1
|
Ni H, Li T, Chen J, Wei Y, Xia M, Wang Q. Store-operated Ca 2+ entry contributes to the ASM phenotype transition in asthma. Exp Lung Res 2025; 51:23-37. [PMID: 40205756 DOI: 10.1080/01902148.2025.2486951] [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: 07/16/2024] [Revised: 01/31/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
AIM OF THE STUDY Phenotype modulation of airway smooth muscle cells (ASMC), characterized by a shift toward a more proliferative and synthetic phenotype from contractile cells, plays a crucial role in airway remodeling in asthma. STIM1 and Orai1, key components of store-operated Ca2+ entry (SOCE), have been demonstrated to enhance ASMC proliferation and migration. This study investigated the impact of STIM1/Orai1-mediated SOCE on ASMC phenotype transition and extracellular matrix (ECM) deposition in asthma. MATERIALS AND METHODS The ASMCs were treated with PDGF-BB and SOCE inhibitors. Immunocytochemistry staining, enzyme-linked immunosorbent assay, and western blot assay were employed to detect the ASMC's proliferation as well as the expressions of contractile proteins, inflammatory cytokines and ECM. Moreover, the effect of SOCE repression in ECM deposition were evaluated in an asthmatic mouse model. RESULTS ASMCs from airways of mice were treated with PDGF-BB to induce the 'proliferative/synthetic' phenotype. We observed elevated expressions of STIM1 and Orai1 in phenotype-switched ASMCs, along with enhanced SOCE. SKF-96365 and RO2959, which target of STIM1/Orai1, could significantly inhibit SOCE activation in ASMCs. Moreover, these SOCE inhibitors mitigated the elevated proliferation rate, decreased the secretion of inflammatory cytokines and restored the reduced levels of contractile proteins in phenotype-switched ASMCs induced by PDGF-BB. Furthermore, we observed that PDGF-BB-induced 'proliferative/synthetic' ASMCs exhibited increased production of ECM components, including collagen I and fibronectin, as well as metalloproteinases (MMPs) such as MMP2 and MMP9, all of which were effectively inhibited by SKF-96365 and RO2959. In vivo experiments also demonstrated that SOCE inhibitors decreased ECM deposition and MMPs production in the asthmatic mouse model. CONCLUSIONS These findings underscored the significant role of STIM1/Orai1-mediated SOCE in ASMC phenotype modulation and its impact on the excessive ECM deposition driven by ASMCs. Thus, our findings suggest that STIM1/Orai1-mediated SOCE may contribute to airway remodeling in asthma.
Collapse
Affiliation(s)
- Hangqi Ni
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, P.R. China
| | - Ting Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, P.R. China
| | - Junjun Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, P.R. China
| | - Yuying Wei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, P.R. China
| | - Mengling Xia
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, P.R. China
| | - Qing Wang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, P.R. China
| |
Collapse
|
2
|
Akenroye A, Boyce JA, Kita H. Targeting alarmins in asthma: From bench to clinic. J Allergy Clin Immunol 2025; 155:1133-1148. [PMID: 39855362 DOI: 10.1016/j.jaci.2025.01.017] [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: 09/20/2024] [Revised: 12/24/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Over the past 2 decades, mechanistic studies of allergic and type 2 (T2)-mediated airway inflammation have led to multiple approved therapies for the treatment of moderate-to-severe asthma. The approval and availability of these monoclonal antibodies targeting IgE, a T2 cytokine (IL-5) and/or cytokine receptors (IL-5Rα, IL-4Rα) has been central to the progresses made in the management of moderate-to-severe asthma over this period. However, there are persistent gaps in clinician's ability to provide precise care, given that many patients with T2-high asthma do not respond to IgE- or T2 cytokine-targeting therapies and that patients with T2-low asthma have few therapeutic options. The new frontier of precision medicine in asthma, as well as in other allergic diseases, includes the targeting of epithelium-derived cytokines known as alarmins, including thymic stromal lymphopoietin, IL-25, IL-33, and their receptors. The effects of these alarmins, which can act upstream of immune cells, involve both the innate and adaptive systems and hold potential for the treatment of both T2-high and -low disease. Tezepelumab, an anti-thymic stromal lymphopoietin antibody, has already been approved for the treatment of severe asthma. In this review, we discuss our current understanding of alarmin biology with a primary focus on allergic airway diseases. We link the mechanistic corollaries to the clinical implications and advances in drug development targeting alarmins, with a particular focus on currently approved treatments, those under study, and future potential targets in alarmin signaling pathways.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| | - Joshua A Boyce
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, the Department of Medicine, and the Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn
| |
Collapse
|
3
|
Chen Z, Liu X, Feng X, Lyu A, Zhou W. A systematic pharmacological strategy-based to decode the synergistic mechanism of 7,4'-dihydroxyflavone in combination with vitamin D3 against asthma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119513. [PMID: 39971017 DOI: 10.1016/j.jep.2025.119513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 7,4'-dihydroxyflavone (74DHF), extracted from Gancao (Rhizoma Glycyrrhizae), has demonstrated to mediate the asthma pathology, while Vitamin D3 (VD3) plays a role in asthma treatment due to its immunomodulatory effects. However, the potential molecular or systems mechanism of 74DHF in combination with VD3 against asthma has not yet been elucidated. AIM OF THE STUDY The current study not only deepens our understanding of the complex synergistic mechanism of 74DHF andVD3 against asthma but also proposes a promising strategy to promote the development of combination therapy. MATERIALS AND METHODS This study employed a systems pharmacology-based approach integrating target fishing, data integration, bioinformatics analysis, network analysis, Gene Ontology (GO) enrichment analysis, pathway analysis, and in vitro experiment validation to elucidate the pharmacological mechanisms of the combination of 74DHF and VD3 for asthma treatment. RESULTS Our investigation revealed 47 overlapping targets, 20 core targets, 10 optimal common GO processes, and 10 key pathways closely associated with asthma in the combination of 74DHF and VD3. The combined treatment of 74DHF and VD3 inhibited the inflammatory response (TNF-α and IL-6) induced by LPS in macrophages and epithelial to mesenchymal transition (EMT) related genes expression (CDH1 and ACTA2) in bronchial epithelial cells under the stimulation of TGF-β1. CONCLUSION The present study deciphered the molecular mechanism of combined therapeutic effect of 74DHF and VD3 on asthma on systemic and cellular level.
Collapse
Affiliation(s)
- Ziyi Chen
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518020, China; Department of Respirology & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen University, Shenzhen, 518020, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518020, China; Department of Respirology & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen University, Shenzhen, 518020, China
| | - Xiaoyang Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Aiping Lyu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong.
| | - Wei Zhou
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518020, China; Department of Respirology & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen University, Shenzhen, 518020, China.
| |
Collapse
|
4
|
Ren K, Niu B, Liang H, Xi C, Song M, Chen J, Zhao F, Cao Z. Zhichuanling injection improves bronchial asthma by attenuation airway inflammation and epithelia-mesenchymal transition. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119540. [PMID: 40020793 DOI: 10.1016/j.jep.2025.119540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhichuanling (ZCL) Injection, is a compound formulation containing extracts of mahuang (Herba Ephedrae, dried stem or aerial part of Ephedra sinica Stapf), bitter almond (Semen Armeniacae Amarum, seeds of Prunus armeniaca var. sibirica (L.) K. Koch), yangjinhua (flower of Datura metel L.) and Fructus Forsythiae (fruits of Forsythia suspensa (Thunb.) Vahl). Intramuscular injection of ZCL has been used in the clinical practice to control asthma. The aerosol inhalation of ZCL has been shown to be effective on allergic bronchial asthma. However, the underlying mechanisms remain established. AIM OF THE STUDY To investigate the underling mechanism by which ZCL inhibits the pathogenesis of bronchial asthma. METHODS The guinea pig tracheal rings and human bronchial epithelial (16HBE) cells were used to assess ZCL's impact on acetylcholine (Ach) induced tracheal contraction, tumor necrosis factor α (TNF-α) induced bronchial inflammation, and transforming growth factor-β1 (TGF-β1) induced airway remodeling. Cell viability and gene expression were assessed using MTT assays, qPCR. RNA-seq (gene expression analysis) was employed to explore the novel mechanisms of ZCL in OVA-induced bronchial asthma. RESULTS In this study, we found that ZCL reduces Ach-induced contraction of isolated guinea pig trachea, suppress TNF-α-induced interleukin (IL)-1β, IL-6, and IL-8 and TGF-β1-induced E-cadherin, α-SMA, Vimentin, N-cadherin mRNA expression in the 16HBE. Transcriptomic analysis of lung tissue from mice with OVA-induced bronchial asthma suggests that ZCL may alleviate asthma symptoms by modulating BPIFA1, HIF3Α, CTXN3, GRFA3, PPEF1, KSR2, and CDSN. CONCLUSION ZCL alleviates asthma by suppressing tracheal contractions, inflammation, and epithelial-to-mesenchymal transition. ZCL effect on asthma is likely through the upregulation of BPIFA1 expression thus providing the molecular insight for the treatment of asthma. The findings suggest that ZCL holds promise as a asthma therapeutic approach, and further research is needed to explore its full clinical potential. Future studies should focus on optimizing dosage, evaluating long-term efficacy, and investigating potential synergistic effects with existing treatments to enhance asthma management and patient outcomes.
Collapse
Affiliation(s)
- Kerui Ren
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Bo Niu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Huaduan Liang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Chuchu Xi
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Mengmeng Song
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jingyi Chen
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Fang Zhao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Lianyungang, 222001, China.
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
5
|
Sun K, Chang Y, Jie J, Wang C, Gu Y. The J-shaped relationship between body roundness index and adult asthma: insights from NHANES 2001-2018. Front Nutr 2025; 12:1516003. [PMID: 40181938 PMCID: PMC11967368 DOI: 10.3389/fnut.2025.1516003] [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: 10/23/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Background Many studies have used Body Mass Index (BMI) to define obesity and examine its potential link to adult asthma. However, BMI overlooks body fat distribution, which may significantly impact health. Unlike BMI, the Body Roundness Index (BRI) can more accurately reflect body fat distribution. Therefore, this study examined BRI's relationship with asthma prevalence in U.S. adults. Methods This study was based on data from the National Health and Nutrition Examination Survey (NHANES) between 2001 and 2018 and covered 40,052 adult participants. Participants were categorized into four quartile groups based on their BRI levels: Quartile 1 (1.05, 3.80); Quartile 2 (3.80, 5.06); Quartile 3 (5.06, 6.61); Quartile 4 (6.61, 23.48). The association between BRI and asthma prevalence was assessed via weighted multivariate logistic regression, smoothed curve fitting, threshold effects, subgroup, and sensitivity analysis. BRI's predictive power was compared to BMI and waist circumference using z-scores. Results Of the study population, 5,605 participants had asthma (13.99% prevalence). After adjusting for possible confounders, the results showed that higher BRI was linked to greater asthma prevalence (OR = 1.41, 95% CI:1.27, 1.56, p < 0.0001). A J-shaped relationship between BRI and asthma prevalence (p-nonlinearity = 0) was found, with asthma prevalence rising significantly when BRI surpassed 4.34. BRI outperformed BMI and waist circumference in predicting asthma (BRI: OR = 1.180; BMI: OR = 1.169; W.C.: OR = 1.166). Subgroup and sensitivity analyses confirmed our results' robustness. Conclusion Adult asthma prevalence increases with increasing BRI levels, showing a J-shaped relationship. Keeping BRI under 4.34 is vital for lowering asthma prevalence, especially for overweight or obese individuals. In addition, BRI outperformed BMI and waist circumference in predicting asthma occurrence.
Collapse
Affiliation(s)
- Kunpeng Sun
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, China
| | - Yiyi Chang
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, China
| | - Jing Jie
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, China
| | - Chunyan Wang
- Department of General Medicine, First Hospital of Jilin University, Changchun, China
| | - Yue Gu
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Adair D, Bagheri A, Yosef M, Khalatbari S, Lewis T, Mohan A, Lugogo N. High Interleukin (IL)-6 is Associated with Lower Lung Function and Increased Likelihood of Metabolic Dysfunction in Asthma. Pulm Ther 2025; 11:41-54. [PMID: 39714726 PMCID: PMC11861817 DOI: 10.1007/s41030-024-00281-z] [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: 07/07/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
INTRODUCTION Asthma is a complex condition characterized by airway inflammation. Interleukin-6 (IL-6) plays a significant role in asthma pathogenesis through its effects on T cells and its association with pro-inflammatory responses. Both lung and circulating IL-6 levels are elevated in asthma. IL-6 is positively associated with disease severity, frequent exacerbations, and impaired lung function, all of which can be observed clinically. We developed an IL-6 cut-off model to examine the association between high IL-6, race, high body mass index (BMI), metabolic disease, and asthma severity as assessed by reduced lung function. METHODS This study utilized the Coronary Artery Risk Development in Young Adults (CARDIA) database, comprised of 5115 adults, to investigate the relationship between IL-6 levels, asthma, race, and metabolic dysfunction. A "healthy" subset of 427 patients was used to compute the IL-6 cut-off. IL-6 levels within detection limits (0.15-12 pg/mL) were analyzed. The IL-6 cut-off was determined using the 95th percentile of log-transformed IL-6 values for lean (BMI < 25) and healthy individuals. Specific cut-offs were established for racial groups. Statistical analyses involved comparing patient characteristics between high and low IL-6 groups, regression analyses, and assessment of factors influencing lung function changes. RESULTS Using an IL-6 cut-off of 4.979 pg/mL, the cohort was divided into high and low IL-6 groups. High IL-6 correlated with Black race, higher BMI, hypertension, and markers of metabolic dysfunction, e.g., elevated HbA1c, C-reactive protein (CRP), and reduced lung function. Multivariable analysis linked high IL-6 with male gender, high BMI, Black race, HbA1c, CRP, and inversely with lung function and total cholesterol. Obesity showed a consistent positive association with elevated IL-6, regardless of the presence or absence of asthma. Patients with asthma and high IL-6 were more likely to be Black and showed increased CRP. Lung function was lowest in non-lean, high IL-6 patients with asthma, with similar trends in non-lean (BMI ≥ 25) patients without asthma. CONCLUSION This study underscores the significant association between IL-6, asthma, obesity, and metabolic dysfunction. Elevated IL-6 correlates with asthma severity, particularly in individuals with obesity. Future research should explore anti-IL-6 therapies for specific phenotypes, such as obesity-related asthma. These findings advance our understanding of asthma and the role of IL-6 in its pathogenesis.
