1
|
Shen G, Yang Y, Wang N, Shi S, Chen Y, Qiao Y, Jia X, Shi X. Association of life's essential 8 and asthma: mediating effect of inflammation and oxidative stress. J Asthma 2025; 62:328-335. [PMID: 39230210 DOI: 10.1080/02770903.2024.2400613] [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: 07/19/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND The association of cardiovascular health (CVH) with asthma risk in U.S. adults remains unclear. This study aimed to explore the association of Life's Essential 8 (LE8), a measurement of CVH, with asthma and investigate the potential mediating effect of inflammation and oxidative stress. METHODS The data was obtained from the National Health and Nutrition Examination Survey (NHANES) in 2005-2018. LE8 score (range 0 ∼ 100) was measured and categorized as low (<50), moderate (50 ∼ <80), and high (≥80) CVH. Survey-weighted logistic regression and restricted cubic spline model were employed to explore the association between LE8 score and asthma. Mediation analyses were conducted to identify the mediating effects of inflammation and oxidative stress biomarkers. RESULTS This study included 10,932 participants aged ≥ 20 years, among whom 890 (8.14%) reported prevalent asthma. After adjusting for all covariates, the odd ratios (OR) for asthma were 0.67 (95% confidence interval (CI): 0.48, 0.94) in the moderate CVH group and 0.52 (95% CI: 0.34, 0.79) in the high CVH group compared with the low CVH group, respectively. The OR for asthma was 0.85 (95% CI: 0.78, 0.93) for every 10 score increase in LE8 score, and linear dose-response relationship was observed (p = 0.0642). Mediation analyses showed that inflammation and oxidative stress mediated 15.97% and 11.50% of the association between LE8 score and asthma, respectively (all p < 0.05). CONCLUSIONS LE8 score was negatively associated with asthma, and inflammation and oxidative stress partially mediated this association. It is recommended that maintaining optimal CVH may prevent asthma.
Collapse
Affiliation(s)
- Guibin Shen
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yongli Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Nana Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shangxin Shi
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yongyue Chen
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Ying Qiao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xiaocan Jia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xuezhong Shi
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| |
Collapse
|
2
|
Luo T, Ji W, Gong Y, Chen L, Liu C, Zhang D, Li X, Lv Y. REDD1 mediates HDM-induced nuclear-cytoplasmic translocation and release of IL-33 in airway epithelial cells by downregulating Nrf2. Respir Res 2025; 26:47. [PMID: 39893427 PMCID: PMC11786574 DOI: 10.1186/s12931-025-03119-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
OBJECTIVE This study aims to investigate whether REDD1 (Regulated in Development and DNA Damage Responses 1) mediates the nuclear-to-cytoplasmic translocation and release of IL-33 in airway epithelial cells induced by house dust mites (HDM). METHODS REDD1 expression levels in bronchial asthma patients were validated using public databases, followed by immunohistochemical analysis of REDD1 protein in airway epithelial cells from these patients. An asthma model was then established using HDM-induced 16HBE cell lines, with REDD1 gene knockout performed. The relationship between varying levels of REDD1 expression, Nrf2, and related inflammatory factors was assessed using Western blot and qPCR. To further investigate the role of the REDD1-Nrf2-IL-33 axis in the development of asthma, we employed Nrf2 activators and inhibitors to reassess the impact of REDD1 on IL-33. RESULTS At both mRNA and protein levels, we found that REDD1 was significantly overexpressed in samples from asthma patients (P < 0.05). In vitro, 24-hour exposure to HDM induced a notable nuclear-to-cytoplasmic translocation of IL-33 and increased its levels in the culture medium of 16HBE cells. In addition, HDM treatment significantly upregulated the expression of both REDD1 and Nrf2. Knockdown of REDD1 markedly suppressed HDM-induced IL-33 release and the expression of TNF-α, IL-6, and IL-1β, while enhancing Nrf2 expression. Moreover, treatment with the Nrf2 agonist curcumin inhibited HDM-induced nuclear-to-cytoplasmic translocation and extracellular secretion of IL-33, whereas the opposite effect was observed when using the Nrf2 antagonist ML385. CONCLUSION This study reveals the crucial regulatory role of the REDD1-Nrf2-IL-33 axis in the pathological process of bronchial asthma. REDD1 modulates the expression of IL-33 and other inflammatory factors through the Nrf2 signaling pathway, thereby influencing the onset and progression of asthma. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Tian Luo
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Wentao Ji
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Yuxin Gong
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Foshan, Guangdong, 510280, China
| | - Lichang Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Foshan, Guangdong, 510280, China
| | - Chao Liu
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Dandan Zhang
- Department of Respiratory and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Xi Li
- Department of Respiratory and Critical Care Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528300, China.
| | - Yanhua Lv
- Department of Respiratory and Critical Care Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528300, China.
| |
Collapse
|
3
|
Wei W, Xie Z, Yan J, Luo R, He J. Progress in research on induced sputum in asthma: a narrative review. J Asthma 2025; 62:189-204. [PMID: 39290080 DOI: 10.1080/02770903.2024.2395383] [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: 05/12/2024] [Revised: 08/01/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE To explore the clinical significance of induced sputum in asthma through a retrospective analysis of induced sputum in patients with asthma. DATA SOURCES The data and references cited in this article were obtained from PubMed, Sci-Hub, and Web of Science. STUDY SELECTION Observational studies with reliable data were selected. CONCLUSIONS The cytological count, -omics, and pathogen detection of induced sputum are helpful for the clinical diagnosis of asthma and in guiding medication choices.
Collapse
Affiliation(s)
- Wenjie Wei
- Department of Respiratory and Critical Care Medicine, Hunan University of Medicine General Hospital, Huaihua, People's Republic of China
| | - Zhihao Xie
- Pediatric Department, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, People's Republic of China
| | - Jun Yan
- Pediatric Department, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, People's Republic of China
| | - Renrui Luo
- Department of Respiratory and Critical Care Medicine, Hunan University of Medicine General Hospital, Huaihua, People's Republic of China
| | - Jianbin He
- Department of Respiratory and Critical Care Medicine, Hunan University of Medicine General Hospital, Huaihua, People's Republic of China
| |
Collapse
|
4
|
Cheng SC, Liou CJ, Wu SJ, Lin CF, Huang TH, Huang WC. Neochlorogenic acid ameliorates allergic airway inflammation by suppressing type 2 immunity and upregulating HO-1 expression. Int Immunopharmacol 2025; 146:113867. [PMID: 39689596 DOI: 10.1016/j.intimp.2024.113867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Neochlorogenic acid is a natural compound isolated from various fruits and vegetables that has anti-inflammation and anti-oxidative effects in macrophages. Inflammatory immune cells and tracheal epithelial cells can stimulate airway hyperresponsiveness, inflammation, and reactive oxygen species. In this study, we investigated the effect of neochlorogenic acid in ameliorating inflammatory and oxidative responses in asthmatic mice. We used an ovalbumin (OVA)-induced mouse model, treating mice with neochlorogenic acid by intraperitoneal injection. We also treated inflammatory human tracheal epithelial (BEAS-2B) cells with neochlorogenic acid to evaluate inflammatory cytokine levels and oxidative responses. The results demonstrate that neochlorogenic acid attenuated airway hyperresponsiveness, eosinophil infiltration, and goblet cell hyperplasia in the lungs of asthmatic mice. Neochlorogenic acid also reduced type 2 cytokine expression in bronchoalveolar lavage fluid and improved oxidative stress in the lung. Neochlorogenic acid effectively blocked monocyte attachment to adherent BEAS-2B cells, and reduced pro-inflammatory cytokine and reactive oxygen species production in inflammatory BEAS-2B cells. These findings suggest that neochlorogenic acid is a potential immunomodulator that can ameliorate airway hyperresponsiveness and airway inflammation in asthmatic mice.
Collapse
Affiliation(s)
- Shu-Chen Cheng
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33303, Taiwan
| | - Chian-Jiun Liou
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City 33303, Taiwan
| | - Shu-Ju Wu
- Department of Nutrition and Health Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33303, Taiwan
| | - Chwan-Fwu Lin
- Department of Cosmetic Science, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan City 33303, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan City 33303, Taiwan
| | - Tse-Hung Huang
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33303, Taiwan.
| | - Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Taoyuan City 33303, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City 33303, Taiwan; Department of Pediatrics, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei 23656, Taiwan.
| |
Collapse
|
5
|
Fawzy MG, Khalil H, Reda A, Bahgat EA. Different manipulations for resolving the overlapped spectra of novel anti-asthmatic combination: Green-blue-white triple evaluation tools. Anal Biochem 2025; 700:115783. [PMID: 39863037 DOI: 10.1016/j.ab.2025.115783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/01/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
This work represents different spectrophotometric techniques for concurrent quantification of Indacaterol (IND) and Mometasone furoate (MOM); co-formulated inhalation capsules to control asthma symptoms. Direct spectrophotometric (D0) approach was applied for IND assay. While, absorption factor (AF), ratio difference (RD), mean centering of the ratio spectra (MC), and continuous wavelet transform (CW) techniques were utilized for MOM quantification. The applied methods' validation was accomplished relative to the ICH guidelines. Within the linearity range of (4-20 μg/mL), these approaches provided accurate and precise assessment of IND and MOM in laboratory-synthetic mixtures and pharmaceutical formulation with determination coefficients higher than 0.9997. Moreover, the proposed methods demonstrated good detection and quantification levels. The respective LOD and LOQ for IND were (0.33 μg/mL) and (1.01 μg/mL). While, the LOD and LOQ for MOM were ranging from (0.16-0.30 μg/mL) and (0.47-0.91 μg/mL), respectively. Statistical comparison revealed no significant differences between the findings derived from the proposed methods and the reported one. The applied methods' environmental effect was cheeked using Analytical Greenness Calculator (AGREE), Blue Applicability Grade Index (BAGI), and Red-Green-Blue 12 (RGB12). All the utilized metrics showed reliable scores, approving that the proposed methods were adhered to the sustainability principles.
Collapse
Affiliation(s)
- Michael Gamal Fawzy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt
| | - Hawa Khalil
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Alaa Reda
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt.
| | - Eman A Bahgat
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
6
|
Bushra SMR, Safuan S, Bakar RA, Andrade LM, Abubakar BD, Nurul AA. Structural elucidation and anti-asthmatic effects of semi-crystalline polysaccharides from Lignosus rhinocerotis (Cooke) Ryvarden. Int J Biol Macromol 2025; 299:140103. [PMID: 39842582 DOI: 10.1016/j.ijbiomac.2025.140103] [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: 10/16/2024] [Revised: 12/15/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Asthma is a chronic respiratory disease characterized by airway inflammation. Lignosus rhinocerotis (LR), a medicinal mushroom rich in polysaccharide, has been traditionally used to treat various diseases, including asthma. This study aimed to fractionate, characterize and evaluate the anti-asthmatic effects of polysaccharides from LR (LRP). LRP was isolated and characterized using high-performance liquid chromatography (HPLC), x-ray diffraction analysis (XRD), fourier transform infrared spectrometry (FTIR) and nuclear magnetic resonance (NMR) spectroscopy. In an OVA-induced asthma model, BALB/c mice were sensitized, challenged, and intranasally treated with LRP. Experimental findings demonstrated that LRP exhibits a semi-crystalline nature with predicted structure of → 4)-α-D-Glcp-(1 → and → 3)-β-D-Glcp-(1→. LRP significantly reduced eosinophilia, Th2 cytokines, and IgE levels. Histological analysis revealed LRP's ability to decrease epithelial damage and epithelial and smooth muscle thickness in lung. Reductions in inflammatory cell infiltration, mucus production, and transforming growth factor (TGF)-β1 expression were observed, although not statistically significant. Gene expression analysis indicated that LRP significantly downregulated the inducible nitric oxide synthase (iNOS) expression. This study highlights a detailed structural analysis of LRP and its potential as an alternative for the management of asthma. Further research is needed to elucidate the precise mechanisms of action and optimize its therapeutic application.
Collapse
Affiliation(s)
| | - Sabreena Safuan
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ruzilawati Abu Bakar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | | | - Bishir Daku Abubakar
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Department of Human Physiology, Faculty of Basic Medical Sciences, Federal University, Dutse, Nigeria
| | - Asma Abdullah Nurul
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
7
|
Garcia FM, de Sousa VP, Silva-Dos-Santos PPE, Fernandes IS, Serpa FS, de Paula F, Mill JG, Bueno MRP, Errera FIV. Copy Number Variation in Asthma: An Integrative Review. Clin Rev Allergy Immunol 2025; 68:4. [PMID: 39755867 DOI: 10.1007/s12016-024-09015-0] [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] [Accepted: 11/13/2024] [Indexed: 01/06/2025]
Abstract
Asthma is a complex disease with varied clinical manifestations resulting from the interaction between environmental and genetic factors. While chronic airway inflammation and hyperresponsiveness are central features, the etiology of asthma is multifaceted, leading to a diversity of phenotypes and endotypes. Although most research into the genetics of asthma focused on the analysis of single nucleotide polymorphisms (SNPs), studies highlight the importance of structural variations, such as copy number variations (CNVs), in the inheritance of complex characteristics, but their role has not yet been fully elucidated in asthma. In this context, an integrative review was conducted to identify the genes and pathways involved, the location, size, and classes of CNVs, as well as their contribution to asthma risk, severity, control, and response to treatment. As a result of the review, 16 articles were analyzed, from different types of observational studies, such as case-control, cohort studies and genotyped-proband or trios design, that have been carried out in populations from different countries, ethnicities, and ages. Chromosomes 12 and 17 were the most studied in three publications each. CNVs located on 12 chromosomes were associated with asthma, the majority being found on chromosome 6p and 17q, of the deletion type, encompassing 30 different coding-protein genes and one pseudogene region. Six genes with CNVs were identified as significant expression quantitative locus (eQTLs) with mean expression in asthma-related tissues, such as the lung and whole blood. The phenotypic variability of asthma may hinder the clinical application of these findings, but the research shows the importance of investigating these genetic variations as possible biomarkers in asthma patients.
