1
|
Mauclin M, Guillien A, Niespodziana K, Boudier A, Schlederer T, Bajic M, Errhalt P, Borochova K, Pin I, Gormand F, Vernet R, Bousquet J, Bouzigon E, Valenta R, Siroux V. Association between asthma and IgG levels specific for rhinovirus and respiratory syncytial virus antigens in children and adults. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100342. [PMID: 39507925 PMCID: PMC11536052 DOI: 10.1016/j.jacig.2024.100342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 11/08/2024]
Abstract
Background Viral infections in childhood, especially to rhinovirus (RV) and respiratory syncytial virus (RSV), are associated with asthma inception and exacerbation. However, little is known about the role of RV- and RSV-specific antibodies in childhood versus adult asthma. Objective We sought to investigate associations between RV- and RSV-specific IgG levels and asthma phenotypes in children and adults. Methods The analysis included 1771 samples from participants of the Epidemiological Study on the Genetics and Environment of Asthma (530 children; age [mean ± SD], 11.1 ± 2.8, and 1241 adults; age [mean ± SD], 43.4 ± 16.7, among whom 274 and 498 had ever asthma, respectively). RSV- and RV-specific IgG levels were determined using microarrayed virus-derived antigens and peptides. Cross-sectional associations between standardized RSV- and RV-specific IgG levels and asthma phenotypes were estimated by multiple regression models. Results In children, ever asthma was associated with higher IgG levels specific to RV, especially to RV-A and RV-C, and to RSV (adjusted odds ratios [95% CI] for a 1 - SD increase in IgG levels were 1.52 [1.16-1.99], 1.42 [1.10-1.83], and 1.24 [0.99-1.54], respectively). These associations were stronger for moderate to severe asthma than for mild asthma. Conversely in adults, ever asthma was associated with lower RV-A, RV-B, and RV-C IgG levels (adjusted odds ratios [95% CI] were 0.86 [0.74-0.99], 0.83 [0.73-0.95], and 0.85 [0.73-0.99], respectively). Conclusions Our results suggest that the association between respiratory virus-specific antibody levels and asthma varies during life, with asthma associated with higher levels of IgG to RSV, RV-A, and RV-C in children and lower levels of IgG responses to RV-A/B/C in adults.
Collapse
Affiliation(s)
- Marion Mauclin
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Alicia Guillien
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Katarzyna Niespodziana
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
| | - Anne Boudier
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
- CHU Grenoble-Alpes, Grenoble, France
| | - Thomas Schlederer
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
| | - Maja Bajic
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
- the Department of Pneumology, University Hospital Krems and Karl Landsteiner University of Health Sciences, Krems
| | - Peter Errhalt
- the Department of Pneumology, University Hospital Krems and Karl Landsteiner University of Health Sciences, Krems
| | - Kristina Borochova
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
| | - Isabelle Pin
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | | | - Raphaël Vernet
- Université Paris Cité, INSERM UMR 1124, Group of Genomic Epidemiology of Multifactorial Diseases, Paris
| | - Jean Bousquet
- Université Paris-Saclay, UVSQ, Université Paris-Sud, INSERM, Equipe d’Epidémiologie Respiratoire Intégrative, CESP, Villejuif
| | - Emmanuelle Bouzigon
- Université Paris Cité, INSERM UMR 1124, Group of Genomic Epidemiology of Multifactorial Diseases, Paris
| | - Rudolf Valenta
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
- Karl Landsteiner University, Krems
| | - Valérie Siroux
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
2
|
Chen K, Lu S, Shi K, Ali MH, Liu J, Yin F, Yin W. Hyperoside attenuates sepsis-induced acute lung injury by Nrf2 activation and ferroptosis inhibition. Int Immunopharmacol 2025; 145:113734. [PMID: 39657533 DOI: 10.1016/j.intimp.2024.113734] [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: 08/21/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening condition associated with high morbidity and mortality rates in intensive care units (ICUs). Emerging evidence from clinical studies suggests that compounds derived from traditional Chinese medicine (TCM) have shown promising therapeutic effects in treating sepsis-induced ALI. Hyperoside is a bioactive compound extracted from TCM. Prior studies reported that hyperoside exhibits potent anti-inflammatory, antioxidant, and organ-protective properties, however, the underlying mechanisms of its effects on ALI remain unclear. Hyperoside pretreatment significantly reduced inflammation, iron accumulation, and lipid peroxidation in the pulmonary tissues of ALI mice induced by CLP and in LPS-stimulated MLE-12 cells. In particular, hyperoside preferentially binds with Keap1 at Arg380 and Arg415, thereby inhibiting the ubiquitin-mediated degradation of nuclear Nrf2, promoting its translocation to the nucleus, and leading to upregulation of anti-ferroptosis gene expression. Moreover, the protective effects of hyperoside were significantly abrogated after Nrf2 expression was silenced or its activity was inhibited by chemical inhibitors, highlighting that Nrf2 is critically involved in the impact of hyperoside. This study confirms that hyperoside exhibits a therapeutically protective effect against sepsis-induced ALI by inhibiting ferroptosis through Nrf2-mediated signaling pathway. Hyperoside acts as an Nrf2 activator by preferentially binding to Arg380 and Arg415 of Keap1 and disrupting the Keap1/Nrf2 interaction.
Collapse
Affiliation(s)
- Kuida Chen
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Shipeng Lu
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Ke Shi
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China; Medical Research Center, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, China
| | - Mustafa Hussein Ali
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jian Liu
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210029, China.
| | - Wu Yin
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
3
|
Wang C, Liu Z, Xie X, Li Y, Sun L. Klotho improves Der p1-induced bronchial epithelial cell damage by inhibiting endoplasmic reticulum stress to regulate mitochondrial function. Tissue Cell 2024; 93:102646. [PMID: 39693897 DOI: 10.1016/j.tice.2024.102646] [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: 09/18/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024]
Abstract
Asthma is a prevalent chronic pediatric lung disease which is commonly perceived as a syndrome of airway inflammation characterized by cough and wheeze in clinic. Klotho is implicated in diverse cellular activities, including inflammation, oxidative stress and apoptosis. This paper aims to explore the role of klotho in asthma and investigate the relevant molecular reaction mechanisms. To this end, we used Der p1 to induce an in vitro asthma model in BEAS-2B cells. Klotho expression was manipulated in Der p1-induced BEAS-2B cells with overexpression and its effects on Der p1-induced pathologies including apoptosis and inflammatory cytokine levels and the expressions of oxidative stress-related markers and major mediators in endoplasmic reticulum stress (ER stress) were investigated. Mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) opening were also detected. Our data demonstrated that Der p1 stimulation decreased klotho expression and klotho overexpression inhibited the Der p1-induced inflammation, oxidative stress and apoptosis. Overexpressing klotho inhibited ER stress to modulate mitochondrial function. The inhibitory effects of klotho overexpression were reversed by ER stress agonist tunicamycin. This paper validated the role of klotho in asthma pathogenies and developed prospective therapeutic targets for asthma treatment.
Collapse
Affiliation(s)
- Caiwen Wang
- Changchun University of Chinese Medicine, Changchun City, Jilin Province 130117, PR China
| | - Zhimei Liu
- Children's Diagnosis and Treatment Center, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun City, Jilin Province 130000, PR China
| | - Xiaofei Xie
- Children's Diagnosis and Treatment Center, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun City, Jilin Province 130000, PR China
| | - Yiquan Li
- Changchun University of Chinese Medicine, Changchun City, Jilin Province 130117, PR China.
| | - Liping Sun
- Children's Diagnosis and Treatment Center, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun City, Jilin Province 130000, PR China.
| |
Collapse
|
4
|
Hao Y, Wang W, Zhang L, Li W. Pyroptosis in asthma: inflammatory phenotypes, immune and non-immune cells, and novel treatment approaches. Front Pharmacol 2024; 15:1452845. [PMID: 39611173 PMCID: PMC11603363 DOI: 10.3389/fphar.2024.1452845] [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: 06/21/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024] Open
Abstract
Pyroptosis is a form of inflammatory programmed cell death, and is activated by pathogen infections or endogenous danger signals. The canonical pyroptosis process is characterized by the inflammasome (typically NLRP3)-mediated activation of caspase-1, which in turn cleaves and activates IL-1β and IL-18, as well as gasdermin D, which is a pore-forming executor protein, leading to cell membrane rupture, and the release of proinflammatory cytokines and damage-associated molecular pattern molecules. Pyroptosis is considered a part of the innate immune response. A certain level of pyroptosis can help eliminate pathogenic microorganisms, but excessive pyroptosis can lead to persistent inflammatory responses, and cause tissue damage. In recent years, pyroptosis has emerged as a crucial contributor to the development of chronic inflammatory respiratory diseases, such as asthma. The present study reviews the involvement of pyroptosis in the development of asthma, in terms of its role in different inflammatory phenotypes of the disease, and its influence on various immune and non-immune cells in the airway. In addition, the potential therapeutic value of targeting pyroptosis for the treatment of specific phenotypes of asthma is discussed.
Collapse
Affiliation(s)
- Yuqiu Hao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Wenrui Wang
- Department of Hepatopancreatobiliary Medicine, Digestive Diseases Center, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lin Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Fernandes AMS, da Silva ES, Silva RC, Silveira EF, Santiago LF, de Andrade Belitardo EMM, Alves VDS, Bôas DSV, de Freitas LAR, Ferreira F, Jacquet A, Pacheco LGC, Alcantara-Neves NM, Pinheiro CS. Therapeutic potential of a novel hybrid protein: Mitigating allergy and airway remodeling in chronic asthma models induced by Dermatophagoides pteronyssinus. Mol Immunol 2024; 175:121-131. [PMID: 39357098 DOI: 10.1016/j.molimm.2024.09.005] [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: 07/17/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND The house-dust mite Dermatophagoides pteronyssinus is a key trigger of allergic asthma. Therefore, it is essential to develop new vaccines that can alter inflammatory processes and airway remodeling. The goal of this study was to test the hypoallergenic and immunogenic characteristics of the hypoallergen rDer p 2231 in a murine model of chronic asthma induced by D. pteronyssinus. METHODS For this, we measured the levels of IgE, IgG1, IgG2a, and cytokines produced by mice receiving the rDer p 2231 protein. Histopathological parameters of the chronic inflammatory response were also investigated by assessing inflammation and airway remodeling. RESULTS rDer p 2231 given as a therapeutic vaccine, led to a reduction in the production of IgE, eosinophils, and neutrophils, a lower activity of eosinophilic peroxidase in the airways, and an increase in the production of IgG1 and IgG2a antibodies. IgG antibodies blocked IgE binding to parental allergens in sera from atopic patients. Splenocytes, BALF, and lung from mice treated with rDer p 2231 secreted higher levels of Th1 and regulatory cytokines, as well as reduced levels of Th2 cytokines. Histopathological investigation of the lower airways demonstrated reductions in the thickness of the bronchiolar smooth muscle layer, in the subepithelial fibrosis, and in the goblet cells hyperplasia. CONCLUSIONS Our preclinical studies suggest that rDer p 2231 is a promising candidate for the treatment of D. pteronyssinus allergy, as the hypoallergen has demonstrated the ability to reduce IgE production, induce specific blocking antibodies, restore and balance Th1/Th2 immune responses, and significantly reduce airway remodeling factors. However, additional clinical studies are needed to more accurately assess the efficacy and safety of rDer p 2231 as a vaccine against D. pteronyssinus-induced allergy.
