Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
1.
Allergol Immunopathol (Madr) ; 51(3): 15-24, 2023.
Article in English | MEDLINE | ID: mdl-37169555

ABSTRACT

BACKGROUND: Airway remodeling, which contributes to the clinical course of childhood asthma, occurs due to airway inflammation and is featured by anomalous biological behaviors of airway smooth muscle cells (ASMCs). microRNA (miRNA) plays an essential role in the etiopathogenesis of asthma. OBJECTIVE: This research was aimed to characterize miR-506 in asthma and uncover potential regulatory machinery. MATERIAL AND METHODS: The asthmatic cell model was established by treating ASMCs with transforming growth factor-beta1 (TGF-ß1) and assessed by the levels of interleukin (IL)-1ß and interferon gamma (IFN-γ). Using real-time quantitative polymerase chain reaction, mRNA expression of miR-506 and polypyrimidine tract-binding protein 1 (PTBP1) was measured. Cell counting kit-8 and Transwell migration tests were used for estimating the capacity of ASMCs to proliferate and migrate. Luciferase reporter assay was used to corroborate whether miR-506 was directly bound to PTBP1. Expression of PTBP1, collagen I and III, and essential proteins of the wingless-related integration (Wnt)/ß-catenin pathway (ß-catenin, c-MYC and cyclin D1) was accomplished by Western blot analysis. The involvement of Wnt/ß-catenin signaling in asthma was confirmed by Wnt signaling pathway inhibitor (IWR-1). RESULTS: miR-506 was poorly expressed in asthmatic tissues and cell model. Functionally, overexpression of miR-506 reduced aberrant proliferation, migration, inflammation and collagen deposition of ASMCs triggered by TGF-ß1. Mechanically, miR-506 directly targeted the 3' untranslated region (3-UTR) of PTBP1 and had a negative regulation on PTBP1 expression. Moreover, overexpression of miR-506 suppressed the induction of Wnt/ß-catenin pathway. The administration of IWR-1 further validated negative correlation between miR-506 and the Wnt/ß-catenin pathway in asthma. CONCLUSION: Our data indicated that targeting miR-506/PTBP1/Wnt/ß-catenin axis might point in a helpful direction for treating asthma in children.


Subject(s)
Airway Remodeling , Asthma , MicroRNAs , Child , Humans , Airway Remodeling/genetics , Airway Remodeling/immunology , Asthma/genetics , Asthma/immunology , Asthma/pathology , beta Catenin/genetics , beta Catenin/metabolism , Cell Proliferation/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Transforming Growth Factor beta1/metabolism , Wnt Signaling Pathway
2.
Int J Mol Sci ; 24(7)2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37047587

ABSTRACT

Mast cells are responsible for IgE-dependent allergic responses, but they also produce various bioactive mediators and contribute to the pathogenesis of various cardiovascular diseases, including pulmonary hypertension (PH). The importance of lipid mediators in the pathogenesis of PH has become evident in recent years, as exemplified by prostaglandin I2, the most central therapeutic target in pulmonary arterial hypertension. New bioactive lipids other than eicosanoids have also been identified that are associated with the pathogenesis of PH. However, it remains largely unknown how mast cell-derived lipid mediators are involved in pulmonary vascular remodeling. Recently, it has been demonstrated that mast cells produce epoxidized n-3 fatty acid (n-3 epoxides) in a degranulation-independent manner, and that n-3 epoxides produced by mast cells regulate the abnormal activation of pulmonary fibroblasts and suppress the progression of pulmonary vascular remodeling. This review summarizes the role of mast cells and bioactive lipids in the pathogenesis of PH. In addition, we introduce the pathophysiological role and therapeutic potential of n-3 epoxides, a mast cell-derived novel lipid mediator, in the pulmonary vascular remodeling in PH. Further knowledge of mast cells and lipid mediators is expected to lead to the development of innovative therapies targeting pulmonary vascular remodeling.