Collapse
Affiliation(s)
- Fernanda Mariano Garcia
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil.
| | - Valdemir Pereira de Sousa
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Priscila Pinto E Silva-Dos-Santos
- Department of Medicine, School of Sciences of Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória, Espírito Santo, Brazil
- Hospital Santa Casa de Misericórdia de Vitória (HSCMV), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Izadora Silveira Fernandes
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Faradiba Sarquis Serpa
- Department of Medicine, School of Sciences of Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória, Espírito Santo, Brazil
- Hospital Santa Casa de Misericórdia de Vitória (HSCMV), Vitória, Espírito Santo, Brazil
| | - Flávia de Paula
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Department of Biological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Maria Rita Passos Bueno
- Department of Genetics and Evolutionary Biology, University of São Paulo (USP), São Paulo, São Paulo, Brazil
- Human Genome and Stem Cell Research Center, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Flávia Imbroisi Valle Errera
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Department of Biological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| |
Collapse
|
8
|
He C, Wang Q, Gao J, Chen H, Tong P. Neuro-immune regulation in allergic Diseases: Role of neuropeptides. Int Immunopharmacol 2025; 145:113771. [PMID: 39667047 DOI: 10.1016/j.intimp.2024.113771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/05/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024]
Abstract
The role of neuro-immune interaction in allergic diseases, a group of common immune system diseases, has garnered increasing attention. Neuropeptides, as a crucial component of neuro-immune crosstalk with local neuroendocrine and signaling functions, play a significant role that must not be overlooked. Neuropeptides are released by neurons and even some immune cells, and mediate neuro-immune crosstalk by activating relevant specific receptors on immune cells. Recent studies have found that neuropeptides have a certain regulatory effect on allergic diseases, which could be beneficial or detrimental for the development of allergic diseases. Nevertheless, the precise mechanism of neuropeptides in allergic diseases remains unclear, particularly in the context of food allergy where their role is poorly understood. This review summarized the interplay between neuropeptides and different immune cells, as well as their current research progress in several common allergic diseases: atopic dermatitis, allergic asthma, and food allergy. It is evident that neuropeptides such as substance P, calcitonin gene-related peptide, vasoactive intestinal peptide, and neuromedin U, exert important regulatory effects on allergic diseases, yet further investigation is required to fully elucidate their mechanisms of action, which may contribute to better understanding of the onset and progression of allergic diseases and finding better immunomodulatory strategies.
Collapse
Affiliation(s)
- Cuiying He
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; College of Food Science & Technology, Nanchang University, Nanchang 330047, China
| | - Qian Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; College of Food Science & Technology, Nanchang University, Nanchang 330047, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; College of Food Science & Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; Sino-German Joint Research Institute (Jiangxi-OAI), Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, China
| |
Collapse
|
9
|
Chuang HC, Yang YCSH, Chou HC, Chen CM. Maternal aspartame exposure alters lung Th1/Th2 cytokine balance in offspring through nuclear factor-κB activation. Int Immunopharmacol 2025; 145:113800. [PMID: 39672024 DOI: 10.1016/j.intimp.2024.113800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/17/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Epidemiological evidence suggests that maternal intake of nonnutritive sweeteners is positively associated with early childhood asthma incidence. We investigated the effects of maternal aspartame exposure during pregnancy and lactation on lung Th1/Th2 cytokine balance and intestinal microbiota in offspring and explored the mechanisms that mediate these effects. METHOD Pregnant BALB/c mice were randomly divided on gestational day 7 into two dietary intervention groups: control (drinking water only) and aspartame (drinking water +0.25 g/L aspartame) groups. The dams nursed their offspring for 3 weeks. On postnatal day 21, heart blood samples were collected, and immunoglobulin E levels were measured. Microorganisms from the lower gastrointestinal tract were sampled using a culture-independent approach. Lung tissues were harvested for biochemical analyses. RESULTS Maternal aspartame exposure increased the body weight of the dams from gestational day 7 to postnatal day 21 and the body weight of the offspring from birth to postnatal day 21. Maternal aspartame exposure significantly increased the levels of Th2 cytokines (interleukin [IL]-4, IL-5, and IL-13) and IL-17 and immunoglobulin E but reduced that of a Th1 cytokine (interferon-γ) in the offspring's lung tissues. The altered Th1/Th2 balance was accompanied by increased lung nuclear factor-κB activation. The bacterial composition and alpha-diversity of the gut microbiota of the offspring did not differ significantly between the control and aspartame groups. CONCLUSION Our findings suggest maternal aspartame exposure influences lung Th1/Th2 cytokine balance in offspring through nuclear factor-κB activation.
Collapse
Affiliation(s)
- Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
10
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2025; 25:41-58. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
11
|
Fucarino A, Pitruzzella A, Burgio S, Intili G, Manna OM, Modica MD, Poma S, Benfante A, Tomasello A, Scichilone N, Bucchieri F. A novel approach to investigate severe asthma and COPD: the 3d ex vivo respiratory mucosa model. J Asthma 2025; 62:110-123. [PMID: 39096201 DOI: 10.1080/02770903.2024.2388781] [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: 03/07/2024] [Revised: 06/23/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Purpose: This article illustrates the replication of asthma and COPD conditions in a laboratory setting and the potential applications of this methodology. Introduction: Biologic drugs have been shown to enhance the treatment of severe asthma and COPD. Monoclonal antibodies against specific targets have dramatically changed the management of these conditions. Although the inflammatory pathways of asthma and COPD have already been clearly outlined, alternative mechanisms of action remain mostly unexplored. They could provide additional insights into these diseases and their clinical management. Aims: In vivo or in vitro models have thus been developed to test alternative hypotheses. This study describes sophisticated ex vivo models that mimic the response of human respiratory mucosa to disease triggers, aiming to narrow the gap between laboratory studies and clinical practice. Results: These models successfully replicate crucial aspects of these diseases, such as inflammatory cell presence, cytokine production, and changes in tissue structure, offering a dynamic platform for investigating disease processes and evaluating potential treatments, such as monoclonal antibodies. The proposed models have the potential to enhance personalized medicine approaches and patient-specific treatments, helping to advance the understanding and management of respiratory diseases.
Collapse
Affiliation(s)
- Alberto Fucarino
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy
| | - Alessandro Pitruzzella
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Stefano Burgio
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
| | - Giorgia Intili
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Olga Maria Manna
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Michele Domenico Modica
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
- Department of Otorhinolaryngology, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Salvatore Poma
- Department of Otorhinolaryngology, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Alida Benfante
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), Division of Respiratory Diseases, University of Palermo, Palermo, Italy
| | - Alessandra Tomasello
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), Division of Respiratory Diseases, University of Palermo, Palermo, Italy
| | - Nicola Scichilone
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), Division of Respiratory Diseases, University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BIND), Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| |
Collapse
|
12
|
Zhang Z, He Y, Liu H, Liu Y, Wu T, Li R, Wang Y, Ma W. NLRP3 regulates ferroptosis via the JAK2/STAT3 pathway in asthma inflammation: Insights from in vivo and in vitro studies. Int Immunopharmacol 2024; 143:113416. [PMID: 39426227 DOI: 10.1016/j.intimp.2024.113416] [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: 07/09/2024] [Revised: 09/15/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Ferroptosis, an iron-dependent form of cell death, plays a pivotal role in the pathologic progression of asthma. Electroacupuncture (EA) has demonstrated considerable efficacy in mitigating asthma airway inflammation, although its underlying mechanisms remain partially elucidated. METHODS We investigated the regulatory effect of NLRP3 on ferroptosis using a lipopolysaccharide (LPS)-induced inflammation model in BEAS-2B cells, where NLRP3 expression was modulated with si-RNA and overexpression plasmids. The levels of inflammatory cytokines TNF-α, IL-1β, and IL-6 were quantified. We also assessed NLRP3 and JAK2/STAT3 pathway-related proteins, and evaluated lipid peroxidation, mitochondrial membrane potential (ΔΨm), and antioxidant system functionality. In vivo, we examined the impact of EA on ferroptosis and airway inflammation by modulating NLRP3 activation. Asthma inflammation severity was evaluated using H&E, Masson, and PAS staining, alongside ELISA. NLRP3 and JAK2/STAT3 pathway-related proteins, as well as ferroptosis indicators, were also analyzed. The mechanism by which NLRP3 activates ferroptosis was investigated through in vitro assays. RESULTS LPS exposure resulted in increased intracellular inflammatory cytokines, and activation of the NLRP3 and JAK2/STAT3 pathways, leading to enhanced lipid peroxidation, decreased ΔΨm, and disruption of antioxidant system balance, ultimately inducing ferroptosis. Si-NLRP3 countered the effects of LPS, whereas oe-NLRP3 exacerbated symptoms. In vivo studies revealed that EA reduced airway inflammation, inhibited NLRP3 activation, and decreased phosphorylation of JAK2/STAT3, effectively lowering ferroptosis-related indicators. Utilizing JAK2/STAT3 activators and inhibitors, we confirmed that NLRP3 mediates ferroptosis via the JAK2/STAT3 pathway. CONCLUSIONS EA alleviates HDM-induced asthma, primarily through the inhibition of NLRP3 activation, which modulates the JAK2/STAT3 pathway and mediates ferroptosis.
Collapse
Affiliation(s)
- Zhengze Zhang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yuewen He
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Hao Liu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yurui Liu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Tong Wu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Ruogen Li
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China.
| | - Wuhua Ma
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China.
| |
Collapse
|
13
|
Gu W, Xie D, Li Q, Feng H, Xue Y, Chen Y, Tang J, Zhou Y, Wang D, Tong S, Liu S. Association of humidity and precipitation with asthma: a systematic review and meta-analysis. FRONTIERS IN ALLERGY 2024; 5:1483430. [PMID: 39713043 PMCID: PMC11659254 DOI: 10.3389/falgy.2024.1483430] [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: 08/21/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction The relationship of asthma with humidity and precipitation remains controversial. The objective of this study was to investigate the association of increased humidity and precipitation with asthma risk. Methods A comprehensive systematic search was conducted across various databases, including PubMed, Embase, Cochrane Library, Web of Science, Chinese Wanfang, CQVIP, and CNKI. A total of 21 studies with 1,052,960 participants from 9 countries or regions were included. The fixed and random effect model were used to analyze the data. Results The study revealed a pooled odds ratio (OR) of 1.0489 [95% confidence interval (CI): 1.0061, 1.0935] for the association between humidity and asthma risk. Specifically, among individuals under 18 years of age, the OR (95% CI) was 1.0898 (1.0290, 1.1541). Furthermore, the OR (95% CI) for developing countries or regions was 1.0927 (1.0220, 1.1684), while it was 1.1298 (0.9502, 1.3433) for regions with a high latitude (41°-51°). The pooled OR for precipitation and asthma risk was 0.9991 (0.9987, 0.9995). The OR (95%CI) values were 0.9991 (0.9987, 0.9995), 0.9991 (0.9987, 0.9995) and 0.9990 (0.9986, 0.9994) in people above the age of 18, developing countries or regions, and middle latitudes (31°-41°), respectively. Discussion The impact of humidity on asthma risk is particularly pronounced among individuals below 18 years of age, people living in developing countries or regions and in regions located in high latitudes. And the influence of precipitation on asthma persons over the age of 18, developing countries or regions, and middle latitudes significantly. Increased humidity appears to elevate asthma risk, and increased precipitation may reduce the risk. In addition, there appears to be a combined effect of humidity and precipitation on asthma. Systematic Review Registration PROSPERO, identifier, CRD42023482446.
Collapse
Affiliation(s)
- Wangyang Gu
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Dan Xie
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Qinpeng Li
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Huike Feng
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Yihao Xue
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Yang Chen
- Department of Epidemiology and Statistics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingyi Tang
- Department of Epidemiology and Statistics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yushi Zhou
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Dan Wang
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Shilu Tong
- Department of Epidemiology and Statistics, School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD, Australia
- Chinese Centers for Disease Control and Prevention, National Institute of Environmental Health, Beijing, China
| | - Shijian Liu
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
- Department of Epidemiology and Statistics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| |
Collapse
|
14
|
Rao X, Luo H, Luo K, Hu C. Silencing SMAD4 inhibits inflammation and ferroptosis in asthma by blocking the IL-17A signaling pathway. Respir Res 2024; 25:429. [PMID: 39643876 PMCID: PMC11622552 DOI: 10.1186/s12931-024-03052-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The TGF-β/SMAD signaling pathway is crucial in the pathogenesis of asthma. However, SMAD family member 4 (SMAD4), a key mediator of TGF-β, its roles and underlying mechanisms in asthma remain unclear. METHODS The in vivo and in vitro roles of SMAD4 in asthma were investigated through an ovalbumin (OVA)-induced mouse model and an interleukin-13 (IL-13)-induced cell model. The molecular mechanism of SMAD4 influenced asthma was examined using transcriptome sequencing, followed by feedback experiments involving recombinant human interleukin 17 A (rhIL-17 A), an IL-17 A signaling pathway activator. RESULTS SMAD4 was highly expressed in the asthma models. SMAD4 silencing alleviated damage to lung tissue and decreased inflammatory infiltration. Expression levels of Caspase-3, IgG, and inflammatory factors were reduced after silencing SMAD4. Silencing SMAD4 suppressed ferroptosis. Silencing SMAD4 also enhanced IL-13-induced BEAS-2B cell proliferation and suppressed apoptosis. Furthermore. IL-17 A signaling pathway was promoted in the asthma models, as evidenced by elevated IL-17RA, IL-17 A, and Act1 protein levels. SMAD4 silencing inhibited the expression levels of these IL-17 A pathway-associated proteins. Moreover, rhIL-17 A treatment notably reversed the impacts of SMAD4 silencing on asthma in the IL-13-induced cell model and OVA-induced mouse model, indicating that silencing SMAD4 inhibited inflammation and ferroptosis in asthma by blocking the IL-17 A signaling pathway. CONCLUSION Silencing SMAD4 prevents inflammation and ferroptosis in asthma by inhibiting the IL-17 pathway, which provides a novel potential approach for asthma therapy.