Collapse
Affiliation(s)
| | - Eduardo Santos da Silva
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Raphael Chagas Silva
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Elisânia Fontes Silveira
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Leonardo Freire Santiago
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | | | - Vítor Dos Santos Alves
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Deise Souza Vilas Bôas
- Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil; Laboratory of Histotechnology, Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Luiz Antônio Rodrigues de Freitas
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FioCruz), Salvador, BA, Brazil; Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador, BA, Brazil.
| | - Fatima Ferreira
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria.
| | - Alain Jacquet
- Department of Biochemistry, Chulalongkorn University, Bangkok, Thailand.
| | - Luis Gustavo Carvalho Pacheco
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil.
| | - Neuza Maria Alcantara-Neves
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| | - Carina Silva Pinheiro
- Laboratory of Allergology and Acarology (LAA), Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil; Post-Graduate Program in Immunology (PPGIm) of the Federal University of Bahia, Salvador, BA, Brazil.
| |
Collapse
|
6
|
Qu G, Liu Y, Ouyang J, Xiao L, Liu X. Effects of MAL gene knockout on lung tissue morphology and on E-cad and α-SMA expression in asthma mouse models. J Asthma 2024; 61:1432-1441. [PMID: 38875021 DOI: 10.1080/02770903.2024.2355982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/12/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVES To investigate the effects of myelin- and lymphocyte-associated protein (MAL) gene knockout on the morphological structure of lung tissue and the expression of E-cadherin (E-cad) and alpha-smooth muscle actin (α-SMA) in an asthmatic mouse model. METHODS Twenty-four specific pathogen-free (SPF) C57BL/6J mice were divided into four groups: the wild-type normal (WT/SAL), wild-type asthmatic (WT/OVA), gene knockout normal (MAL-/-/SAL), and gene knockout asthmatic (MAL-/-/OVA) groups. The establishment of the asthma mouse models was confirmed by evaluating behavioral symptoms and histopathological H&E and Masson staining. Western blotting and RT-qPCR were used to measure E-cad and α-SMA expression levels in lung tissues. RESULTS H&E staining of mouse lung tissues from WT/OVA, MAL-/-/SAL, and MAL-/-/OVA groups revealed a thickened bronchial wall, irregular lumen edge, locally fallen mucosal epithelium, and inflammatory cell infiltration compared with those of the WT/SAL group. In the WT and MAL-/- groups, the proportion of Masson-stained tissues in the OVA group was greater than that in the SAL group (p < 0.05). Compared with those in the WT/SAL group, the expression levels of α-SMA mRNA and protein were increased, while those of E-cad were decreased in the WT/OVA group (p < 0.01). Similarly, compared with those in the MAL-/-/SAL group, the expression levels of E-cad mRNA and protein were increased, while those of α-SMA were decreased in the MAL-/-/OVA group (p < 0.01). All these differences were statistically significant (p < 0.01). CONCLUSIONS The MAL gene contributes to EMT inhibition and the stability of the airway barrier under normal physiological conditions by regulating E-cad and α-SMA expression.
Collapse
Affiliation(s)
- Gaojie Qu
- The First Affiliated Hospital, Department of Traditional Chinese Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Third Affiliated Hospital, Department of Respiratory and Critical Care Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yulu Liu
- Department of Respiratory and Critical Care Medicine, Xiangdong Hospital Affiliated with Hunan Normal University, Zhuzhou, Hunan, China
| | - Jieyuan Ouyang
- The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linlin Xiao
- The First Affiliated Hospital, Department of Respiratory and Critical Care Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin Liu
- The First Affiliated Hospital, Department of Traditional Chinese Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
8
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Zhang F, Weng X, Zhu J, Tang Q, Lei M, Zhou W. Identification and validation of three potential biomarkers and immune microenvironment for in severe asthma in microarray and single-cell datasets. J Asthma 2024; 61:1252-1264. [PMID: 38647226 DOI: 10.1080/02770903.2024.2335562] [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: 03/10/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
Objective: The aim of this study was to identify genetic biomarkers and cellular communications associated with severe asthma in microarray data sets and single cell data sets. The potential gene expression levels were verified in a mouse model of asthma.Methods: We identified differentially expressed genes from the microarray datasets (GSE130499 and GSE63142) of severe asthma, and then constructed models to screen the most relevant biomarkers to severe asthma by machine learning algorithms (LASSO and SVM-RFE), with further validation of the results by GSE43696. Single-cell datasets (GSE193816 and GSE227744) were identified for potential biomarker-specific expression and intercellular communication. Finally, The expression levels of potential biomarkers were verified with a mouse model of asthma.Results: The 73 genes were differentially expressed between severe asthma and normal control. LASSO and SVM-RFE recognized three genes BCL3, DDIT4 and S100A14 as biomarkers of severe asthma and had good diagnostic effect. Among them, BCL3 transcript level was down-regulated in severe asthma, while S100A14 and DDIT4 transcript levels were up-regulated. The transcript levels of the three genes were confirmed in the mouse model. Infiltration of neutrophils and mast cells were found to be increased in severe asthma and may be associated with bronchial epithelial cells through BMP and NRG signalingConclusions: We identified three differentially expressed genes (BCL3, DDIT4 and S100A14) of diagnostic significance that may be involved in the development of severe asthma and these gene expressions could be serviced as biomarker of severe asthma and investigating the function roles could bring new insights into the underlying mechanisms.
Collapse
Affiliation(s)
- Fuying Zhang
- Zhangjiajie Hospital Affiliated to Hunan Normal University, Zhangjiajie, Hunan, China
| | - Xiang Weng
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiabao Zhu
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qin Tang
- Zhangjiajie Hospital Affiliated to Hunan Normal University, Zhangjiajie, Hunan, China
| | - Mingsheng Lei
- Zhangjiajie Hospital Affiliated to Hunan Normal University, Zhangjiajie, Hunan, China
- Zhangjiajie College, Zhangjiajie, Hunan, China
| | - Weimin Zhou
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Gohal G, Moni SS, Bakkari MA, Elmobark ME. A Review on Asthma and Allergy: Current Understanding on Molecular Perspectives. J Clin Med 2024; 13:5775. [PMID: 39407835 PMCID: PMC11476424 DOI: 10.3390/jcm13195775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Asthma, a complex disease characterized by persistent airway inflammation, remains an urgent global health concern. We explored the critical role of allergic biomarkers and dysregulated immune system in asthma through an extensive literature review in databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. This review summarizes the growing data on the pivotal role of allergic biomarkers and dysregulated immune system in the development and evolution of asthma. Recent studies have uncovered several biomarkers that elucidate intrinsic allergic mechanisms in individuals with asthma. This article highlights these biomarkers' potential in predicting asthma onset, assessing its intensity, guiding therapeutic interventions, and tracking disease progression. We also explore the innovative therapeutic prospects arising from the convergence of allergy and dysregulated immune system in asthma and emphasize the potential for precision medicine approaches. Understanding allergic biomarkers intertwined with a dysregulated immune system heralds a new era in asthma treatment and points to improved and individualized treatment modalities.
Collapse
Affiliation(s)
- Gassem Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Sivakumar S. Moni
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
11
|
Ebrahimi Samani S, Tatsukawa H, Hitomi K, Kaartinen MT. Transglutaminase 1: Emerging Functions beyond Skin. Int J Mol Sci 2024; 25:10306. [PMID: 39408635 PMCID: PMC11476513 DOI: 10.3390/ijms251910306] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Transglutaminase enzymes catalyze Ca2+- and thiol-dependent posttranslational modifications of glutamine-residues that include esterification, hydrolysis and transamidation, which results in covalent protein-protein crosslinking. Among the eight transglutaminase family members in mammals, transglutaminase 1 (TG1) plays a crucial role in skin barrier formation via crosslinking and insolubilizing proteins in keratinocytes. Despite this established function in skin, novel functions have begun merging in normal tissue homeostasis as well as in pathologies. This review summarizes our current understanding of the structure, activation, expression and activity patterns of TG1 and discusses its putative novel role in other tissues, such as in vascular integrity, and in diseases, such as cancer and fibrosis.
Collapse
Affiliation(s)
- Sahar Ebrahimi Samani
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada;
| | - Hideki Tatsukawa
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan; (H.T.); (K.H.)
| | - Kiyotaka Hitomi
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan; (H.T.); (K.H.)
| | - Mari T. Kaartinen
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada;
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
12
|
Panganiban RA, Nadeau KC, Lu Q. Pyroptosis, gasdermins and allergic diseases. Allergy 2024; 79:2380-2395. [PMID: 39003568 PMCID: PMC11368650 DOI: 10.1111/all.16236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
Pyroptosis is an inflammatory form of programmed cell death that is distinct from necrosis and apoptosis. Pyroptosis is primarily mediated by the gasdermin family of proteins (GSDMA-E and PVJK), which, when activated by proteolytic cleavage, form pores in the plasma membrane, leading to cell death. While much of the past research on pyroptosis has focused on its role in cancer, metabolic disorders, and infectious diseases, recent experimental and observational studies have begun to implicate pyroptosis in allergic diseases. These studies suggest that gasdermin-mediated pyroptosis contributes to the development of allergic conditions and could offer novel targets for therapy. Here, we review our current understanding of pyroptosis with an emphasis on the role of gasdermins as executioners of pyroptosis and potential mediators to allergic disease. We highlight new discoveries that establish a mechanistic link between the biochemical actions of gasdermins and the onset of allergic diseases. Additionally, we discuss how pyroptosis and gasdermins might contribute to the dysfunction of epithelial barrier, a key factor believed to initiate the progression of various allergic diseases.
Collapse
Affiliation(s)
- Ronald Allan Panganiban
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
14
|
Sasidharan A, Grosche A, Xu X, Kinane TB, Angoli D, Vidyasagar S. Select amino acids recover cytokine-altered ENaC function in human bronchial epithelial cells. PLoS One 2024; 19:e0307809. [PMID: 39052685 PMCID: PMC11271875 DOI: 10.1371/journal.pone.0307809] [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: 10/01/2023] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
The airway epithelium plays a pivotal role in regulating mucosal immunity and inflammation. Epithelial barrier function, homeostasis of luminal fluid, and mucociliary clearance are major components of mucosal defense mechanisms. The epithelial sodium channel (ENaC) is one of the key players in controlling airway fluid volume and composition, and characteristic cytokines cause ENaC and barrier dysfunctions following pulmonary infections or allergic reactions. Given the limited understanding of the requisite duration and magnitude of cytokines to affect ENaC and barrier function, available treatment options for restoring normal ENaC activity are limited. Previous studies have demonstrated that distinct amino acids can modulate epithelial ion channel activities and barrier function in intestines and airways. Here, we have investigated the time- and concentration-dependent effect of representative cytokines for Th1- (IFN-γ and TNF-α), Th2- (IL-4 and IL-13), and Treg-mediated (TGF-β1) immune responses on ENaC activity and barrier function in human bronchial epithelial cells. When cells were exposed to Th1 and Treg cytokines, ENaC activity decreased gradually while barrier function remained largely unaffected. In contrast, Th2 cytokines had an immediate and profound inhibitory effect on ENaC activity that was subsequently followed by epithelial barrier disruption. These functional changes were associated with decreased membrane protein expression of α-, β-, and γ-ENaC, and decreased mRNA levels of β- and γ-ENaC. A proprietary blend of amino acids was developed based on their ability to prevent Th2 cytokine-induced ENaC dysfunction. Exposure to the select amino acids reversed the inhibitory effect of IL-13 on ENaC activity by increasing mRNA levels of β- and γ-ENaC, and protein expression of γ-ENaC. This study indicates the beneficial effect of select amino acids on ENaC activity in an in vitro setting of Th2-mediated inflammation suggesting these amino acids as a novel therapeutic approach for correcting this condition.
Collapse
Affiliation(s)
- Anusree Sasidharan
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Astrid Grosche
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Xiaodong Xu
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - T. Bernard Kinane
- Pediatric Pulmonary Division, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Damiano Angoli
- Pediatric Pulmonary Division, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
15
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
16
|
Steffan BN, Townsend EA, Denlinger LC, Johansson MW. Eosinophil-Epithelial Cell Interactions in Asthma. Int Arch Allergy Immunol 2024; 185:1033-1047. [PMID: 38885626 PMCID: PMC11534548 DOI: 10.1159/000539309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Eosinophils have numerous roles in type 2 inflammation depending on their activation states in the blood and airway or after encounter with inflammatory mediators. Airway epithelial cells have a sentinel role in the lung and, by instructing eosinophils, likely have a foundational role in asthma pathogenesis. SUMMARY In this review, we discuss various topics related to eosinophil-epithelial cell interactions in asthma, including the influence of eosinophils and eosinophil products, e.g., granule proteins, on epithelial cell function, expression, secretion, and plasticity; the effects of epithelial released factors, including oxylipins, cytokines, and other mediators on eosinophils, e.g., on their activation, expression, and survival; possible mechanisms of eosinophil-epithelial cell adhesion; and the role of intra-epithelial eosinophils in asthma. KEY MESSAGES We suggest that eosinophils and their products can have both injurious and beneficial effects on airway epithelial cells in asthma and that there are bidirectional interactions and signaling between eosinophils and airway epithelial cells in asthma.