Collapse
Affiliation(s)
- Dionne Adair
- Division of Pediatric Pulmonary, Medical College of Georgia, Augusta, GA, USA
| | - AmirBehzad Bagheri
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Matheos Yosef
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI, USA
| | - Shokoufeh Khalatbari
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI, USA
| | - Toby Lewis
- Division of Pediatric Pulmonology, University of Michigan, Ann Arbor, MI, USA
| | - Arjun Mohan
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Njira Lugogo
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Zhu W, Han L, He L, Peng W, Li Y, Tian W, Qi H, Wei S, Shen J, Song Y, Shen Y, Zhu Q, Zhou J. Lsm2 is critical to club cell proliferation and its inhibition aggravates COPD progression. Respir Res 2025; 26:71. [PMID: 40022153 PMCID: PMC11871738 DOI: 10.1186/s12931-025-03126-8] [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: 09/22/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory condition, with its severity inversely related to the levels of Club cell 10 kDa secretory protein (CC10). The gene Lsm2, involved in RNA metabolism and cell proliferation, has an unclear role in COPD development. METHODS An in vitro COPD model was developed by stimulating 16HBE cells with cigarette smoke extract (CSE). To establish an in vivo COPD model, mice with defective Lsm2 gene expression in lung or club cells were exposed to cigarette smoke for 3 months. Multiplexed immunohistochemistry (mIHC) was employed to identify the specific cells where Lsm2 gene expression is predominant. RNA sequencing and single-nucleus RNA sequencing were conducted to investigate the role of Lsm2 in the pathogenesis of COPD. RESULTS In this study, we found that cigarette smoke extract increases Lsm2 expression, and knocking down Lsm2 in 16HBE cells significantly reduces cell viability in vitro. mIHC showed that Lsm2 is primarily expressed in Club cells. Knockout of Lsm2, either in the lungs or specifically in Club cells, exacerbated lung injury and inflammation caused by cigarette smoke exposure in vivo. Single-nucleus RNA sequencing analysis revealed that Club cell-specific knockout of Lsm2 leads to a reduction in the Club cell population, particularly those expressing Chia1+/Crb1+. This decrease in Club cells subsequently reduces the number of ciliated epithelial cells. CONCLUSION Knocking out Lsm2 in Club cells results in a significant decrease in Club cell numbers, which subsequently leads to a reduction in ciliated epithelial cells. This increased lung vulnerability to cigarette smoke and accelerating the progression of COPD. Our findings highlight that Lsm2 is critical to club cell proliferation and its inhibition aggravates COPD progression.
Collapse
Affiliation(s)
- Wensi Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China
| | - Linxiao Han
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China
| | - Ludan He
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China
| | - Wenjun Peng
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China
| | - Ying Li
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Weibin Tian
- Department of Respiratory and Critial Care Medicine, Shanghai Pudong Hospital, 2800 Gongwei Rd, Shanghai, 201399, China
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050091, Hebei, China
| | - Shuoyan Wei
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China
- Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China
| | - Jie Shen
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Fudan University, Shanghai, 200540, China
| | - Yuanlin Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China
| | - Yao Shen
- Department of Respiratory and Critial Care Medicine, Shanghai Pudong Hospital, 2800 Gongwei Rd, Shanghai, 201399, China.
| | - Qiaoliang Zhu
- Department of Thoracic Surgery, Shanghai Geriatric Medical Center, 2560 Chunshen Road, Shanghai, 201104, China.
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center of Internet of Things for Respiratory Medicine, 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, 180 Fenglin Road, Shanghai, 200032, China.
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Fudan University, Shanghai, 200540, China.
| |
Collapse
|
8
|
Magen I, Biton R, Khateeb M, Magen E. Possible Resolution of Food Allergies Following Tocilizumab-Induced Remission of Juvenile Idiopathic Arthritis. Cureus 2025; 17:e79442. [PMID: 40130131 PMCID: PMC11931585 DOI: 10.7759/cureus.79442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2025] [Indexed: 03/26/2025] Open
Abstract
This case report describes a unique instance of a four-year-old Ashkenazi Jewish male with systemic juvenile idiopathic arthritis (JIA) and severe IgE-mediated food allergies who achieved complete remission of both conditions following treatment with tocilizumab, an IL-6 receptor antagonist. The patient initially presented with anaphylactic reactions to peanuts, eggs, and cow's milk, confirmed by positive skin prick tests and elevated specific IgE (sIgE) levels. Systemic JIA symptoms, including daily fever spikes, evanescent rash, and polyarticular arthritis, developed shortly thereafter. After inadequate response to standard therapies, tocilizumab was initiated, resulting in complete remission of JIA symptoms and unexpected resolution of food allergies. Repeat allergological evaluations, including skin prick tests, sIgE levels, and controlled oral food challenges, confirmed sustained tolerance to previously allergenic foods. The dual remission observed in this case suggests that interleukin-6 (IL-6) inhibition may modulate shared immunological pathways underlying autoimmune and allergic diseases. IL-6 is a key cytokine in both conditions, promoting Th17-mediated inflammation in JIA and Th2-mediated allergic responses, including IgE production and mast cell activation. While the spontaneous resolution of food allergies, particularly to milk and eggs, is well-documented in pediatric populations, the resolution of peanut allergy, in this case, is highly unusual and temporally associated with tocilizumab treatment. This case raises the possibility that IL-6 inhibition may play a role in modulating autoimmune and allergic responses, warranting further investigation into the interplay between these pathways.
Collapse
Affiliation(s)
- Israel Magen
- Medicine, Assuta Ashdod University Medical Center, Ashdod, ISR
| | - Rey Biton
- Internal Medicine, Assuta Ashdod University Medical Center, Ashdod, ISR
| | - Majd Khateeb
- Internal Medicine, Assuta Ashdod University Medical Center, Ashdod, ISR
| | - Eli Magen
- Medicine, Assuta Ashdod University Medical Center, Ashdod, ISR
| |
Collapse
|
9
|
Pourfarzam S, Ardestani SK, Jamali T, Ghazanfari H, Naghizadeh MM, Faghihzadeh S, Yaraee R, Ghazanfari Z, Ghazanfari T. Distinct inflammatory profiles in mustard lung: A study of sulfur mustard-exposed patients with serious pulmonary complications. Int Immunopharmacol 2025; 146:113832. [PMID: 39689604 DOI: 10.1016/j.intimp.2024.113832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/08/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024]
Abstract
Mustard Lung (ML) refers to respiratory complications caused by sulfur mustard (SM) exposure, a chemical warfare agent. This study explores the inflammatory profile of SM-exposed veterans with serious ML, aiming to distinguish it from other respiratory diseases. The aim is also to comprehend the role of inflammatory markers in disease severity. A study was conducted on 257 male SM-exposed veterans and 64 unexposed control subjects. Inflammatory markers in serum and sputum samples were measured using the ELISA technique. Clinical evaluations identified the SM-exposed group as the patients with serious problems. Statistical analyses were conducted to compare groups. SM-exposed veterans exhibited decreased serum levels of IL-1α, IL-8, IL-17, GM-CSF, and NO compared to controls, while other markers did not differ significantly. Intriguingly, inflammatory patterns in ML subgroups, including those resembling Bronchiolitis Obliterans (BO), Chronic Bronchitis (CB), and asthma, differed from the corresponding cases of these diseases as reported in prior literature. Additionally, certain factors demonstrated notable decreases in concentration in severe ML cases compared to controls. The study reveals substantial changes in inflammatory markers in ML patients, highlighting their distinctive profile compared to other respiratory conditions. Despite some similarities with asthma, BO, and CB, the ML shows discernible variations in the levels of most inflammatory markers. Furthermore, the severity of ML impacted the serum levels of specific factors. These findings support the notion of classifying ML as a separate pulmonary condition, distinct from CB, BO, and asthma, opening avenues for further research and tailored treatment strategies.
Collapse
Affiliation(s)
| | | | - Tahereh Jamali
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Hadi Ghazanfari
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Soghrat Faghihzadeh
- Department of Biostatistics and Social Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; Faculty of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Roya Yaraee
- Department of Immunology, Shahed University, Tehran, Iran
| | - Zeinab Ghazanfari
- Department of Health Education and Promotion, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Immunology, Shahed University, Tehran, Iran.
| |
Collapse
|
10
|
Khalid Salah Al-Sheakly B, Saheb Sharif-Askari F, Saheb Sharif-Askari N, Hundt JE, Halwani R. The potential role of nanobodies in asthma therapy. Front Pharmacol 2025; 15:1510806. [PMID: 39902079 PMCID: PMC11788342 DOI: 10.3389/fphar.2024.1510806] [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: 10/13/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Asthma is a chronic inflammatory disease of the airways characterized by bronchoconstriction, airway hyperresponsiveness, and mucus production. The pathophysiology of asthma involves a complex interplay of immune cells and mediators, including cytokines, chemokines, and other inflammatory molecules. Despite advances in asthma management, many patients continue to experience symptoms due to the limitations of current therapies. Monoclonal antibodies (mAbs) targeting specific inflammatory mediators have improved treatment outcomes for some patients, but challenges such as poor tissue penetration and high costs remain. Nanobodies (Nbs), a novel class of single-domain antibodies, offer a promising alternative due to their small size, stability, and potential for enhanced tissue penetration. This review discusses the key mediators involved in asthma, challenges in current treatments, and the potential of Nbs as a new therapeutic strategy. We also explore current studies and innovations in nanobody technology.
Collapse
Affiliation(s)
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jennifer E. Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pediatrics, Faculty of Medicine, Prince Abdullah Ben Khaled Celiac Disease Chair, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Wu Y, Zhang Y, Wang J, Gan Q, Su X, Zhang S, Ding Y, Yang X, Zhang N, Wu K. Genetic evidence for the causal effects of air pollution on the risk of respiratory diseases. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117602. [PMID: 39740427 DOI: 10.1016/j.ecoenv.2024.117602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Epidemiological studies have consistently demonstrated a robust association between long-term exposure to air pollutants and respiratory diseases. However, establishing causal relationships remains challenging due to residual confounding in observational studies. In this study, Mendelian randomization (MR) analysis was used to explore the causal and epigenetic relationships between various air pollutants and common respiratory diseases. METHODS We utilized a two-sample Mendelian randomization (TSMR) approach to explore the impact of PM2.5, PM2.5-10, PM10, NO2, and NOX on the incidence of nine respiratory diseases using data from large-scale European GWAS datasets (N = 423,796-456,380 for exposures; N = 162,962-486,484 for outcomes). The primary analytical method was inverse variance weighting (IVW), which explored the exposure-outcome relationship using single nucleotide polymorphisms (SNPs) associated with air pollution. Sensitivity analyses, including MR-Egger regression and leave-one-out analyses, were employed to ensure result consistency. Multivariate MR (MVMR) was performed to adjust for potential smoking-related confounders, such as cigarettes per day, household smoking, exposure to tobacco smoke at home, ever smoked, second-hand smoke, smoking initiation, and age at smoking initiation, as well as the independent effects of each air pollutant. Additionally, methylation and enrichment analyses were conducted to further elucidate the potential effects of air pollution on respiratory diseases. RESULTS TSMR analysis revealed that exposure to PM2.5 increased the risk of early-onset chronic obstructive pulmonary disease (COPD), pneumonia, pulmonary embolism and lung cancer. PM2.5-10 exposure was associated with an increased risk of lung cancer, while PM10 exposure increased the risk of pneumonia and bronchiectasis. NO2 exposure was associated with increased risks of lung cancer and adult asthma. Importantly, these associations remained robust even after controlling for potential tobacco-related confounders in the MVMR analyses. In the MVMR analysis adjusting for other pollutants, significant associations persisted between PM2.5 and early-onset COPD, and between PM10 and pneumonia. Genetic co-localization analyses confirmed that methylation of PM2.5-associated CpG loci (cg11386376 near c1orf175, cg11846064 near rfx2, cg18612040 near rptor, and cg19765378 near c7orf50) was associated with an increased risk of early-onset COPD. Finally, SNPs significantly associated with exposure and outcome were selected for enrichment analysis. CONCLUSIONS Our findings suggest that exposure to air pollutants may play a causal role in the development of respiratory diseases, with a potential role of epigenomic modifications emphasized. Strengthening comprehensive air pollution regulations by relevant authorities could potentially mitigate the risk of these diseases.
Collapse
Affiliation(s)
- Yanjuan Wu
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Yuting Zhang
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Jingcun Wang
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Qiming Gan
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Xiaofen Su
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Sun Zhang
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Yutong Ding
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Xinyan Yang
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China
| | - Nuofu Zhang
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China.
| | - Kang Wu
- Sleep Medicine Center, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510160, China.
| |
Collapse
|
12
|
Guo D, Huang K, Guan X, Ding R, Zhu D, Zhao Y, Yang T, He P. Association between chronic respiratory diseases and frailty in Chinese elderly: a population-based longitudinal study. BMJ Open Respir Res 2025; 12:e002171. [PMID: 39753242 PMCID: PMC11752046 DOI: 10.1136/bmjresp-2023-002171] [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: 11/03/2023] [Accepted: 12/05/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Chronic respiratory diseases (CRDs) have been shown to be associated with frailty, but these findings have not yet reached a consensus. The aim of this study was to investigate the association between CRDs and frailty in the elderly using a nationally representative data from China. METHODS Data from the China Health and Retirement Longitudinal Study (CHARLS) were analysed, including 3309 frailty-free participants followed for three waves from 2011. Frailty was assessed using the physical frailty phenotype scale, and CRDs were conformed by self-reported physician diagnoses. Cox proportional hazard models were used to examine the association between baseline CRDs and subsequent frailty. RESULTS Among participants (mean age 67.07 years, 51.53% male), 497 (15.02%) had CRDs. During a mean follow-up of 46 months, 273 (8.25%) participants developed frailty. The incidence rate of frailty was significantly higher in the CRDs group (37.17% per 1000 person-years vs 18.41% per 1000 person-years, p<0.01). Adjusted for covariables, participants with CRDs had a 44% higher risk of developing frailty (HR = 1.44, 95% CI: 1.08 to 1.91). Specifically, asthma only (HR=1.89, 95% CI: 1.07 to 3.33) and asthma-chronic obstructive pulmonary disease (COPD) overlap (ACO) (HR=1.79, 95% CI: 1.19 to 2.69) were associated with a higher risk of frailty among the elderly, while COPD only was not (HR=1.11, 95% CI: 0.73 to 1.65). CONCLUSION This study shows a significant association between CRDs, particularly asthma only and ACO, and frailty in the elderly. We need to pay attention to the frailty status of CRDs patients and consider routine screening among them in both clinical practice and community settings. Active treatment and control of CRDs are necessary to avoid frailty caused by primary lung disease progression or exacerbation.
Collapse
Affiliation(s)
- Dan Guo
- Aerospace Science and Industry Corporation 731 Hospital, Beijing, China
- School of Public Health, Peking University, Beijing, China
| | - Ke Huang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- National Center for Respiratory Medicine, Beijing, China
| | | | | | | | - Yanan Zhao
- Faculty of Health and Wellness, City University of Macau, Taipa, Macau, China
| | - Ting Yang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences, Beijing, China
| | - Ping He
- Peking University, Beijing, China
| |
Collapse
|
13
|
Tashiro H, Kuwahara Y, Kurihara Y, Takahashi K. Molecular mechanisms and clinical impact of biologic therapies in severe asthma. Respir Investig 2025; 63:50-60. [PMID: 39642687 DOI: 10.1016/j.resinv.2024.11.015] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Severe asthma is a critical condition for patients with asthma, characterized by frequent exacerbations, decreased pulmonary function, and unstable symptoms related to asthma. Consequently, the administration of systemic corticosteroids, which cause secondary damage because of their adverse effects, is considered. Recently, several types of molecular-targeted biological therapies have become available for patients with severe asthma, and they have a capacity to improve the pathophysiology of severe asthma. However, several clinical reports indicate that the effects differ depending on the biological targets of asthma in individual patients. In this review, the molecular mechanisms and clinical impact of biologic therapies in severe asthma are described. In addition, molecules targeted by possible future biologics are also addressed. Better understanding of the mechanistic basis for the role of biologics in severe asthma could lead to new therapeutic options for these patients.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan.
| |
Collapse
|
14
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. Cell Rep 2024; 43:115045. [PMID: 39661516 DOI: 10.1016/j.celrep.2024.115045] [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: 07/30/2024] [Revised: 10/01/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential environmental dangers. However, this function can be detrimental during allergic reactions, as vagal nociceptors contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we investigate the changes in the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identify a specific class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of allergic airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the neuropeptide Y (NPY) receptor Npy1r. A screening of cytokines and neurotrophins reveals that interleukin 1β (IL-1β), IL-13, and brain-derived neurotrophic factor (BDNF) drive part of this reprogramming. IL-13 triggers Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, NPY is released into the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells reveals that a cell-specific knockout of NPY1R in nociceptor neurons in asthmatic mice altered T cell infiltration. Opposite findings are observed in asthmatic mice in which nociceptor neurons are chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits the activity of nociceptor neurons.