Collapse
Affiliation(s)
- Xingyu Rao
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Hong Luo
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Kaiyuan Luo
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Chaohua Hu
- Department of Surgery I, The Third Affiliated Hospital of Gannan Medical University/Affiliated Stomatological Hospital, No. 46, Jingjiu Road, Zhanggong District, Ganzhou, Jiangxi Province, 341000, China.
| |
Collapse
|
15
|
Hattori K, Tanaka S, Hashiba D, Tamura J, Etori K, Kageyama T, Ito T, Meguro K, Iwata A, Suto A, Suzuki K, Nakamura J, Ohtori S, Ziegler SF, Nakajima H. Synovial regulatory T cells expressing ST2 deteriorate joint inflammation through the suppression of immunoregulatory eosinophils. J Autoimmun 2024; 149:103333. [PMID: 39509740 DOI: 10.1016/j.jaut.2024.103333] [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: 05/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic polyarthritis. It is well-established that helper T cells play crucial roles in the development and deterioration of RA. Recent studies also revealed the significant roles of regulatory T (Treg) cells in this context. Although Treg cells distributed in peripheral tissues exhibit various functions, the characteristics of synovial Treg cells remain unknown. In this study, we demonstrate that synovial Treg cells exacerbate synovial inflammation by reducing the number of immunoregulatory eosinophils through competitive consumption of IL-33. Synovial Treg cells expressed ST2 in a murine arthritis model, and surprisingly, Treg-specific ST2 knockout (ST2ΔTreg) mice exhibited attenuated arthritis. In ST2ΔTreg mice, an increase in immunoregulatory synovial eosinophils was observed. Additionally, immunoregulatory eosinophils were found to express ST2, and ST2-expressing Treg cells controlled the abundance of immunoregulatory eosinophils, possibly by consuming IL-33. Our results highlight that a subset of synovial Treg cells possesses the machinery to worsen arthritis by suppressing eosinophils. In the future landscape where Treg cell-based therapies are employed for autoimmune diseases, it is important to comprehend the characteristics of disease-related Treg cells. Understanding these aspects is crucial for ensuring safer treatment modalities that do not inadvertently worsen the diseases.
Collapse
MESH Headings
- Animals
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Eosinophils/immunology
- Eosinophils/metabolism
- Mice
- Interleukin-1 Receptor-Like 1 Protein/metabolism
- Interleukin-1 Receptor-Like 1 Protein/genetics
- Mice, Knockout
- Interleukin-33/metabolism
- Interleukin-33/immunology
- Interleukin-33/genetics
- Synovial Membrane/immunology
- Synovial Membrane/pathology
- Synovial Membrane/metabolism
- Disease Models, Animal
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Humans
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Koto Hattori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Daisuke Hashiba
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan; Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Jun Tamura
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Keishi Etori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Kazuyuki Meguro
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Kotaro Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Junichi Nakamura
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Seiji Ohtori
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Steven F Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101-2795, USA.
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| |
Collapse
|
16
|
Kotrba J, Müller I, Pausder A, Hoffmann A, Camp B, Boehme JD, Müller AJ, Schreiber J, Bruder D, Kahlfuss S, Dudeck A, Stegemann-Koniszewski S. Innate players in Th2 and non-Th2 asthma: emerging roles for the epithelial cell, mast cell, and monocyte/macrophage network. Am J Physiol Cell Physiol 2024; 327:C1373-C1383. [PMID: 39401422 DOI: 10.1152/ajpcell.00488.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 11/12/2024]
Abstract
Asthma is one of the most common chronic respiratory diseases and is characterized by airway inflammation, increased mucus production, and structural changes in the airways. Recently, there is increasing evidence that the disease is much more heterogeneous than expected, with several distinct asthma endotypes. Based on the specificity of T cells as the best-known driving force in airway inflammation, bronchial asthma is categorized into T helper cell 2 (Th2) and non-Th2 asthma. The most studied effector cells in Th2 asthma include T cells and eosinophils. In contrast to Th2 asthma, much less is known about the pathophysiology of non-Th2 asthma, which is often associated with treatment resistance. Besides T cells, the interaction of myeloid cells such as monocytes/macrophages and mast cells with the airway epithelium significantly contributes to the pathogenesis of asthma. However, the underlying molecular regulation and particularly the specific relevance of this cellular network in certain asthma endotypes remain to be understood. In this review, we summarize recent findings on the regulation of and complex interplay between epithelial cells and the "nonclassical" innate effector cells mast cells and monocytes/macrophages in Th2 and non-Th2 asthma with the ultimate goal of providing the rationale for future research into targeted therapy regimens.
Collapse
Affiliation(s)
- Johanna Kotrba
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ilka Müller
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Alexander Pausder
- Research Group Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Aaron Hoffmann
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Belinda Camp
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Julia D Boehme
- Research Group Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Research Group Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas J Müller
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Dunja Bruder
- Research Group Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Research Group Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sascha Kahlfuss
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Anne Dudeck
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
17
|
Cheng L, Xiang S, Yu Q, Yu T, Sun P, Ye C, Xue H. Paeoniflorin inhibits PRAS40 interaction with Raptor to activate mTORC1 to reverse excessive autophagy in airway epithelial cells for asthma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155946. [PMID: 39276538 DOI: 10.1016/j.phymed.2024.155946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Bronchial asthma is a chronic condition characterized by airway inflammation and remodeling, which pose complex pathophysiological challenges. Autophagy has been identified as a practical strategy to regulate inflammation and remodeling processes in chronic inflammatory diseases with pathological characteristics, such as asthma. PF (Paeoniflorin) is a potential new autophagy regulatory compound. Previous studies have reported that PF can inhibit airway inflammation to alleviate allergic asthma, but whether this is mediated through the regulation of autophagy and the molecular mechanism of action remains unclear. PURPOSE The aim of this study was to evaluate the inhibitory effect of natural small molecule PF on asthma by regulating epithelial autophagy. METHODS The rat asthma model was established through intraperitoneal injection of OVA and aluminum hydroxide suspension, followed by atomized inhalation of OVA for a period of two weeks. Following treatment with PF, histopathology was observed using Masson and H&E staining, while airway Max Rrs was evaluated using a pulmonary function apparatus. Levels of inflammatory cells in BALF were detected using a blood cell analyzer, and levels of inflammatory factors in BALF were detected through Elisa. Expressions of p-PRAS40 and p-Raptor were observed through immunohistochemistry, and levels of Beclin1 and LC3B were observed through immunofluorescence. The structure and quantity of autophagosomes and autophagolysosomal were observed through TEM. An autophagy model of 16HBE cells was established after treatment with 10ng/mL IL13 for 30 minutes. PRAS40 (AKT1S1) overexpression and mutation of PF and Raptor binding site (K207M& L302I& Q417H) were introduced in 16HBE cells. Autophagy in cells was measured by mFRP-GFP-LC3 ADV fluorescent tracer. The binding sites of PF and Raptor were analyzed using the Autodock Tool. The p-mTOR, p-Raptor, p-PRAS40, LC3II/LC3I were detected through Western blot, and interaction between PRAS40-Raptor and Raptor-mTOR was detected through Co-IP. RESULTS The results showed that PF effectively reduced airway inflammation, improved airway pathological changes and remodeling, and maintained lung function. Additionally, PF was found to reverse excessive autophagy in airway epithelial cells. Interestingly, PF activated the mTORC1 subunit PRAS40 and Raptor in airway epithelial cells by regulating their phosphorylation. PRAS40 is an endogenous mTOR inhibitor that promotes autophagy. PF competitively binds Raptor to PRAS40, promoting Raptor-mTOR interactions to activate mTORC1, an outcome that can be reversed by PRAS40 overexpression and site-specific amino acid codon mutations in Raptor. CONCLUSION These findings suggest that PF intervention and inhibition of PRAS40-Raptor interaction are effective treatments for bronchial asthma. By activating mTORC1, PF effectively reverses excessive autophagy in airway epithelial cells, leading to improved airway function and reduced inflammation.
Collapse
Affiliation(s)
- Linhui Cheng
- Jiangxi University of Chinese Medicine, Nanchang,330000, China
| | - Shuangdi Xiang
- Jiangxi University of Chinese Medicine, Nanchang,330000, China.
| | - Qiangqiang Yu
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang,330000, China
| | - Tao Yu
- Jiangxi University of Chinese Medicine, Nanchang,330000, China
| | - Peng Sun
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang,330000, China
| | - Chao Ye
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang,330000, China
| | - Hanrong Xue
- Jiangxi University of Chinese Medicine, Nanchang,330000, China.
| |
Collapse
|
18
|
Wang S, Li D, Sun L. Weight-adjusted waist index is an independent predictor of all-cause and cause-specific mortality in patients with asthma. Heart Lung 2024; 68:166-174. [PMID: 39003963 DOI: 10.1016/j.hrtlng.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND There is a close relationship between obesity and the occurrence of asthma.The weight-adjusted waist index (WWI) is a relatively novel anthropometric parameter that reflects obesity. OBJECTIVE We aimed to explore the association between WWI and mortality in the asthma population. METHODS We included adult with asthma from NHANES 1999-2018. WWI = Waist circumference (cm)/square root of body weight (kg). Current asthma was determined by the participant's responses in standardized questionnaires. All-cause, cardiovascular disease (CVD), cancer, and respiratory disease mortality information was obtained by prospectively matching these data to the National Death Index. Multivariate-adjusted Cox proportional hazards regression analyses, Kaplan Meier survival analyses, restricted cubic spline (RCS) analyses, stratified analyses, and sensitivity analyses were used to clarify these associations. RESULTS A total of 101,316 participants were included in the study, and 3223 were diagnosed with asthma.WWI was independently and positively associated with all-cause and all factor-specific mortality in asthma. In fully adjusted models, each unit increase in WWI was associated with 43 % (hazard ratio [HR] and 95 % confidence interval [CI] = 1.43 [1.25,1.64], p < 0.0001), 58 % (1.58 [1.25, 1.99], p < 0.001), 50 % (1.50 [1.19, 1.90], p < 0.001), and 79 % (1.79 [1.34, 2.39], p < 0.0001) increased all-cause, CVD, cancer, and respiratory disease mortality, respectively. RCS analyses showed largely linear associations between WWI and all mortality risks. Stratified analyses indicated that these associations were influenced by multiple factors, and that age was consistently the effect modifier across all associations. CONCLUSIONS WWI is an independent predictor of all-cause, CVD, cancer, and respiratory-related mortality in the adult asthma population. These findings highlight that WWI may have novel prognostic value as a simple and easily accessible obesity parameter in asthma patients.
Collapse
Affiliation(s)
- Shidong Wang
- Department of Respiratory Medicine, Shaoxing Second Hospital, Zhejiang, China
| | - Dai Li
- Department of Respiratory Medicine, Shaoxing Second Hospital, Zhejiang, China
| | - Liping Sun
- Department of Nutrition, Shaoxing Second Hospital, Zhejiang, China.
| |
Collapse
|
19
|
Xue J, Liu Z, Xie B, Dong R, Wu J, Wu Y, Xu Z, Tian Y, Wei Y, Geng Z, Lu L, Liu Y, Xie J, Yang P. Probiotic nucleotides increase IL-10 expression in airway macrophages to mitigate airway allergy. Inflamm Res 2024; 73:1919-1930. [PMID: 39235607 DOI: 10.1007/s00011-024-01940-2] [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: 05/13/2024] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Dysfunctional immune regulation plays a crucial role in the pathogenesis of airway allergies. Macrophages are one of the components of the immune regulation cells. The aim of this study is to elucidate the role of lysine demethylase 5 A (KDM5A) in maintaining macrophages' immune regulatory ability. METHODS DNA was extracted from Lactobacillus rhamnosus GG to be designated as LgDNA. LgDNA was administered to the mice through nasal instillations. M2 macrophages (M2 cells) were isolated from the airway tissues using flow cytometry. RESULTS We found that airway M2 cells of mice with airway Th2 polarization had reduced amounts of IL-10 and KDM5A. Mice with Kdm5a deficiency in M2 cells showed the airway Th2 polarization. The expression of Kdm5a in airway M2 cells was enhanced by nasal instillations containing LgDNA. KDM5A mediated the effects of LgDNA on inducing the Il10 expression in airway M2 cells. Administration of LgDNA mitigated experimental airway allergy. CONCLUSIONS M2 macrophages in the airway tissues of mice with airway allergy show low levels of KDM5A. By upregulating KDM5A expression, LgDNA can increase Il10 expression and reconcile airway Th2 polarization.