Collapse
Affiliation(s)
- Breanne N. Steffan
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Elizabeth A. Townsend
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
- Department of Anesthesiology, University of Wisconsin, Madison, Wisconsin, USA
| | - Loren C. Denlinger
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Mats W. Johansson
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
17
|
Xu L, Wang Y, Chen Q, Zhu X, Hong J. Propofol modulates Nrf2/NLRP3 signaling to ameliorate cigarette smoke-induced damage in human bronchial epithelial cells. Tissue Cell 2024; 88:102341. [PMID: 38479189 DOI: 10.1016/j.tice.2024.102341] [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: 12/26/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 06/17/2024]
Abstract
Cigarette smoke extract (CSE) is known as a significant contributor to chronic obstructive pulmonary disease (COPD). Propofol, an anesthetic agent, has been studied for its potential protective effects against lung damage. This study aimed to elucidate the protective mechanisms of propofol against CSE-induced damage in human bronchial epithelial 16HBE cells. In CSE-induced 16HBE cells treated by propofol with or without transfection of nuclear factor erythroid 2-related factor 2 (Nrf2) interference plasmids, CCK-8 assay and lactate dehydrogenase (LDH) assay evaluated cytotoxicity. TUNEL assay and Western blot appraised cell apoptosis. ELISA and relevant assay kits severally measured inflammatory and oxidative stress levels. DCFH-DA fluorescent probe detected intracellular reactive oxygen species (ROS) activity. Immunofluorescence staining and Western blot estimated pyroptosis. Also, Western blot analyzed the expression of Nrf2/NLR family pyrin domain containing 3 (NLRP3) signaling-related proteins. Propofol was found to enhance the viability, reduce LDH release, and alleviate the apoptosis, inflammatory response, oxidative stress and pyroptosis in CSE-induced 16HBE cells in a concentration-dependent manner. Meanwhile, propofol decreased NLRP3 expression while raised Nrf2 expression. Further, after Nrf2 was silenced, the impacts of propofol on Nrf2/NLRP3 signaling, LDH release, apoptosis, inflammatory response, oxidative stress and pyroptosis in CSE-exposed 16HBE cells were eliminated. Conclusively, propofol may exert protective effects against CSE-induced damage in 16HBE cells, partly through the modulation of the Nrf2/NLRP3 signaling pathway, suggesting a potential therapeutic role for propofol in CSE-induced bronchial epithelial cell damage.
Collapse
Affiliation(s)
- Linmei Xu
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, Fujian 361000, China; The School of Clinical Medicine, Fujian Medical University, Fujian 361000, China
| | - Yanbin Wang
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, Fujian 361000, China
| | - Qumin Chen
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, Fujian 361000, China
| | - Xiaolei Zhu
- Department of Thoracic Surgery, the First Affiliated Hospital of Xiamen University, Fujian 361000, China
| | - Jiageng Hong
- Department of Anesthesiology, the First Affiliated Hospital of Xiamen University, Fujian 361000, China; The School of Clinical Medicine, Fujian Medical University, Fujian 361000, China.
| |
Collapse
|
18
|
Canè L, Poto R, Palestra F, Pirozzi M, Parashuraman S, Iacobucci I, Ferrara AL, La Rocca A, Mercadante E, Pucci P, Marone G, Monti M, Loffredo S, Varricchi G. TSLP is localized in and released from human lung macrophages activated by T2-high and T2-low stimuli: relevance in asthma and COPD. Eur J Intern Med 2024; 124:89-98. [PMID: 38402021 DOI: 10.1016/j.ejim.2024.02.020] [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: 01/08/2024] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Macrophages are the predominant immune cells in the human lung and play a central role in airway inflammation, including asthma and chronic obstructive pulmonary disease (COPD). Thymic stromal lymphopoietin (TSLP), a pleiotropic cytokine mainly expressed by bronchial epithelial cells, plays a key role in asthma and COPD pathobiology. TSLP exists in two variants: the long form (lfTSLP) and a shorter TSLP isoform (sfTSLP). We aimed to localize TSLP in human lung macrophages (HLMs) and investigate the mechanisms of its release from these cells. We also evaluated the effects of the two variants of TSLP on the release of angiogenic factor from HLMs. METHODS We employed immunofluorescence and Western blot to localize intracellular TSLP in HLMs purified from human lung parenchyma. HLMs were activated by T2-high (IL-4, IL-13) and T2-low (lipopolysaccharide: LPS) immunological stimuli. RESULTS TSLP was detected in HLMs and subcellularly localized in the cytoplasm. IL-4 and LPS induced TSLP release from HLMs. Preincubation of macrophages with brefeldin A, known to disrupt the Golgi apparatus, inhibited TSLP release induced by LPS and IL-4. lfTSLP concentration-dependently induced the release of vascular endothelial growth factor-A (VEGF-A), the most potent angiogenic factor, from HLMs. sfTSLP neither activated nor interfered with the activating property of lfTSLP on macrophages. CONCLUSIONS Our results highlight a novel immunologic circuit between HLMs and TSLP. Given the central role of macrophages in airway inflammation, this autocrine loop holds potential translational relevance in understanding innovative aspects of the pathobiology of asthma and chronic inflammatory lung disorders.
Collapse
Affiliation(s)
- Luisa Canè
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Francesco Palestra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Marinella Pirozzi
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Seetharaman Parashuraman
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Ilaria Iacobucci
- CEINGE Advanced Biotechnologies, Naples, Italy; Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Antonello La Rocca
- Thoracic Surgery Unit - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Edoardo Mercadante
- Thoracic Surgery Unit - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Piero Pucci
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Maria Monti
- CEINGE Advanced Biotechnologies, Naples, Italy; Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| |
Collapse
|
19
|
Bradding P, Porsbjerg C, Côté A, Dahlén SE, Hallstrand TS, Brightling CE. Airway hyperresponsiveness in asthma: The role of the epithelium. J Allergy Clin Immunol 2024; 153:1181-1193. [PMID: 38395082 DOI: 10.1016/j.jaci.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Airway hyperresponsiveness (AHR) is a key clinical feature of asthma. The presence of AHR in people with asthma provides the substrate for bronchoconstriction in response to numerous diverse stimuli, contributing to airflow limitation and symptoms including breathlessness, wheeze, and chest tightness. Dysfunctional airway smooth muscle significantly contributes to AHR and is displayed as increased sensitivity to direct pharmacologic bronchoconstrictor stimuli, such as inhaled histamine and methacholine (direct AHR), or to endogenous mediators released by activated airway cells such as mast cells (indirect AHR). Research in in vivo human models has shown that the disrupted airway epithelium plays an important role in driving inflammation that mediates indirect AHR in asthma through the release of cytokines such as thymic stromal lymphopoietin and IL-33. These cytokines upregulate type 2 cytokines promoting airway eosinophilia and induce the release of bronchoconstrictor mediators from mast cells such as histamine, prostaglandin D2, and cysteinyl leukotrienes. While bronchoconstriction is largely due to airway smooth muscle contraction, airway structural changes known as remodeling, likely mediated in part by epithelial-derived mediators, also lead to airflow obstruction and may enhance AHR. In this review, we outline the current knowledge of the role of the airway epithelium in AHR in asthma and its implications on the wider disease. Increased understanding of airway epithelial biology may contribute to better treatment options, particularly in precision medicine.
Collapse
Affiliation(s)
- Peter Bradding
- Department of Respiratory Sciences, Leicester Respiratory National Institute for Health and Care Research Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Andréanne Côté
- Quebec Heart and Lung Institute, Université Laval, Laval, Quebec, Canada; Department of Medicine, Université Laval, Laval, Quebec, Canada
| | - Sven-Erik Dahlén
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Teal S Hallstrand
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Wash; Center for Lung Biology, University of Washington, Seattle, Wash.
| | - Christopher E Brightling
- Department of Respiratory Sciences, Leicester Respiratory National Institute for Health and Care Research Biomedical Research Centre, Glenfield Hospital, University of Leicester, Leicester, United Kingdom.
| |
Collapse
|
20
|
Bellomo A, Herbert J, Kudlak MJ, Laskin JD, Gow AJ, Laskin DL. Identification of early events in nitrogen mustard pulmonary toxicity that are independent of infiltrating inflammatory cells using precision cut lung slices. Toxicol Appl Pharmacol 2024; 486:116941. [PMID: 38677601 DOI: 10.1016/j.taap.2024.116941] [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: 01/20/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Nitrogen mustard (NM; mechlorethamine) is a cytotoxic vesicant known to cause acute lung injury which can progress to chronic disease. Due to the complex nature of NM injury, it has been difficult to analyze early responses of resident lung cells that initiate inflammation and disease progression. To investigate this, we developed a model of acute NM toxicity using murine precision cut lung slices (PCLS), which contain all resident lung cell populations. PCLS were exposed to NM (1-100 μM) for 0.5-3 h and analyzed 1 and 3 d later. NM caused a dose-dependent increase in cytotoxicity and a reduction in metabolic activity, as measured by LDH release and WST-1 activity, respectively. Optimal responses were observed with 50 μM NM after 1 h incubation and these conditions were used in further experiments. Analysis of PCLS bioenergetics using an Agilent Seahorse showed that NM impaired both glycolytic activity and mitochondrial respiration. This was associated with injury to the bronchial epithelium and a reduction in methacholine-induced airway contraction. NM was also found to cause DNA damage in bronchial epithelial cells in PCLS, as measured by expression of γ-H2AX, and to induce oxidative stress, which was evident by a reduction in glutathione levels and upregulation of the antioxidant enzyme catalase. Cleaved caspase-3 was also upregulated in airway smooth muscle cells indicating apoptotic cell death. Characterizing early events in NM toxicity is key in identifying therapeutic targets for the development of efficacious countermeasures.
Collapse
Affiliation(s)
- Alyssa Bellomo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Julia Herbert
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Melissa J Kudlak
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
21
|
Sompunga P, Rodprasert W, Srisuwatanasagul S, Techangamsuwan S, Jirajessada S, Hanchaina R, Kangsamaksin T, Yodmuang S, Sawangmake C. Preparation of Decellularized Tissue as Dual Cell Carrier Systems: A Step Towards Facilitating Re-epithelization and Cell Encapsulation for Tracheal Reconstruction. Ann Biomed Eng 2024; 52:1222-1239. [PMID: 38353908 DOI: 10.1007/s10439-024-03448-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/09/2024] [Indexed: 04/06/2024]
Abstract
Surgical treatment of tracheal diseases, trauma, and congenital stenosis has shown success through tracheal reconstruction coupled with palliative care. However, challenges in surgical-based tracheal repairs have prompted the exploration of alternative approaches for tracheal replacement. Tissue-based treatments, involving the cultivation of patient cells on a network of extracellular matrix (ECM) from donor tissue, hold promise for restoring tracheal structure and function without eliciting an immune reaction. In this study, we utilized decellularized canine tracheas as tissue models to develop two types of cell carriers: a decellularized scaffold and a hydrogel. Our hypothesis posits that both carriers, containing essential biochemical niches provided by ECM components, facilitate cell attachment without inducing cytotoxicity. Canine tracheas underwent vacuum-assisted decellularization (VAD), and the ECM-rich hydrogel was prepared through peptic digestion of the decellularized trachea. The decellularized canine trachea exhibited a significant reduction in DNA content and major histocompatibility complex class II, while preserving crucial ECM components such as collagen, glycosaminoglycan, laminin, and fibronectin. Scanning electron microscope and fluorescent microscope images revealed a fibrous ECM network on the luminal side of the cell-free trachea, supporting epithelial cell attachment. Moreover, the ECM-rich hydrogel exhibited excellent viability for human mesenchymal stem cells encapsulated for 3 days, indicating the potential of cell-laden hydrogel in promoting the development of cartilage rings of the trachea. This study underscores the versatility of the trachea in producing two distinct cell carriers-decellularized scaffold and hydrogel-both containing the native biochemical niche essential for tracheal tissue engineering applications.
Collapse
Affiliation(s)
- Pensuda Sompunga
- Medical Sciences Program, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Watchareewan Rodprasert
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sayamon Srisuwatanasagul
- Department of Anatomy, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Somporn Techangamsuwan
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand
| | - Sirinee Jirajessada
- Biology Program, Faculty of Science, Buriram Rajabhat University, Muang, Buriram, 31000, Thailand
| | - Rattanavinan Hanchaina
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Thaned Kangsamaksin
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Supansa Yodmuang
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Ananda Mahidol Building, 1873 Rama 4 Rd, Pathumwan, Bangkok, 10330, Thailand.
- Center of Excellence in Biomaterial Engineering for Medical and Health, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand.