Subject(s)
Airway Remodeling , Fatty Acids, Unsaturated , Hypertension, Pulmonary , Lysophospholipids , Mast Cells , Pulmonary Artery , Mast Cells/metabolism , Airway Remodeling/immunology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/pathology , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Lysophospholipids/biosynthesis , Fatty Acids, Unsaturated/biosynthesis , Humans , Animals
3.
Can Respir J ; 2022: 8437348, 2022.
Article in English | MEDLINE | ID: mdl-36091328

ABSTRACT

Introduction: Vascular smooth muscle cells (VSMCs) are highly involved in airway vascular remodeling in asthma. Objectives: This study aimed to investigate the mechanisms underlying the effects of a disintegrin and metalloproteinase-33 (ADAM33) gene on the migration capacity and inflammatory cytokine secretion of VSMCs. Methods: Human aortic smooth muscle cells (HASMCs) were transfected with lentiviral vectors carrying short hairpin RNA (shRNA) targeting ADAM33 or negative control vectors. The migration capacity of HASMCs was evaluated by a transwell assay. The levels of secreted inflammatory cytokines were measured using enzyme-linked immunosorbent assay (ELISA) kits. Reverse transcription-quantitative polymerase chain reaction and Western blot assays were performed to detect mRNA and protein expression levels. Results: Silencing of ADAM33 significantly inhibited the migration of HASMCs. The expression of tumor necrosis factor alpha (TNF-α) in the supernatant of HASMCs was decreased, while that of interferon gamma (IFN-γ) was increased after the transfection of shRNA targeting ADAM33. Insufficient ADAM33 expression also suppressed the expression levels of phosphatidylinositol 3-kinase (PI3K), phospho-protein kinase B (AKT), phospho-mammalian target of rapamycin (mTOR), Rho-associated protein kinases, phospho-forkhead box protein O1 (FOXO1), and cyclin D1, but it did not affect the levels of AKT, mTOR, or Rho. Conclusion: Silencing of the ADAM33 gene inhibited HASMC migration and regulated inflammatory cytokine secretion via targeting the PI3K/AKT/mTOR pathway and its downstream signaling. These data contribute to a better understanding of the regulatory mechanisms of airway vascular remodeling in asthma.


Subject(s)
ADAM Proteins , Airway Remodeling , Asthma , Gene Silencing , Muscle, Smooth, Vascular , Vascular Remodeling , ADAM Proteins/genetics , ADAM Proteins/immunology , Airway Remodeling/genetics , Airway Remodeling/immunology , Asthma/genetics , Asthma/immunology , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Gene Silencing/physiology , Humans , Muscle, Smooth, Vascular/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , RNA, Small Interfering/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Vascular Remodeling/genetics , Vascular Remodeling/immunology
4.
Int Arch Allergy Immunol ; 183(2): 142-152, 2022.
Article in English | MEDLINE | ID: mdl-34587616

ABSTRACT

INTRODUCTION: Inhalation of fungal allergens induces airway epithelial damage following airway inflammation and excessive mucus secretion, which can lead to severe asthma with fungal sensitization (SAFS). Comprehensive gene expression analysis in Alternaria-exposed mouse airways, a model of SAFS, has not been conducted. METHODS: BALB/c mice received intranasal administration of Alternaria extract or phosphate-buffered saline twice a week for 6 weeks. Lung sections and bronchoalveolar lavage fluid were obtained to assess airway inflammation. RNA-Seq in the central airway was performed, and gene ontology (GO) analysis and gene set enrichment analysis (GSEA) were conducted for pathway analyses. An in vitro experiment using human airway epithelial cell 16HBE14o- was performed to validate the RNA-Seq findings. RESULTS: Eosinophilic airway inflammation with mucus overproduction and airway remodeling was observed in mice exposed to Alternaria. RNA-Seq analysis revealed 403 upregulated and 108 downregulated genes in airways of Alternaria-exposed mice. In GO analysis, the functions of immunoglobulin (Ig) receptor binding, Ig production, inflammatory response, and T-cell activation were upregulated, while those of keratinization and defense response to other organisms were downregulated. GSEA revealed positive enrichment in T-cell receptor complex, immunological synapse, antigen binding, mast cell activation, and Ig receptor binding, and negative enrichment in keratinization and cornification in Alternaria-exposed mice relative to control. Alternaria exposure to 16HBE14o- cells validated the downregulation of epithelial keratinization-related genes, including SPRR1A, SPRR1B, and KRT6B. CONCLUSION: RNA-Seq analysis showed that Alternaria exposure induced inflammatory response and impaired defense mechanisms in mice airway epithelium, which might be therapeutic targets for SAFS.