Collapse
Affiliation(s)
- Jinmei Xue
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Bailing Xie
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy and Immunology, Shenzhen University School of Medicine, Room A7-509 at Lihu Campus. 1066 Xueyuan Blvd., Shenzhen, China
| | - Rui Dong
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Juan Wu
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yisha Wu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Zhihan Xu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yuhe Tian
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yao Wei
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Zhigang Geng
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Lei Lu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yu Liu
- Department of General Practice Medicine, Third Hospital of Shenzhen University, Shenzhen, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China.
| | - Pingchang Yang
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy and Immunology, Shenzhen University School of Medicine, Room A7-509 at Lihu Campus. 1066 Xueyuan Blvd., Shenzhen, China.
| |
Collapse
|
20
|
Lin CC, Law BF, Hettick JM. Circular RNA hsa_circ_0008726 Targets the hsa-miR-206-3p/KLF4 Axis to Modulate 4,4'-Methylene Diphenyl Diisocyanate-Glutathione Conjugate-Induced Chemokine Transcription in Macrophages. Cells 2024; 13:1725. [PMID: 39451243 PMCID: PMC11505732 DOI: 10.3390/cells13201725] [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: 09/06/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Exposure to 4,4'-methylene diphenyl diisocyanate (MDI) in the workplace may lead to the development of occupational asthma (OA). However, the specific mechanism(s) by which MDI induces OA are poorly understood. Previous reports have demonstrated that MDI and MDI-glutathione (GSH) conjugate exposure downregulates endogenous human/murine (hsa/mmu)-microRNA(miR)-206-3p, resulting in the activation of mmu/hsa-miR-206-3p-regulated signaling pathways in macrophages. Circular RNAs (circRNAs) regulate many important biological processes by targeting endogenous miRs; however, whether MDI/MDI-GSH exposure may influence circRNA expressions is unknown. Several circRNAs have been identified that regulate hsa-miR-206-3p. We hypothesize that MDI-GSH conjugate exposure induces endogenous circRNA(s) to regulate hsa-miR-206-3p in macrophages. The expression of candidate hsa-miR-206-3p-binding circRNAs was determined from MDI-GSH conjugate-treated differentiated THP-1 macrophages using RT-qPCR. MDI-GSH exposures induced hsa_circ_0008726 and its host gene transcript DNAJB6, whereas other circRNA(s) examined were either not detected or unchanged. RNA-induced silencing complex-immunoprecipitation (RISC-IP) experiments confirm that hsa-miR-206-3p can bind to hsa_circ_0008726. The expressions of endogenous hsa-miR-206-3p, hsa-miR-206-3p-regulated KLF4, and KLF4-activated M2 macrophage-associated markers and chemokines were up-/down-regulated by transfection of hsa_circ_0008726 siRNAs or hsa_circ_0008726 overexpression plasmid in macrophages, respectively. These results suggest MDI-GSH exposure downregulates hsa-miR-206-3p via induction of endogenous hsa_circ_0008726/DNAJB6, resulting in the upregulation of hsa-miR-206-3p-mediated regulations in macrophages.
Collapse
Affiliation(s)
- Chen-Chung Lin
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA; (B.F.L.); (J.M.H.)
| | | | | |
Collapse
|
21
|
Qin L, Yao Y, Wang W, Qin Q, Liu J, Liu H, Yuan L, Yuan Y, Du X, Zhao B, Wu X, Qing B, Huang L, Wang G, Xiang Y, Qu X, Zhang X, Yang M, Xia Z, Liu C. Airway epithelial overexpressed cathepsin K induces airway remodelling through epithelial-mesenchymal trophic unit activation in asthma. Br J Pharmacol 2024; 181:3700-3716. [PMID: 38853468 DOI: 10.1111/bph.16423] [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: 06/15/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Airway epithelial cells (AECs) regulate the activation of epithelial-mesenchymal trophic units (EMTUs) during airway remodelling through secretion of signalling mediators. However, the major trigger and the intrinsic pathogenesis of airway remodelling is still obscure. EXPERIMENTAL APPROACH The differing expressed genes in airway epithelia related to airway remodelling were screened and verified by RNA-sequencing and signalling pathway analysis. Then, the effects of increased cathepsin K (CTSK) in airway epithelia on airway remodelling and EMTU activation were identified both in vitro and in vivo, and the molecular mechanism was elucidated in the EMTU model. The potential of CTSK as an an effective biomarker of airway remodelling was analysed in an asthma cohort of differing severity. Finally, an inhibitor of CTSK was administered for potential therapeutic intervention for airway remodelling in asthma. KEY RESULTS The expression of CTSK in airway epithelia increased significantly along with the development of airway remodelling in a house dust mite (HDM)-stressed asthma model. Increased secretion of CTSK from airway epithelia induced the activation of EMTUs by activation of the PAR2-mediated pathway. Blockade of CTSK inhibited EMTU activation and alleviated airway remodelling as an effective intervention target of airway remodelling. CONCLUSION AND IMPLICATIONS Increased expression of CTSK in airway epithelia is involved in the development of airway remodelling in asthma through EMTU activation, mediated partly through the PAR2-mediated signalling pathway. CTSK is a potential biomarker for airway remodelling, and may also be a useful intervention target for airway remodelling in asthma patients.
Collapse
Affiliation(s)
- Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, Hunan, China
| | - Ye Yao
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Weijie Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Qingwu Qin
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jingjing Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Yunchang Yuan
- Department of Thoracic Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Bingrong Zhao
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xinyu Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Bei Qing
- Department of Thoracic Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Leng Huang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Gang Wang
- Department of Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xuewei Zhang
- Department of Health Management, Xiangya Hospital, Cental South University, Changsha, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Zhenkun Xia
- Department of Thoracic Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, Hunan, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
Lu K, Li C, Men J, Xu B, Chen Y, Yan P, Gai Z, Zhang Q, Zhang L. Traditional Chinese medicine to improve immune imbalance of asthma: focus on the adjustment of gut microbiota. Front Microbiol 2024; 15:1409128. [PMID: 39411430 PMCID: PMC11473343 DOI: 10.3389/fmicb.2024.1409128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Asthma, being the prevailing respiratory ailment globally, remains enigmatic in terms of its pathogenesis. In recent times, the advancement of traditional Chinese medicine pertaining to the intestinal microbiota has yielded a plethora of investigations, which have substantiated the potential of traditional Chinese medicine in disease prevention and treatment through modulation of the intestinal microbiota. Both animal models and clinical trials have unequivocally demonstrated the indispensable role of the intestinal microbiota in the pathogenesis of asthma. This article presents a summary of the therapeutic effects of traditional Chinese medicine in the context of regulating gut microbiota and its metabolites, thereby achieving immune regulation and inhibiting airway inflammation associated with asthma. It elucidates the mechanism by which traditional Chinese medicine modulates the gut microbiota to enhance asthma management, offering a scientific foundation for the utilization of traditional Chinese medicine in the treatment of asthma.
Collapse
Affiliation(s)
- Ke Lu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingwen Men
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bin Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizheng Yan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhibo Gai
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingxiang Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lu Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
23
|
Jung JH, Kang SA, Park JH, Kim SD, Yu HS, Mun SJ, Cho KS. Immunomodulatory Effect of Adipose Stem Cell-Derived Extra-Cellular Vesicles on Cytokine Expression and Regulatory T Cells in Patients with Asthma. Int J Mol Sci 2024; 25:10524. [PMID: 39408853 PMCID: PMC11477288 DOI: 10.3390/ijms251910524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Although mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are as effective as MSCs in the suppression of allergic airway inflammation, few studies have evaluated the immunomodulatory capacity of MSC-derived EVs in patients with asthma. Thus, we assessed the effects of adipose stem cell (ASC)-derived EVs on cytokine expression and regulatory T cells (Tregs) in peripheral blood mononuclear cells (PBMCs) of asthmatic patients. PBMCs (1 × 106 cells/mL) were isolated from asthmatic patient and healthy controls and co-cultured with 1 μg/mL of ASC-derived EVs. Th (T helper) 1-, Th2-, and Treg-related cytokine expression, fluorescence-activated cell sorting analysis of CD4+CD25+FOXP3+ T cells, and co-stimulatory molecules were analyzed before and after ASC-derived EV treatment. The expression levels of IL-4 and costimulatory molecules such as CD83 and CD86 were significantly higher in PBMCs of asthmatic patients than in control PBMCs. However, ASC-derived EV treatment significantly decreased the levels of interleukin (IL)-4 and co-stimulatory molecules such as CD83 and CD86 in the phytohemagglutinin (PHA)-stimulated PBMC of asthmatic patients. Furthermore, ASC-derived EVs remarkably increased the transforming growth factor-β (TGF-β) levels and expression of Tregs in the PBMC of asthmatic patients. ASC-derived EVs induce Treg expansion and have immunomodulatory effects by downregulating IL-4 and upregulating TGF-β in PBMCs of asthmatic patients.
Collapse
Affiliation(s)
- Jae Hoon Jung
- Department of Otorhinolaryngology, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Shin Ae Kang
- Department of Environmental Medical Biology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea;
| | - Ji-Hwan Park
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| | - Sung-Dong Kim
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| | - Hak Sun Yu
- Department of Parasitology and Tropical medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Sue Jean Mun
- Department of Otorhinolaryngology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| |
Collapse
|
24
|
Zeng Z, Ruan Y, Ying H, Wang J, Wang H, Chen S. Baicalin Attenuates Type 2 Immune Responses in a Mouse Allergic Asthma Model through Inhibiting the Production of Thymic Stromal Lymphopoietin. Int Arch Allergy Immunol 2024:1-9. [PMID: 39299223 DOI: 10.1159/000541100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
INTRODUCTION Baicalin is a flavonoid chemical extracted and purified from the traditional Chinese medicine named Scutellaria baicalensis Georgi, which possesses broad pharmacological properties. Our work aimed to explore the protective role of baicalin in allergic asthma and its potential mechanisms on regulating type 2 immune response. METHODS Mice were injected intraperitoneally with ovalbumin (OVA) twice, further challenged with OVA aerosol for continuous 5 days. For baicalin group, mice were pre-administrated with baicalin. After the final challenge, the immune cells in bronchoalveolar lavage fluid (BALF) and blood were examined. The cytokines were evaluated by ELISA. Histological inspections were examined by hematoxylin and eosin staining and Periodic Acid-Schiff staining. Thymic stromal lymphopoietin (TSLP) expression in lungs were detected using immunohistochemistry and Western blotting. RESULTS The eosinophils infiltrating in BALF were reduced remarkably in baicalin-treated asthmatic mice. Baicalin decreased OVA-induced inflammatory cytokines and total serum immunoglobulin E secretion significantly. Moreover, baicalin alleviated the asthmatic pathological changes and substantially suppressed TSLP expression in the lung tissues. CONCLUSION Our study indicates that baicalin attenuates OVA-induced allergic asthma in mice effectively by suppressing type 2 immune responses, which might provide a novel insight into the anti-asthmatic activity of baicalin.
Collapse
Affiliation(s)
- Zhisen Zeng
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Key Laboratory of Respiratory Diseases, Xiamen Medical College, Xiamen, China
- Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Yaoxin Ruan
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, China
| | - Haoran Ying
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Key Laboratory of Respiratory Diseases, Xiamen Medical College, Xiamen, China
- Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Jie Wang
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Key Laboratory of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Huangbin Wang
- Department of Microbiology and Immunology, Xiamen Medical College, Xiamen, China
| | - Shuzhen Chen
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Key Laboratory of Respiratory Diseases, Xiamen Medical College, Xiamen, China
- Department of Microbiology and Immunology, Xiamen Medical College, Xiamen, China
| |
Collapse
|
25
|
Zhu L, Li C, Gong X, Xu Z, Zhang H. Nebulized Budesonide Prevents Airway Inflammation in Children with High Total IgE Levels After Open Heart Surgery with Cardiopulmonary Bypass: A Prospective Randomized Controlled Trial. Pediatr Cardiol 2024:10.1007/s00246-024-03649-9. [PMID: 39292258 DOI: 10.1007/s00246-024-03649-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Cardiopulmonary bypass (CPB) is a crucial technique used to repair congenital heart defects (CHD); however, it may induce inflammatory response, leading to airway inflammation and need for prolonged mechanical ventilation. In this study, we aimed to evaluate the effect of budesonide nebulization in children with high serum total immunoglobulin E (tIgE) levels undergoing surgical repair of CHD via CPB. We conducted a randomized, single-center, controlled trial at a tertiary teaching hospital. One-hundred and one children with high tIgE were enrolled and randomized into the budesonide nebulization group (BUD group, n = 50) or the normal saline nebulization group (NS group, n = 51) between January 2020 and December 2020. Budesonide or normal saline was administered through a vibrating mesh nebulizer during mechanical ventilation every 8 h. Blood and bronchoalveolar lavage fluid (BALF) samples were examined and data on airway mechanics and clinical outcomes were recorded. IL-6 and IL-8 levels in the blood and BALF samples significantly increased after CPB in both groups. Budesonide inhalation reduced IL-6 and IL-8 levels in the blood and BALF samples in children with high tIgE (P < 0.05). The mean airway pressure, PCO2, and oxygen index in the BUD group were significantly lower than those in the NS group after the first inhalation dose and persisted until almost 24 h after surgery. The peak inspiratory pressure and drive pressure were lower in the BUD group than in the NS group at nearly 24 h after surgery, with no significant difference at other time points. Additionally, the duration of mechanical ventilation, number of noninvasive ventilations after extubation, and number of patients using aerosol-inhaled bronchodilators after CICU in the BUD group were significantly lower than those in the NS group (P < 0.05). Children with high preoperative tIgE levels are at risk of airway inflammation after cardiopulmonary bypass. Inhaling budesonide during postoperative mechanical ventilation can reduce the intensity of inflammatory reactions, shorten the duration of mechanical ventilation, reduce airway pressure and the utilization of NIV after extubation.