- Clinical Excellence Center for Advanced Therapy Medicinal Products, King Chulalongkorn Memorial Hospital, Pathumwan, Bangkok, 10330, Thailand.
- Avatar Biotech for Oral Health & Healthy Longevity Research Unit, Chulalongkorn University, Pathumwan, Bangkok, 10330, Thailand.
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
22
|
Su D, Luo X, Chen J, Lu N, Zhao J, Wan Y, Gao Y, Liu Q, Luo Z. Construction of a three-dimensional inflammation model of human bronchial epithelial cells BEAS-2B by using the self-assembling D-form peptide Sciobio-Ⅲ. Biochem Biophys Res Commun 2024; 704:149701. [PMID: 38408415 DOI: 10.1016/j.bbrc.2024.149701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Human bronchial epithelial cells in the airway system, as the primary barrier between humans and the surrounding environment, assume a crucial function in orchestrating the processes of airway inflammation. Target to develop a new three-dimensional (3D) inflammatory model to airway system, and here we report a strategy by using self-assembling D-form peptide to cover the process. By testing physicochemical properties and biocompatibility of Sciobio-Ⅲ, we confirmed that it can rapidly self-assembles under the trigger of ions to form a 3D nanonetwork-like scaffold, which supports 3D cell culture including the cell strains like BEAS-2B cells. Subsequently, inflammation model was established by lipopolysaccharide (LPS), the expression of some markers of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-8 (IL-8), the levels of relevant inflammatory factors were measured by RT-qPCR and the secretion profile of inflammatory cytokines by ELISA, are obtained the quite difference effects in 2D and 3D microenvironment, which suggested Sciobio-Ⅲ hydrogel is an ideal scaffold that create the microenvironment for 3D cell culture. Here we are success to establish a 3D inflammation model for airway system. This innovative model allows for rapid and accurate evaluation of drug metabolism and toxicological side effects, hope to use in drug screening for airway inflammatory diseases and beyond.
Collapse
Affiliation(s)
- Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China; Department of Materials Science and Engineering, University of California, Irvine, CA, 92697, USA
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Jiawei Zhao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Yuan Wan
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, IA, 52242, USA
| | - Yu Gao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Qichen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
23
|
Lv X, Zheng L, Zhang T, Wang W, Chen Y, Li J, Cai Z, Guo X, Song L. CLCA1 exacerbates lung inflammation via p38 MAPK pathway in acute respiratory distress syndrome. Exp Lung Res 2024; 50:85-95. [PMID: 38597420 DOI: 10.1080/01902148.2024.2334262] [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: 08/14/2023] [Accepted: 03/19/2024] [Indexed: 04/11/2024]
Abstract
Recent research has revealed that airway epithelial calcium-activated chloride channel-1 (CLCA1) is implicated in the inflammation of multiple human respiratory diseases, but the specific role in acute respiratory distress syndrome (ARDS) remains unknown. To investigate the role of CLCA1 in ARDS, 80 participants, including 26 ARDS patients, 26 patients with community-acquired pneumonia (CAP) and 28 control subjects, were enrolled in this study. As the result shows, the level of CLCA1 was significantly increased in ARDS patients and positively correlated with neutrophil infiltration and the poor prognosis of ARDS. Then, the level of CLCA1 also elevated in the LPS-induced ARDS mouse model, and the administration of CLCA1 significantly regulated the phenotypes of ARDS in mice, such as lung injury score, BALF protein concentration, neutrophils infiltration and the secretions of inflammatory factors. Furthermore, administration of CLCA1 substantially altered the phosphorylation of p38 in the ARDS mouse model, whereas repressing the expression of CLCA1 or inhibiting the activation of p38 both alleviated the inflammatory response of ARDS. In summary, CLCA1 was notably correlated with ARDS and exacerbated the ARDS phenotypes through the p38 MAPK pathway.
Collapse
Affiliation(s)
- Xing Lv
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Long Zheng
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | - Weijia Wang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuanyuan Chen
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Li
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhigui Cai
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xingxing Guo
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liqiang Song
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
24
|
Canè L, Poto R, Palestra F, Iacobucci I, Pirozzi M, Parashuraman S, Ferrara AL, Illiano A, La Rocca A, Mercadante E, Pucci P, Marone G, Spadaro G, Loffredo S, Monti M, Varricchi G. Thymic Stromal Lymphopoietin (TSLP) Is Cleaved by Human Mast Cell Tryptase and Chymase. Int J Mol Sci 2024; 25:4049. [PMID: 38612858 PMCID: PMC11012384 DOI: 10.3390/ijms25074049] [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: 02/22/2024] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Thymic stromal lymphopoietin (TSLP), mainly expressed by epithelial cells, plays a central role in asthma. In humans, TSLP exists in two variants: the long form TSLP (lfTSLP) and a shorter TSLP isoform (sfTSLP). Macrophages (HLMs) and mast cells (HLMCs) are in close proximity in the human lung and play key roles in asthma. We evaluated the early proteolytic effects of tryptase and chymase released by HLMCs on TSLP by mass spectrometry. We also investigated whether TSLP and its fragments generated by these enzymes induce angiogenic factor release from HLMs. Mass spectrometry (MS) allowed the identification of TSLP cleavage sites caused by tryptase and chymase. Recombinant human TSLP treated with recombinant tryptase showed the production of 1-97 and 98-132 fragments. Recombinant chymase treatment of TSLP generated two peptides, 1-36 and 37-132. lfTSLP induced the release of VEGF-A, the most potent angiogenic factor, from HLMs. By contrast, the four TSLP fragments generated by tryptase and chymase failed to activate HLMs. Long-term TSLP incubation with furin generated two peptides devoid of activating property on HLMs. These results unveil an intricate interplay between mast cell-derived proteases and TSLP. These findings have potential relevance in understanding novel aspects of asthma pathobiology.
Collapse
Affiliation(s)
- Luisa Canè
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- CEINGE Advanced Biotechnologies F. Salvatore, 80131 Naples, Italy; (I.I.); (P.P.)
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Francesco Palestra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Ilaria Iacobucci
- CEINGE Advanced Biotechnologies F. Salvatore, 80131 Naples, Italy; (I.I.); (P.P.)
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Marinella Pirozzi
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; (M.P.); (S.P.)
| | - Seetharaman Parashuraman
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; (M.P.); (S.P.)
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Amalia Illiano
- Thoracic Surgery Unit—Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy; (A.I.); (A.L.R.); (E.M.)
| | - Antonello La Rocca
- Thoracic Surgery Unit—Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy; (A.I.); (A.L.R.); (E.M.)
| | - Edoardo Mercadante
- Thoracic Surgery Unit—Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy; (A.I.); (A.L.R.); (E.M.)
| | - Piero Pucci
- CEINGE Advanced Biotechnologies F. Salvatore, 80131 Naples, Italy; (I.I.); (P.P.)
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; (M.P.); (S.P.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; (M.P.); (S.P.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Maria Monti
- CEINGE Advanced Biotechnologies F. Salvatore, 80131 Naples, Italy; (I.I.); (P.P.)
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (F.P.); (A.L.F.); (G.M.); (G.S.); (S.L.)
- World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; (M.P.); (S.P.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
25
|
Andreasson LM, Dyhre-Petersen N, Hvidtfeldt M, Jørgensen GØ, Von Bülow A, Klein DK, Uller L, Erjefält J, Porsbjerg C, Sverrild A. Airway hyperresponsiveness correlates with airway TSLP in asthma independent of eosinophilic inflammation. J Allergy Clin Immunol 2024; 153:988-997.e11. [PMID: 38081546 DOI: 10.1016/j.jaci.2023.11.915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Thymic stromal lymphopoietin (TSLP) is released from the airway epithelium in response to various environmental triggers, inducing a type-2 inflammatory response, and is associated with airway inflammation, airway hyperresponsiveness (AHR), and exacerbations. TSLP may also induce AHR via a direct effect on airway smooth muscle and mast cells, independently of type-2 inflammation, although association between airway TSLP and AHR across asthma phenotypes has been described sparsely. OBJECTIVES This study sought to investigate the association between AHR and levels of TSLP in serum, sputum, and bronchoalveolar lavage in patients with asthma with and without type-2 inflammation. METHODS A novel ultrasensitive assay was used to measure levels of TSLP in patients with asthma (serum, n = 182; sputum, n = 81; bronchoalveolar lavage, n = 85) and healthy controls (serum, n = 47). The distribution and association among airway and systemic TSLP, measures of AHR, type-2 inflammation, and severity of disease were assessed. RESULTS TSLP in sputum was associated with AHR independently of levels of eosinophils and fractional exhaled nitric oxide (ρ = 0.49, P = .005). Serum TSLP was higher in both eosinophil-high and eosinophil-low asthma compared to healthy controls: geometric mean: 1600 fg/mL (95% CI: 1468-1744 fg/mL) and 1294 fg/mL (95% CI: 1167-1435 fg/mL) versus 846 fg/mL (95% CI: 661-1082 fg/mL), but did not correlate with the level of AHR. Increasing age, male sex, and eosinophils in blood were associated with higher levels of TSLP in serum, whereas lung function, inhaled corticosteroid dose, and symptom score were not. CONCLUSIONS The association between TSLP in sputum and AHR to mannitol irrespective of markers of type-2 inflammation further supports a role of TSLP in AHR that is partially independent of eosinophilic inflammation.
Collapse
Affiliation(s)
- Louise Munkholm Andreasson
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Nanna Dyhre-Petersen
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Morten Hvidtfeldt
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Gustav Ørting Jørgensen
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Anna Von Bülow
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Ditte Kjærsgaard Klein
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Lena Uller
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - Jonas Erjefält
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, University Hospital Bispebjerg, Copenhagen, Denmark.
| |
Collapse
|
26
|
Russell RJ, Boulet LP, Brightling CE, Pavord ID, Porsbjerg C, Dorscheid D, Sverrild A. The airway epithelium: an orchestrator of inflammation, a key structural barrier and a therapeutic target in severe asthma. Eur Respir J 2024; 63:2301397. [PMID: 38453256 PMCID: PMC10991852 DOI: 10.1183/13993003.01397-2023] [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: 08/18/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Asthma is a disease of heterogeneous pathology, typically characterised by excessive inflammatory and bronchoconstrictor responses to the environment. The clinical expression of the disease is a consequence of the interaction between environmental factors and host factors over time, including genetic susceptibility, immune dysregulation and airway remodelling. As a critical interface between the host and the environment, the airway epithelium plays an important role in maintaining homeostasis in the face of environmental challenges. Disruption of epithelial integrity is a key factor contributing to multiple processes underlying asthma pathology. In this review, we first discuss the unmet need in asthma management and provide an overview of the structure and function of the airway epithelium. We then focus on key pathophysiological changes that occur in the airway epithelium, including epithelial barrier disruption, immune hyperreactivity, remodelling, mucus hypersecretion and mucus plugging, highlighting how these processes manifest clinically and how they might be targeted by current and novel therapeutics.
Collapse
Affiliation(s)
- Richard J Russell
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | | | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ian D Pavord
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| | - Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
27
|
Shigemasa R, Masuko H, Oshima H, Hyodo K, Kitazawa H, Kanazawa J, Yatagai Y, Iijima H, Naito T, Saito T, Konno S, Hirota T, Tamari M, Sakamoto T, Hizawa N. The primary ciliary dyskinesia-related genetic risk score is associated with susceptibility to adult-onset asthma. PLoS One 2024; 19:e0300000. [PMID: 38457400 PMCID: PMC10923447 DOI: 10.1371/journal.pone.0300000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/19/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Disturbance of mucociliary clearance is an important factor in the pathogenesis of asthma. We hypothesized that common variants in genes responsible for ciliary function may contribute to the development of asthma with certain phenotypes. METHODS Three independent adult Japanese populations (including a total of 1,158 patients with asthma and 2,203 non-asthmatic healthy participants) were studied. First, based on the ClinVar database (https://www.ncbi.nlm.nih.gov/clinvar/), we selected 12 common single-nucleotide polymorphisms (SNPs) with molecular consequences (missense, nonsense, and 3'-untranslated region mutation) in 5 primary ciliary dyskinesia (PCD)-related genes and calculated a PCD-genetic risk score (GRS) as a cumulative effect of these PCD-related genes. Second, we performed a two-step cluster analysis using 3 variables, including PCD-GRS, forced expiratory volume in 1 second (%predicted FEV1), and age of asthma onset. RESULTS Compared to adult asthma clusters with an average PCD-GRS, clusters with high and low PCD-GRS had similar overall characteristics: adult-onset, female predominance, preserved lung function, and fewer features of type 2 immunity as determined by IgE reactivity and blood eosinophil counts. The allele frequency of rs1530496, a SNP representing an expression quantitative trait locus (eQTL) of DNAH5 in the lung, showed the largest statistically significant difference between the PCD-GRS-High and PCD-GRS-Low asthma clusters (p = 1.4 x 10-15). CONCLUSION Genes associated with PCD, particularly the common SNPs associated with abnormal expression of DNAH5, may have a certain influence on the development of adult-onset asthma, perhaps through impaired mucociliary clearance.