Collapse
Affiliation(s)
- Theo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Herbert Herzog
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l'Apprentissage, Université de Montréal, Montreal, QC, Canada; Département de Physique, Université de Montréal, Montreal, QC, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
15
|
Santibáñez M, Ruiz-Cubillán JJ, Expósito A, Agüero J, García-Rivero JL, Abascal B, Amado CA, Ruiz-Azcona L, Lopez-Hoyos M, Irure J, Robles Y, Berja A, Barreiro E, Núñez-Robainas A, Cifrián JM, Fernandez-Olmo I. Association Between Oxidative Potential of Particulate Matter Collected by Personal Samplers and Systemic Inflammation Among Asthmatic and Non-Asthmatic Adults. Antioxidants (Basel) 2024; 13:1464. [PMID: 39765793 PMCID: PMC11673029 DOI: 10.3390/antiox13121464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
With the rationale that the oxidative potential of particulate matter (PM-OP) may induce oxidative stress and inflammation, we conducted the ASTHMA-FENOP study in which 44 asthmatic patients and 37 matched controls wore a personal sampler for 24 h, allowing the collection of fine and coarse PM fractions separately, to determine PM-OP by the dithiothreitol (DTT) and ascorbic acid (AA) methods. The levels of Interleukin 6 (IL-6) and the IL-6/IL-10 ratio, as indicators of pro- and anti-inflammatory statuses, were determined by calculating the mean differences (MDs), odds ratios (ORs) and p-trends adjusted for sex, age, study level and body mass index. Positive associations for IL-6 levels in the form of adjusted MDs and ORs were obtained for all PM-OP metrics, reaching statistical significance for both OP-DTT and OP-AA in the fine fraction, with adjusted OR = 5.66; 95%CI (1.46 to 21.92) and 3.32; 95%CI (1.07 to 10.35), respectively, along with statistically significant dose-response patterns when restricting to asthma and adjusted also for clinical variables (adjusted p-trend = 0.029 and 0.01). Similar or stronger associations and dose-response patterns were found for the IL-6/IL-10 ratio. In conclusion, our findings on the effect of PM-OP on systemic inflammation support that asthma is a heterogeneous disease at the molecular level, with PM-OP potentially playing an important role.
Collapse
Affiliation(s)
- Miguel Santibáñez
- Global Health Research Group, Faculty of Nursing, Universidad de Cantabria-Valdecilla Research Institute (IDIVAL), Avenida Valdecilla, s/n, 39008 Santander, Cantabria, Spain;
| | - Juan José Ruiz-Cubillán
- Division of Pneumology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (J.J.R.-C.); (J.A.); (J.L.G.-R.); (B.A.); (C.A.A.); (J.M.C.)
| | - Andrea Expósito
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Avenida Los Castros, s/n, 39005 Santander, Cantabria, Spain; (A.E.); (I.F.-O.)
| | - Juan Agüero
- Division of Pneumology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (J.J.R.-C.); (J.A.); (J.L.G.-R.); (B.A.); (C.A.A.); (J.M.C.)
| | - Juan Luis García-Rivero
- Division of Pneumology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (J.J.R.-C.); (J.A.); (J.L.G.-R.); (B.A.); (C.A.A.); (J.M.C.)
| | - Beatriz Abascal
- Division of Pneumology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (J.J.R.-C.); (J.A.); (J.L.G.-R.); (B.A.); (C.A.A.); (J.M.C.)
| | - Carlos Antonio Amado
- Division of Pneumology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (J.J.R.-C.); (J.A.); (J.L.G.-R.); (B.A.); (C.A.A.); (J.M.C.)
| | - Laura Ruiz-Azcona
- Global Health Research Group, Faculty of Nursing, Universidad de Cantabria-Valdecilla Research Institute (IDIVAL), Avenida Valdecilla, s/n, 39008 Santander, Cantabria, Spain;
| | - Marcos Lopez-Hoyos
- Division of Immunology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (M.L.-H.); (J.I.)
| | - Juan Irure
- Division of Immunology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (M.L.-H.); (J.I.)
| | - Yolanda Robles
- Division of Biochemistry, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (Y.R.); (A.B.)
| | - Ana Berja
- Division of Biochemistry, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (Y.R.); (A.B.)
| | - Esther Barreiro
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer, IMIM-Hospital del Mar, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB), 08003 Barcelona, Spain; (E.B.); (A.N.-R.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08034 Barcelona, Spain
| | - Adriana Núñez-Robainas
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer, IMIM-Hospital del Mar, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB), 08003 Barcelona, Spain; (E.B.); (A.N.-R.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08034 Barcelona, Spain
| | - José Manuel Cifrián
- Division of Pneumology, Hospital Universitario Marqués de Valdecilla, IDIVAL, 39008 Santander, Cantabria, Spain; (J.J.R.-C.); (J.A.); (J.L.G.-R.); (B.A.); (C.A.A.); (J.M.C.)
| | - Ignacio Fernandez-Olmo
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Avenida Los Castros, s/n, 39005 Santander, Cantabria, Spain; (A.E.); (I.F.-O.)
| |
Collapse
|
16
|
Ali A, Rasheed HM, Ansari SA, Ansari SA, Alkahtani HM. Network Pharmacology and Molecular Docking Reveal Anti-Asthmatic Potential of Zephyranthes rosea Lindl. in an Ovalbumin-Induced Asthma Model. Pharmaceuticals (Basel) 2024; 17:1558. [PMID: 39598467 PMCID: PMC11597469 DOI: 10.3390/ph17111558] [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: 09/27/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Background: This study aimed to evaluate the anti-inflammatory effects of a Zephyranthes rosea in an ovalbumin-induced asthma model. Methods: Allergic asthma was induced in mice via intraperitoneal injection, followed by intranasal ovalbumin challenge. Methanolic extract of Z. rosea bulb was orally administered to asthmatic mice for 14 days. Hematological parameters for bronchoalveolar lavage fluid (BALF) and blood were analyzed. The mRNA expression levels of interleukins and transforming growth factor beta (TGF-β1) in lung tissues were determined using reverse transcriptase-polymerase chain reaction (RT-PCR). Network pharmacology analysis was used to find possible Z. rosea targets. After building a protein-protein interaction network to find hub genes, GO and KEGG enrichment analyses were carried out to determine the potential mechanism. In silico analysis was performed by Molecular Operating Environment. Results: GC-MS analysis of Z. rosea extract detected major classes of phytochemicals. Hematological parameters in blood and BALF from Z. rosea extract-treated animals were significantly reduced in a dose-dependent fashion. Histopathology revealed that Z. rosea bulb had an ameliorative effect on lung tissues. Moreover, treatment with Z. rosea bulb extract significantly restored the normal levels of IL-4, IL-6, IL-1β, IL-10, IL-13, and TGF-β1 in allergic asthmatic mice compared to the diseased group. In silico analysis, particularly of the binding affinities of Z. rosea bulb phytoconstituents for IL6, AKT1, and Src, supported in vivo results. Conclusions: These findings indicated that Z. rosea bulb extract significantly ameliorates cellular and molecular biomarkers of bronchial inflammation and could be a potential candidate for treating allergic asthma.
Collapse
Affiliation(s)
- Amir Ali
- Faculty of Pharmacy, The University of Lahore, Lahore 54590, Pakistan
| | | | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.A.); (H.M.A.)
| | - Shoeb Anwar Ansari
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy;
| | - Hamad M. Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (S.A.A.); (H.M.A.)
| |
Collapse
|
17
|
Lin YC, Wang WC, Lee HL, Tsai JJ, Kao SH. House dust mite allergen Der f 2 drives IL-6 and GM-CSF expression in airway epithelial cells via p38 MAPK/NF-κB signaling. J Asthma 2024; 61:1449-1458. [PMID: 38748873 DOI: 10.1080/02770903.2024.2356685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Der f 2, a major allergen derived from Dermatophagoides farinae, is a leading cause of allergic asthma. IL-6 and GM-CSF play essential roles in the exacerbation of asthma. However, the mechanical act by which Der f 2 mediates the expression of IL-6, IL-8, and GM-CSF in airway epithelial cells remains incompletely elucidated. Herein, we aimed to explore the effect of Der f 2 on IL-6 and GM-CSF expression in the human airway epithelial cell BEAS-2B and A549. METHODS Recombinant Der f 2 (rDf2) was acquired using Pichia pastoris. BEAS-2B and A549 cells were used as cell model. The expression of genes and proteins and the involvement of the signaling cascade were assessed using RT-PCR, quantitative real-time PCR (qPCR), Western blotting, and ELISA, respectively. RESULTS Our findings showed that rDf2 significantly induced mRNA expression and protein production of IL-6 and GM-CSF in BEAS-2B and A549 cells. In contrast, rDf2 did not influence IL-8 expression or production in both cells. Mechanistic studies revealed that rDf2 triggered activation of the p38 MAPK and JNK. Inhibition of p38, but not JNK, significantly attenuated rDf2-induced IL-6 and GM-CSF expression and production. CONCLUSION This study demonstrates that Der f 2 promotes the expression and production of the pro-inflammatory cytokines IL-6 and GM-CSF in airway epithelial cells via activation of the p38 signaling pathway. These findings provide insights into the molecular mechanisms that Der f 2 may exacerbate airway inflammation.
Collapse
Affiliation(s)
- Yu-Cian Lin
- Division of Cardiovascular Surgery, Department of Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Wang
- Institute of Biochemistry and Biotechnology (Institute of Medicine), College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jaw-Ji Tsai
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan
| | - Shao-Hsuan Kao
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
18
|
Kosuru R, Romito O, Sharma GP, Ferraresso F, Ghadrdoost Nakhchi B, Yang K, Mammoto T, Mammoto A, Kastrup CJ, Zhang DX, Goldspink PH, Trebak M, Chrzanowska M. Rap1A Modulates Store-Operated Calcium Entry in the Lung Endothelium: A Novel Mechanism Controlling NFAT-Mediated Vascular Inflammation and Permeability. Arterioscler Thromb Vasc Biol 2024; 44:2271-2287. [PMID: 39324266 PMCID: PMC11495542 DOI: 10.1161/atvbaha.124.321458] [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: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Store-operated calcium entry mediated by STIM (stromal interaction molecule)-1-Orai1 (calcium release-activated calcium modulator 1) is essential in endothelial cell (EC) functions, affecting signaling, NFAT (nuclear factor for activated T cells)-induced transcription, and metabolic programs. While the small GTPase Rap1 (Ras-proximate-1) isoforms, including the predominant Rap1B, are known for their role in cadherin-mediated adhesion, EC deletion of Rap1A after birth uniquely disrupts lung endothelial barrier function. Here, we elucidate the specific mechanisms by which Rap1A modulates lung vascular integrity and inflammation. METHODS The role of EC Rap1A in lung inflammation and permeability was examined using in vitro and in vivo approaches. RESULTS We explored Ca2+ signaling in human ECs following siRNA-mediated knockdown of Rap1A or Rap1B. Rap1A knockdown, unlike Rap1B, significantly increased store-operated calcium entry in response to a GPCR (G-protein-coupled receptor) agonist, ATP (500 µmol/L), or thapsigargin (250 nmol/L). This enhancement was attenuated by Orai1 channel blockers 10 μmol/L BTP2 (N-[4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-4-methyl-1,2,3-thiadiazole-5-carboxamide), 10 μmol/L GSK-7975A, and 5 μmol/L Gd3+. Whole-cell patch clamp measurements revealed enhanced Ca2+ release-activated Ca2+ current density in siRap1A ECs. Rap1A depletion in ECs led to increased NFAT1 nuclear translocation and activity and elevated levels of proinflammatory cytokines (CXCL1 [C-X-C motif chemokine ligand 1], CXCL11 [C-X-C motif chemokine 11], CCL5 [chemokine (C-C motif) ligand 5], and IL-6 [interleukin-6]). Notably, reducing Orai1 expression in siRap1A ECs normalized store-operated calcium entry, NFAT activity, and endothelial hyperpermeability in vitro. EC-specific Rap1A knockout (Rap1AiΔEC) mice displayed an inflammatory lung phenotype with increased lung permeability and inflammation markers, along with higher Orai1 expression. Delivery of siRNA against Orai1 to lung endothelium using lipid nanoparticles effectively normalized Orai1 levels in lung ECs, consequently reducing hyperpermeability and inflammation in Rap1AiΔEC mice. CONCLUSIONS Our findings uncover a novel role of Rap1A in regulating Orai1-mediated Ca2+ entry and expression, crucial for NFAT-mediated transcription and endothelial inflammation. This study distinguishes the unique function of Rap1A from that of the predominant Rap1B isoform and highlights the importance of normalizing Orai1 expression in maintaining lung vascular integrity and modulating endothelial functions.
Collapse
Affiliation(s)
- Ramoji Kosuru
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | - Olivier Romito
- Department of Pharmacology and Chemical Biology (O.R., M.T.), University of Pittsburgh School of Medicine, PA
| | - Guru Prasad Sharma
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | - Francesca Ferraresso
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | | | - Kai Yang
- Data Science Institute (K.Y.), Medical College of Wisconsin, Milwaukee
| | - Tadanori Mammoto
- Department of Pediatrics (T.M., A.M.), Medical College of Wisconsin, Milwaukee
| | - Akiko Mammoto
- Department of Pediatrics (T.M., A.M.), Medical College of Wisconsin, Milwaukee
| | - Christian J. Kastrup
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | - David X. Zhang
- Department of Medicine (D.X.Z.), Medical College of Wisconsin, Milwaukee
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, University of Illinois Chicago (P.H.G.)
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology (O.R., M.T.), University of Pittsburgh School of Medicine, PA
- Vascular Medicine Institute (M.T.), University of Pittsburgh School of Medicine, PA
- UPMC Hillman Cancer Center (M.T.), University of Pittsburgh School of Medicine, PA
| | - Magdalena Chrzanowska
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
- Department of Pharmacology and Toxicology (M.C.), Medical College of Wisconsin, Milwaukee
- Cardiovascular Center (M.C.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
19
|
Seker A, Qirko-Gurakuqi A, Tabaku M, Javate KRP, Rathwell I. Maternal atopic conditions and autism spectrum disorder: a systematic review. Eur Child Adolesc Psychiatry 2024; 33:3727-3737. [PMID: 37661216 PMCID: PMC11588786 DOI: 10.1007/s00787-023-02285-7] [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: 11/27/2022] [Accepted: 08/14/2023] [Indexed: 09/05/2023]
Abstract
Autism spectrum disorder (ASD) is a disabling neurodevelopmental condition with complex etiology. Emerging evidence has pointed to maternal atopy as a possible risk factor. It is hypothesized that maternal atopic disease during pregnancy can lead to increased levels of inflammatory cytokines in fetal circulation via placental transfer or increased production. These cytokines can then pass through the immature blood-brain barrier, causing aberrant neurodevelopment via mechanisms including premature microglial activation. The objective of this study is to systematically review observational studies that investigate whether a maternal history of atopic disease (asthma, allergy, or eczema/atopic dermatitis) is associated with a diagnosis of ASD in offspring. A search was conducted in Ovid MEDLINE, PsycINFO, and Embase databases for relevant articles up to November 2021; this was later updated in January 2022. Observational studies published in peer-reviewed journals were included. Data were synthesized and qualitatively analyzed according to the specific atopic condition. Quality assessment was done using the Newcastle-Ottawa Scale. Nine articles were identified, with all including asthma as an exposure, alongside four each for allergy and eczema. Findings were inconsistent regarding the association between a maternal diagnosis of either asthma, allergy, or eczema, and ASD in offspring, with variations in methodology contributing to the inconclusiveness. More consistent associations were demonstrated regarding maternal asthma that was treated or diagnosed during pregnancy. Evidence suggests that symptomatic maternal asthma during pregnancy could be associated with ASD in offspring, underscoring the importance of effective management of atopic conditions during pregnancy. Further research is needed, particularly longitudinal studies that use gold-standard assessment tools and correlate clinical outcomes with laboratory and treatment data.PROSPERO Registration Number and Date: CRD42018116656, 26.11.2018.