Collapse
Affiliation(s)
- Limin Zhu
- Cardiac Intensive Care Unit, Department of Cardiovascular and Thoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Chunxiang Li
- Cardiac Intensive Care Unit, Department of Cardiovascular and Thoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Xiaolei Gong
- Cardiac Intensive Care Unit, Department of Cardiovascular and Thoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Zhuoming Xu
- Cardiac Intensive Care Unit, Department of Cardiovascular and Thoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Haibo Zhang
- Department of Cardiovascular and Thoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
26
|
Park JY, Lee JW, Oh ES, Song YN, Kang MJ, Ryu HW, Kim DY, Oh SR, Lee J, Choi J, Kim N, Kim MO, Hong ST, Lee SU. Daphnetin alleviates allergic airway inflammation by inhibiting T-cell activation and subsequent JAK/STAT6 signaling. Eur J Pharmacol 2024; 979:176826. [PMID: 39033840 DOI: 10.1016/j.ejphar.2024.176826] [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: 03/23/2024] [Revised: 06/21/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Allergic asthma is a major health burden on society as a chronic respiratory disease characterized by inflammation and muscle tightening around the airways in response to inhaled allergens. Daphne kiusiana Miquel is a medicinal plant that can suppress allergic airway inflammation; however, its specific molecular mechanisms of action are unclear. In this study, we aimed to elucidate the mechanisms by which D. kiusiana inhibits allergic airway inflammation. We evaluated the anti-inflammatory effects of the ethyl acetate (EA) fraction of D. kiusiana and its major compound, daphnetin, on murine T lymphocyte EL4 cells stimulated with phorbol 12-myristate 13-acetate and ionomycin in vitro and on asthmatic mice stimulated with ovalbumin in vivo. The EA fraction and daphnetin inhibited T-helper type 2 (Th2) cytokine secretion, serum immunoglobulin E production, mucus secretion, and inflammatory cell recruitment in vivo. In vitro, daphnetin suppressed intracellular Ca2+ mobilization (a critical regulator of nuclear factor of activated T cells) and functions of the activator protein 1 transcription factor to reduce interleukin (IL)-4 and IL-13 expression. Daphnetin effectively suppressed the IL-4/-13-induced activation of Janus kinase (JAK)/signal transducer and activator of transcription 6 (STAT6) signaling in vitro and in vivo, thereby inhibiting the expression of GATA3 and PDEF, two STAT6-target genes responsible for producing Th2 cytokines and mucins. These findings indicate that daphnetin suppresses allergic airway inflammation by stabilizing intracellular Ca2+ levels and subsequently inactivating the JAK/STAT6/GATA3/PDEF pathway, suggesting that daphnetin is a promising alternative to existing asthma treatments.
Collapse
Affiliation(s)
- Ji-Yoon Park
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; Department of Anatomy & Cell Biology, Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Jae-Won Lee
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Eun Sol Oh
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Yu Na Song
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Myung-Ji Kang
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Hyung Won Ryu
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Doo-Young Kim
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Sei-Ryang Oh
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Juhyun Lee
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Jinseon Choi
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Namho Kim
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; Department of Anatomy & Cell Biology, Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Mun-Ock Kim
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Sung-Tae Hong
- Department of Anatomy & Cell Biology, Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Su Ui Lee
- Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| |
Collapse
|
27
|
Wang C, Zhong J, Hu J, Cao C, Qi S, Ma R, Fu W, Zhang X, Akdis CA, Gao Y. IL-37 protects against house dust mite-induced airway inflammation and airway epithelial barrier dysfunction via inhibiting store-operated calcium entry. Int Immunopharmacol 2024; 138:112525. [PMID: 38941668 DOI: 10.1016/j.intimp.2024.112525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Airway epithelial barrier dysfunction has been proved to contribute to the development of type 2 inflammation of asthma. Interleukin (IL)-37 is a negative regulator of immune responses and allergic airway inflammation. However, whether IL-37 has any effect on airway epithelial barrier has been unknown. METHODS We evaluated the role of IL-37 in both mouse model and cultured 16HBE cells. Histology and ELISA assays were used to evaluate airway inflammation. FITC-dextran permeability assay was used to evaluate the airway epithelial barrier function. Immunofluorescence, western blot and quantitative Real-Time PCR (RT-PCR) were used to evaluate the distribution and expression of tight junction proteins. RT-PCR and Ca2+ fluorescence measurement were used to evaluate the mRNA expression and activity of store-operated calcium entry (SOCE). RESULTS IL-37 inhibited house dust mite (HDM)-induced airway inflammation and decreased the levels of IgE in serum and type 2 cytokines in bronchoalveolar lavage fluid (BALF) compared to asthmatic mice. IL-37 protected against HDM-induced airway epithelial barrier dysfunction, including reduced leakage of FITC-dextran, enhanced expression of TJ proteins, and restored the membrane distribution of TJ proteins. Moreover, IL-37 decreased the level of IL-33 in the BALF of asthmatic mice and the supernatants of HDM-treated 16HBE cells. IL-37 decreased the peak level of Ca2+ fluorescence induced by thapsigargin and HDM, and inhibited the mRNA expression of Orai1, suggesting an inhibiting effect of IL-37 on SOCE in airway epithelial cells. CONCLUSION IL-37 plays a protective role in airway inflammation and HDM-induced airway epithelial barrier dysfunction by inhibiting SOCE.
Collapse
Affiliation(s)
- Changchang Wang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jian Zhong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaqian Hu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Can Cao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shiquan Qi
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ruxue Ma
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Fu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolian Zhang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yadong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Allergy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
28
|
Lin CC, Law BF, Hettick JM. MicroRNA-mediated Krüppel-like factor 4 upregulation induces alternatively activated macrophage-associated marker and chemokine transcription in 4,4'-methylene diphenyl diisocyanate exposed macrophages. Xenobiotica 2024; 54:730-748. [PMID: 38568505 PMCID: PMC11489325 DOI: 10.1080/00498254.2024.2334329] [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: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/20/2024]
Abstract
1. Occupational exposure to 4,4'-methylene diphenyl diisocyanate (MDI) is associated with occupational asthma (OA) development. Alveolar macrophage-induced recruitment of immune cells to the lung microenvironment plays an important role during asthma pathogenesis. Previous studies identified that MDI/MDI-glutathione (GSH)-exposure downregulates endogenous hsa-miR-206-3p/hsa-miR-381-3p. Our prior report shows that alternatively activated (M2) macrophage-associated markers/chemokines are induced by MDI/MDI-GSH-mediated Krüppel-Like Factor 4 (KLF4) upregulation in macrophages and stimulates immune cell chemotaxis. However, the underlying molecular mechanism(s) by which MDI/MDI-GSH upregulates KLF4 remain unclear. 2. Following MDI-GSH exposure, microRNA(miR)-inhibitors/mimics or plasmid transfection, endogenous hsa-miR-206-3p/hsa-miR-381-3p, KLF4, or M2 macrophage-associated markers (CD206, TGM2), and chemokines (CCL17, CCL22, CCL24) were measured by either RT-qPCR, western blot, or luciferase assay. 3. MDI-GSH exposure downregulates hsa-miR-206-3p/hsa-miR-381-3p by 1.46- to 9.75-fold whereas upregulates KLF4 by 1.68- to 1.99-fold, respectively. In silico analysis predicts binding between hsa-miR-206-3p/hsa-miR-381-3p and KLF4. Gain- and loss-of-function, luciferase reporter assays and RNA-induced silencing complex-immunoprecipitation (RISC-IP) studies confirm the posttranscriptional regulatory roles of hsa-miR-206-3p/hsa-miR-381-3p and KLF4 in macrophages. Furthermore, hsa-miR-206-3p/hsa-miR-381-3p regulate the expression of M2 macrophage-associated markers and chemokines via KLF4. 4. In conclusion, hsa-miR-206-3p/hsa-miR-381-3p play a major role in regulation of MDI/MDI-GSH-induced M2 macrophage-associated markers and chemokines by targeting the KLF4 transcript, and KLF4-mediated regulation in macrophages.
Collapse
Affiliation(s)
- Chen-Chung Lin
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Brandon F. Law
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Justin M. Hettick
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| |
Collapse
|
29
|
Kong Y, Wang Z, Yu H, Dong A, Song Y, Guo L, Zhu J, Sun L, Guo Y. YiQi GuBen formula alleviates airway inflammation and airway remodeling in OVA-induced asthma mice through TLR4/NF-κB signaling pathway. J Pharm Pharmacol 2024; 76:1028-1037. [PMID: 38824434 DOI: 10.1093/jpp/rgae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND We aim to investigate the effect of YiQi GuBen formula (YQGB) on airway inflammation and airway remodeling in the ovalbumin (OVA)-induced asthma model to further explore the potential mechanisms of YQGB in treating allergic asthma. METHODS Mice were divided into five groups randomly (n = 10): the control group, OVA group, OVA + Dex (0.1 mg/kg) group, OVA + low-dose (1.1 g/kg) YQGB group, and OVA + high-dose (2.2 g/kg) YQGB group. Inflammatory cell count and IgE were detected in bronchoalveolar lavage fluid (BALF). Lung tissue histopathology was observed by using H&E, PAS, Masson, and immunohistochemistry staining. qRT-PCR and western blot were applied to analyze key genes and proteins associated with TLR4 and NF-κB signaling pathways. RESULTS In OVA-induced asthma mice, YQGB decreased eosinophils and IgE in BALF. YQGB alleviated the OVA-induced inflammatory infiltration and declined IL-4, IL-5, IL-13, Eotaxin, ECP, GM-CSF, LTC4, and LTD4. YQGB attenuated the OVA-induced goblet cell metaplasia and mucus hypersecretion. YQGB mitigated the OVA-induced subepithelial fibrosis and lowered TGF-β1, E-Cadherin, Vimentin, and Fibronectin. YQGB ameliorated the OVA-induced airway smooth muscle thickening and lessened α-SMA and PDGF levels. YQGB reduced the expression of TLR4, MyD88, TRAF6, IκBα, and p65 mRNAs, and IκBα and p-p65 protein levels were also reduced. CONCLUSION YQGB exhibits the anti-asthma effect by reducing airway inflammation and airway remodeling through suppressing TLR4/NF-κB signaling pathway, and is worth promoting clinically.
Collapse
Affiliation(s)
- Yibu Kong
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Zhongtian Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
| | - Hongjun Yu
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Aiai Dong
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Yongfu Song
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
| | - Lei Guo
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Jinpu Zhu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
| | - Liping Sun
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| | - Yinan Guo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medical, No.1035, Boshuo Road, Jingyue District, Changchun 130017, China
- Department of Pediatrics, The Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Road, Chaoyang District, Changchun 130021, Jilin, China
| |
Collapse
|
30
|
Chen S, Gao J, Zhang T. From mesenchymal stem cells to their extracellular vesicles: Progress and prospects for asthma therapy. Asian J Pharm Sci 2024; 19:100942. [PMID: 39253613 PMCID: PMC11382190 DOI: 10.1016/j.ajps.2024.100942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 12/18/2023] [Accepted: 05/20/2024] [Indexed: 09/11/2024] Open
Abstract
Asthma is a widespread public health concern, with an increasing incidence. Despite the implementation of current treatment strategies, asthma control, particularly for severe cases, remains suboptimal. Recent research has revealed the encouraging prospects of extracellular vesicles (EVs) secreted by mesenchymal stem cells (MSCs) as a viable therapeutic option for alleviating asthma symptoms. Therefore, the present review aims to provide an overview of the current progress and the therapeutic mechanisms of using MSC-derived EVs (MSC-EVs) for asthma treatment. Additionally, different administration approaches for EVs and their impacts on biodistribution and the curative outcomes of EVs are summarized. Notably, the potential benefits of nebulized inhalation of MSC-EVs are addressed. Also, the possibilities and challenges of using MSC-EVs for asthma treatment in clinics are highlighted. Overall, this review is intended to give new insight into the utilization of MSC-EVs as a potential biological drug for asthma treatment.
