC expression. Moreover, cockroach allergen induced epithelial NLRP3 inflammasome activation (e.g., NLRP3, Caspase-1, and IL-1β), which was enhanced by AhR knockdown or the antagonist CH223191. Furthermore, AhR deletion in HBECs led to enhanced ROS generation, particularly Mito-ROS, and inhibition of ROS or Mito-ROS subsequently suppressed the inflammasome activation. Importantly, inhibition of the inflammasome with MCC950, a NLRP3-specifc inhibitor, attenuated allergic airway inflammation and Muc5ac expression. IL-1β generated by the activated inflammasomes mediated cockroach allergen-induced Muc5ac expression in HBECs. Conclusions These results reveal a previously unidentified functional axis of AhR-ROS-NLRP3 inflammasome in regulating Muc5ac expression and airway inflammation.
Collapse
Affiliation(s)
- Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yingchun Shen
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yilin Zhao
- Department of Respiratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Ji Wang
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, Sichuan University, Chengdu, China
| | - Xin Zhang
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - William Kaufman
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Juntao Feng
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|