Collapse
Affiliation(s)
- Asilay Seker
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- South London and Maudsley NHS Foundation Trust, London, UK.
| | - Anxhela Qirko-Gurakuqi
- Department of Biomedical and Experimental Subjects, University of Medicine, Tirana, Albania
| | - Mirela Tabaku
- Paediatric Department, University of Medicine, Tirana, Albania
| | - Kenneth Ross P Javate
- Department of Psychiatry, The Medical City Hospital, Manila, Philippines
- School of Medicine and Public Health, Ateneo de Manila University, Manila, Philippines
| | - Iris Rathwell
- South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
20
|
Jiang Y, Wang Y, Guo J, Wang Z, Wang X, Yao X, Yang H, Zou Y. Exploring potential therapeutic targets for asthma: a proteome-wide Mendelian randomization analysis. J Transl Med 2024; 22:978. [PMID: 39472987 PMCID: PMC11520847 DOI: 10.1186/s12967-024-05782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Asthma poses a significant global health challenge, characterized by high rates of morbidity and mortality. Despite available treatments, many severe asthma patients remain poorly managed, highlighting the need for novel therapeutic strategies. This study aims to identify potential drug targets for asthma by examining the influence of circulating plasma proteins on asthma risk. METHODS This study employs summary-data-based Mendelian randomization (MR) and two-sample MR methods to investigate the association between 2940 plasma proteins from the UK Biobank study and asthma. The analysis includes discovery (FinnGen cohort) and replication (GERA cohort) phases, with Bayesian colocalization used to validate the relationships between proteins and asthma. Furthermore, protein-protein interaction and druggability assessments were conducted on high-evidence strength protein biomarkers, and candidate drug prediction and molecular docking were performed for proteins without targeted drugs. Given the complexity of asthma pathogenesis, the study also explores the relationships between plasma proteins and asthma-related endpoints (e.g., obesity-related asthma, infection-related asthma, childhood asthma) to identify potential therapeutic targets for different subtypes. RESULTS In the discovery cohort, 75 plasma proteins were associated with asthma, including IL1RAP, IL1RL1, IL6, CXCL5, and CXCL8. Additionally, 6 proteins (IL4R, LTB, CASP8, MAX, PCDH12, and SCLY) were validated through co-localization analysis and validation cohort. The assessment of drug targetability revealed potential drug targets for IL4R, CASP8, and SCLY, while candidate drugs were predicted for LTB and MAX proteins. MAX exhibited strong binding affinity with multiple small molecules indicating a highly stable interaction and significant druggability potential. Analysis of the 75 proteins with 9 asthma-related endpoints highlighted promising targets such as DOK2, ITGAM, CA1, BTN2A1, and GZMB. CONCLUSION These findings elucidate the link between asthma, its related endpoints, and plasma proteins, advancing our understanding of molecular pathogenesis and treatment strategies. The discovery of potential therapeutic targets offers new insights into asthma drug target research.
Collapse
Affiliation(s)
- Yuhan Jiang
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China
| | - Yifan Wang
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China
| | - Ju Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zixuan Wang
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China
| | - Xuelin Wang
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China
| | - Xueming Yao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hongxi Yang
- Department of Bioinformatics, School of Basic Medical Science, Tianjin Medical University, Tianjin, 300070, China.
| | - Yingxue Zou
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin, China.
- Department of Pulmonology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Machang Compus, 225 Machang Road, Hexi District, Tianjin, 300074, China.
| |
Collapse
|
21
|
Ghosh A, Rogers KL, Gallant SC, Kim YH, Rager JE, Gilmour MI, Randell SH, Jaspers I. Effects of simulated smoke condensate generated from combustion of selected military burn pit contents on human airway epithelial cells. Part Fibre Toxicol 2024; 21:41. [PMID: 39380034 PMCID: PMC11460082 DOI: 10.1186/s12989-024-00604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Exposure to military burn pit smoke during deployment is associated with different respiratory and non-respiratory diseases. However, information linking smoke exposure to human pulmonary health is lacking. This study examined the effects of simulated burn pit smoke condensates on human airway epithelial cells (HAECs) from twelve donors (smokers/non-smokers, biological female/male) cultured at an air-liquid interface and exposed to condensates from three simulated burn pit waste materials (cardboard, plywood, and plastic) incinerated at two combustion conditions: smoldering and flaming. Cellular gene expression was analyzed using bulk RNA sequencing, and basolateral media cytokine levels were assessed using multiplex immunoassay. RESULTS Flaming smoke condensates caused more significant differentially expressed genes (DEGs) with plywood flaming smoke being the most potent in altering gene expression and modulating cytokine release. Cardboard and plywood flaming condensates primarily activated detoxification pathways, whereas plastic flaming affected genes related to anti-microbial and inflammatory responses. Correlation analysis between smoke condensate chemicals and gene expression to understand the underlying mechanism revealed crucial role of oxygenated polycyclic aromatic hydrocarbons (PAHs) and aluminum, molybdenum, and silicon elements; IL6 expression was positively correlated with most PAHs. Stratification of data based on HAEC donor demographics suggests that these affect gene expression changes. Enrichment analysis indicated similarity with several deployment-related presumptive and reported diseases, including asthma, emphysema, and cancer of different organs. CONCLUSIONS This study highlights that simulated burn pit smoke exposure of HAECs causes gene expression changes indicative of deployment-related diseases with more pronounced effects seen in smokers and females. Future studies are needed to further characterize how sex and smoking status affect deployment-related diseases.
Collapse
Affiliation(s)
- Arunava Ghosh
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina, School of Medicine, 104 Mason Farm Road, Chapel Hill, NC, 27599-7310, USA
| | - Keith L Rogers
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC, 27599-7310, USA
| | - Samuel C Gallant
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Yong Ho Kim
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Julia E Rager
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina, School of Medicine, 104 Mason Farm Road, Chapel Hill, NC, 27599-7310, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC, 27599-7310, USA
- Department of Environmental Sciences and Engineering (ESE), Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - M Ian Gilmour
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina, School of Medicine, 104 Mason Farm Road, Chapel Hill, NC, 27599-7310, USA.
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC, 27599-7310, USA.
- Department of Environmental Sciences and Engineering (ESE), Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
22
|
Cai SY, Liu A, Xie WX, Zhang XQ, Su B, Mao Y, Weng DG, Chen ZY. Esketamine mitigates mechanical ventilation-induced lung injury in chronic obstructive pulmonary disease rats via inhibition of the MAPK/NF-κB signaling pathway and reduction of oxidative stress. Int Immunopharmacol 2024; 139:112725. [PMID: 39059100 DOI: 10.1016/j.intimp.2024.112725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE To investigate esketamine's impact on inflammation and oxidative stress in ventilated chronic obstructive pulmonary disease (COPD) rats, examining its regulatory mechanisms. METHODS Rats were divided into four groups: control group (Con), COPD model group (M), COPD model with saline treatment group (M+S), and COPD model with esketamine treatment group (M+K), with 12 rats in each group. After two months, all rats underwent anesthesia and mechanical ventilation. Group M+K received 5 mg/kg esketamine intravenously, while Group M+S received the same volume of saline. Lung tissues were collected for analysis two hours later, including airway peak pressure, wet-to-dry(W/D) ratio, lung permeability index(LPI), hematoxylin and eosin(H&E) staining, and transmission electron microscopy(TEM). Tumor necrosis factor-alpha(TNF-α), interleukin-6(IL-6), interleukin-8(IL-8), and interleukin-10(IL-10) levels were determined by enzyme-linked immunosorbent assay(ELISA); phosphorylated Nuclear Factor Kappa B(p-NF-κB), mitogen-activated protein kinase 14(p38), phosphorylated p38 (p-p38), c-Jun N-terminal kinase(JNK), and phosphorylated JNK (p-JNK) expressions by Western blotting and immunohistochemistry; and malondialdehyde(MDA), myeloperoxidase(MPO), and superoxide dismutase(SOD) levels were also measured by corresponding biochemical assays. RESULTS Lung specimens from groups M, M+S, and M+K manifested hallmark histopathological features of COPD. Compared with group Con, group M displayed increased peak airway pressure, W/D ratio, and LPI. In group M+K, compared with group M, esketamine significantly reduced the W/D ratio, LPI, and concentrations of pro-inflammatory cytokines TNF-α, IL-6, and IL-8 while concurrently elevating IL-10 levels. Furthermore, the treatment attenuated the activation of the NF-κB and MAPK pathways, indicated by decreased levels of p-NF-κB, p-p38, and p-JNK.Additionally, compared to group M, group M+K showed decreased MDA and MPO levels and increased SOD levels in lung tissue. CONCLUSION Esketamine attenuates mechanical ventilation-induced lung injury in COPD rat models by inhibiting the MAPK/NF-κB signaling pathway and reducing oxidative stress.
Collapse
Affiliation(s)
- San-Ying Cai
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China; Department of Anesthesiology, Mindong Hospital Affiliated to Fujian Medical University, Fuan 355000, Fujian, China
| | - Ang Liu
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China; Department of Anesthesiology, Heze Municipal Hospital, Heze 274000, China
| | - Wen-Xi Xie
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Xiao-Qi Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Bin Su
- Department of Anesthesiology, Heze Municipal Hospital, Heze 274000, China
| | - Yu Mao
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Di-Gui Weng
- Department of Anesthesiology, Mindong Hospital Affiliated to Fujian Medical University, Fuan 355000, Fujian, China.
| | - Zhi-Yuan Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China.
| |
Collapse
|
23
|
Crosson T, Bhat S, Wang JC, Salaun C, Fontaine E, Roversi K, Herzog H, Rafei M, Blunck R, Talbot S. Cytokines reprogram airway sensory neurons in asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.26.525731. [PMID: 39345572 PMCID: PMC11429693 DOI: 10.1101/2023.01.26.525731] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Nociceptor neurons play a crucial role in maintaining the body's homeostasis by detecting and responding to potential dangers in the environment. However, this function can be detrimental during allergic reactions, since vagal nociceptors can contribute to immune cell infiltration, bronchial hypersensitivity, and mucus imbalance, in addition to causing pain and coughing. Despite this, the specific mechanisms by which nociceptors acquire pro-inflammatory characteristics during allergic reactions are not yet fully understood. In this study, we aimed to investigate the molecular profile of airway nociceptor neurons during allergic airway inflammation and identify the signals driving such reprogramming. Using retrograde tracing and lineage reporting, we identified a unique class of inflammatory vagal nociceptor neurons that exclusively innervate the airways. In the ovalbumin mouse model of airway inflammation, these neurons undergo significant reprogramming characterized by the upregulation of the NPY receptor Npy1r. A screening of cytokines and neurotrophins revealed that IL-1β, IL-13 and BDNF drive part of this reprogramming. IL-13 triggered Npy1r overexpression in nociceptors via the JAK/STAT6 pathway. In parallel, sympathetic neurons and macrophages release NPY in the bronchoalveolar fluid of asthmatic mice, which limits the excitability of nociceptor neurons. Single-cell RNA sequencing of lung immune cells has revealed that a cell-specific knockout of Npy1r in nociceptor neurons in asthmatic mice leads to an increase in airway inflammation mediated by T cells. Opposite findings were observed in asthmatic mice in which nociceptor neurons were chemically ablated. In summary, allergic airway inflammation reprograms airway nociceptor neurons to acquire a pro-inflammatory phenotype, while a compensatory mechanism involving NPY1R limits nociceptor neurons' activity.
Collapse
Affiliation(s)
- Théo Crosson
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Shreyas Bhat
- Centre Interdisciplinaire sur le Cerveau et l’Apprentissage, Université de Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Jo-Chiao Wang
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Clara Salaun
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Eleanne Fontaine
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Katiane Roversi
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | | | - Moutih Rafei
- Département de Pharmacologie et Physiologie, Université de Montréal, Canada
| | - Rikard Blunck
- Centre Interdisciplinaire sur le Cerveau et l’Apprentissage, Université de Montréal, Canada
- Département de Physique, Université de Montréal, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet. Sweden
- Department of Biomedical and Molecular Sciences, Queen’s University. Canada
| |
Collapse
|
24
|
Suraya R, Nagano T, Yumura M, Hara T, Akashi M, Yamamoto M, Tachihara M, Nishimura Y, Kobayashi K. Loss of JCAD/KIAA1462 Protects the Lung from Acute and Chronic Consequences of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2024; 25:9492. [PMID: 39273437 PMCID: PMC11394678 DOI: 10.3390/ijms25179492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Even with recent advances in pathobiology and treatment options, chronic obstructive pulmonary disease (COPD) remains a major contributor to morbidity and mortality. To develop new ways of combating this disease, breakthroughs in our understanding of its mechanisms are sorely needed. Investigating the involvement of underanalyzed lung cell types, such as endothelial cells (ECs), is one way to further our understanding of COPD. JCAD is a junctional protein in endothelial cells (ECs) arising from the KIAA1462 gene, and a mutation in this gene has been implicated in the risk of developing COPD. In our study, we induced inflammation and emphysema in mice via the global knockout of KIAA1462/JCAD (JCAD-KO) and confirmed it in HPMECs and A549 to examine how the loss of JCAD could affect COPD development. We found that KIAA1462/JCAD loss reduced acute lung inflammation after elastase treatment. Even after 3 weeks of elastase, JCAD-KO mice demonstrated a preserved lung parenchymal structure and vasculature. In vitro, after KIAA1462 expression is silenced, both endothelial and epithelial cells showed alterations in pro-inflammatory gene expression after TNF-α treatment. We concluded that JCAD loss could ameliorate COPD through its anti-inflammatory and anti-angiogenic effects, and that KIAA1462/JCAD could be a novel target for COPD therapy.