Collapse
Affiliation(s)
- Rie Shigemasa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hironori Masuko
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hisayuki Oshima
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kentaro Hyodo
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruna Kitazawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jun Kanazawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yohei Yatagai
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | | | - Takefumi Saito
- National Hospital Organization Ibaraki Higashi National Hospital, Tokai, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomomitsu Hirota
- Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Mayumi Tamari
- Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Tohru Sakamoto
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
28
|
Sverrild A, Cerps S, Nieto-Fontarigo JJ, Ramu S, Hvidtfeldt M, Menzel M, Kearley J, Griffiths JM, Parnes JR, Porsbjerg C, Uller L. Tezepelumab decreases airway epithelial IL-33 and T2-inflammation in response to viral stimulation in patients with asthma. Allergy 2024; 79:656-666. [PMID: 37846599 DOI: 10.1111/all.15918] [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: 06/19/2023] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Respiratory virus infections are main triggers of asthma exacerbations. Tezepelumab, an anti-TSLP mAb, reduces exacerbations in patients with asthma, but the effect of blocking TSLP on host epithelial resistance and tolerance to virus infection is not known. AIM To examine effects of blocking TSLP in patients with asthma on host resistance (IFNβ, IFNλ, and viral load) and on the airway epithelial inflammatory response to viral challenge. METHODS Bronchoalveolar lavage fluid (BALF, n = 39) and bronchial epithelial cells (BECs) were obtained from patients with uncontrolled asthma before and after 12 weeks of tezepelumab treatment (n = 13) or placebo (n = 13). BECs were cultured in vitro and exposed to the viral infection mimic poly(I:C) or infected by rhinovirus (RV). Alarmins, T2- and pro-inflammatory cytokines, IFNβ IFNλ, and viral load were analyzed by RT-qPCR and multiplex ELISA before and after stimulation. RESULTS IL-33 expression in unstimulated BECs and IL-33 protein levels in BALF were reduced after 12 weeks of tezepelumab. Further, IL-33 gene and protein levels decreased in BECs challenged with poly(I:C) after tezepelumab whereas TSLP gene expression remained unaffected. Poly(I:C)-induced IL-4, IL-13, and IL-17A release from BECs was also reduced with tezepelumab whereas IFNβ and IFNλ expression and viral load were unchanged. CONCLUSION Blocking TSLP with tezepelumab in vivo in asthma reduced the airway epithelial inflammatory response including IL-33 and T2 cytokines to viral challenge without affecting anti-viral host resistance. Our results suggest that blocking TSLP stabilizes the bronchial epithelial immune response to respiratory viruses.
Collapse
Affiliation(s)
- A Sverrild
- Department of Respiratory Medicine, University Hospital Bispebjerg, Copenhagen, Denmark
| | - S Cerps
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - J J Nieto-Fontarigo
- Department of Experimental Medicine, Lund University, Lund, Sweden
- BioLympho Research group, Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research in Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - S Ramu
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - M Hvidtfeldt
- Department of Respiratory Medicine, University Hospital Bispebjerg, Copenhagen, Denmark
| | - M Menzel
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - J Kearley
- Bioscience, Research & Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - J M Griffiths
- Translational Science and Experimental Medicine, Research & Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - J R Parnes
- Translational Medicine, Amgen, Thousand Oaks, California, USA
| | - C Porsbjerg
- Department of Respiratory Medicine, University Hospital Bispebjerg, Copenhagen, Denmark
| | - L Uller
- Department of Experimental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
29
|
Tang H, Guo Y, Gan S, Chen Z, Dong M, Lin L, Chen H, Ji X, Xian M, Shi X, Tao A, Lv Y, Yao L, Chen R, Li S, Li J. GLUT1 mediates the release of HMGB1 from airway epithelial cells in mixed granulocytic asthma. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167040. [PMID: 38281711 DOI: 10.1016/j.bbadis.2024.167040] [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: 08/11/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Asthma is quite heterogenous and can be categorized as eosinophilic, mixed granulocytic (presence of both eosinophils and neutrophils in the airways) and neutrophilic. Clinically, mixed granulocytic asthma (MGA) often tends to be severe and requires large doses of corticosteroids. High mobility group box 1 (HMGB1) is one of the epithelium-derived alarmins that contributes to type 2 inflammation and asthma. This study was aimed to investigate the role of glucose transporter 1 (GLUT1) in modulation of airway epithelial HMGB1 production in MGA. Induced sputum and bronchial biopsy specimens were obtained from healthy subjects and asthma patients. BALB/c mice, the airway epithelial cell line BEAS-2B, or primary human bronchial epithelial cells (HBECs) were immunized with allergens. Intracellular and extracellular HMGB1 were both detected. The role of GLUT1 was assessed by using a pharmacological antagonist BAY876. MGA patients have a significant higher sputum HMGB1 level than the health and subjects with other inflammatory phenotypes. Nuclear-to-cytoplasmic translocation of HMGB1 was also observed in the bronchial epithelia. Allergen exposure markedly induced GLUT1 expression in murine lungs and cultured epithelial cells. Pharmacological antagonism of GLUT1 with BAY876 dramatically decreased airway hyperresponsiveness, neutrophil and eosinophil accumulation, as well as type 2 inflammation in murine models of MGA. Besides, the allergen-induced up-regulation of HMGB1 was also partly recovered by BAY876, accompanied by inhibited secretion into the airway lumen. In vitro, treatment with BAY876 relieved the allergen-induced over-expression and secretion of HMGB1 in airway epithelia. Taken together, our data indicated that GLUT1 mediates bronchial epithelial HMGB1 release in MGA.
Collapse
Affiliation(s)
- Haixiong Tang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yubiao Guo
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sudan Gan
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zemin Chen
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meihua Dong
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liqin Lin
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huifang Chen
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Xiaolong Ji
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mo Xian
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu Shi
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ailin Tao
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yanhua Lv
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Gongdong, China
| | - Lihong Yao
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruchong Chen
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Shiyue Li
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Jing Li
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Lu HF, Zhou YC, Yang LT, Zhou Q, Wang XJ, Qiu SQ, Cheng BH, Zeng XH. Involvement and repair of epithelial barrier dysfunction in allergic diseases. Front Immunol 2024; 15:1348272. [PMID: 38361946 PMCID: PMC10867171 DOI: 10.3389/fimmu.2024.1348272] [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: 12/02/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
The epithelial barrier serves as a critical defense mechanism separating the human body from the external environment, fulfilling both physical and immune functions. This barrier plays a pivotal role in shielding the body from environmental risk factors such as allergens, pathogens, and pollutants. However, since the 19th century, the escalating threats posed by environmental pollution, global warming, heightened usage of industrial chemical products, and alterations in biodiversity have contributed to a noteworthy surge in allergic disease incidences. Notably, allergic diseases frequently exhibit dysfunction in the epithelial barrier. The proposed epithelial barrier hypothesis introduces a novel avenue for the prevention and treatment of allergic diseases. Despite increased attention to the role of barrier dysfunction in allergic disease development, numerous questions persist regarding the mechanisms underlying the disruption of normal barrier function. Consequently, this review aims to provide a comprehensive overview of the epithelial barrier's role in allergic diseases, encompassing influencing factors, assessment techniques, and repair methodologies. By doing so, it seeks to present innovative strategies for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Hui-Fei Lu
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Yi-Chi Zhou
- Department of Gastroenterology, Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Li-Tao Yang
- Clinical Laboratory Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People’s Hospital of Shenzhen, Shenzhen, China
| | - Qian Zhou
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xi-Jia Wang
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Shu-Qi Qiu
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Bao-Hui Cheng
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Xian-Hai Zeng
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| |
Collapse
|
31
|
Esmat K, Jamil B, Kheder RK, Kombe Kombe AJ, Zeng W, Ma H, Jin T. Immunoglobulin A response to SARS-CoV-2 infection and immunity. Heliyon 2024; 10:e24031. [PMID: 38230244 PMCID: PMC10789627 DOI: 10.1016/j.heliyon.2024.e24031] [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: 06/13/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
The novel coronavirus disease (COVID-19) and its infamous "Variants" of the etiological agent termed Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) has proven to be a global health concern. The three antibodies, IgA, IgM, and IgG, perform their dedicated role as main workhorses of the host adaptive immune system in virus neutralization. Immunoglobulin-A (IgA), also known as "Mucosal Immunoglobulin", has been under keen interest throughout the viral infection cycle. Its importance lies because IgA is predominant mucosal antibody and SARS family viruses primarily infect the mucosal surfaces of human respiratory tract. Therefore, IgA can be considered a diagnostic and prognostic marker and an active infection biomarker for SARS CoV-2 infection. Along with molecular analyses, serological tests, including IgA detection tests, are gaining ground in application as an early detectable marker and as a minimally invasive detection strategy. In the current review, it was emphasized the role of IgA response in diagnosis, host defense strategies, treatment, and prevention of SARS-CoV-2 infection. The data analysis was performed through almost 100 published peer-reviewed research reports and comprehended the importance of IgA in antiviral immunity against SARS-CoV-2 and other related respiratory viruses. Taken together, it is concluded that secretory IgA- Abs can serve as a promising detection tool for respiratory viral diagnosis and treatment parallel to IgG-based therapeutics and diagnostics. Vaccine candidates that target and trigger mucosal immune response may also be employed in future dimensions of research against other respiratory viruses.
Collapse
Affiliation(s)
- Khaleqsefat Esmat
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Baban Jamil
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, KRG, Erbil, Iraq
| | - Ramiar Kaml Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Weihong Zeng
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Huan Ma
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| |
Collapse
|
32
|
Hoyer A, Chakraborty S, Lilienthal I, Konradsen JR, Katayama S, Söderhäll C. The functional role of CST1 and CCL26 in asthma development. Immun Inflamm Dis 2024; 12:e1162. [PMID: 38270326 PMCID: PMC10797655 DOI: 10.1002/iid3.1162] [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: 06/28/2023] [Revised: 12/18/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Asthma is the most common chronic disease in children with an increasing prevalence. Its development is caused by genetic and environmental factors and allergic sensitization is a known trigger. Dog allergens affect up to 30% of all children and dog dander-sensitized children show increased expression of cystatin-1 (CST1) and eotaxin-3 (CCL26) in nasal epithelium. The aim of our study was to investigate the functional mechanism of CST1 and CCL26 in the alveolar basal epithelial cell line A549. METHODS A549 cells were transfected with individual overexpression vectors for CST1 and CCL26 and RNA sequencing was performed to examine the transcriptomics. edgeR was used to identify differentially expressed genes (= DEG, |log2 FC | ≥ 2, FDR < 0.01). The protein expression levels of A549 cells overexpressing CST1 and CCL26 were analyzed using the Target 96 inflammation panel from OLINK (antibody-mediated proximity extension-based assay; OLINK Proteomics). Differentially expressed proteins were considered with a |log2 FC| ≥ 1, p < .05. RESULTS The overexpression of CST1 resulted in a total of 27 DEG (1 upregulated and 26 downregulated) and the overexpression of CCL26 in a total of 137 DEG (0 upregulated and 137 downregulated). The gene ontology enrichment analysis showed a significant downregulation of type I and III interferon signaling pathway genes as well as interferon-stimulated genes. At the protein level, overexpression of CST1 induced a significantly increased expression of CCL3, whereas CCL26 overexpression led to increased expression of HGF, and a decrease of CXCL11, CCL20, CCL3 and CXCL10. CONCLUSION Our results indicate that an overexpression of CST1 and CCL26 cause a downregulation of interferon related genes and inflammatory proteins. It might cause a higher disease susceptibility, mainly for allergic asthma, as CCL26 is an agonist for CCR-3-carrying cells, such as eosinophils and Th2 lymphocytes, mostly active in allergic asthma.