Subject(s)
Allergens/immunology , Asthma/etiology , Fungi/immunology , RNA-Seq , Transcriptome , Airway Remodeling/immunology , Alternaria/immunology , Animals , Asthma/diagnosis , Asthma/metabolism , Bronchoalveolar Lavage Fluid/cytology , Computational Biology/methods , Disease Models, Animal , Eosinophils/pathology , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Immunization , Immunohistochemistry , Mice , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
5.
Environ Toxicol Pharmacol ; 89: 103782, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34883242

ABSTRACT

Simultaneous exposure to both BaP and house dust mites (HDM) has been shown to exacerbate pulmonary inflammation and hyperresponsiveness in a murine asthma model. The mechanistic insight into epigenetic inheritance for this effect, however, remains to be clarified. As such, in this study, we explore the molecular basis for the enhancement of asthma. Female BAL/C mice were intranasally administered HDM (25 µg in 25 µL saline) and/or BaP (10 µg/kg) every other day for 9 weeks. RNA sequencing and DNA methylation assessment were used to explore the underlying mechanism. Following simultaneous exposure to HDM and BaP, mice exhibited pulmonary inflammation and the transcript level of IL4i1b, muc4 and IL22ra2 that were associated with altered DNA methylation, suggesting that there may be an epigenetic basis for BaP-induced asthma exacerbation. Our data suggest that DNA methylation is a major epigenetic modification that accompanies airway remodeling associated with changes in the allergic mice.


Subject(s)
Airway Remodeling/drug effects , Benzo(a)pyrene/toxicity , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Airway Remodeling/immunology , Animals , Asthma/chemically induced , Asthma/immunology , Disease Models, Animal , Female , Inflammation/pathology , Mice, Inbred BALB C , Pyroglyphidae/immunology , Sequence Analysis, RNA
6.
Immunobiology ; 226(5): 152124, 2021 09.
Article in English | MEDLINE | ID: mdl-34333403

ABSTRACT

Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling as well as hyper-responsiveness. Thymic stromal lymphopoietin (TSLP), which is a crucial inflammatory cytokine in immune homeostasis, consists of two isoforms, the long isoform lfTSLP and short isoform sfTSLP. The lfTSLP promotes inflammation and plays a pivotal role in asthma pathogenesis, while sfTSLP had been reported to have anti-asthma effects. Experiments have shown that lfTSLP could induce autophagy in hepatocytes. It is unknown whether lfTSLP or sfTSLP could influence autophagy and affect the progression of asthma. Using house dust mite (HDM)-stimulated airway smooth muscle cells as an in vitro model and HDM-induced asthma mice as in vivo model, we found that lfTSLP could induce autophagy and remodeling, while sfTSLP has the reverse effect. Strikingly, sfTSLP treatment in vivo reversed HDM-mediated activation of inflammation and airway remodeling, partly determined by autophagy change. These findings may help us understand the function of TSLP isoforms in the pathogenesis of asthma, and they support the use of drugs targeting sfTSLP and TSLP for asthma treatment.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Cytokines/immunology , Allergens/immunology , Animals , Asthma/blood , Asthma/pathology , Autophagy , Bronchoalveolar Lavage Fluid/cytology , Cell Line , Cytokines/blood , Female , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Lung/pathology , Mice, Inbred C57BL , Myocytes, Smooth Muscle/immunology , Protein Isoforms/immunology , Pyroglyphidae/immunology
7.
Am J Physiol Lung Cell Mol Physiol ; 321(3): L533-L544, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34231388