Collapse
Affiliation(s)
- Ratoe Suraya
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| | - Masako Yumura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| | - Tetsuya Hara
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe 658-8558, Japan;
| | - Masaya Akashi
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.S.); (T.N.); (M.Y.); (M.Y.); (Y.N.); (K.K.)
| |
Collapse
|
25
|
Hufnagel M, Rademaekers A, Weisert A, Häberlein H, Franken S. Pharmacological profile of dicaffeoylquinic acids and their role in the treatment of respiratory diseases. Front Pharmacol 2024; 15:1371613. [PMID: 39239645 PMCID: PMC11374715 DOI: 10.3389/fphar.2024.1371613] [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: 01/16/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Dicaffeoylquinic acids (DCQAs) are polyphenolic compounds found in various medicinal plants such as Echinacea species and Hedera helix, whose multi-constituent extracts are used worldwide to treat respiratory diseases. Besides triterpenes, saponins, alkamides, and other constituents, DCQAs are an important group of substances for the pharmacological activity of plant-derived extracts. Therefore, the pharmacological properties of DCQAs have been studied over the last decades, suggesting antioxidative, anti-inflammatory, antimicrobial, hypoglycaemic, cardiovascular protective, neuroprotective, and hepatoprotective effects. However, the beneficial pharmacological profile of DCQAs has not yet been linked to their use in treating respiratory diseases such as acute or even chronic bronchitis. The aim of this review was to assess the potential of DCQAs for respiratory indications based on published in vitro and in vivo pharmacological and pre-clinical data, with particular focus on antioxidative, anti-inflammatory, and respiratory-related effects such as antitussive or antispasmodic properties. A respective literature search revealed a large number of publications on the six DCQA isoforms. Based on this search, a focus was placed on 1,3-, 3,4-, 3,5-, and 4,5-DCQA, as the publications focused mainly on these isomers. Based on the available pre-clinical data, DCQAs trigger cellular mechanisms that are important in the treatment of respiratory diseases such as decreasing NF-κB activation, reducing oxidative stress, or activating the Nrf2 pathway. Taken together, these data suggest an essential role for DCQAs within herbal medicines used for the treatment of respiratory diseases and highlights the need for the identifications of DCQAs as lead substances within such extracts.
Collapse
Affiliation(s)
| | | | - Anika Weisert
- Engelhard Arzneimittel GmbH & Co. KG, Niederdorfelden, Germany
| | - Hanns Häberlein
- Medical Faculty, Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Sebastian Franken
- Medical Faculty, Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
26
|
Lazzara V, Pinto P, Di Vincenzo S, Ferraro M, Catalano F, Provinzano P, Pace E, Bonsignore MR. In vitro evidence of antioxidant and anti-inflammatory effects of a new nutraceutical formulation explains benefits in a clinical setting of COPD patients. Front Pharmacol 2024; 15:1439835. [PMID: 39228520 PMCID: PMC11368797 DOI: 10.3389/fphar.2024.1439835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024] Open
Abstract
Background and Aim: Increased oxidative stress within the airways is associated to epithelial damage and amplification of inflammatory responses that in turn contribute to Chronic Obstructive Pulmonary Disease (COPD) progression. This study was aimed to identify whether a new formulation of N-acetylcisteine (NAC), carnitine, curcumin and B2 vitamin could counteract oxidative stress and downstream pro-inflammatory events promoted by cigarette smoke extract (CSE) exposure in primary bronchial epithelial cells (PBEC), both submerged/undifferentiated (S-PBEC) and cultured at the air-liquid interface (ALI-PBEC). Methods: PBEC were exposed to CSE with/without the new formulation or NAC alone and ROS production, IL-8 and IL-6 gene expression and protein release were evaluated. Results: CSE increased ROS, IL-8 and IL-6 gene expression and protein release and the new formulation counteracted these effects. NAC alone was not effective on IL-8 and IL-6 release. The effects of a similar nutraceutical formulation were evaluated in COPD patients treated for six months. The results showed that the treatment reduced the concentration of IL-8 in nasal wash and improved quality of life. Conclusion: The tested formulation, exerting antioxidant and anti-inflammatory effects, can preserve airway epithelial homeostasis and improve clinical symptoms in COPD.
Collapse
Affiliation(s)
- Valentina Lazzara
- Dipartimento Promozione della Salute Materno-Infantile di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro” (PROMISE), Università degli Studi di Palermo, Palermo, Italy
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
| | - Paola Pinto
- Dipartimento Promozione della Salute Materno-Infantile di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro” (PROMISE), Università degli Studi di Palermo, Palermo, Italy
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
- PhD National Program in One Health Approaches to Infectious Diseases and Life Science Research, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Serena Di Vincenzo
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
| | - Maria Ferraro
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
| | - Filippo Catalano
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Pietro Provinzano
- Dipartimento Promozione della Salute Materno-Infantile di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro” (PROMISE), Università degli Studi di Palermo, Palermo, Italy
| | - Elisabetta Pace
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
| | - Maria Rosaria Bonsignore
- Dipartimento Promozione della Salute Materno-Infantile di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro” (PROMISE), Università degli Studi di Palermo, Palermo, Italy
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
- Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| |
Collapse
|
27
|
Lozano-Paniagua D, Parrón T, Alarcón R, Requena M, Lacasaña M, Hernández AF. A Th2-type immune response and low-grade systemic inflammatory reaction as potential immunotoxic effects in intensive agriculture farmers exposed to pesticides. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 938:173545. [PMID: 38802022 DOI: 10.1016/j.scitotenv.2024.173545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 05/03/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Pesticides are chemicals widely used in agriculture to keep crops healthy and prevent them from being destroyed by pests, thus contributing to a sustainable food and feed production. However, long-term exposure to these compounds may be harmful to human health as they can affect the function of various organs systems, including the immune system. There is growing evidence that pesticides may increase the risk of developing immune-based diseases and inflammation. This study assessed whether greenhouse farmers occupationally exposed to pesticides presented alterations in immunoregulatory proteins, used as surrogate biomarkers of immune function. The study population consisted of 175 greenhouse workers occupationally exposed to pesticides and 91 non-exposed controls. Serum levels of 27 cytokines, chemokines and growth factors were measured using a magnetic bead-based immunoassay in a subpopulation of 111 greenhouse workers and 79 non-exposed controls. Since analytical determinations were performed in two periods of the same crop season with different use of pesticides (period of high and low pesticide exposure), linear mixed models for repeated measures were used to optimize statistical inference. The increase in IL-13, IL-4 and IL-6 observed in greenhouse workers compared to controls, and in the period of high exposure to pesticides relative to that of low exposure, suggest an altered Th1/Th2 balance towards the Th2 response. This finding points to a type-2 inflammation commonly presented as allergic inflammation, which has often been reported in farm-workers and in which pesticide exposure is considered a risk factor. Furthermore, the increase in IL-1β and VEGF, mediators of inflammation and angiogenesis, may suggest a low-grade systemic inflammation that might underlie chronic pathological conditions linked to pesticide exposure.
Collapse
Affiliation(s)
| | - Tesifón Parrón
- University of Almería School of Health Sciences, Almería, Spain
| | - Raquel Alarcón
- University of Almería School of Health Sciences, Almería, Spain
| | - Mar Requena
- University of Almería School of Health Sciences, Almería, Spain
| | - Marina Lacasaña
- Escuela Andaluza de Salud Pública, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto de Investigación Biosanitaria, Granada (ibs.GRANADA), Spain; Andalusian Health and Environment Observatory (OSMAN), Granada, Spain
| | - Antonio F Hernández
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain; Instituto de Investigación Biosanitaria, Granada (ibs.GRANADA), Spain; Department of Legal Medicine and Toxicology, University of Granada School of Medicine, Granada, Spain.
| |
Collapse
|
28
|
Ishmael L, Casale T, Cardet JC. Molecular Pathways and Potential Therapeutic Targets of Refractory Asthma. BIOLOGY 2024; 13:583. [PMID: 39194521 DOI: 10.3390/biology13080583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024]
Abstract
Asthma is a chronic inflammatory lung disease. Refractory asthma poses a significant challenge in management due to its resistance to standard therapies. Key molecular pathways of refractory asthma include T2 inflammation mediated by Th2 and ILC2 cells, eosinophils, and cytokines including IL-4, IL-5, and IL-13. Additionally, non-T2 mechanisms involving neutrophils, macrophages, IL-1, IL-6, and IL-17 mediate a corticosteroid resistant phenotype. Mediators including alarmins (IL-25, IL-33, TSLP) and OX40L have overlap between T2 and non-T2 inflammation and may signify unique pathways of asthma inflammation. Therapies that target these pathways and mediators have proven to be effective in reducing exacerbations and improving lung function in subsets of severe asthma patients. However, there are patients with severe asthma who do not respond to approved therapies. Small molecule inhibitors, such as JAK-inhibitors, and monoclonal antibodies targeting mast cells, IL-1, IL-6, IL-33, TNFα, and OX40L are under investigation for their potential to modulate inflammation involved in refractory asthma. Understanding refractory asthma heterogeneity and identifying mediators involved are essential in developing therapeutic interventions for patients unresponsive to currently approved biologics. Further investigation is needed to develop personalized treatments based on these molecular insights to potentially offer more effective treatments for this complex disease.
Collapse
Affiliation(s)
- Leah Ishmael
- Division of Pulmonary, Allergy, and Sleep Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Thomas Casale
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Juan Carlos Cardet
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
29
|
Almestica-Roberts M, Nguyen ND, Sun L, Serna SN, Rapp E, Burrell-Gerbers KL, Memon TA, Stone BL, Nkoy FL, Lamb JG, Deering-Rice CE, Rower JE, Reilly CA. The Cytochrome P450 2C8*3 Variant (rs11572080) Is Associated with Improved Asthma Symptom Control in Children and Altered Lipid Mediator Production and Inflammatory Response in Human Bronchial Epithelial Cells. Drug Metab Dispos 2024; 52:836-846. [PMID: 38772712 PMCID: PMC11257687 DOI: 10.1124/dmd.124.001684] [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: 02/03/2024] [Revised: 04/16/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024] Open
Abstract
This study investigated an association between the cytochrome P450 (CYP) 2C8*3 polymorphism with asthma symptom control in children and changes in lipid metabolism and pro-inflammatory signaling by human bronchial epithelial cells (HBECs) treated with cigarette smoke condensate (CSC). CYP genes are inherently variable in sequence, and while such variations are known to produce clinically relevant effects on drug pharmacokinetics and pharmacodynamics, the effects on endogenous substrate metabolism and associated physiologic processes are less understood. In this study, CYP2C8*3 was associated with improved asthma symptom control among children: Mean asthma control scores were 3.68 (n = 207) for patients with one or more copies of the CYP2C8*3 allele versus 4.42 (n = 965) for CYP2C8*1/*1 (P = 0.0133). In vitro, CYP2C8*3 was associated with an increase in montelukast 36-hydroxylation and a decrease in linoleic acid metabolism despite lower mRNA and protein expression. Additionally, CYP2C8*3 was associated with reduced mRNA expression of interleukin-6 (IL-6) and C-X-C motif chemokine ligand 8 (CXCL-8) by HBECs in response to CSC, which was replicated using the soluble epoxide hydrolase inhibitor, 12-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]-dodecanoic acid. Interestingly, 9(10)- and 12(13)- dihydroxyoctadecenoic acid, the hydrolyzed metabolites of 9(10)- and 12(13)- epoxyoctadecenoic acid, increased the expression of IL-6 and CXCL-8 mRNA by HBECs. This study reveals previously undocumented effects of the CYP2C8*3 variant on the response of HBECs to exogenous stimuli. SIGNIFICANCE STATEMENT: These findings suggest a role for CYP2C8 in regulating the epoxyoctadecenoic acid:dihydroxyoctadecenoic acid ratio leading to a change in cellular inflammatory responses elicited by environmental stimuli that exacerbate asthma.
Collapse
Affiliation(s)
- Marysol Almestica-Roberts
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Nam D Nguyen
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Lili Sun
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Samantha N Serna
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Emmanuel Rapp
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Katherine L Burrell-Gerbers
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Tosifa A Memon
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Bryan L Stone
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Flory L Nkoy
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - John G Lamb
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Cassandra E Deering-Rice
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Joseph E Rower
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Christopher A Reilly
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| |
Collapse
|
30
|
Santus P, Signorello JC, Danzo F, Lazzaroni G, Saad M, Radovanovic D. Anti-Inflammatory and Anti-Oxidant Properties of N-Acetylcysteine: A Fresh Perspective. J Clin Med 2024; 13:4127. [PMID: 39064168 PMCID: PMC11278452 DOI: 10.3390/jcm13144127] [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: 05/30/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
N-acetyl-L-cysteine (NAC) was initially introduced as a treatment for mucus reduction and widely used for chronic respiratory conditions associated with mucus overproduction. However, the mechanism of action for NAC extends beyond its mucolytic activity and is complex and multifaceted. Contrary to other mucoactive drugs, NAC has been found to exhibit antioxidant, anti-infective, and anti-inflammatory activity in pre-clinical and clinical reports. These properties have sparked interest in its potential for treating chronic lung diseases, including chronic obstructive pulmonary disease (COPD), bronchiectasis (BE), cystic fibrosis (CF), and idiopathic pulmonary fibrosis (IPF), which are associated with oxidative stress, increased levels of glutathione and inflammation. NAC's anti-inflammatory activity is noteworthy, and it is not solely secondary to its antioxidant capabilities. In ex vivo models of COPD exacerbation, the anti-inflammatory effects have been observed even at very low doses, especially with prolonged treatment. The mechanism involves the inhibition of the activation of NF-kB and neurokinin A production, resulting in a reduction in interleukin-6 production, a cytokine abundantly present in the sputum and breath condensate of patients with COPD and correlates with the number of exacerbations. The unique combination of mucolytic, antioxidant, anti-infective, and anti-inflammatory properties positions NAC as a safe, cost-effective, and efficacious therapy for a plethora of respiratory conditions.
Collapse
Affiliation(s)
- Pierachille Santus
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Juan Camilo Signorello
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Fiammetta Danzo
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Giada Lazzaroni
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Marina Saad
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20122 Milano, Italy;
| | - Dejan Radovanovic
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20122 Milano, Italy;
| |
Collapse
|
31
|
Kita K, Gawinowska M, Chełmińska M, Niedoszytko M. The Role of Exhaled Breath Condensate in Chronic Inflammatory and Neoplastic Diseases of the Respiratory Tract. Int J Mol Sci 2024; 25:7395. [PMID: 39000502 PMCID: PMC11242091 DOI: 10.3390/ijms25137395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are among the most common chronic respiratory diseases. Chronic inflammation of the airways leads to an increased production of inflammatory markers by the effector cells of the respiratory tract and lung tissue. These biomarkers allow the assessment of physiological and pathological processes and responses to therapeutic interventions. Lung cancer, which is characterized by high mortality, is one of the most frequently diagnosed cancers worldwide. Current screening methods and tissue biopsies have limitations that highlight the need for rapid diagnosis, patient differentiation, and effective management and monitoring. One promising non-invasive diagnostic method for respiratory diseases is the assessment of exhaled breath condensate (EBC). EBC contains a mixture of volatile and non-volatile biomarkers such as cytokines, leukotrienes, oxidative stress markers, and molecular biomarkers, providing significant information about inflammatory and neoplastic states in the lungs. This article summarizes the research on the application and development of EBC assessment in diagnosing and monitoring respiratory diseases, focusing on asthma, COPD, and lung cancer. The process of collecting condensate, potential issues, and selected groups of markers for detailed disease assessment in the future are discussed. Further research may contribute to the development of more precise and personalized diagnostic and treatment methods.