Collapse
Affiliation(s)
- Shihan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
31
|
Wang Y, Liu L. Immunological factors, important players in the development of asthma. BMC Immunol 2024; 25:50. [PMID: 39060923 PMCID: PMC11282818 DOI: 10.1186/s12865-024-00644-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Asthma is a heterogeneous disease, and its development is the result of a combination of factors, including genetic factors, environmental factors, immune dysfunction and other factors. Its specific mechanism has not yet been fully investigated. With the improvement of disease models, research on the pathogenesis of asthma has made great progress. Immunological disorders play an important role in asthma. Previously, we thought that asthma was mainly caused by an imbalance between Th1 and Th2 immune responses, but this theory cannot fully explain the pathogenesis of asthma. Recent studies have shown that T-cell subsets such as Th1 cells, Th2 cells, Th17 cells, Tregs and their cytokines contribute to asthma through different mechanisms. For the purpose of the present study, asthma was classified into distinct phenotypes based on airway inflammatory cells, such as eosinophilic asthma, characterized by predominant eosinophil aggregates, and neutrophilic asthma, characterized by predominant neutrophil aggregates. This paper will examine the immune mechanisms underlying different types of asthma, and will utilize data from animal models and clinical studies targeting specific immune pathways to inform more precise treatments for this condition.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pediatric Respiratory, Children's Medical Center,The First Hospital of Jilin University, Changchun, 130021, China
| | - Li Liu
- Department of Pediatric Respiratory, Children's Medical Center,The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
32
|
Li Y, Wang J, Zhao Q, Jin F, Liu G, Pan L. The impact of inflammatory cells on lung function in asthmatics in a cross sectional retrospect study. Heliyon 2024; 10:e33691. [PMID: 39071553 PMCID: PMC11283163 DOI: 10.1016/j.heliyon.2024.e33691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Background Limited data exists on the impact of inflammatory cells and clinical characteristics on lung function in individuals with asthma. Objective The objective is to examine the correlation between increased inflammatory cells, asthma symptoms, and lung function in patients with asthma in a clinical setting. Methods A retrospective cohort study was conducted on 234 individuals suspected of having asthma in Xian, China between January 2008 and December 2021. Of those, 143 patients with complete clinical feature and lung function data were enrolled to examine the relationship between increased inflammatory cells, asthma symptoms, and lung function. Basic characteristics, blood eosinophil count, blood neutrophil count, blood platelet count, blood C-reactive protein (CRP), and comprehensive lung function analysis were evaluated at each inpatient for the 143 adult asthmatics. The association between inflammatory cells and clinical parameters with pulmonary function was compared. Results The results of the study showed that individuals in the alcohol intake group had elevated blood eosinophil count compared to those in the non-alcohol intake group (P = 0.024). Long-acting inhaled beta 2 agonists and antibiotic therapy were associated with lower blood eosinophil count (P = 0.021 and P = 0.049, respectively) compared to other therapy. There was a independent association between blood eosinophil counts and FEV1 pre- and post-therapy in asthma but there was a markedly correlation between blood eosinophil counts and FEV1/FVC pre-and post-therapy in Asthma (P = 0.007). Blood neutrophil counts were inversely correlated with FEV1/FVC after treatment (P = 0.032). Night onset in asthma was positively correlated with blood neutrophil counts, while fever was negatively correlated with blood CRP (P = 0.028). Platelet counts >300 × 109/L after treatment were significantly associated with a decline in FEV (<0.001) in patients with asthma. Elevated blood eosinophil count was independently associated with clinical features in asthma. Conclusions Based on the study's findings, there is a significant decline in FEV1/FVC among individuals with elevated blood eosinophil count, both pre- and post-bronchodilator while there was a independent relationship between blood eosinophil counts and FEV1 pre-and post-therapy in asthma. This suggests that increased levels of eosinophils may independently associated contribute to reduced lung function in asthma patients.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Jingjing Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Qi Zhao
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Faguang Jin
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Gang Liu
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Lei Pan
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
33
|
Ma H, Wang T, Wang J, Wang P, Shu Q, Qin R, Li S, Xu H. Formaldehyde exacerbates inflammation and biases T helper cell lineage commitment through IFN-γ/STAT1/T-bet pathway in asthma. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116534. [PMID: 38823345 DOI: 10.1016/j.ecoenv.2024.116534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024]
Abstract
The correlation between formaldehyde (FA) exposure and prevalence of asthma has been widely reported. However, the underlying mechanism is still not fully understood. FA exposure at 2.0 mg/m3 was found to exacerbate asthma in OVA-induced murine models. IFN-γ, the cytokine produced by T helper 1 (Th1) cells, was significantly induced by FA in serum and bronchoalveolar lavage fluid (BALF) of asthmatic mice, which was different from cytokines secreted by other Th cells. The observation was also confirmed by mRNA levels of Th marker genes in CD4+ T cells isolated from BALF. In addition, increased production of IFN-γ and expression of T-bet in Jurkat T cells primed with phorbol ester and phytohaemagglutinin were also observed with 100 μM FA treatment in vitro. Upregulated STAT1 phosphorylation, T-bet expression and IFN-γ production induced by FA was found to be restrained by STAT1 inhibitor fludarabine, indicating that FA promoted Th1 commitment through the autocrine IFN-γ/STAT1/T-bet pathway in asthma. This work not only revealed that FA could bias Th lineage commitment to exacerbate allergic asthma, but also identified the signaling mechanism of FA-induced Th1 differentiation, which may be utilized as the target for development of interfering strategies against FA-induced immune disorders.
Collapse
Affiliation(s)
- Huijuan Ma
- School of Public Health, Anhui University of Science and Technology, Hefei, Anhui Province 231131, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Tingqian Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Junfeng Wang
- School of Public Health, Anhui University of Science and Technology, Hefei, Anhui Province 231131, China
| | - Peiyao Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Shu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ruilin Qin
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Sijia Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Huan Xu
- School of Public Health, Anhui University of Science and Technology, Hefei, Anhui Province 231131, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
34
|
Ghorbanzadeh S, Khojini JY, Abouali R, Alimardan S, Zahedi M, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Clearing the Path: Exploring Apoptotic Cell Clearance in Inflammatory and Autoimmune Disorders for Therapeutic Advancements. Mol Biotechnol 2024:10.1007/s12033-024-01222-6. [PMID: 38935260 DOI: 10.1007/s12033-024-01222-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 06/28/2024]
Abstract
Inflammatory and autoimmune disorders, characterized by dysregulated immune responses leading to tissue damage and chronic inflammation, present significant health challenges. This review uniquely focuses on efferocytosis-the phagocyte-mediated clearance of apoptotic cells-and its pivotal role in these disorders. We delve into the intricate mechanisms of efferocytosis' four stages and their implications in disease pathogenesis, distinguishing our study from previous literature. Our findings highlight impaired efferocytosis in conditions like atherosclerosis and asthma, proposing its targeting as a novel therapeutic strategy. We discuss the therapeutic potential of efferocytosis in modulating immune responses and resolving inflammation, offering a new perspective in treating inflammatory disorders.
Collapse
Affiliation(s)
- Shadi Ghorbanzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran
| | - Reza Abouali
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, IR, Iran.
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
35
|
Zhang L, Huang FY, Dai SZ, Wang L, Zhou X, Zheng ZY, Li Q, Tan GH, Wang CC. Rosuvastatin attenuates airway inflammation and remodeling in a chronic allergic asthma model through modulation of the AMPKα signaling pathway. PLoS One 2024; 19:e0305863. [PMID: 38913666 PMCID: PMC11195969 DOI: 10.1371/journal.pone.0305863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/05/2024] [Indexed: 06/26/2024] Open
Abstract
The efficacy of rosuvastatin in reducing allergic inflammation has been established. However, its potential to reduce airway remodeling has yet to be explored. This study aimed to evaluate the efficacy of rosuvastatin in reducing airway inflammation and remodeling in a mouse model of chronic allergic asthma induced by sensitization and challenge with OVA. Histology of the lung tissue and the number of inflammatory cells in bronchoalveolar lavage fluid (BALF) showed a marked decrease in airway inflammation and remodeling in mice treated with rosuvastatin, as evidenced by a decrease in goblet cell hyperplasia, collagen deposition, and smooth muscle hypertrophy. Furthermore, levels of inflammatory cytokines, angiogenesis-related factors, and OVA-specific IgE in BALF, plasma, and serum were all reduced upon treatment with rosuvastatin. Western blotting was employed to detect AMPK expression, while immunohistochemistry staining was used to observe the expression of remodeling signaling proteins such as α-SMA, TGF-β, MMP-9, and p-AMPKα in the lungs. It was found that the activity of 5'-adenosine monophosphate-activated protein kinase alpha (AMPKα) was significantly lower in the lungs of OVA-induced asthmatic mice compared to Control mice. However, the administration of rosuvastatin increased the ratio of phosphorylated AMPK to total AMPKα, thus inhibiting the formation of new blood vessels, as indicated by CD31-positive staining mainly in the sub-epithelial region. These results indicate that rosuvastatin can effectively reduce airway inflammation and remodeling in mice with chronic allergic asthma caused by OVA, likely due to the reactivation of AMPKα and a decrease in angiogenesis.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, China
| | - Feng-Ying Huang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Shu-Zhen Dai
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Lin Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, China
| | - Zhen-You Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Cai-Chun Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University & Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, China
| |
Collapse
|
36
|
Zhang J, Zheng X, Luo W, Sun B. Cross-domain microbiomes: the interaction of gut, lung and environmental microbiota in asthma pathogenesis. Front Nutr 2024; 11:1346923. [PMID: 38978703 PMCID: PMC11229079 DOI: 10.3389/fnut.2024.1346923] [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: 12/01/2023] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Recent experimental and epidemiological studies underscore the vital interaction between the intestinal microbiota and the lungs, an interplay known as the "gut-lung axis". The significance of this axis has been further illuminated following the identification of intestinal microbial metabolites, such as short-chain fatty acids (SCFA), as key mediators in setting the tone of the immune system. Through the gut-lung axis, the gut microbiota and its metabolites, or allergens, are directly or indirectly involved in the immunomodulation of pulmonary diseases, thereby increasing susceptibility to allergic airway diseases such as asthma. Asthma is a complex outcome of the interplay between environmental factors and genetic predispositions. The concept of the gut-lung axis may offer new targets for the prevention and treatment of asthma. This review outlines the relationships between asthma and the respiratory microbiome, gut microbiome, and environmental microbiome. It also discusses the current advancements and applications of microbiomics, offering novel perspectives and strategies for the clinical management of chronic respiratory diseases like asthma.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Xianhui Zheng
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Wenting Luo
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| |
Collapse
|
37
|
Taheri MM, Javan F, Poudineh M, Athari SS. CAR-NKT Cells in Asthma: Use of NKT as a Promising Cell for CAR Therapy. Clin Rev Allergy Immunol 2024; 66:328-362. [PMID: 38995478 DOI: 10.1007/s12016-024-08998-0] [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] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
NKT cells, unique lymphocytes bridging innate and adaptive immunity, offer significant potential for managing inflammatory disorders like asthma. Activating iNKT induces increasing IFN-γ, TGF-β, IL-2, and IL-10 potentially suppressing allergic asthma. However, their immunomodulatory effects, including granzyme-perforin-mediated cytotoxicity, and expression of TIM-3 and TRAIL warrant careful consideration and targeted approaches. Although CAR-T cell therapy has achieved remarkable success in treating certain cancers, its limitations necessitate exploring alternative approaches. In this context, CAR-NKT cells emerge as a promising approach for overcoming these challenges, potentially achieving safer and more effective immunotherapies. Strategies involve targeting distinct IgE-receptors and their interactions with CAR-NKT cells, potentially disrupting allergen-mast cell/basophil interactions and preventing inflammatory cytokine release. Additionally, targeting immune checkpoints like PDL-2, inducible ICOS, FASL, CTLA-4, and CD137 or dectin-1 for fungal asthma could further modulate immune responses. Furthermore, artificial intelligence and machine learning hold immense promise for revolutionizing NKT cell-based asthma therapy. AI can optimize CAR-NKT cell functionalities, design personalized treatment strategies, and unlock a future of precise and effective care. This review discusses various approaches to enhancing CAR-NKT cell efficacy and longevity, along with the challenges and opportunities they present in the treatment of allergic asthma.
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
| | - Seyyed Shamsadin Athari
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
38
|
Wang FX, Jin LW. Research on the Mechanism and Application of Acupuncture Therapy for Asthma: A Review. J Asthma Allergy 2024; 17:495-516. [PMID: 38828396 PMCID: PMC11144428 DOI: 10.2147/jaa.s462262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024] Open
Abstract
Asthma is a high-risk disease based on airway hyperresponsiveness (AHR). In this review, we found that there are many studies on clinical therapy for asthma that focus on the efficacy of acupuncture therapy and its mechanisms, including the functional connectivity of different brain regions, with the aid of functional magnetic resonance imaging (fMRI), immune responses/cell recognition (innate lymphoid cells and balance of Th1/Th2 and Treg/Th17), intracellular mechanism (autophagy, endoplasmic reticulum stress, and epigenetic alteration), and ligand-receptor/chemical signaling pathway (neurotransmitter, hormone, and small molecules). In this review, we summarized the clinical and experimental evidence for the mechanisms of acupuncture therapy in asthma to offer insights into drug discovery and clinical therapy. Given the paucity of clinical studies on the mechanisms of acupuncture in the treatment of asthma, this review notably included studies based on animal models to investigate the mechanisms of acupuncture in the treatment of asthma.