ABSTRACT

Store-operated calcium entry (SOCE) is involved in the pathogenesis of airway inflammation and remodeling in asthma. Store-operated calcium entry-associated regulatory factor (SARAF) can downregulate SOCE. We sought to investigate the role of SARAF in the regulation of airway inflammation and remodeling in asthma mice models, as well as in the functional regulation of human airway smooth muscle cells (hASMCs). Balb/c mice were sensitized and challenged with ovalbumin to establish the asthma mice models. Mice were transfected with lentivirus, which expressed the SARAF gene + GFP (green fluorescence protein) or the negative control gene + GFP. Airway resistance was measured with the animal pulmonary function system. Airway inflammation and remodeling were evaluated via histological staining. In vitro cultured hASMCs were transfected with scrambled small interfering RNA (siRNA) or SARAF-specific siRNA, respectively. The proliferation, migration rate, hypertrophy, and SOCE activity of hASMCs were examined with Cell Counting Kit-8, wound healing test, bright field imaging, and Ca2+ fluorescence imaging, respectively. SARAF expression was measured by quantitative real-time PCR. Asthma mice models showed decreased SARAF mRNA expression in the lungs. SARAF overexpression attenuated airway inflammation, resistance, and also remodeling. Downregulation of SARAF expression with siRNA promoted the proliferation, migration, hypertrophy, and SOCE activity in hASMCs. SARAF plays a protective role against airway inflammation and remodeling in asthma mice models by blunting SOCE; SARAF may also be a functional regulating factor of hASMCs.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Calcium-Binding Proteins/immunology , Gene Expression Regulation/immunology , Lung/immunology , Membrane Proteins/immunology , Myocytes, Smooth Muscle/immunology , Airway Remodeling/drug effects , Airway Remodeling/genetics , Airway Resistance/drug effects , Airway Resistance/genetics , Airway Resistance/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Calcium-Binding Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Lung/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myocytes, Smooth Muscle/pathology
8.
J Allergy Clin Immunol ; 147(6): 1983-1995, 2021 06.
Article in English | MEDLINE | ID: mdl-34092351

ABSTRACT

The airway smooth muscle (ASM) cell plays a central role in the pathogenesis of asthma and constitutes an important target for treatment. These cells control muscle tone and thus regulate the opening of the airway lumen and air passage. Evidence indicates that ASM cells participate in the airway hyperresponsiveness as well as the inflammatory and remodeling processes observed in asthmatic subjects. Therapeutic approaches require a comprehensive understanding of the structure and function of the ASM in both the normal and disease states. This review updates current knowledge about ASM and its effects on airway narrowing, remodeling, and inflammation in asthma.


Subject(s)
Asthma/etiology , Asthma/metabolism , Disease Susceptibility , Muscle, Smooth/metabolism , Airway Remodeling/genetics , Airway Remodeling/immunology , Animals , Biomarkers , Bronchoconstriction/genetics , Bronchoconstriction/immunology , Gene Expression Regulation , Humans , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/metabolism
9.
Int Immunopharmacol ; 97: 107690, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33940323

ABSTRACT

It has recently been shown that expression levels of tissue factor (TF) are high in the serum and peripheral blood mononuclear cells of patients with asthma. However, whether TF impacts airway inflammation and remodelling in asthma remains unknown. The aim of this study was to investigate the effect of TF in asthma airway inflammation and remodelling using a house dust mite (HDM)-induced chronic asthma model and human bronchial epithelial (16HBE) cells. A chronic asthma model was constructed in BALB/c mice by the intranasal instillation of HDM. Mice were treated with short hairpin TF (shTF), and airway inflammation and remodelling features of asthma and epithelial-mesenchymal transition (EMT) were assessed. 16HBE cells were induced by transforming growth factor-ß1 (TGF-ß1) and HDM in the presence or absence of shTF; then, EMT markers and invasion and migration ability were determined. TF expression increased in the lung tissue and 16HBE cells when exposed to HDM. TF downregulation in the lung significantly reduced airway hyperresponsiveness, eosinophil inflammation, the EMT process, and levels of interleukin (IL)-4, IL-6, IL-13, and TGF-ß1 in bronchoalveolar lavage fluid of asthmatic mice. Moreover, TF downregulation inhibited migration and incursion and decreased the expression levels of fibronectin 1 and TGF-ß1, but increased the expression of E-cadherin in HDM- and TGF-ß1-stimulated 16HBE cells. These results demonstrated that TF promoted airway pathological features by enhancing the EMT of bronchial epithelial cells both in vitro and in mice with house dust mite-induced asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Dermatophagoides pteronyssinus/immunology , Thromboplastin/metabolism , Airway Remodeling/immunology , Animals , Asthma/pathology , Bronchi/cytology , Bronchi/immunology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/immunology , HEK293 Cells , Humans , Mice , Specific Pathogen-Free Organisms , Thromboplastin/genetics , Up-Regulation/immunology
10.
Acta Trop ; 220: 105934, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33895144