Collapse
Affiliation(s)
- Karolina Kita
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Marika Gawinowska
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Marta Chełmińska
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Marek Niedoszytko
- Department of Allergology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
32
|
Song XY, Sun Q, Wei SZ, Wang HR, Wang Y, Zhang WB, Ren C, Song XC, Mou YK. IL-6 mediates olfactory dysfunction in a mouse model of allergic rhinitis. Brain Res 2024; 1833:148885. [PMID: 38531465 DOI: 10.1016/j.brainres.2024.148885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Immune-inflammatory response is a key element in the occurrence and development of olfactory dysfunction (OD) in patients with allergic rhinitis (AR). As one of the core factors in immune-inflammatory responses, interleukin (IL)-6 is closely related to the pathogenesis of allergic diseases. It may also play an important role in OD induced by diseases, such as Sjögren's syndrome and coronavirus disease 2019. However, there is no study has reported its role in OD in AR. Thus, this study aimed to investigate the role of IL-6 in AR-related OD, in an attempt to discover a new target for the prevention and treatment of OD in patients with AR. METHODS Differential expression analysis was performed using the public datasets GSE52804 and GSE140454 for AR, and differentially expressed genes (DEGs) were obtained by obtaining the intersection points between these two datasets. IL-6, a common differential factor, was obtained by intersecting the DEGs with the General Olfactory Sensitivity Database (GOSdb) again. A model of AR mice with OD was developed by sensitizing with ovalbumin (OVA) to verify the reliability of IL-6 as a key factor of OD in AR and explore the potential mechanisms. Furthermore, a supernatant and microglia co-culture model of nasal mucosa epithelial cells stimulated by the allergen house dust mite extract Derp1 was established to identify the cellular and molecular mechanisms of IL-6-mediated OD in AR. RESULTS The level of IL-6 in the nasal mucosa and olfactory bulb of AR mice with OD significantly increased and showed a positive correlation with the expression of olfactory bulb microglia marker Iba-1 and the severity of OD. In-vitro experiments showed that the level of IL-6 significantly increased in the supernatant after the nasal mucosa epithelial cells were stimulated by Derp1, along with significantly decreased barrier function of the nasal mucosa. The expression levels of neuroinflammatory markers IL-1β and INOS increased after a conditioned culture of microglia with the supernatant including IL-6. Then knockdown (KD) of IL-6R by small interfering RNA (siRNA), the expression of IL-1β and INOS significantly diminished. CONCLUSION IL-6 plays a key role in the occurrence and development of OD in AR, which may be related to its effect on olfactory bulb microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Xiao-Yu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Qi Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Shi-Zhuang Wei
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Han-Rui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Yao Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Wen-Bin Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Chao Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| | - Xi-Cheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| | - Ya-Kui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
33
|
Fong A, Rochus CM, Shandilya UK, Muniz MMM, Sharma A, Schenkel FS, Karrow NA, Baes CF. The role of interleukin-10 receptor alpha (IL10Rα) in Mycobacterium avium subsp. paratuberculosis infection of a mammary epithelial cell line. BMC Genom Data 2024; 25:58. [PMID: 38867147 PMCID: PMC11167801 DOI: 10.1186/s12863-024-01234-w] [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: 01/29/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Johne's disease is a chronic wasting disease caused by the bacterium Mycobacterium avium subspecies paratuberculosis (MAP). Johne's disease is highly contagious and MAP infection in dairy cattle can eventually lead to death. With no available treatment for Johne's disease, genetic selection and improvements in management practices could help reduce its prevalence. In a previous study, the gene coding interleukin-10 receptor subunit alpha (IL10Rα) was associated with Johne's disease in dairy cattle. Our objective was to determine how IL10Rα affects the pathogenesis of MAP by examining the effect of a live MAP challenge on a mammary epithelial cell line (MAC-T) that had IL10Rα knocked out using CRISPR/cas9. The wild type and the IL10Rα knockout MAC-T cell lines were exposed to live MAP bacteria for 72 h. Thereafter, mRNA was extracted from infected and uninfected cells. Differentially expressed genes were compared between the wild type and the IL10Rα knockout cell lines. Gene ontology was performed based on the differentially expressed genes to determine which biological pathways were involved. RESULTS Immune system processes pathways were targeted to determine the effect of IL10Rα on the response to MAP infection. There was a difference in immune response between the wild type and IL10Rα knockout MAC-T cell lines, and less difference in immune response between infected and not infected IL10Rα knockout MAC-T cells, indicating IL10Rα plays an important role in the progression of MAP infection. Additionally, these comparisons allowed us to identify other genes involved in inflammation-mediated chemokine and cytokine signalling, interleukin signalling and toll-like receptor pathways. CONCLUSIONS Identifying differentially expressed genes in wild type and ILR10α knockout MAC-T cells infected with live MAP bacteria provided further evidence that IL10Rα contributes to mounting an immune response to MAP infection and allowed us to identify additional potential candidate genes involved in this process. We found there was a complex immune response during MAP infection that is controlled by many genes.
Collapse
Affiliation(s)
- Aisha Fong
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Christina M Rochus
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada.
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| | - Umesh K Shandilya
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Maria M M Muniz
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ankita Sharma
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Flavio S Schenkel
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Niel A Karrow
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Christine F Baes
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON, N1G 2W1, Canada.
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, 3002, Switzerland.
| |
Collapse
|
34
|
Shahdab N, Ward C, Hansbro PM, Cummings S, Young JS, Moheimani F. Distinct Effects of Respiratory Viral Infection Models on miR-149-5p, IL-6 and p63 Expression in BEAS-2B and A549 Epithelial Cells. Cells 2024; 13:919. [PMID: 38891051 PMCID: PMC11172188 DOI: 10.3390/cells13110919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Respiratory viruses cause airway inflammation, resulting in epithelial injury and repair. miRNAs, including miR-149-5p, regulate different pathological conditions. We aimed to determine how miR-149-5p functions in regulating pro-inflammatory IL-6 and p63, key regulators of airway epithelial wound repair, in response to viral proteins in bronchial (BEAS-2B) and alveolar (A549) epithelial cells. BEAS-2B or A549 cells were incubated with poly (I:C, 0.5 µg/mL) for 48 h or SARS-CoV-2 spike protein-1 or 2 subunit (S1 or S2, 1 μg/mL) for 24 h. miR-149-5p was suppressed in BEAS-2B challenged with poly (I:C), correlating with IL-6 and p63 upregulation. miR-149-5p was down-regulated in A549 stimulated with poly (I:C); IL-6 expression increased, but p63 protein levels were undetectable. miR-149-5p remained unchanged in cells exposed to S1 or S2, while S1 transfection increased IL-6 expression in BEAS-2B cells. Ectopic over-expression of miR-149-5p in BEAS-2B cells suppressed IL-6 and p63 mRNA levels and inhibited poly (I:C)-induced IL-6 and p63 mRNA expressions. miR-149-5p directly suppressed IL-6 mRNA in BEAS-2B cells. Hence, BEAS-2B cells respond differently to poly (I:C), S1 or S2 compared to A549 cells. Thus, miR-149-5p dysregulation may be involved in poly (I:C)-stimulated but not S1- or S2-stimulated increased IL-6 production and p63 expression in BEAS-2B cells.
Collapse
Affiliation(s)
- Nafeesa Shahdab
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - Christopher Ward
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia;
| | - Stephen Cummings
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - John S. Young
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - Fatemeh Moheimani
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK
| |
Collapse
|
35
|
Ghosh A, Payton A, Gallant SC, Rogers KL, Mascenik T, Hickman E, Love CA, Schichlein KD, Smyth TR, Kim YH, Rager JE, Gilmour MI, Randell SH, Jaspers I. Burn Pit Smoke Condensate-Mediated Toxicity in Human Airway Epithelial Cells. Chem Res Toxicol 2024; 37:791-803. [PMID: 38652897 PMCID: PMC11251002 DOI: 10.1021/acs.chemrestox.4c00064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Burn pits are a method of open-air waste management that was common during military operations in Iraq, Afghanistan, and other regions in Southwest Asia. Veterans returning from deployment have reported respiratory symptoms, potentially from exposure to burn pit smoke, yet comprehensive assessment of such exposure on pulmonary health is lacking. We have previously shown that exposure to condensates from burn pit smoke emissions causes inflammation and cytotoxicity in mice. In this study, we explored the effects of burn pit smoke condensates on human airway epithelial cells (HAECs) to understand their impact on cellular targets in the human lung. HAECs were cultured at the air-liquid interface (ALI) and exposed to burn pit waste smoke condensates (plywood, cardboard, plastic, mixed, and mixed with diesel) generated under smoldering and flaming conditions. Cytotoxicity was evaluated by measuring transepithelial electrical resistance (TEER) and lactate dehydrogenase (LDH) release; toxicity scores (TSs) were quantified for each exposure. Pro-inflammatory cytokine release and modulation of gene expression were examined for cardboard and plastic condensate exposures. Burn pit smoke condensates generated under flaming conditions affected cell viability, with flaming mixed waste and plywood exhibiting the highest toxicity scores. Cardboard and plastic smoke condensates modulated cytokine secretion, with GM-CSF and IL-1β altered in more than one exposure group. Gene expression of detoxifying enzymes (ALDH1A3, ALDH3A1, CYP1A1, CYP1B1, NQO1, etc.), mucins (MUC5AC, MUC5B), and cytokines was affected by several smoke condensates. Particularly, expression of IL6 was elevated following exposure to all burn pit smoke condensates, and polycyclic aromatic hydrocarbon acenaphthene was positively associated with the IL-6 level in the basolateral media of HAECs. These observations demonstrate that exposure to smoke condensates of materials present in burn pits adversely affects HAECs and that aberrant cytokine secretion and altered gene expression profiles following burn pit material smoke exposure could contribute to the development of airway disease.
Collapse
Affiliation(s)
- Arunava Ghosh
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Alexis Payton
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Environmental Sciences and Engineering (ESE), Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Samuel C. Gallant
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Keith L. Rogers
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599-7310 USA
| | - Teresa Mascenik
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Elise Hickman
- Department of Environmental Sciences and Engineering (ESE), Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599-7310 USA
| | - Charlotte A. Love
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Kevin D. Schichlein
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Timothy R. Smyth
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Yong Ho Kim
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Julia E. Rager
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Environmental Sciences and Engineering (ESE), Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599-7310 USA
| | - M. Ian Gilmour
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Scott H. Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Environmental Sciences and Engineering (ESE), Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599-7310 USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
36
|
Xue H, Chen Q, Lan X, Xu H, Yang H, Lin C, Xue Q, Xie B. Preventing CXCL12 elevation helps to reduce acute exacerbation of COPD in individuals co-existing type-2 diabetes: A bioinformatics and clinical pharmacology study. Int Immunopharmacol 2024; 132:111894. [PMID: 38569426 DOI: 10.1016/j.intimp.2024.111894] [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: 01/06/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
AIMS To investigate the immunology shared mechanisms underlying chronic obstructive pulmonary disease (COPD) and type 2 diabetes mellitus (T2DM) and examine the impact of anti-diabetic drugs on acute exacerbation of COPD (AECOPD). METHODS We analyzed GSE76925, GSE76894, GSE37768, and GSE25724 to identify differentially expressed genes. Hub-genes were identified through protein-protein interaction network analysis and evaluated by the receiver operating characteristic curve. CXCL12 emerged as a robust biomarker, and its correlation with lung function and CD8+ T cells were further quantified and validated. The activated signaling pathways were inferred through Gene set enrichment analysis (GSEA). The retrospective clinical analysis was executed to identify the influence of dipeptidyl peptidase-4 inhibitors (DPP-4i) on CXCL12 and evaluate the drug's efficacy in AECOPD. RESULTS The significant up-regulation of CXCL12 expression in patients with two diseases were revealed. CXCL12 exhibited a negative correlation with pulmonary function (r = -0.551, p < 0.05). Consistent with analysis in GSE76925 and GSE76894, the positive correlation between the proportion of CD8+ T cells was demonstrated(r=0.469, p<0.05). GSEA identified "cytokines interaction" as an activated signaling pathway, and the clinical study revealed the correlation between CXCL12 and IL-6 (r=0.668, p<0.05). In patients with COPD and T2DM, DDP-4i treatment exhibited significantly higher serum CXCL12, compared to GLP-1RA. Analysis of 187 COPD patients with T2DM indicated that the DPP-4i group had a higher frequency of AECOPD compared to the GLP-1RA group (OR 1.287, 95%CI [1.018-2.136]). CONCLUSIONS CXCL12 may represent a therapeutic target for COPD and T2DM. GLP-1RA treatment may be associated with lower CXCL12 levels and a lower risk of AECOPD compared to DPP-4i treatment. CLINICAL TRIAL REGISTRATION China Clinical Trial Registration Center(ChiCTR2200055611).
Collapse
Affiliation(s)
- Hong Xue
- Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Qianshun Chen
- Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Thoracic Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Xiuyan Lan
- Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Hang Xu
- Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Haitao Yang
- Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Changjian Lin
- Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Qing Xue
- The Third Clinical Medical College, Fujian Medical University, Ningde Municipal Hospital, Ningde 352100, Fujian, China; Ningde Municipal Hospital of Ningde Normal University, Ningde 352100, Fujian, China.
| | - Baosong Xie
- Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China; Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China.
| |
Collapse
|
37
|
Li Y, Lei H, Wen X, Cao H. A powerful approach to identify replicable variants in genome-wide association studies. Am J Hum Genet 2024; 111:966-978. [PMID: 38701746 PMCID: PMC11080610 DOI: 10.1016/j.ajhg.2024.04.004] [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: 08/19/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Replicability is the cornerstone of modern scientific research. Reliable identifications of genotype-phenotype associations that are significant in multiple genome-wide association studies (GWASs) provide stronger evidence for the findings. Current replicability analysis relies on the independence assumption among single-nucleotide polymorphisms (SNPs) and ignores the linkage disequilibrium (LD) structure. We show that such a strategy may produce either overly liberal or overly conservative results in practice. We develop an efficient method, ReAD, to detect replicable SNPs associated with the phenotype from two GWASs accounting for the LD structure. The local dependence structure of SNPs across two heterogeneous studies is captured by a four-state hidden Markov model (HMM) built on two sequences of p values. By incorporating information from adjacent locations via the HMM, our approach provides more accurate SNP significance rankings. ReAD is scalable, platform independent, and more powerful than existing replicability analysis methods with effective false discovery rate control. Through analysis of datasets from two asthma GWASs and two ulcerative colitis GWASs, we show that ReAD can identify replicable genetic loci that existing methods might otherwise miss.
Collapse
Affiliation(s)
- Yan Li
- School of Computer Science and Technology, Changchun University of Science and Technology, Changchun, Jilin 130022, China; School of Mathematics, Jilin University, Changchun, Jilin 130012, China
| | - Haochen Lei
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hongyuan Cao
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
38
|
Khan RN, Saporito AF, Zenon J, Goodman L, Zelikoff JT. Traffic-related air pollution in marginalized neighborhoods: a community perspective. Inhal Toxicol 2024; 36:343-354. [PMID: 38618680 DOI: 10.1080/08958378.2024.2331259] [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: 11/15/2023] [Accepted: 03/10/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVES Marginalized communities are exposed to higher levels of traffic-related air pollution (TRAP) than the general population. TRAP exposure is linked to pulmonary toxicity, neurotoxicity, and cardiovascular toxicity often through mechanisms of inflammation and oxidative stress. Early life exposure to TRAP is also implicated in higher rates of asthma in these same communities. There is a critical need for additional epidemiological, in vivo, and in vitro studies to define the health risks of TRAP exposure affecting the most vulnerable groups to set strict, protective air pollution standards in these communities. MATERIALS AND METHODS A literature review was conducted to summarize recent findings (2010-2024) concerning TRAP exposure and toxic mechanisms that are relevant to the most affected underserved communities. CONCLUSIONS Guided by the perspectives of NYC community scientists, this contemporary review of toxicological and epidemiological studies considers how the exposome could lead to disproportionate exposures and health effects in underserved populations.