Collapse
Affiliation(s)
- Angela Hoyer
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| | - Sandip Chakraborty
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| | - Ingrid Lilienthal
- Childhood Cancer Research Unit, Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
| | - Jon R. Konradsen
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| | - Shintaro Katayama
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
- Stem Cells and Metabolism Research ProgramUniversity of HelsinkiHelsinkiFinland
- Folkhälsan Research CenterHelsinkiFinland
| | - Cilla Söderhäll
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| |
Collapse
|
33
|
Albrecht M, Garn H, Buhl T. Epithelial-immune cell interactions in allergic diseases. Eur J Immunol 2024; 54:e2249982. [PMID: 37804068 DOI: 10.1002/eji.202249982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Epithelial/immune interactions are characterized by the different properties of the various epithelial tissues, the mediators involved, and the varying immune cells that initiate, sustain, or abrogate allergic diseases on the surface. The intestinal mucosa, respiratory mucosa, and regular skin feature structural differences according to their primary function and surroundings. In the context of these specialized functions, the active role of the epithelium in shaping immune responses is increasingly recognizable. Crosstalk between epithelial and immune cells plays an important role in maintaining homeostatic conditions. While cells of the myeloid cell lineage, mainly macrophages, are the dominating immune cell population in the skin and the respiratory tract, lymphocytes comprise most intraepithelial immune cells in the intestine under healthy conditions. Common to all surface epithelia is the fact that innate immune cells represent the first line of immunosurveillance that either directly defeats invading pathogens or initiates and coordinates more effective successive immune responses involving adaptive immune cells and effector cells. Pharmacological approaches for the treatment of allergic and chronic inflammatory diseases involving epithelial barriers target immunological mediators downstream of the epithelium (such as IL-4, IL-5, IL-13, and IgE). The next generation of therapeutics involves upstream events of the inflammatory cascade, such as epithelial-derived alarmins and related mediators.
Collapse
Affiliation(s)
- Melanie Albrecht
- Molecular Allergology, Vice President´s Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University of Marburg, Marburg, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
34
|
Song J, Zhang H, Tong Y, Wang Y, Xiang Q, Dai H, Weng C, Wang L, Fan J, Shuai Y, Lai C, Fang X, Chen M, Bao J, Zhang W. Molecular mechanism of interleukin-17A regulating airway epithelial cell ferroptosis based on allergic asthma airway inflammation. Redox Biol 2023; 68:102970. [PMID: 38035662 PMCID: PMC10711239 DOI: 10.1016/j.redox.2023.102970] [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: 10/20/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
Interleukin-17A (IL-17A) levels are elevated in patients with asthma. Ferroptosis has been identified as the non-apoptotic cell death type associated with asthma. Data regarding the relation of ferroptosis with asthma and the effect of IL-17A on modulating ferroptosis in asthma remain largely unclear. The present work focused on investigating the role of IL-17A in allergic asthma-related ferroptosis and its associated molecular mechanisms using public datasets, clinical samples, human bronchial epithelial cells, and an allergic asthma mouse model. We found that IL-17A was significantly upregulated within serum in asthma cases. Adding IL-17A significantly increased ferroptosis within human bronchial epithelial cells (BEAS-2B). In ovalbumin (OVA)-induced allergic asthmatic mice, IL-17A regulated and activated lipid peroxidation induced ferroptosis, whereas IL-17A knockdown effectively inhibited ferroptosis in vivo by protection of airway epithelial cells via the xCT-GSH-GPX4 antioxidant system and reduced airway inflammation. Mouse mRNA sequencing results indicated that the tumor necrosis factor (TNF) pathway was the differential KEGG pathway in the OVA group compared to healthy controls and the OVA group compared to the IL-17A knockout OVA group. We further used N-acetylcysteine (TNF inhibitor) to inhibit the TNF signaling pathway, which was found to protect BEAS-2B cells from IL-17A induced lipid peroxidation and ferroptosis damage. Our findings reveal a novel mechanism for the suppression of ferroptosis in airway epithelial cells, which may represent a new strategy for the use of IL-17A inhibitors against allergic asthma.
Collapse
Affiliation(s)
- Jingjing Song
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Hui Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yu Tong
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yufei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qiangwei Xiang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Huan Dai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Cuiye Weng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Lei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Junwen Fan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yilong Shuai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chuqiao Lai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoxiao Fang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Mingxin Chen
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiali Bao
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
35
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
36
|
Holmdahl I, Chakraborty S, Hoyer A, Filiou A, Asarnoj A, Sjölander A, Borres MP, van Hage M, Hedlin G, Konradsen JR, Söderhäll C. Inflammatory related plasma proteins involved in acute preschool wheeze. Clin Transl Allergy 2023; 13:e12308. [PMID: 38006384 PMCID: PMC10618892 DOI: 10.1002/clt2.12308] [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: 05/19/2023] [Revised: 10/01/2023] [Accepted: 10/16/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Preschool wheeze is a risk factor for asthma development. However, the molecular mechanism behind a wheezing episode is not well understood. OBJECTIVE Our aims were to assess the association of plasma proteins with acute preschool wheeze and to study the proteins with differential expression at the acute phase at revisit after 3 months. Additionally, to investigate the relationship between protein expression and clinical parameters. METHOD We measured 92 inflammatory proteins in plasma and clinical parameters from 145 children during an episode of preschool wheeze (PW) and at the revisit after 3 months (PW-R, n = 113/145) and 101 healthy controls (HC) aged 6-48 months in the GEWAC cohort using the antibody-mediated proximity extension-based assay (Olink Proteomics, Uppsala). RESULTS Of the 74 analysed proteins, 52 were differentially expressed between PW and HC. The expression profiles of the top 10 proteins, Oncostatin M (OSM), IL-10, IL-6, Fibroblast growth factor 21 (FGF21), AXIN1, CXCL10, SIRT2, TNFSF11, Tumour necrosis factor β (TNF-β) and CASP8, could almost entirely separate PW from HC. Five out of 10 proteins were associated with intake of oral corticosteroids (OCS) 24 h preceding blood sampling (OSM, CASP8, IL-10, TNF-β and CXCL10). No differences in protein expression were seen between PWs with or without OCS in comparison to HC. At the revisit after 3 months, differential protein expressions were still seen between PW-R and HC for three (IL-10, SIRT2 and FGF21) of the 10 proteins. CONCLUSION Our results contribute to unravelling potential immunopathological pathways shared between preschool wheeze and asthma.
Collapse
Affiliation(s)
- Idun Holmdahl
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Sandip Chakraborty
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Angela Hoyer
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Anastasia Filiou
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Anna Asarnoj
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | | | - Magnus P. Borres
- Thermo Fisher ScientificUppsalaSweden
- Department of Women's and Children's HealthUppsala UniversityUppsalaSweden
| | - Marianne van Hage
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska Institutet and Karolinska University HospitalStockholmSweden
| | - Gunilla Hedlin
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Jon R. Konradsen
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Cilla Söderhäll
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| |
Collapse
|
37
|
Kim HS, Oh H, Kim B, Ji Y, Holzapfel WH, Kang H, Arellano-Ayala K. Multifunctional effects of Lactobacillus sakei HEM 224 on the gastrointestinal tract and airway inflammation. Sci Rep 2023; 13:17918. [PMID: 37864021 PMCID: PMC10589218 DOI: 10.1038/s41598-023-45043-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/15/2023] [Indexed: 10/22/2023] Open
Abstract
Mucosal tissues serve as the first defense line and their commensal microbiota play a role in sustaining of host health. This study aimed to isolate and evaluate a putative probiotic strain on various mucosal regions. Lactobacillus sakei HEM 224 was isolated from traditional Korean kimchi and identified. In the safety assessment L. sakei HEM 224 showed negative results for hemolysis, biogenic amine production and transferable antibiotic resistance. The probiotic potential of strain HEM 224 in diverse mucosal areas was shown in two different models, viz. a murine model with colitis induced by dextran sulfate sodium (DSS) and an allergic airway inflammation model induced by ovalbumin (OVA). In the colitis model, oral administration of L. sakei HEM 224 improved colitis physiology with immunomodulation, enhancing barrier components and gut microbiota alteration. In the allergic airway inflammation model, the intranasal administration of the strain decreased type 2 inflammation and enhanced epithelial barrier integrity from the airways. These results demonstrate that L. sakei HEM 224 can ameliorate inflammatory conditions in both the gastrointestinal and respiratory tracts through the reinforcement of the epithelial barrier and immunomodulation.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, 558, Handong-ro, Pohang, Gyeongbuk, 37554, Republic of Korea
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Hanna Oh
- Department of Advanced Convergence, Handong Global University, 558, Handong-ro, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Bobae Kim
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Yosep Ji
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Convergence, Handong Global University, 558, Handong-ro, Pohang, Gyeongbuk, 37554, Republic of Korea.
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea.
| | - Hyeji Kang
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea.
- Global Green Research and Development Institute, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea.
| | - Karina Arellano-Ayala
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, Vienna, 1030, Austria
| |
Collapse
|
38
|
Rynne J, Ortiz-Zapater E, Bagley DC, Zanin O, Doherty G, Kanabar V, Ward J, Jackson DJ, Parsons M, Rosenblatt J, Adcock IM, Martinez-Nunez RT. The RNA binding proteins ZFP36L1 and ZFP36L2 are dysregulated in airway epithelium in human and a murine model of asthma. Front Cell Dev Biol 2023; 11:1241008. [PMID: 37928904 PMCID: PMC10624177 DOI: 10.3389/fcell.2023.1241008] [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: 06/15/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction: Asthma is the most common chronic inflammatory disease of the airways. The airway epithelium is a key driver of the disease, and numerous studies have established genome-wide differences in mRNA expression between health and asthma. However, the underlying molecular mechanisms for such differences remain poorly understood. The human TTP family is comprised of ZFP36, ZFP36L1 and ZFP36L2, and has essential roles in immune regulation by determining the stability and translation of myriad mRNAs encoding for inflammatory mediators. We investigated the expression and possible role of the tristetraprolin (TTP) family of RNA binding proteins (RBPs), poorly understood in asthma. Methods: We analysed the levels of ZFP36, ZFP36L1 and ZFP36L2 mRNA in several publicly available asthma datasets, including single cell RNA-sequencing. We also interrogated the expression of known targets of these RBPs in asthma. We assessed the lung mRNA expression and cellular localization of Zfp36l1 and Zfp36l2 in precision cut lung slices in murine asthma models. Finally, we determined the expression in airway epithelium of ZFP36L1 and ZFP36L2 in human bronchial biopsies and performed rescue experiments in primary bronchial epithelium from patients with severe asthma. Results: We found ZFP36L1 and ZFP36L2 mRNA levels significantly downregulated in the airway epithelium of patients with very severe asthma in different cohorts (5 healthy vs. 8 severe asthma; 36 moderate asthma vs. 37 severe asthma on inhaled steroids vs. 26 severe asthma on oral corticoids). Integrating several datasets allowed us to infer that mRNAs potentially targeted by these RBPs are increased in severe asthma. Zfp36l1 was downregulated in the lung of a mouse model of asthma, and immunostaining of ex vivo lung slices with a dual antibody demonstrated that Zfp36l1/l2 nuclear localization was increased in the airway epithelium of an acute asthma mouse model, which was further enhanced in a chronic model. Immunostaining of human bronchial biopsies showed that airway epithelial cell staining of ZFP36L1 was decreased in severe asthma as compared with mild, while ZFP36L2 was upregulated. Restoring the levels of ZFP36L1 and ZFP36L2 in primary bronchial epithelial cells from patients with severe asthma decreased the mRNA expression of IL6, IL8 and CSF2. Discussion: We propose that the dysregulation of ZFP36L1/L2 levels as well as their subcellular mislocalization contributes to changes in mRNA expression and cytoplasmic fate in asthma.