ABSTRACT

Allergic bronchial asthma is characterized by chronic inflammation of the respiratory airways mediated by T-helper 2 (Th2), Th17 and their cytokines. Although most asthmatic patients suffer from allergic airway remodeling (AAR), aggressive anti-allergic treatment failed to reverse it. The hygiene hypothesis illuminated the counter relationship between allergy and helminthic infections. The immune system is modulated by Trichinella spiralis (T. spiralis) infection to maintain homeostasis. Therefore, this work aimed to investigate the impact of chronic T. spiralis infection on induced AAR in C57BL/6 mice sensitized by house dust mites (HDM) allergens. Forty mice were divided into 3 groups: I (10 healthy mice), IΙ (15 HDM sensitized mice), and ΙΙI (15 T. spiralis chronically infected mice and sensitized with HDM allergens). The assessment aimed to evaluate the effects of regulatory CD4+CD25+FOXP3+ cells (Tregs) and their cytokines comparative to hypersensitivity mediated cytokines. Chronic T. spiralis infection effectively prevented the host's AAR. This result was evidenced by upregulated Tregs in blood by flow cytometric analysis and increased interleukin-10 (IL-10) levels in bronchoalveolar lavage (BAL) by Enzyme linked immunosorbent assay (ELISA) as well as improved lung histopathological changes. Also, serum HDM specific immunoglobulin E (IgE), BAL eosinophils, BAL IL-5 levels, and IL-17 gene expression in lung tissues were significantly reduced in T. spiralis chronically infected mice. In conclusion, the immune response in chronic T. spiralis infection could provide a promising mechanistic tool for protection against AAR, which paves the way for innovative preventive measures of other immunological disorders.


Subject(s)
Airway Remodeling/immunology , Pyroglyphidae/immunology , Trichinellosis/immunology , Allergens/immunology , Allergens/pharmacology , Animals , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Humans , Immunoglobulin E/blood , Inflammation/immunology , Interleukins/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Trichinella spiralis
11.
Int Immunopharmacol ; 97: 107637, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33895479

ABSTRACT

Asthma is characterized by airway remodeling. Glucocorticoid induced transcript 1 (GLCCI1) was reported to be associated with the development of asthma, while its exact mechanism is still not clear. In our study, ovalbumin (OVA) combined with aluminum hydroxide were used to establish asthmatic mouse model. ELISA assay was fulfilled to ensure the concentration of inflammatory factors in both bronchoalveolar lavage fluid and serum. The pathological changes and collagen deposition in lung tissues were analyzed using H&E staining and Masson staining, respectively. The expression of proteins was measured using western blot, and the expression of GLCCI1 mRNA was ensured by qRT-PCR. Here, we demonstrated that OVA-induced inflammation, lung structural remodeling and collagen deposition in asthmatic mice was notably improved by hydroprednisone treatment or GLCCI1 overexpressing. The expression of GLCCI1 was decreased, while IL-13, periostin and TGF-ß1 were increased in the lung tissue of asthmatic mice. Importantly, upregulation of GLCCI1 suppressed the expression of IL-13, periostin and TGF-ß1, phosphorylation of Smad2 and Smad3, and extracellular matrix (ECM) deposition-related proteins expression. IL-13-induced upregulation of periostin and TGF-ß1 expression, phosphorylation of Smad2 and Smad3, and ECM deposition in airway epithelial cells (AECs) was repressed by GLCCI1 increasing. Furthermore, our results showed that overexpression of GLCCI1 repressed the effect of IL-13 on AECs via inhibiting periostin expression. Overall, our data revealed that GLCCI1 limited the airway remodeling in mice with asthma through inhibiting IL-13/periostin/TGF-ß1 signaling pathway. Our data provided a novel target for asthma treatment.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Lung/pathology , Receptors, Glucocorticoid/metabolism , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/toxicity , Animals , Asthma/chemically induced , Asthma/drug therapy , Asthma/pathology , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Female , Humans , Interleukin-13/metabolism , Lung/drug effects , Lung/immunology , Mice , Ovalbumin/administration & dosage , Ovalbumin/immunology , Prednisone/administration & dosage , Receptors, Glucocorticoid/agonists , Signal Transduction/immunology , Transforming Growth Factor beta1/metabolism
12.
J Allergy Clin Immunol ; 148(2): 645-651.e11, 2021 08.
Article in English | MEDLINE | ID: mdl-33819511