Collapse
Affiliation(s)
- Fei-xuan Wang
- Department of Clinical Medical College, Qilu Medical College, Zibo, Shandong, People’s Republic of China
| | - Lu-wei Jin
- Department of Acupuncture and Tuina, Wenzhou Hospital of Traditional Chinese Medicine, Wenzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
39
|
Wang Y, Cui J, Jiang Y, Zhang S, Chen L, Ma Z, Yang D, Zhang Z, Huang X, Yang Y, Guo J, Lu Z, Li C. Jiawei Yanghe Decoction attenuate allergic airway inflammation by suppressing group 2 innate lymphoid cells responses. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117927. [PMID: 38373665 DOI: 10.1016/j.jep.2024.117927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei Yanghe Decoction (JWYHD) is modified Yanghe Decoction (YHD). YHD historically utilized as a potent medicinal solution for addressing chronic inflammatory conditions, holds promising therapeutic potential in the treatment of asthma. However, the mechanisms underlying JWYHD's effects on allergic asthma remain unclear. AIM OF THE STUDY To investigate the therapeutic effect as well as the underlying mechanisms of JWYHD on asthmatic mice. MATERIALS AND METHODS The ovalbumin (OVA)-induced mouse model was utilized, followed by the administration of JWYHD to allergic asthmatic mice. Subsequently, inflammatory cells in the bronchoalveolar lavage fluid (BALF) and lung tissues were conducted. The levels of various cytokines including interleukin (IL)-4, IL-5, IL-13, IL-33, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ in BALF, as well as the total immunoglobulin E (IgE) content in serum, were assessed. Lung function and tissue pathology examinations were performed to assess the protective impacts of JWYHD. The chemical components of JWYHD and its lung prototype compounds (referred to the chemical components present in JWYHD that were observed in the lung) were explored by ultra-high performance liquid chromatography coupled to quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). RNA-seq analysis revealed the regulation mechanisms of JWYHD treating asthma. Furthermore, the effect of JWYHD on type 2 innate lymphoid cells (ILC2s) in asthmatic mice was detected by flow cytometry and Smart-RNA-seq analysis. Then molecular docking analysis was used to show the interaction between identified compounds and key targets. RESULTS JWYHD significantly attenuated the airway inflammation of asthmatic mice, reduced the levels of inflammatory cells in BALF, as well the levels of the cytokines IL-4, IL-5, IL-13, IL-33, and TNF-α in BALF and IgE in serum. Airway hyperresponsiveness (AHR) and lung inflammation infiltration were also alleviated by JWYHD. Moreover, RNA-seq analysis revealed that JWYHD attenuated airway inflammation in asthmatic mice via regulating immunity. Flow cytometry confirmed that JWYHD could inhibit ILC2 responses. ILC2 Smart-RNA-seq analysis showed that JWYHD impaired the inflammation reaction-related signaling pathways in ILC2s, and neuropilin-1 (Nrp1), endothelial transcription factor 3 (GATA3) and interleukin 1 receptor like protein 1 (ST2) might be the key targets. The molecular docking analysis investigating the connection between the primary targets and JWYHD's prototype compounds in the lung demonstrated that liquiritin apioside, icariin, glycyrrhizic acid, and uralsaponin B, identified through UPLC-Q-TOF/MS, exhibited significant affinity in binding to the mentioned key targets. CONCLUSION Our results suggested that the mechanism of JWYHD in treating asthma might be related to limiting ILC2 responses. Our findings provided some pharmacological evidence for the clinical application of JWYHD in the treatment of asthma.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Jie Cui
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuwei Jiang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shaoyan Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Linjin Chen
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zifeng Ma
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Di Yang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhengyi Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xing Huang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongqing Yang
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinglei Guo
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhenhui Lu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Cui Li
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
40
|
Abdelmawgood IA, Kotb MA, Ashry H, Ebeed BW, Mahana NA, Mohamed AS, Eid JI, Ramadan MA, Rabie NS, Mohamed MY, Saed NT, Yasser N, Essam D, Zaki YY, Saeed S, Mahmoud A, Eladawy MM, Badr AM. β-glucan mitigates ovalbumin-induced airway inflammation by preventing oxidative stress and CD8 + T cell infiltration. Int Immunopharmacol 2024; 132:111985. [PMID: 38603862 DOI: 10.1016/j.intimp.2024.111985] [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: 12/16/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Bronchial asthma is a severe respiratory condition characterized by airway inflammation, remodeling, and oxidative stress. β-Glucan (BG) is a polysaccharide found in fungal cell walls with powerful immunomodulatory properties. This study examined and clarified the mechanisms behind BG's ameliorativeactivitiesin an allergic asthma animal model. METHOD BG was extracted from Chaga mushroom and characterized using FT-IR, UV-visible, zeta potential, and 1H NMR analysis. The mice were divided into five groups, including control, untreated asthmatic, dexamethasone (Dexa)-treated (1 mg/kg), and BG (30 and 100 mg/kg)-treated groups. RESULTS BG treatment reduced nasal scratching behavior, airway-infiltrating inflammatory cells, and serum levels of IgE significantly. Additionally, BG attenuated oxidative stress biomarkers by lowering malonaldehyde (MDA) concentrations and increasing the levels of reduced glutathione (GSH), glutathione peroxidase (GPx), and catalase (CAT). Immunohistochemical and flow cytometric analyses have confirmed the suppressive effect of BG on the percentage of airway-infiltrating cytotoxic CD8+ T cells. CONCLUSION The findings revealed the role of CD8+ T cells in the pathogenesis of asthma and the role of BG as a potential therapeutic agent for asthma management through the suppression of airway inflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Mohamed A Kotb
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Hamid Ashry
- Biochemistry Branch, Chemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Bassam W Ebeed
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Noha A Mahana
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | | | - Jehane I Eid
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Marwa A Ramadan
- Department of Laser Application in Metrology, Photochemistry, and Agriculture National Institute of Laser-Enhanced Science (NILES), Cairo University, Giza, Egypt
| | - Nahla S Rabie
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Mariam Y Mohamed
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Nermeen Th Saed
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Nada Yasser
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Dina Essam
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Youssef Y Zaki
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Samar Saeed
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Asmaa Mahmoud
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Marwan M Eladawy
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Abeer Mahmoud Badr
- Zoology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| |
Collapse
|
41
|
Akenroye A, Nopsopon T, Hacker JJ, Laidlaw TM. Ratio of plasma IL-13/TNF- ∝ and CXCL10/CCL17 predicts mepolizumab and omalizumab response in asthma better than eosinophil count or immunoglobulin E level. Sci Rep 2024; 14:10404. [PMID: 38710930 PMCID: PMC11074109 DOI: 10.1038/s41598-024-60864-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
To date, most studies to identify biomarkers associated with response to the anti-interleukin 5 agent, mepolizumab, and to the anti-immunoglobulin E agent, omalizumab have focused on clinically available biomarkers, such as the peripheral blood eosinophil counts (BEC) and total immunoglobulin E (IgE). However, these biomarkers often have low predictive accuracy, with many patients with eosinophilic or allergic asthma failing to demonstrate clinical response to mepolizumab or omalizumab respectively. In this study, we evaluated the association of baseline pre-biologic plasma levels of 26 cytokines and chemokines, including T-helper 1 (Th1)-, Th2-, Th17-related cytokines, and their ratios with subsequent clinical response to mepolizumab or omalizumab. We defined clinical response as a reduction in the baseline annual exacerbation rate by half or more over the one-year period following initiation of the biologic. Baseline levels of plasma IL-13 were differentially elevated in responders versus non-responders to mepolizumab and plasma CXCL10 levels were differentially elevated in responders to omalizumab. The ratio of IL-13/TNF-α had the best sensitivity and specificity in predicting response to mepolizumab and CXCL10/CCL17 to omalizumab, and these performed better as predictive biomarkers of response than BEC and IgE. Cytokines and chemokines associated with airway eosinophilia, allergic inflammation, or Th2 inflammation, such as IL-13 and CXCL10, may be better predictors of clinical response to mepolizumab and omalizumab, than IL-5 or IgE, the targets of mepolizumab and omalizumab.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Tanawin Nopsopon
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Jonathan J Hacker
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Tanya M Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| |
Collapse
|
42
|
Yang S, Guo R, Meng X, Zhang Y. AIM2 participates in house dust mite (HDM)-induced epithelial dysfunctions and ovalbumin (OVA)-induced allergic asthma in infant mice. J Asthma 2024; 61:479-490. [PMID: 38078661 DOI: 10.1080/02770903.2023.2289157] [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: 07/28/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 01/13/2024]
Abstract
Objective: Allergen sensitization and high rates of concomitant allergic diseases are characteristic of severe pediatric asthma. The present study was aimed to explore the mechanism of allergic asthma via bioinformatics and experiment investigation. Methods: The GSE27011 dataset contained the expression profiles of normal and pediatric asthma white blood cells was downloaded for analyzing the different expression genes and function enrichment. The allergic asthma model in infant mice was established by ovalbumin (OVA) stimulation. The cellular model was established by house dust mite (HDM)-stimulation in human bronchial epithelial cells. The absent in melanoma 2 (AIM2) knockdown was achieved by intranasal lentivirus injection or cell infection. The bronchoalveolar lavage fluid (BALF) was collected for cell counting and ELISA assessment of cytokines. Lung tissues were collected for HE staining and immunohistochemical (IHC) staining. Real-time PCR and immunoblotting were used for the determination of key gene expressions in mouse and cell models. Results: upregulation of AIM2 gene expression was observed in pediatric asthma patients based on GSE27011 and OVA-induced infant mouse allergic asthma model. AIM2 knockdown ameliorated OVA caused elevation in airway hyper-responsiveness (AHR), elevation in cell quantities (eosinophils, neutrophils, lymphocytes), and levels of cytokines (IL-4, IL-13, TNF-α, and OVA-specific IgE) in BALF. Moreover, AIM2 knockdown relieved OVA-caused histopathological alterations in mouse lungs, up-regulation of AIM2 levels, and NOD1 and receptor-interacting protein 2 (RIP2) protein levels, as well as p65 phosphorylation. In the cell model, AIM2 knockdown partially ameliorated HDM-induced epithelial dysfunctions by promoting cell viability, down-regulating inflammatory cytokines levels, and decreasing the protein levels of AIM2, NOD1, RIP2, and phosphorylated p65. Conclusion: AIM2 participates in HDM-induced epithelial dysfunctions and OVA-induced allergic asthma progression. AIM2 could be a promising target for pediatric allergic asthma treatment regimens, which warrants further in vivo investigations.
Collapse
Affiliation(s)
- Shengzhi Yang
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Ru Guo
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Xianmei Meng
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| | - Yunhong Zhang
- Department of Pediatrics, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, PR China
| |
Collapse
|
43
|
Lu L, Li J, Jiang X, Bai R. CXCR4/CXCL12 axis: "old" pathway as "novel" target for anti-inflammatory drug discovery. Med Res Rev 2024; 44:1189-1220. [PMID: 38178560 DOI: 10.1002/med.22011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/25/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024]
Abstract
Inflammation is the body's defense response to exogenous or endogenous stimuli, involving complex regulatory mechanisms. Discovering anti-inflammatory drugs with both effectiveness and long-term use safety is still the direction of researchers' efforts. The inflammatory pathway was initially identified to be involved in tumor metastasis and HIV infection. However, research in recent years has proved that the CXC chemokine receptor type 4 (CXCR4)/CXC motif chemokine ligand 12 (CXCL12) axis plays a critical role in the upstream of the inflammatory pathway due to its chemotaxis to inflammatory cells. Blocking the chemotaxis of inflammatory cells by CXCL12 at the inflammatory site may block and alleviate the inflammatory response. Therefore, developing CXCR4 antagonists has become a novel strategy for anti-inflammatory therapy. This review aimed to systematically summarize and analyze the mechanisms of action of the CXCR4/CXCL12 axis in more than 20 inflammatory diseases, highlighting its crucial role in inflammation. Additionally, the anti-inflammatory activities of CXCR4 antagonists were discussed. The findings might help generate new perspectives for developing anti-inflammatory drugs targeting the CXCR4/CXCL12 axis.
Collapse
Affiliation(s)
- Liuxin Lu
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junjie Li
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoying Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
44
|
Hosomi Y, Okamura T, Sakai K, Yuge H, Yoshimura T, Majima S, Okada H, Senmaru T, Ushigome E, Nakanishi N, Satoh T, Akira S, Hamaguchi M, Fukui M. IL-33 Reduces Saturated Fatty Acid Accumulation in Mouse Atherosclerotic Foci. Nutrients 2024; 16:1195. [PMID: 38674885 PMCID: PMC11054828 DOI: 10.3390/nu16081195] [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: 03/08/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The cellular and molecular mechanisms of atherosclerosis are still unclear. Type 2 innate lymphocytes (ILC2) exhibit anti-inflammatory properties and protect against atherosclerosis. This study aimed to elucidate the pathogenesis of atherosclerosis development using atherosclerosis model mice (ApoE KO mice) and mice deficient in IL-33 receptor ST2 (ApoEST2 DKO mice). Sixteen-week-old male ApoE KO and ApoEST2 DKO mice were subjected to an 8-week regimen of a high-fat, high-sucrose diet. Atherosclerotic foci were assessed histologically at the aortic valve ring. Chronic inflammation was assessed using flow cytometry and real-time polymerase chain reaction. In addition, saturated fatty acids (palmitic acid) and IL-33 were administered to human aortic endothelial cells (HAECs) to assess fatty acid metabolism. ApoEST2 DKO mice with attenuated ILC2 had significantly worse atherosclerosis than ApoE KO mice. The levels of saturated fatty acids, including palmitic acid, were significantly elevated in the arteries and serum of ApoEST2 DKO mice. Furthermore, on treating HAECs with saturated fatty acids with or without IL-33, the Oil Red O staining area significantly decreased in the IL-33-treated group compared to that in the non-treated group. IL-33 potentially prevented the accumulation of saturated fatty acids within atherosclerotic foci.