Collapse
Affiliation(s)
- Jennifer Rynne
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Elena Ortiz-Zapater
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Dustin C. Bagley
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
| | - Onofrio Zanin
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - George Doherty
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Varsha Kanabar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Jon Ward
- Histochemistry Research Unit, University of Southampton, Southampton, United Kingdom
| | - David J. Jackson
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Maddy Parsons
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
| | - Jody Rosenblatt
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
| | - Ian M. Adcock
- National Heart and Lung Institute and Data Science Institute, Imperial College London, London, United Kingdom
| | - Rocio T. Martinez-Nunez
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
39
|
Bravo M, Diaz-Chamorro S, Garrido-Jiménez S, Blanco J, Simón I, García W, Montero MJ, Gonçalves P, Martínez C, Cumplido-Laso G, Benítez DA, Mulero-Navarro S, Centeno F, Román ÁC, Fernández-Llario P, Cerrato R, Carvajal-González JM. Immunomodulatory effects of inactivated Ligilactobacillus salivarius CECT 9609 on respiratory epithelial cells. Vet Res 2023; 54:91. [PMID: 37845774 PMCID: PMC10580541 DOI: 10.1186/s13567-023-01228-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/07/2023] [Indexed: 10/18/2023] Open
Abstract
The microbiota in humans and animals play crucial roles in defense against pathogens and offer a promising natural source for immunomodulatory products. However, the development of physiologically relevant model systems and protocols for testing such products remains challenging. In this study, we present an experimental condition where various natural products derived from the registered lactic acid bacteria Ligilactobacillus salivarius CECT 9609, known for their immunomodulatory activity, were tested. These products included live and inactivated bacteria, as well as fermentation products at different concentrations and culture times. Using our established model system, we observed no morphological changes in the airway epithelium upon exposure to Pasteurella multocida, a common respiratory pathogen. However, early molecular changes associated with the innate immune response were detected through transcript analysis. By employing diverse methodologies ranging from microscopy to next-generation sequencing (NGS), we characterized the interaction of these natural products with the airway epithelium and their potential beneficial effects in the presence of P. multocida infection. In particular, our discovery highlights that among all Ligilactobacillus salivarius CECT 9609 products tested, only inactivated cells preserve the conformation and morphology of respiratory epithelial cells, while also reversing or altering the natural immune responses triggered by Pasteurella multocida. These findings lay the groundwork for further exploration into the protective role of these bacteria and their derivatives.
Collapse
Affiliation(s)
| | - Selene Diaz-Chamorro
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | - Sergio Garrido-Jiménez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | | | | | | | | | | | | | - Guadalupe Cumplido-Laso
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | - Dixan Agustín Benítez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | - Sonia Mulero-Navarro
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | - Francisco Centeno
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | - Ángel Carlos Román
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain
| | | | | | - José María Carvajal-González
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071, Badajoz, Spain.
| |
Collapse
|
40
|
Wang Z. Role of transforming growth factor-β in airway remodelling in bronchiolitis obliterans. Growth Factors 2023; 41:192-209. [PMID: 37487145 DOI: 10.1080/08977194.2023.2239356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Airway remodelling is the main pathological mechanism of bronchiolitis obliterans (BO). Several studies have found that transforming growth factor-β (TGF-β) expression is increased in BO during airway remodelling, where it plays an important role in various biological processes by binding to its receptor complex to activate multiple signalling proteins and pathways. This review examines the role of TGF-β in airway remodelling in BO and its potential as a therapeutic target, highlighting the mechanisms of TGF-β activation and signalling, cellular targets of TGF-β actions, and research progress in TGF-β signalling and TGF-β-mediated processes.
Collapse
Affiliation(s)
- Ziwei Wang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Gerayeli FV, Milne S, Yang CX, Li X, Guinto E, Yang JSW, Cheung CY, Stach TR, Shaipanich T, Leung JM, Sin DD. Single-cell RNA sequencing of bronchoscopy specimens: development of a rapid minimal handling protocol. Biotechniques 2023; 75:157-167. [PMID: 37815826 DOI: 10.2144/btn-2023-0017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is an important tool for understanding disease pathophysiology, including airway diseases. Currently, the majority of scRNA-seq studies in airway diseases have used invasive methods (airway biopsy, surgical resection), which carry inherent risks and thus present a major limitation to scRNA-seq investigation of airway pathobiology. Bronchial brushing, where the airway mucosa is sampled using a cytological brush, is a viable, less invasive method of obtaining airway cells for scRNA-seq. Here we describe the development of a rapid and minimal handling protocol for preparing single-cell suspensions from bronchial brush specimens for scRNA-seq. Our optimized protocol maximizes cell recovery and cell quality and facilitates large-scale profiling of the airway transcriptome at single-cell resolution.
Collapse
Affiliation(s)
- Firoozeh V Gerayeli
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Milne
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Chen Xi Yang
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Xuan Li
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth Guinto
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Julia Shun-Wei Yang
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Chung Yan Cheung
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Tara R Stach
- Biomedical Research Center, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Tawimas Shaipanich
- Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Janice M Leung
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
42
|
Frey A, Lunding LP, Wegmann M. The Dual Role of the Airway Epithelium in Asthma: Active Barrier and Regulator of Inflammation. Cells 2023; 12:2208. [PMID: 37759430 PMCID: PMC10526792 DOI: 10.3390/cells12182208] [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: 07/13/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic airway inflammation is the cornerstone on which bronchial asthma arises, and in turn, chronic inflammation arises from a complex interplay between environmental factors such as allergens and pathogens and immune cells as well as structural cells constituting the airway mucosa. Airway epithelial cells (AECs) are at the center of these processes. On the one hand, they represent the borderline separating the body from its environment in order to keep inner homeostasis. The airway epithelium forms a multi-tiered, self-cleaning barrier that involves an unstirred, discontinuous mucous layer, the dense and rigid mesh of the glycocalyx, and the cellular layer itself, consisting of multiple, densely interconnected cell types. On the other hand, the airway epithelium represents an immunologically highly active tissue once its barrier has been penetrated: AECs play a pivotal role in releasing protective immunoglobulin A. They express a broad spectrum of pattern recognition receptors, enabling them to react to environmental stressors that overcome the mucosal barrier. By releasing alarmins-proinflammatory and regulatory cytokines-AECs play an active role in the formation, strategic orientation, and control of the subsequent defense reaction. Consequently, the airway epithelium is of vital importance to chronic inflammatory diseases, such as asthma.
Collapse
Affiliation(s)
- Andreas Frey
- Division of Mucosal Immunology and Diagnostics, Research Center Borstel, 23845 Borstel, Germany;
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
| | - Lars P. Lunding
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
- Division of Lung Immunology, Research Center Borstel, 23845 Borstel, Germany
| | - Michael Wegmann
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
- Division of Lung Immunology, Research Center Borstel, 23845 Borstel, Germany
| |
Collapse
|
43
|
Goossens J, Jonckheere AC, Seys SF, Dilissen E, Decaesteker T, Goossens C, Peers K, Vanbelle V, Stappers J, Aertgeerts S, De Wilde B, Leus J, Verelst S, Raes M, Dupont L, Bullens DM. Activation of epithelial and inflammatory pathways in adolescent elite athletes exposed to intense exercise and air pollution. Thorax 2023; 78:775-783. [PMID: 36927754 PMCID: PMC10359548 DOI: 10.1136/thorax-2022-219651] [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: 09/19/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023]
Abstract
RATIONALE Participation in high-intensity exercise in early life might act as stressor to the airway barrier. OBJECTIVES To investigate the effect of intense exercise and associated exposure to air pollution on the airway barrier in adolescent elite athletes compared with healthy controls and to study exercise-induced bronchoconstriction (EIB) in this population. METHODS Early-career elite athletes attending 'Flemish-Elite-Sports-Schools' (12-18 years) of 4 different sport disciplines (n=90) and control subjects (n=25) were recruited. Presence of EIB was tested by the eucapnic voluntary hyperventilation (EVH) test. Markers at mRNA and protein level; RNA-sequencing; carbon load in airway macrophages were studied on induced sputum samples. RESULTS 444 genes were differentially expressed in sputum from athletes compared with controls, which were related to inflammation and epithelial cell damage and sputum samples of athletes contained significantly more carbon loaded airway macrophages compared with controls (24%, 95% CI 20% to 36%, p<0.0004). Athletes had significantly higher substance P (13.3 pg/mL, 95% CI 2.0 to 19.2) and calprotectin (1237 ng/mL, 95% CI 531 to 2490) levels as well as IL-6, IL-8 and TNF-α mRNA levels compared with controls (p<0.05). The incidence of EIB in athletes was 9%. The maximal fall in forced expiratory volume in 1 s (%) after EVH test in athletes was significantly associated with prior PM10 and PM2.5 exposure. CONCLUSION Early-career elite athletes showed increased markers of air pollution exposure, epithelial damage and airway inflammation compared with controls. Acute exposure to increased air pollution PM10 levels was linked to increased airway hyper-reactivity. TRIAL REGISTRATION NUMBER NCT03587675.
Collapse
Affiliation(s)
- Janne Goossens
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Anne-Charlotte Jonckheere
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Sven F Seys
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Ellen Dilissen
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Tatjana Decaesteker
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Camille Goossens
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Koen Peers
- Sport Medical Advice Centre, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Sven Aertgeerts
- Academic Centre for General Practitioners, Catholic University, Leuven, Belgium
| | | | - Jasmine Leus
- Pediatric Allergy, AZ Maria Middelares, Sint-Niklaas, Belgium
- Clinical Division of Paediatrics, Katholieke Universiteit, Leuven, Flanders, Belgium
| | - Sophie Verelst
- Clinical Division of Paediatrics, Katholieke Universiteit, Leuven, Flanders, Belgium
- Pediatrics, Jessa Hospital Campus Virga Jesse, Hasselt, Belgium
| | - Marc Raes
- Clinical Division of Paediatrics, Katholieke Universiteit, Leuven, Flanders, Belgium
- Pediatrics, Jessa Hospital Campus Virga Jesse, Hasselt, Belgium
| | - Lieven Dupont
- Department of Respiratory Medicine, University Hospital Gasthuisberg, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Dominique M Bullens
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Clinical Division of Paediatrics, Katholieke Universiteit, Leuven, Flanders, Belgium
| |
Collapse
|
44
|
Fang L, Zhou L, Kulić Ž, Lehner MD, Tamm M, Roth M. EPs ® 7630 Stimulates Tissue Repair Mechanisms and Modifies Tight Junction Protein Expression in Human Airway Epithelial Cells. Int J Mol Sci 2023; 24:11230. [PMID: 37446408 DOI: 10.3390/ijms241311230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
Airway epithelium repair after infection consists of wound repair, re-synthesis of the extracellular matrix (ECM), and tight junction proteins. In humans, EPs® 7630 obtained from Pelargonium sidoides roots reduces the severity and duration of acute respiratory tract infections. The effect of EPs® 7630 on tissue repair of rhinovirus-16 (RV-16) infected and control human airway epithelial cells was assessed for: (i) epithelial cell proliferation by manual cell counts, (ii) epithelial wound repair by "scratch assay", (iii) ECM composition by Western-blotting and cell-based ELISA, and (iv) epithelial tight junction proteins by Western-blotting. EPs® 7630 stimulated cell proliferation through cAMP, CREB, and p38 MAPK. EPs® 7630 significantly improved wound repair. Pro-inflammatory collagen type-I expression was reduced by EPs® 7630, while fibronectin was increased. Virus-binding tight junction proteins desmoglein2, desmocollin2, ZO-1, claudin1, and claudin4 were downregulated by EPs® 7630. The RV16-induced shift of the ECM towards the pro-inflammatory type was prevented by EPs® 7630. Most of the effects of EPs® 7630 on tissue repair and regeneration were sensitive to inhibition of cAMP-induced signaling. The data suggest that EPs® 7630-dependent modification of epithelial cell metabolism and function might underlie the faster recovery time from viral infections, as reported by others in clinical studies.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Liang Zhou
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Žarko Kulić
- Preclinical Research and Development, Dr. Willmar Schwabe GmbH & Co. KG, D-76227 Karlsruhe, Germany
| | - Martin D Lehner
- Preclinical Research and Development, Dr. Willmar Schwabe GmbH & Co. KG, D-76227 Karlsruhe, Germany
| | - Michael Tamm
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| |
Collapse
|
45
|
Wu YC, Moon HG, Bindokas VP, Phillips EH, Park GY, Lee SSY. Multiresolution 3D Optical Mapping of Immune Cell Infiltrates in Mouse Asthmatic Lung. Am J Respir Cell Mol Biol 2023; 69:13-21. [PMID: 37017484 PMCID: PMC10324044 DOI: 10.1165/rcmb.2022-0353ma] [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: 09/06/2022] [Accepted: 04/04/2023] [Indexed: 04/06/2023] Open
Abstract
Asthma is a chronic inflammatory airway disease driven by various infiltrating immune cell types into the lung. Optical microscopy has been used to study immune infiltrates in asthmatic lungs. Confocal laser scanning microscopy (CLSM) identifies the phenotypes and locations of individual immune cells in lung tissue sections by employing high-magnification objectives and multiplex immunofluorescence staining. In contrast, light-sheet fluorescence microscopy (LSFM) can visualize the macroscopic and mesoscopic architecture of whole-mount lung tissues in three dimensions (3D) by adopting an optical tissue-clearing method. Despite each microscopy method producing image data with unique resolution from a tissue sample, CLSM and LSFM have not been applied together because of different tissue-preparation procedures. Here, we introduce a new approach combining LSFM and CLSM into a sequential imaging pipeline. We built a new optical tissue clearing workflow in which the immersion clearing agent can be switched from an organic solvent to an aqueous sugar solution for sequential 3D LSFM and CLSM of mouse lungs. This sequential combination microscopy offered quantitative 3D spatial analyses of the distribution of immune infiltrates in the same mouse asthmatic lung tissue at the organ, tissue, and cell levels. These results show that our method facilitates multiresolution 3D fluorescence microscopy as a new imaging approach providing comprehensive spatial information for a better understanding of inflammatory lung diseases.