ABSTRACT

BACKGROUND: Bronchial remodeling is a key feature of asthma that is already present in preschoolers with wheezing. Moreover, bronchial smooth muscle (BSM) remodeling at preschool age is predictive of asthma at school age. However, the mechanism responsible for BSM remodeling in preschoolers with wheezing remains totally unknown. In contrast, in adult asthma, BSM remodeling has been associated with an increase in BSM cell proliferation related to increased mitochondrial mass and biogenesis triggered by an altered calcium homeostasis. Indeed, BSM cell proliferation was decreased in vitro by the calcium channel blocker gallopamil. OBJECTIVE: Our aim was to investigate the mechanisms involved in BSM cell proliferation in preschoolers with severe wheezing, with special attention to the role of mitochondria and calcium signaling. METHODS: Bronchial tissue samples obtained from 12 preschool controls without wheezing and 10 preschoolers with severe wheezing were used to measure BSM mass and establish primary BSM cell cultures. BSM cell proliferation was assessed by manual counting and flow cytometry, ATP content was assessed by bioluminescence, mitochondrial respiration was assessed by using either the Seahorse or Oroboros technique, mitochondrial mass and biogenesis were assessed by immunoblotting, and calcium response to carbachol was assessed by confocal microscopy. The effect of gallopamil was also evaluated. RESULTS: BSM mass, cell proliferation, ATP content, mitochondrial respiration, mass and biogenesis, and calcium response were all increased in preschoolers with severe wheezing compared with in the controls. Gallopamil significantly decreased BSM mitochondrial biogenesis and mass, as well as cell proliferation. CONCLUSION: Mitochondria are key players in BSM cell proliferation in preschoolers with severe wheezing and could represent a potential target to treat BSM remodeling at an early stage of the disease.


Subject(s)
Airway Remodeling/immunology , Bronchi/immunology , Mitochondria, Muscle/immunology , Muscle, Smooth/immunology , Respiratory Sounds/immunology , Asthma/etiology , Asthma/immunology , Asthma/pathology , Bronchi/pathology , Calcium Signaling/drug effects , Calcium Signaling/immunology , Cells, Cultured , Child, Preschool , Female , Gallopamil/pharmacology , Humans , Infant , Male , Mitochondria, Muscle/pathology , Muscle, Smooth/pathology
13.
Int Immunopharmacol ; 94: 107488, 2021 May.
Article in English | MEDLINE | ID: mdl-33640857

ABSTRACT

MicroRNA-181b (miR-181b) has been well noted with anti-inflammatory properties in several pathological conditions. It has also been suggested to be downregulated in patients with asthma. In this study, we explored the function of miR-181b in airway remodeling in asthmatic mice and the molecular mechanism. A mouse model with asthma was induced by ovalbumin (OVA) challenge, and miR-181b was found to be downregulated in lung tissues in the OVA-challenged mice. Overexpression of miR-181b was introduced in mice, after which the respiratory resistance, inflammatory infiltration, mucus production, and epithelial-mesenchymal transition (EMT) and fibrosis in mouse airway tissues were decreased. The integrated bioinformatics analysis suggested long non-coding RNA (lncRNA) TUG1 as a sponge for miR-181b. miR-181 directly targeted high mobility group box 1 (HMGB1) mRNA. HMGB1 was suggested to enhance activation of the nuclear factor kappa B (NF-κB) signaling. Further upregulation of lncRNA TUG1 blocked the protective functions of miR-181b in asthmatic mice. To conclude, this study evidenced that lncRNA TUG1 reinforces HMGB1 expression through sequestering microRNA-181b, which activates the NF-κB signaling pathway and promotes airway remodeling in asthmatic mice. This study may provide novel ideas in asthma management.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , HMGB1 Protein/immunology , MicroRNAs/immunology , RNA, Long Noncoding/immunology , Airway Remodeling/genetics , Allergens , Animals , Asthma/genetics , Asthma/pathology , Cells, Cultured , Disease Models, Animal , Female , HMGB1 Protein/genetics , Lung/pathology , Mice, Inbred BALB C , Mucus/immunology , NF-kappa B/immunology , Ovalbumin , Signal Transduction
14.
J Immunol Res ; 2021: 6645072, 2021.
Article in English | MEDLINE | ID: mdl-33628848