Collapse
Affiliation(s)
- Yukako Hosomi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Kimiko Sakai
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Hiroki Yuge
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Takashi Yoshimura
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Saori Majima
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Takashi Satoh
- Department of Immune Regulation, Graduate School and Faculty of Medicine, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan;
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Suita 565-0871, Japan;
- Laboratory of Host Defense, World Premier Institute Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (Y.H.); (T.O.); (K.S.); (H.Y.); (T.Y.); (S.M.); (H.O.); (T.S.); (E.U.); (N.N.); (M.F.)
| |
Collapse
|
45
|
Brandao-Rangel MAR, Moraes-Ferreira R, Silva-Reis A, Souza-Palmeira VH, Almeida FM, da Silva Olimpio FR, Oliveira CR, Damaceno-Rodrigues NR, Pesquero JB, Martin L, Aimbire F, Albertini R, Faria SS, Vieira RP. Aerobic physical training reduces severe asthma phenotype involving kinins pathway. Mol Biol Rep 2024; 51:499. [PMID: 38598121 DOI: 10.1007/s11033-024-09474-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION Aerobic physical training (APT) reduces eosinophilic airway inflammation, but its effects and mechanisms in severe asthma remain unknown. METHODS An in vitro study employing key cells involved in the pathogenesis of severe asthma, such as freshly isolated human eosinophils, neutrophils, and bronchial epithelial cell lineage (BEAS-2B) and lung fibroblasts (MRC-5 cells), was conducted. Additionally, an in vivo study using male C57Bl/6 mice, including Control (Co; n = 10), Trained (Exe; n = 10), house dust mite (HDM; n = 10), and HDM + Trained (HDM + Exe; n = 10) groups, was carried out, with APT performed at moderate intensity, 5x/week, for 4 weeks. RESULTS HDM and bradykinin, either alone or in combination, induced hyperactivation in human neutrophils, eosinophils, BEAS-2B, and MRC-5 cells. In contrast, IL-10, the primary anti-inflammatory molecule released during APT, inhibited these inflammatory effects, as evidenced by the suppression of numerous cytokines and reduced mRNA expression of the B1 receptor and ACE-2. The in vivo study demonstrated that APT decreased bronchoalveolar lavage levels of bradykinin, IL-1β, IL-4, IL-5, IL-17, IL-33, TNF-α, and IL-13, while increasing levels of IL-10, klotho, and IL-1RA. APT reduced the accumulation of polymorphonuclear cells, lymphocytes, and macrophages in the peribronchial space, as well as collagen fiber accumulation, epithelial thickness, and mucus accumulation. Furthermore, APT lowered the expression of the B1 receptor and ACE-2 in lung tissue and reduced bradykinin levels in the lung tissue homogenate compared to the HDM group. It also improved airway resistance, tissue resistance, and tissue damping. On a systemic level, APT reduced total leukocytes, eosinophils, neutrophils, basophils, lymphocytes, and monocytes in the blood, as well as plasma levels of IL-1β, IL-4, IL-5, IL-17, TNF-α, and IL-33, while elevating the levels of IL-10 and IL-1RA. CONCLUSION These findings indicate that APT inhibits the severe asthma phenotype by targeting kinin signaling.
Collapse
Affiliation(s)
- Maysa Alves Rodrigues Brandao-Rangel
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos, SP, 11060-001, Brazil
| | - Renilson Moraes-Ferreira
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos, SP, 11060-001, Brazil
| | - Anamei Silva-Reis
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos, SP, 11060-001, Brazil
| | - Victor Hugo Souza-Palmeira
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos, SP, 11060-001, Brazil
| | - Francine Maria Almeida
- Laboratory of Experimental Therapeutic (LIM 20), School of Medicine, University of São Paulo, Avenida Doutor Arnaldo 455, São Paulo, SP, 01246-903, Brazil
| | - Fabiana Regina da Silva Olimpio
- Post-graduate Program in Translational Medicine, Department of Medicine, Federal University of São Paulo (UNIFESP), Rua Pedro De Toledo 720, 2º Andar, São Paulo, SP, 04039-002, Brazil
| | - Carlos Rocha Oliveira
- School of Medicine, Anhembi Morumbi University, Avenida Deputado Benedito Matarazzo 6070, São José dos Campos, SP, 12230-002, Brazil
- Post-graduate Program in Biomedical Enginnering, Federal University of São Paulo (UNIFESP), Rua Talim 330, São José dos Campos, SP, 12231-280, Brazil
- GAP Biotech, Rua Comendador Remo Cesaroni 223, São José dos Campos, SP, 12243-020, Brazil
| | - Nilsa Regina Damaceno-Rodrigues
- Laboratory of Cellular Biology (LIM 59 HCFMUSP), School of Medicine, University of São Paulo, Avenida Doutor Arnaldo 455, São Paulo, SP, 01246-903, Brazil
| | - João Bosco Pesquero
- Department of Biophysics, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, São Paulo, SP, 04023-062, Brazil
| | - Leonardo Martin
- Department of Biophysics, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, São Paulo, SP, 04023-062, Brazil
- Division of Medical Sciences, Laboratory of Transcriptional Regulation, Institute of Medical Biology of Polish, Academy of Sciences (IMB-PAS), Lodowa 106, Lodz, 93-232, Poland
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Flavio Aimbire
- Post-graduate Program in Translational Medicine, Department of Medicine, Federal University of São Paulo (UNIFESP), Rua Pedro De Toledo 720, 2º Andar, São Paulo, SP, 04039-002, Brazil
| | - Regiane Albertini
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos, SP, 11060-001, Brazil
| | - Sara Socorro Faria
- Post-graduate Programs in Humam Movement and Rehabilitation and in Pharmaceutical Sciences, Evangelical University of Goias (UniEvavngelica), Avenida Universitária Km3,5, Anápolis, GO, 75083-515, Brazil
| | - Rodolfo P Vieira
- Post-graduate Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Avenida Ana Costa 95, Santos, SP, 11060-001, Brazil.
- Post-graduate Programs in Humam Movement and Rehabilitation and in Pharmaceutical Sciences, Evangelical University of Goias (UniEvavngelica), Avenida Universitária Km3,5, Anápolis, GO, 75083-515, Brazil.
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), Rua Pedro Ernesto 240, São José dos Campos, SP, 12245-520, Brazil.
| |
Collapse
|
46
|
Gutiérrez-Vera C, García-Betancourt R, Palacios PA, Müller M, Montero DA, Verdugo C, Ortiz F, Simon F, Kalergis AM, González PA, Saavedra-Avila NA, Porcelli SA, Carreño LJ. Natural killer T cells in allergic asthma: implications for the development of novel immunotherapeutical strategies. Front Immunol 2024; 15:1364774. [PMID: 38629075 PMCID: PMC11018981 DOI: 10.3389/fimmu.2024.1364774] [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: 01/03/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024] Open
Abstract
Allergic asthma has emerged as a prevalent allergic disease worldwide, affecting most prominently both young individuals and lower-income populations in developing and developed countries. To devise effective and curative immunotherapy, it is crucial to comprehend the intricate nature of this condition, characterized by an immune response imbalance that favors a proinflammatory profile orchestrated by diverse subsets of immune cells. Although the involvement of Natural Killer T (NKT) cells in asthma pathology is frequently implied, their specific contributions to disease onset and progression remain incompletely understood. Given their remarkable ability to modulate the immune response through the rapid secretion of various cytokines, NKT cells represent a promising target for the development of effective immunotherapy against allergic asthma. This review provides a comprehensive summary of the current understanding of NKT cells in the context of allergic asthma, along with novel therapeutic approaches that leverage the functional response of these cells.
Collapse
Affiliation(s)
- Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Richard García-Betancourt
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - David A. Montero
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carlos Verdugo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisca Ortiz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Noemi A. Saavedra-Avila
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Steven A. Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
47
|
Sha J, Zhang M, Feng J, Shi T, Li N, Jie Z. Promyelocytic leukemia zinc finger controls type 2 immune responses in the lungs by regulating lineage commitment and the function of innate and adaptive immune cells. Int Immunopharmacol 2024; 130:111670. [PMID: 38373386 DOI: 10.1016/j.intimp.2024.111670] [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: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
Type 2 immune responses are critical for host defense, mediate allergy and Th2-high asthma. The transcription factor, promyelocytic leukemia zinc finger (PLZF), has emerged as a significant regulator of type 2 inflammation in the lung; however, its exact mechanism remains unclear. In this review, we summarized recent findings regarding the ability of PLZF to control the development and function of innate lymphoid cells (ILCs), iNKT cells, memory T cells, basophils, and other immune cells that drive type 2 responses. We discussed the important role of PLZF in the pathogenesis of Th2-high asthma. Collectively, prior studies have revealed the critical role of PLZF in the regulation of innate and adaptive immune cells involved in type 2 inflammation in the lung. Therefore, targeting PLZF signaling represents a promising therapeutic approach to suppress Th2-high asthma.
Collapse
Affiliation(s)
- Jiafeng Sha
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jingjing Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China.
| |
Collapse
|
48
|
Tang M, Da X, Xu Z, Zhao X, Zhou H. UHPLC/MS-based metabolomics of asthmatic mice reveals metabolic changes in group 2 innate lymphoid cells. Int Immunopharmacol 2024; 130:111775. [PMID: 38430805 DOI: 10.1016/j.intimp.2024.111775] [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: 12/07/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Helper Th2-type immune responses are essential in allergic airway diseases, including asthma and allergic rhinitis. Recent studies have indicated that group 2 innate lymphoid cells (ILC2s) play a crucial role in the occurrence and development of asthma. However, the metabolic profile of ILC2s and their regulatory mechanisms in asthma remain unclear. Therefore, we established two asthma mouse models: an ovalbumin (OVA)-induced asthma model and an IL-33-induced asthma model. We then used ultra-high-performance liquid chromatography/mass spectrometry (UHPLC/MS) to conduct high-throughput untargeted metabolic analysis of ILC2s in the lung tissues of the asthma models. The identified metabolites primarily consisted of lipids, lipid-like molecules, benzene, organic acids, derivatives, and organic oxidation compounds. Specifically, 34 differentially accumulated metabolites influenced the metabolic profiles of the control and OVA-induced asthma model groups. Moreover, the accumulation of 39 metabolites significantly differed between the Interleukin 33 (IL-33) and control groups. These differentially accumulated metabolites were mainly involved in pathways such as sphingolipid, oxidative phosphorylation, and fatty acid metabolism. This metabolomic study revealed, for the first time, the key metabolites and metabolic pathways of ILC2s, revealing new aspects of cellular metabolism in the context of airway inflammation. These findings not only contribute to unraveling the pathogenesis of asthma but also provide a crucial theoretical foundation for the future development of therapeutic strategies targeting ILC2s.
Collapse
Affiliation(s)
- Min Tang
- Department of Pediatrics, Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Xianzong Da
- Department of Pediatrics, Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Zhiwei Xu
- Department of Pediatrics, Bengbu Medical College, Bengbu, China
| | - Xiaoman Zhao
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Haoquan Zhou
- Department of Pediatrics, Provincial Hospital affiliated to Anhui Medical University, Hefei, China; Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
49
|
Karkout R, Gaudreault V, Labrie L, Aldossary H, Azalde Garcia N, Shan J, Fixman ED. Female-specific enhancement of eosinophil recruitment and activation in a type 2 innate inflammation model in the lung. Clin Exp Immunol 2024; 216:13-24. [PMID: 37607041 PMCID: PMC10929703 DOI: 10.1093/cei/uxad100] [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: 03/07/2023] [Revised: 07/19/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023] Open
Abstract
A sex disparity in asthma prevalence and severity exists in humans. Multiple studies have highlighted the role of innate cells in shaping the adaptive immune system in chronic asthma. To explore the sex bias in the eosinophilic response, we delivered IL-33 to the lungs of mice and delineated the kinetics by which the inflammatory response was induced. Our data demonstrate that females recruited more eosinophils capable of responding to IL-33. Eosinophil activation occurred selectively in the lung tissue and was enhanced in females at all time points. This increase was associated with increased ex vivo type 2 cytokine and chemokine production and female-specific expansion of group 2 innate lymphoid cells lacking expression of the killer-cell lectin-like receptor G1. Our findings suggest that the enhanced eosinophilic response in females is due, firstly, to a greater proportion of eosinophils recruited to the lungs in females that can respond to IL-33; and secondly, to an enhanced production of type 2 cytokines in females. Our data provide insight into the mechanisms that guide the female-specific enhancement of eosinophil activation in the mouse and form the basis to characterize these responses in human asthmatics.
Collapse
Affiliation(s)
- Rami Karkout
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Véronique Gaudreault
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Lydia Labrie
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Haya Aldossary
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Noelia Azalde Garcia
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Jichuan Shan
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Elizabeth D Fixman
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
50
|
Zhang M, Han Y. MicroRNAs in chronic pediatric diseases (Review). Exp Ther Med 2024; 27:100. [PMID: 38356668 PMCID: PMC10865459 DOI: 10.3892/etm.2024.12388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/15/2023] [Indexed: 02/16/2024] Open
Abstract
MicroRNAs are small non-coding RNAs with a length of 20-24 nucleotides. They bind to the 3'-untranslated region of target genes to induce the degradation of target mRNAs or inhibit their translation. Therefore, they are involved in the regulation of development, apoptosis, proliferation, differentiation and other biological processes (including hormone secretion, signaling and viral infections). Chronic diseases in children may be difficult to treat and are often associated with malnutrition resulting from a poor diet. Consequently, further complications, disease aggravation and increased treatment costs impose a burden on patients and their families. Existing evidence suggests that microRNAs are involved in various chronic non-neoplastic diseases in children. The present review discusses the roles of microRNAs in five major chronic diseases in children, namely, diabetes mellitus, congenital heart diseases, liver diseases, bronchial asthma and epilepsy, providing a theoretical basis for them to become therapeutic biomarkers in chronic pediatric diseases.
Collapse
Affiliation(s)
- Mingyao Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yanhua Han
- Department of Pediatrics, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|