Collapse
Affiliation(s)
- Rahanna N Khan
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Antonio F Saporito
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Jania Zenon
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Judith T Zelikoff
- Division of Environmental Medicine, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
39
|
Chung C, Park SY, Huh JY, Kim NH, Shon C, Oh EY, Park YJ, Lee SJ, Kim HC, Lee SW. Fine particulate matter aggravates smoking induced lung injury via NLRP3/caspase-1 pathway in COPD. J Inflamm (Lond) 2024; 21:13. [PMID: 38654364 DOI: 10.1186/s12950-024-00384-z] [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: 10/22/2022] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Exposure to noxious particles, including cigarette smoke and fine particulate matter (PM2.5), is a risk factor for chronic obstructive pulmonary disease (COPD) and promotes inflammation and cell death in the lungs. We investigated the combined effects of cigarette smoking and PM2.5 exposure in patients with COPD, mice, and human bronchial epithelial cells. METHODS The relationship between PM2.5 exposure and clinical parameters was investigated in patients with COPD based on smoking status. Alveolar destruction, inflammatory cell infiltration, and pro-inflammatory cytokines were monitored in the smoking-exposed emphysema mouse model. To investigate the mechanisms, cell viability and death and pyroptosis-related changes in BEAS-2B cells were assessed following the exposure to cigarette smoke extract (CSE) and PM2.5. RESULTS High levels of ambient PM2.5 were more strongly associated with high Saint George's respiratory questionnaire specific for COPD (SGRQ-C) scores in currently smoking patients with COPD. Combined exposure to cigarette smoke and PM2.5 increased mean linear intercept and TUNEL-positive cells in lung tissue, which was associated with increased inflammatory cell infiltration and inflammatory cytokine release in mice. Exposure to a combination of CSE and PM2.5 reduced cell viability and upregulated NLRP3, caspase-1, IL-1β, and IL-18 transcription in BEAS-2B cells. NLRP3 silencing with siRNA reduced pyroptosis and restored cell viability. CONCLUSIONS PM2.5 aggravates smoking-induced airway inflammation and cell death via pyroptosis. Clinically, PM2.5 deteriorates quality of life and may worsen prognosis in currently smoking patients with COPD.
Collapse
Affiliation(s)
- Chiwook Chung
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Department of Pulmonary and Critical Care Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Suk Young Park
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
| | - Jin-Young Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Chung- Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Na Hyun Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
| | - ChangHo Shon
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Efficacy Evaluation Center, WOOJUNGBIO Inc, Hwaseong, Republic of Korea
| | - Eun Yi Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Jun Park
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Jin Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hwan-Cheol Kim
- Department of Occupational and Environmental Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Wang E, Wroblewski KE, McClintock MK, Pinto JM, Witt LJ. Olfactory decline develops in parallel with frailty in older US adults with obstructive lung diseases. Int Forum Allergy Rhinol 2024; 14:819-827. [PMID: 37747949 PMCID: PMC10961252 DOI: 10.1002/alr.23273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Frailty is prevalent among older adults with asthma or chronic obstructive pulmonary disease (obstructive lung diseases [OLDs]). Frailty and OLD's co-occurrence is associated with increased hospitalization/mortality. Chemosensory dysfunction is closely connected to both OLD and frailty. We evaluated the utility of olfactory decline as a biomarker of frailty in the setting of OLD. METHODS We performed a prospective, longitudinal, nationally representative study of community-dwelling older US adults in the National Social Life, Health and Aging Project, an omnibus in-home survey. Respondents reported a physician's diagnosis of OLD. Decline in odor identification and sensitivity over 5 years and frailty (adapted fried frailty phenotype criteria) were measured using standard tools. Multivariate logistic regressions evaluated the association between OLD status, olfactory decline, and frailty. RESULTS We compared individuals with OLD (n = 98; mean age 71.2 years, 59.2% women) and those without OLD (n = 1036; mean age 69.5 years, 58.9% women). Olfactory identification decline was associated with developing frailty over the 5-year follow-up period in individuals with OLD (odds ratio [OR] = 9.1, 95% confidence interval [CI] = 2.1-38.6, p = 0.003). Olfactory decline predicted incidence of frailty in individuals with OLD (identification: OR = 4.8, 95% CI = 1.3-17.5, P = 0.018; sensitivity: OR = 6.1, 95%CI = 1.2-31.0, p = 0.030) but not in those without OLD adjusting for demographics, heavy alcohol use, current smoking, and comorbidity. Results were robust to different thresholds for olfactory decline and frailty development. CONCLUSIONS Older adults with OLD who experience olfactory decline face higher odds of developing frailty. Use of olfactory decline as a biomarker to identify frailty could allow earlier intervention and decrease adverse outcomes for high-risk older adults with OLD.
Collapse
Affiliation(s)
- Esther Wang
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Kristen E Wroblewski
- Department of Public Health Sciences, The University of Chicago, Chicago, Illinois, USA
| | - Martha K McClintock
- Department of Psychology and The Institute for Mind and Biology, The University of Chicago, Chicago, Illinois, USA
| | - Jayant M Pinto
- Department of Surgery, Section of Otolaryngology, The University of Chicago Medicine, Chicago, Illinois, USA
| | - Leah J Witt
- Divisions of Geriatrics and Pulmonary, Critical Care, Allergy and Sleep Medicine, The University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
41
|
Agarwal CD, Palka JM, Gajewski AJ, Khan DA, Brown ES. The efficacy of citalopram or escitalopram in patients with asthma and major depressive disorder. Ann Allergy Asthma Immunol 2024; 132:374-382. [PMID: 37952772 DOI: 10.1016/j.anai.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Major depressive disorder is common in people with asthma. Yet, few studies have evaluated depression treatment in those with asthma. OBJECTIVE To explore the relationship between antidepressant use, depressive symptoms, and asthma control, pooled data from 3 randomized trials of either citalopram or escitalopram were assessed. METHODS Linear fixed effects and binary logistic regression analyses were conducted with between-subject covariates including treatment group, (original) study, and demographics. The within-subject effect of visit, and a treatment group-visit (between-within) interaction effect, were also evaluated. Analyses were repeated in a high asthma exacerbation subgroup having at least 3 oral corticosteroid bursts in the previous 12 months. Outcomes included the Hamilton rating scale for depression (HAM-D17), the 7-item asthma control questionnaire (ACQ), and oral corticosteroid use (yes or no). RESULTS In the pooled sample (n = 255), the antidepressant treatment group exhibited lower HAM-D17 overall (P ≤ .001) and a lower likelihood for oral corticosteroid use (P ≤ .001) relative to the placebo group. In the high-exacerbation subgroup (n = 96), treatment group participants had lower overall asthma control questionnaire (P = .004) and HAM-D17 scores (P ≤ .001), and a lower likelihood of oral corticosteroid use (P = .003), relative to placebo participants. All treatment group interaction effects were not significant. CONCLUSION Citalopram or escitalopram exhibited efficacy in reducing depressive symptoms and the need for rescue oral corticosteroids in patients with asthma and major depressive disorder. Future work should determine whether selective serotonin reuptake inhibitors are effective at improving asthma outcomes in those with asthma who are not depressed. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT00621946 and NCT01324700 (one study was conducted before ClinicalTrials.gov requirements).
Collapse
Affiliation(s)
- Catherine D Agarwal
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jayme M Palka
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexander J Gajewski
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - David A Khan
- Division of Allergy and Immunology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - E Sherwood Brown
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas; The Altshuler Center for Education and Research, Metrocare Services, Dallas, Texas.
| |
Collapse
|
42
|
Al-Ahmad M, Ali A, Maher A, Haider MZ. Association between interleukin-6-174G/C gene polymorphism and asthma severity: exploring the role of total serum IgE, blood eosinophils, and FeNO as markers of type 2 inflammation. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:15. [PMID: 38388670 PMCID: PMC10885618 DOI: 10.1186/s13223-024-00880-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND While a connection has been established between serum interleukin-6 (IL-6) levels and the IL-6 gene (- 174G/C) polymorphism in allergic diseases such as asthma, its specific association with severe asthma remains unexplored. This study examined the relationship between the IL-6 (- 174G/C) gene polymorphism and mild and severe asthma, focusing on its influence on type 2 inflammation. METHODS Our study comprised 98 patients with mild asthma and 116 with severe asthma. Additionally, we recruited 121 healthy participants to serve as controls for comparative analyses. The IL-6 gene (- 174G/C) polymorphism was assessed utilizing the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. RESULTS In our study, the risk of mild asthma exhibited a significant fourfold increase in individuals with the GG genotype pattern compared to healthy controls, yielding an odds ratio (OR) of 4.4 (p < 0.001). Conversely, we found no significant correlation between the IL-6 - 174G/C gene polymorphism and severe asthma when compared to the healthy control group. However, a noteworthy pattern emerged when we compared subgroups of mild and severe asthma. The risk of severe asthma increased fivefold in individuals with the GC polymorphism pattern, with an OR of 4.99 (p < 0.001), while the likelihood of mild asthma showed a similar fourfold increase with the GG polymorphism pattern, OR = 4.4 (p < 0.001). Consequently, we observed a significantly higher frequency of the C allele in patients with severe asthma, whereas the G allele was more prevalent in individuals with mild asthma (p = 0.05). Additionally, the correlation between markers of type 2 inflammation and the dominant model of the IL-6 gene -174G/C polymorphism (CC + CG vs GG) revealed a significant increase in total serum immunoglobulin E (IgE), Blood Eosinophil Counts (BEC), and Fractional Exhaled Nitric Oxide (FeNO) levels in asthmatic patients with the CC + CG gene pattern compared to those with GG, with p-values of 0.04, 0.03, and 0.04, respectively. Furthermore, after adjusting for other risk factors, the likelihood of developing severe asthma increased from fourfold to eightfold, with an OR of 8.12 (p = 0.01) with (CC + CG) gene pattern. Other predictors for severe asthma included older age and childhood-onset disease (OR = 1.13 and 19.19, p < 0.001). Allergic rhinitis (AR) and nasal polyps (NP) also demonstrated a substantial association with an increased risk of severe asthma, with odds ratios of 5 and 32.29 (p = 0.01 and < 0.001), respectively. Additionally, elevated Body Mass Index (BMI), BEC, and FeNO were linked to severe asthma, with ORs of 1.11, 1.00, and 1.04, respectively (p = 0.04, 0.05, and 0.001). CONCLUSION This study illuminated the intricate relationship between the IL-6 gene polymorphism, type 2 inflammation markers, and diverse risk factors in shaping asthma severity. As a significant association between the GG polymorphism of the IL-6 gene (- 174G/C) and mild asthma was found, while possessing at least one C allele, whether in a homozygous (CC) or heterozygous (CG) combination, independently predicts the likelihood of severe asthma.
Collapse
Affiliation(s)
- Mona Al-Ahmad
- Department of Microbiology, College of Medicine, Kuwait University, Safat, P.O. Box 24923, 13110, Kuwait City, Kuwait.
- Department of Allergy, Al-Rashed Allergy Center, Ministry of Health, Kuwait City, Kuwait.
| | - Asmaa Ali
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
- Department of Allergy, Al-Rashed Allergy Center, Ministry of Health, Kuwait City, Kuwait
- Department of Pulmonary Medicine, Abbassia Chest Hospital, Ministry of Health, Cairo, Egypt
| | - Ahmed Maher
- Department of Allergy, Al-Rashed Allergy Center, Ministry of Health, Kuwait City, Kuwait
| | - Mohammad Z Haider
- Department of Pediatrics, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
43
|
Sousa de Almeida M, Lee A, Itel F, Maniura-Weber K, Petri-Fink A, Rothen-Rutishauser B. The Effect of Substrate Properties on Cellular Behavior and Nanoparticle Uptake in Human Fibroblasts and Epithelial Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:342. [PMID: 38392715 PMCID: PMC10892529 DOI: 10.3390/nano14040342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
The delivery of nanomedicines into cells holds enormous therapeutic potential; however little is known regarding how the extracellular matrix (ECM) can influence cell-nanoparticle (NP) interactions. Changes in ECM organization and composition occur in several pathophysiological states, including fibrosis and tumorigenesis, and may contribute to disease progression. We show that the physical characteristics of cellular substrates, that more closely resemble the ECM in vivo, can influence cell behavior and the subsequent uptake of NPs. Electrospinning was used to create two different substrates made of soft polyurethane (PU) with aligned and non-aligned nanofibers to recapitulate the ECM in two different states. To investigate the impact of cell-substrate interaction, A549 lung epithelial cells and MRC-5 lung fibroblasts were cultured on soft PU membranes with different alignments and compared against stiff tissue culture plastic (TCP)/glass. Both cell types could attach and grow on both PU membranes with no signs of cytotoxicity but with increased cytokine release compared with cells on the TCP. The uptake of silica NPs increased more than three-fold in fibroblasts but not in epithelial cells cultured on both membranes. This study demonstrates that cell-matrix interaction is substrate and cell-type dependent and highlights the importance of considering the ECM and tissue mechanical properties when designing NPs for effective cell targeting and treatment.
Collapse
Affiliation(s)
- Mauro Sousa de Almeida
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
| | - Aaron Lee
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
- Department of Bioengineering, Imperial College London, South Kensington, London SW7 2BP, UK
| | - Fabian Itel
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biomimetic Membranes and Textiles, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland;
| | - Katharina Maniura-Weber
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Biointerfaces, Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland;
| | - Alke Petri-Fink
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Barbara Rothen-Rutishauser
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; (M.S.d.A.); (A.L.); (A.P.-F.)
| |
Collapse
|
44
|
Jung DY, Park SM, Lim GH, Seo KW, Oh YI, Youn HY. Assessment of MMP-9 and clinical characteristics in dogs with tracheal collapse based on cough severity and fluoroscopic findings: a cross-sectional study. BMC Vet Res 2024; 20:52. [PMID: 38341543 PMCID: PMC10858467 DOI: 10.1186/s12917-023-03872-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/28/2023] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Tracheal collapse (TC), a common disease in dogs, is characterized by cough; however, little is known about the serum biomarkers that can objectively evaluate the severity of cough in canine TC. Furthermore, studies elucidating the relationship of fluoroscopic characteristics with the severity of cough are lacking. Therefore, this study aimed to evaluate the relationship between cough severity and clinical characteristics, fluoroscopic images, and new serum biomarkers in canine TC. RESULTS Fifty-one client-owned dogs diagnosed with TC based on fluoroscopic and clinical signs were enrolled in this study and divided into three groups according to the severity of cough (grade of cough: 0, 1, and 2). Signalments, comorbidities, and fluoroscopic characteristics were compared among the groups retrospectively. The serum matrix metalloproteinase-9 (MMP-9), interleukin-6 (IL-6), surfactant protein-A (SP-A), and syndecan-1 (SDC-1) levels were measured in all groups. No significant differences in age, breed, sex, or clinical history were observed among the groups. Concomitant pharyngeal collapse increased significantly with the severity of cough (p = .031). Based on the fluoroscopic characteristics, the TC grade of the carinal region increased significantly and consistently with the grade of cough (p = .03). The serum MMP-9 level was significantly higher in the grade 2 group than that in the grade 0 group (p = .014). The serum IL-6 level was significantly lower in the grade 1 group than that in the grade 0 group (p = .020). The serum SP-A and SDC-1 levels did not differ significantly among the groups. CONCLUSIONS The severity of cough with the progression of TC can be predicted with the fluoroscopic TC grade at the carinal region. MMP-9 may be used as an objective serum biomarker that represents cough severity to understand the pathogenesis.