Collapse
Affiliation(s)
| | - Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
| | - Vytautas P. Bindokas
- Integrated Light Microscopy Facility, The University of Chicago, Chicago, Illinois; and
| | | | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | | |
Collapse
|
46
|
Ginebaugh SP, Hagner M, Ray A, Erzurum SC, Comhair SAA, Denlinger LC, Jarjour NN, Castro M, Woodruff PG, Christenson SA, Bleecker ER, Meyers DA, Hastie AT, Moore WC, Mauger DT, Israel E, Levy BD, Wenzel SE, Camiolo MJ. Bronchial epithelial cell transcriptional responses to inhaled corticosteroids dictate severe asthmatic outcomes. J Allergy Clin Immunol 2023; 151:1513-1524. [PMID: 36796454 PMCID: PMC10257752 DOI: 10.1016/j.jaci.2023.01.028] [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: 08/18/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Inhaled corticosteroids (CSs) are the backbone of asthma treatment, improving quality of life, exacerbation rates, and mortality. Although effective for most, a subset of patients with asthma experience CS-resistant disease despite receiving high-dose medication. OBJECTIVE We sought to investigate the transcriptomic response of bronchial epithelial cells (BECs) to inhaled CSs. METHODS Independent component analysis was performed on datasets, detailing the transcriptional response of BECs to CS treatment. The expression of these CS-response components was examined in 2 patient cohorts and investigated in relation to clinical parameters. Supervised learning was used to predict BEC CS responses using peripheral blood gene expression. RESULTS We identified a signature of CS response that was closely correlated with CS use in patients with asthma. Participants could be separated on the basis of CS-response genes into groups with high and low signature expression. Patients with low expression of CS-response genes, particularly those with a severe asthma diagnosis, showed worse lung function and quality of life. These individuals demonstrated enrichment for T-lymphocyte infiltration in endobronchial brushings. Supervised machine learning identified a 7-gene signature from peripheral blood that reliably identified patients with poor CS-response expression in BECs. CONCLUSIONS Loss of CS transcriptional responses within bronchial epithelium was related to impaired lung function and poor quality of life, particularly in patients with severe asthma. These individuals were identified using minimally invasive blood sampling, suggesting these findings may enable earlier triage to alternative treatments.
Collapse
Affiliation(s)
- Scott P Ginebaugh
- Integrative Systems Biology, University of Pittsburgh, Pittsburgh, Pa
| | | | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | | | | | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, Mo
| | - Prescott G Woodruff
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | | | - Eugene R Bleecker
- Division for Genetics, Genomics and Personalized Medicine, University of Arizona College of Medicine, Tucson, Ariz
| | - Deborah A Meyers
- Division for Genetics, Genomics and Personalized Medicine, University of Arizona College of Medicine, Tucson, Ariz
| | | | - Wendy C Moore
- Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Elliot Israel
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Bruce D Levy
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Environmental Medicine and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | | |
Collapse
|
47
|
Li N, Liu B, Xiong R, Li G, Wang B, Geng Q. HDAC3 deficiency protects against acute lung injury by maintaining epithelial barrier integrity through preserving mitochondrial quality control. Redox Biol 2023; 63:102746. [PMID: 37244125 DOI: 10.1016/j.redox.2023.102746] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/12/2023] [Indexed: 05/29/2023] Open
Abstract
Sepsis is one common cause of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which is closely associated with high mortality in intensive care units (ICU). Histone deacetylase 3 (HDAC3) serves as an important epigenetic modifying enzyme which could affect chromatin structure and transcriptional regulation. Here, we explored the effects of HDAC3 in type II alveolar epithelial cells (AT2) on lipopolysaccharide (LPS)-induced ALI and shed light on potential molecular mechanisms. We generated ALI mouse model with HDAC3 conditional knockout mice (Sftpc-cre; Hdac3f/f) in AT2 and the roles of HDAC3 in ALI and epithelial barrier integrity were investigated in LPS-treated AT2. The levels of HDAC3 were significantly upregulated in lung tissues from mice with sepsis and in LPS-treated AT2. HDAC3 deficiency in AT2 not only decreased inflammation, apoptosis, and oxidative stress, but also maintained epithelial barrier integrity. Meanwhile, HDAC3 deficiency in LPS-treated AT2 preserved mitochondrial quality control (MQC), evidenced by the shift of mitochondria from fission into fusion, decreased mitophagy, and improved fatty acid oxidation (FAO). Mechanically, HDAC3 promoted the transcription of Rho-associated protein kinase 1 (ROCK1) in AT2. In the context of LPS stimulation, the upregulated ROCK1 elicited by HDAC3 could be phosphorylated by Rho-associated (RhoA), thus disturbing MQC and triggering ALI. Furthermore, we found that forkhead box O1 (FOXO1) was one of transcription factors of ROCK1. HDAC3 directly decreased the acetylation of FOXO1 and promoted its nuclear translocation in LPS-treated AT2. Finally, HDAC3 inhibitor RGFP966 alleviated epithelial damage and improved MQC in LPS-treated AT2. Altogether, HDAC3 deficiency in AT2 alleviated sepsis-induced ALI by preserving mitochondrial quality control via FOXO1-ROCK1 axis, which provided a potential strategy for the treatment of sepsis and ALI.
Collapse
Affiliation(s)
- Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
48
|
Zajac D, Wojciechowski P. The Role of Vitamins in the Pathogenesis of Asthma. Int J Mol Sci 2023; 24:ijms24108574. [PMID: 37239921 DOI: 10.3390/ijms24108574] [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: 02/28/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Vitamins play a crucial role in the proper functioning of organisms. Disturbances of their levels, seen as deficiency or excess, enhance the development of various diseases, including those of the cardiovascular, immune, or respiratory systems. The present paper aims to summarize the role of vitamins in one of the most common diseases of the respiratory system, asthma. This narrative review describes the influence of vitamins on asthma and its main symptoms such as bronchial hyperreactivity, airway inflammation, oxidative stress, and airway remodeling, as well as the correlation between vitamin intake and levels and the risk of asthma in both pre- and postnatal life.
Collapse
Affiliation(s)
- Dominika Zajac
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warszawa, Poland
| | - Piotr Wojciechowski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warszawa, Poland
| |
Collapse
|
49
|
Yang H, Liang J, Li X, Yan L, Zhang Y. Inhibition of lincRNA-Cox2 alleviates apoptosis and inflammatory injury of lipopolysaccharide-stimulated human bronchial epithelial cells via the Nrf2/HO-1 axis. J Clin Biochem Nutr 2023; 72:234-241. [PMID: 37251964 PMCID: PMC10209602 DOI: 10.3164/jcbn.22-102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/28/2022] [Indexed: 05/31/2023] Open
Abstract
This study mainly explored the role and mechanism of lincRNA-Cox2 in inflammatory injury of human bronchial epithelial cells. BEAS-2B cells were stimulated with lipopolysaccharide to establish an in vitro inflammatory injury model. Real-time polymerase chain reaction was used to detect lincRNA-Cox2 expression in LPS-stimulated BEAS-2B. Cell viability and apoptosis of cells were assessed using CCK-8 and Annexin V-PI double staining. The contents of inflammatory factors were determined by enzyme-linked immunosorbent assay kits. The protein levels of nuclear factor erythrocyte 2-related factor 2 and haem oxygenase 1 protein levels were measured by Western blot. The results showed that lincRNA-Cox2 was upregulated in LPS-stimulated BEAS-2B cells. lincRNA-Cox2 knockdown inhibited apoptosis and the release of tumour necrosis factor alpha, interleukin 1beta (IL-1β), IL-4, IL-5, and IL-13 in BEAS-2B cells. lincRNA-Cox2 overexpression had the opposite effect. lincRNA-Cox2 knockdown also inhibited LPS-induced oxidative damage in BEAS-2B cells. Further mechanistic studies showed that inhibition of lincRNA-Cox2 upregulated the levels of Nrf2 and HO-1, and si-Nrf2 reversed the effects of si-lincRNA-Cox2. In conclusion, lincRNA-Cox2 knockdown inhibited BEAS-2B apoptosis and the level of inflammatory factors by activating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Hua Yang
- Department of Pediatrics, Xi’an Children’s Hospital, Xi’an, Shaanxi 710003, China
| | - Jing Liang
- Department of Children Healthcare, Xi’an Fourth Hospital, Xi’an, Shaanxi 710004, China
| | - Xiangni Li
- Department of Pediatrics, Xi’an Children’s Hospital, Xi’an, Shaanxi 710003, China
| | - Liping Yan
- Department of Pediatrics, Xi’an Children’s Hospital, Xi’an, Shaanxi 710003, China
| | - Yi Zhang
- Department of Pediatrics, Xi’an Children’s Hospital, Xi’an, Shaanxi 710003, China
| |
Collapse
|
50
|
Pandher U, Kirychuk S, Schneberger D, Thompson B, Aulakh G, Sethi RS, Singh B. Adhesion Molecules in Lung Inflammation from Repeated Glyphosate Exposures. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:ijerph20085484. [PMID: 37107767 PMCID: PMC10138447 DOI: 10.3390/ijerph20085484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/25/2023] [Accepted: 04/03/2023] [Indexed: 05/11/2023]
Abstract
Glyphosate is an active ingredient in herbicides. Exposure to glyphosate-based herbicides has been associated with respiratory dysfunctions in agricultural workers. The ability of inhaled glyphosate to induce lung inflammation is not well understood. Further, the role of adhesion molecules in glyphosate-induced lung inflammation has not been studied. We evaluated lung inflammatory responses from single and repeated glyphosate exposures. Male C57BL/6 mice were intranasally exposed to glyphosate (1 μg/40 μL) for 1 day or once daily for 5 days or 10 days. Lung tissue and bronchoalveolar lavage (BAL) fluid were collected and analyzed. Repeated exposure to glyphosate for 5 days and 10 days resulted in an increase in neutrophils in BAL fluid and higher eosinophil peroxidase levels in lungs, with leukocyte infiltration further confirmed through lung histology. Repetitive exposure to glyphosate increased IL-33 and Th2 cytokines IL-5 and IL-13. A single glyphosate treatment revealed expression for ICAM-1, VCAM-1, and vWF adhesion molecules in the perivascular region of lung sections; with repeated treatment (5 and 10 days), adhesion molecule expression was found in the perivascular, peribronchiolar, and alveolar regions of the lungs. Repetitive exposure to glyphosate induced cellular inflammation in which adhesion molecules may be important to the lung inflammatory process.
Collapse
Affiliation(s)
- Upkardeep Pandher
- Health Sciences Graduate Program, University of Saskatchewan, 107 Wiggins Road, P.O. Box 23, Saskatoon, SK S7N 5E5, Canada
- Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, 104 Clinic Place, P.O. Box 23, Saskatoon, SK S7N 2Z4, Canada
| | - Shelley Kirychuk
- Department of Medicine, College of Medicine, Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, 104 Clinic Place, P.O. Box 23, Saskatoon, SK S7N 2Z4, Canada
- Correspondence:
| | - David Schneberger
- Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, 104 Clinic Place, P.O. Box 23, Saskatoon, SK S7N 2Z4, Canada
| | - Brooke Thompson
- Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, 104 Clinic Place, P.O. Box 23, Saskatoon, SK S7N 2Z4, Canada
| | - Gurpreet Aulakh
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, P.O. Box 23, Saskatoon, SK S7N 5B4, Canada
| | - R. S. Sethi
- College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, India
| | - Baljit Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, P.O. Box 23, Saskatoon, SK S7N 5B4, Canada
| |
Collapse
|