Collapse
Affiliation(s)
- Da-Yeon Jung
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 00826, Republic of Korea
| | - Su-Min Park
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 00826, Republic of Korea
| | - Ga-Hyun Lim
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 00826, Republic of Korea
| | - Kyoung-Won Seo
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 00826, Republic of Korea
| | - Ye-In Oh
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 00826, Republic of Korea.
| |
Collapse
|
45
|
Lu Y, Xu J, Tang R, Zeng P, Li Z, You J, Li T, Zhang T, Ma X, He Y, Chen N, Deng X, Wu J. Edible pueraria lobata-derived exosome-like nanovesicles ameliorate dextran sulfate sodium-induced colitis associated lung inflammation through modulating macrophage polarization. Biomed Pharmacother 2024; 170:116098. [PMID: 38154276 DOI: 10.1016/j.biopha.2023.116098] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/17/2023] [Accepted: 12/26/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Inflammatory bowel diseases (IBD), such as severe colitis, are associated with the development of lung inflammation and tissue damage. Pueraria lobata (P. lobata) plays an essential role in controlling cytokines. However, the exact mechanism of the inflammation response is still unknown. PURPOSE To investigate the effects of the P. lobata-derived exosomes-like nanovesicles (PLDENs) on colitis and their role in the lung inflammatory response. METHODS In this study, we investigated the effects of PLDENs on the dextran sulfate sodium (DSS)-induced colitis and explored the mechanisms by forming the gut-lung axis. PLDENs were characterized by mass spectrometry-based proteomic analysis. RESULTS The results showed that PLDENs had significant preventive effects in DSS-induced colitis and pathological changes in colons in a dose-dependent manner. Simultaneously, the treatment of PLDENs could effectively reduce inflammatory changes in the lung. PLDENs could selectively regulate the composition of gut microbiota. CONCLUSION These data suggested that the treatment of PLDENs could 'attenuate DSS-induced colitis and lung inflammation, providing an efficacious supplement for reducing co-morbidities in IBD patients.
Collapse
Affiliation(s)
- Yu Lu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Jin Xu
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Rui Tang
- Department of Pathology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Peiyuan Zeng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Ziyu Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Jingcan You
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Tian Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Tao Zhang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Xiaoyu Ma
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Yuqian He
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Ni Chen
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Xin Deng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China
| | - Jianbo Wu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, China; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
46
|
Wu Y, Ni Z, Wang S, Sun Y, Luo X, Wang X, Liu J. The mechanism of Sanzi Yangqin decoction for asthma treatment based on network pharmacology and experimental verification. BMC Complement Med Ther 2023; 23:452. [PMID: 38093206 PMCID: PMC10717567 DOI: 10.1186/s12906-023-04272-6] [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: 11/22/2022] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Asthma is a chronic airway inflammatory disease characterized by airway inflammation, mucus hypersecretion, airway hyper-reactivity. Sanzi Yangqin Decoction (SZYQD) is widely prescribed for asthma treatment. Its anti-asthma activities have been reported in animal model, but the exact mechanism and targets of SZYQD in asthma treatment have not been fully elucidated. METHODS A network pharmacological approach was used to predict the active components, targets, and signalling pathways of SZYQD in asthma, including potential target prediction, protein‒protein interaction (PPI) network construction and analysis, and Gene Ont (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The active ingredients were identified from the SZYQD, and were molecular docked according to the results of network pharmacology. A mouse model of asthma induced by ovalbumin (OVA) and lipopolysaccharide (LPS) was constructed to evaluate the therapeutic effect of SZYQD. Furthermore, the effects of SZYQD and its active ingredients were tested in vitro for regulating inflammation and MUC5AC expression (two main pathophysiologic abnormalities of asthma) in macrophages and airway epithelial cells by using Real-time PCR and western blotting. RESULTS A total of 28 active ingredients and 111 HUB genes were screened in the relevant databases, including three key ingredients (luteolin, β-carotene, and Sinapine) and nine core target genes (JUN, CTNNB1, IL10, TP53, AKT1, STAT3, TNF, IL6 and EGFR). KEGG and GO analysis indicated that the potential anti-asthmatic mechanisms of SZYQD were related to PI3K-Akt signalling pathway and response to lipopolysaccharide, etc. In the in vivo asthmatic model, our findings demonstrated that SZYQD exerted a protective effect against asthmatic mice induced by OVA and LPS through the inhibition of inflammation and mucus overproduction. Consistently, cell experiments showed that the SZYQD extract or the key active ingredients luteolin significantly decreased lipopolysaccharide (LPS)-induced IL-6 expression and activation of the NF-κB pathway in macrophages. In addition, SZYQD extract or luteolin inhibited activation of the AKT pathway and expression of MUC5AC induced by EGF in airway epithelial cells. CONCLUSION The anti-asthmatic mechanism of SZYQD might be associated with inhibiting inflammation and airway mucus hypersecretion by regulating the NF-κB and AKT signalling pathways as predicted by network pharmacology, which provides more evidence for the application of SZYQD in asthma treatment.
Collapse
Affiliation(s)
- Yue Wu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Zhenhua Ni
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- Central lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Shiqiang Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yipeng Sun
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Xuming Luo
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Jinjin Liu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
47
|
Akkenepally SV, Yombo DJK, Yerubandi S, Reddy GB, Deshpande DA, McCormack FX, Madala SK. Interleukin 31 receptor α promotes smooth muscle cell contraction and airway hyperresponsiveness in asthma. Nat Commun 2023; 14:8207. [PMID: 38081868 PMCID: PMC10713652 DOI: 10.1038/s41467-023-44040-1] [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: 02/08/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by airway hyperresponsiveness (AHR), inflammation, and goblet cell hyperplasia. Multiple cytokines, including IFNγ, IL-4, and IL-13 are associated with asthma; however, the mechanisms underlying the effects of these cytokines remain unclear. Here, we report a significant increase in the expression of IL-31RA, but not its cognate ligand IL-31, in mouse models of allergic asthma. In support of this, IFNγ, IL-4, and IL-13 upregulated IL-31RA but not IL-31 in both human and mice primary airway smooth muscle cells (ASMC) isolated from the airways of murine and human lungs. Importantly, the loss of IL-31RA attenuated AHR but had no effect on inflammation and goblet cell hyperplasia in mice challenged with allergens or treated with IL-13 or IFNγ. We show that IL-31RA functions as a positive regulator of muscarinic acetylcholine receptor 3 expression, augmenting calcium levels and myosin light chain phosphorylation in human and murine ASMC. These findings identify a role for IL-31RA in AHR that is distinct from airway inflammation and goblet cell hyperplasia in asthma.
Collapse
Affiliation(s)
- Santhoshi V Akkenepally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Biochemistry, National Institute of Nutrition, Hyderabad, Telangana, India
| | - Dan J K Yombo
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sanjana Yerubandi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Deepak A Deshpande
- Division of Pulmonary, Allergy, and Critical Care Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Satish K Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
48
|
El-Husseini ZW, Khalenkow D, Lan A, van der Molen T, Brightling C, Papi A, Rabe KF, Siddiqui S, Singh D, Kraft M, Beghe B, van den Berge M, van Gosliga D, Nawijn MC, Rose-John S, Koppelman GH, Gosens R. An epithelial gene signature of trans-IL-6 signaling defines a subgroup of type 2-low asthma. Respir Res 2023; 24:308. [PMID: 38062491 PMCID: PMC10704725 DOI: 10.1186/s12931-023-02617-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Asthma is stratified into type 2-high and type 2-low inflammatory phenotypes. Limited success has been achieved in developing drugs that target type 2-low inflammation. Previous studies have linked IL-6 signaling to severe asthma. IL-6 cooperates with soluble-IL-6Rα to activate cell signaling in airway epithelium. OBJECTIVE We sought to study the role of sIL-6Rα amplified IL-6 signaling in airway epithelium and to develop an IL-6+ sIL-6Rα gene signature that may be used to select asthma patients who potentially respond to anti-IL-6 therapy. METHODS Human airway epithelial cells were stimulated with combinations of IL-6, sIL-6Rα, and inhibitors, sgp130 (Olamkicept), and anti-IL-6R (Tocilizumab), to assess effects on pathway activation, epithelial barrier integrity, and gene expression. A gene signature was generated to identify IL-6 high patients using bronchial biopsies and nasal brushes. RESULTS Soluble-IL-6Rα amplified the activation of the IL-6 pathway, shown by the increase of STAT3 phosphorylation and stronger gene induction in airway epithelial cells compared to IL-6 alone. Olamkicept and Tocilizumab inhibited the effect of IL-6 + sIL-6Rα on gene expression. We developed an IL-6 + sIL-6Rα gene signature and observed enrichment of this signature in bronchial biopsies but not nasal brushes from asthma patients compared to healthy controls. An IL-6 + sIL-6Rα gene signature score was associated with lower levels of sputum eosinophils in asthma. CONCLUSION sIL-6Rα amplifies IL-6 signaling in bronchial epithelial cells. Higher local airway IL-6 + sIL-6Rα signaling is observed in asthma patients with low sputum eosinophils.
Collapse
Affiliation(s)
- Zaid W El-Husseini
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Dmitry Khalenkow
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Andy Lan
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Thys van der Molen
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Chris Brightling
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - Alberto Papi
- Department of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F Rabe
- Department of Medicine, Christian Albrechts University Kiel, Kiel and Lungen Clinic Grosshansdorf (Members of the German Center for Lung Research (DZL)), Grosshansdorf, Germany
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College and Imperial NIHR Biomedical Research Centre, London, UK
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, University of Manchester, Manchester, UK
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Bianca Beghe
- University of Modena and Reggio Emilia, AOU of Modena, Modena, Italy
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Djoke van Gosliga
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pathology and Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Martijn C Nawijn
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pathology and Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands.
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
49
|
Tahir A, Akhtar MF, Saleem A, Naveed M, Ajiboye BO, Anwar F, Khan A. Curative potential of Populus ciliata Wall ex. Royle extract against adjuvant-induced arthritis and peripheral neuropathy in Wistar rats. Inflammopharmacology 2023; 31:3081-3100. [PMID: 37266813 DOI: 10.1007/s10787-023-01248-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/06/2023] [Indexed: 06/03/2023]
Abstract
Populus ciliata (PCCR) is traditionally used to treat muscular swelling, inflammation, pain, and fever. The current study was designed to validate the potential of aqueous ethanolic extract of the plant against inflammation, peripheral neuropathy, and pain in arthritic rats. The PCCR was chemically characterized by gas chromatography-mass spectroscopy and high-performance liquid chromatography. In vitro antioxidant, and in vitro anti-inflammatory assays were carried out on PCCR. For anti-arthritic potential, Wistar rats' rear paws were injected with 0.1 ml Complete Freund's Adjuvant using methotrexate (3 mg/kg/week) as standard control. PCCR at 100, 200, and 400 mg/kg was given orally to arthritic rats for 21 days. The PCCR exhibited significant inhibition of bovine serum albumin denaturation (IC-50: 202.1 µg/ml), egg albumin denaturation (IC-50:553.5 mg/ml) and RBC membrane stabilization (IC-50: 122.5 µg/ml) and antioxidant (IC-50 = 49.43 µg/ml) activities. The PCCR notably decreased the paw diameter and increased body weight of treated arthritic animals as equated to diseased control. The treatment notably (p < 0.05-0.0001) decreased malondialdehyde, and increased superoxide dismutase, reduced glutathione, and catalase in the liver and sciatic nerve homogenate in compared to diseased rats. The PCCR treatment remarkably (p < 0.05-0.0001) regulated the levels of nor-adrenaline and serotonin in sciatic nerve in contrast to diseased rats. Treatment with PCCR improved the motor activity, pain, ligament degeneration, and synovial hyperplasia in arthritic rats. Moreover, PCCR significantly (p < 0.01-0.0001) decreased the IL-6 and TNF-α. It is evident from the current study that PCCR had ameliorated polyarthritis and peripheral neuropathy through reduction of inflammatory markers, and improvement of oxidative stress might be due to presence of phenolic acids, flavonoids, phytosterols, and other fatty acids.
Collapse
Affiliation(s)
- Amna Tahir
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan.
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Muhammad Naveed
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Basiru Olaitan Ajiboye
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti, Ekiti State, Nigeria
| | - Fareeha Anwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan
| | - Aslam Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan
| |
Collapse
|
50
|
Akar-Ghibril N, Greco KF, Jackson-Browne M, Phipatanakul W, Permaul P. High plasma IL-6 levels may enhance the adverse effects of mouse allergen exposure in urban schools on asthma morbidity in children. J Allergy Clin Immunol 2023; 152:1677-1682. [PMID: 37541489 PMCID: PMC10837306 DOI: 10.1016/j.jaci.2023.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/25/2023] [Accepted: 06/16/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Few data on the relationships between environmental exposures, asthma morbidity, and systemic IL-6 inflammation exist. OBJECTIVE We sought to determine whether baseline plasma IL-6 level is associated with increased asthma morbidity in children exposed to mouse allergen in inner-city classrooms. METHODS Data from the longitudinal School Inner-City Asthma Studies of 215 children with asthma, aged 4 to 14 years and recruited from urban elementary schools, were analyzed. Given the unknown threshold of IL-6 risk levels and skewness of the distribution, the children were stratified into tertiles as follows: low baseline IL-6 level (<0.013 pg/mL), moderate baseline IL-6 level (0.013-0.302 pg/mL), and high baseline IL-6 level (>0.302 pg/mL). Relationships between plasma IL-6 level and body mass index (BMI) percentile, inflammatory markers, lung function, mouse allergen exposure, and asthma outcomes were assessed. RESULTS Cross-sectional analysis demonstrated that increasing IL-6 level was associated with higher BMI percentile (P < .0001), C-reactive protein level (P = .0006), and blood neutrophil count (P = .0024). IL-6 was not associated with type 2 inflammatory markers, including blood eosinophil count, allergic sensitization, or fractional exhaled nitric oxide level. Longitudinal analysis showed that children with high IL-6 levels had a higher number of days with asthma symptoms than did those children with moderate (incidence rate ratio = 1.74 [95% CI = 1.10-2.77]; P = .0187) or low (incidence rate ratio =1.83 [95% CI = 1.21-2.77]; P = .0043) IL-6 levels. Children with high IL-6 levels who were exposed to increasing levels of mouse allergen exhibited lower ratios of FEV1 value to forced vital capacity than did children with moderate IL-6 levels (β = -0.0044 [95% CI = -0.0073 to -0.0015]; pairwise interaction P = .0028) or low IL-6 levels (β = -0.0042 [95% CI = - 0.0070 to -0.0013]; pairwise interaction P = .0039). CONCLUSIONS Inner-city children with asthma and high plasma IL-6 levels are more likely to have an increased BMI, elevated C-reactive protein level, elevated blood neutrophil count, and greater asthma symptoms. High IL-6 level appears to increase susceptibility to the effects of classroom exposure to mouse allergen on lung function in urban children.
Collapse
Affiliation(s)
- Nicole Akar-Ghibril
- Division of Immunology, Allergy, and Rheumatology, Joe DiMaggio Children's Hospital, Hollywood, Fla
| | - Kimberly F Greco
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | | | - Wanda Phipatanakul
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass.
| | - Perdita Permaul
- Division of Pulmonology, Allergy and Immunology, Weill Cornell Medicine/New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY
| |
Collapse
|