ABSTRACT

Asthma is a chronic airway disorder associated with aberrant inflammatory and remodeling responses. Angiogenesis and associated vascular remodeling are one of the pathological hallmarks of asthma. The mechanisms underlying angiogenesis in asthmatic airways and its clinical relevance represent a relatively nascent field in asthma when compared to other airway remodeling features. Matrix metalloproteinases (MMPs) are proteases that play an important role in both physiological and pathological conditions. In addition to facilitating extracellular matrix turnover, these proteolytic enzymes cleave bioactive molecules, thereby regulating cell signaling. MMPs have been implicated in the pathogenesis of asthma by interacting with both the airway inflammatory cells and the resident structural cells. MMPs also cover a broad range of angiogenic functions, from the degradation of the vascular basement membrane and extracellular matrix remodeling to the release of a variety of angiogenic mediators and growth factors. This review focuses on the contribution of MMPs and the regulatory role exerted by them in angiogenesis and vascular remodeling in asthma as well as addresses their potential as therapeutic targets in ameliorating angiogenesis in asthma.


Subject(s)
Asthma/metabolism , Asthma/pathology , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic/metabolism , Airway Remodeling/genetics , Airway Remodeling/immunology , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Asthma/etiology , Asthma/therapy , Biomarkers , Disease Susceptibility , Extracellular Matrix/metabolism , Humans , Immunomodulation , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors/therapeutic use , Matrix Metalloproteinases/genetics , Molecular Targeted Therapy , Neovascularization, Pathologic/genetics
15.
Allergol Immunopathol (Madr) ; 49(2): 191-207, 2021.
Article in English | MEDLINE | ID: mdl-33641309

ABSTRACT

Eosinophils are the major inflammatory cells which play a crucial role in the development of allergic and non-allergic asthma phenotypes. Eosinophilic asthma is the most heterogeneous phenotype where activated eosinophils are reported to be significantly associated with asthma severity. Activated eosinophils display an array of cell adhesion molecules that not only act as an activation marker, suitable for assessing severity, but also secrete several tissue factors, cytokines and chemokines which modulate the clinical severity. Eosinophil activations are also strictly associated with activation of other hetero cellular populations like neutrophils, macrophages, mast cells, and platelets which culminate in the onset and progression of abnormal phenotypes such as bronchoconstriction, allergic response, fibrosis instigated by tissue inflammation, epithelial injury, and oxidative stress. During the activated state, eosinophils release several potent toxic signaling molecules such as major basic proteins, eosinophil peroxidase, eosinophil cationic protein (ECP), and lipid mediators, rendering tissue damage and subsequently leading to allergic manifestation. The tissue mediators render a more complex manifestation of a severe phenotype by activating prominent signaling cross-talk. Here, in the current review with the help of search engines of PubMed, Medline, etc, we have tried to shed light and explore some of the potent determinants regulating eosinophil activation leading to asthma phenotype.


Subject(s)
Asthma/immunology , Cell Communication/immunology , Eosinophils/immunology , Airway Remodeling/immunology , Animals , Asthma/blood , Asthma/diagnosis , Asthma/pathology , Blood Platelets/immunology , Bronchi/immunology , Bronchi/pathology , Bronchoconstriction/immunology , Disease Models, Animal , Eosinophils/metabolism , Fibrosis , Humans , Leukocyte Count , Macrophages/immunology , Mast Cells/immunology , Mice , Neutrophils/immunology , Oxidative Stress/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Severity of Illness Index
16.
Int J Mol Sci ; 22(2)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33477617

ABSTRACT

Chronic rhinosinusitis (CRS) is a chronic inflammatory condition of the nasal and paranasal sinus mucosa that affects up to 10% of the population worldwide. CRS is the most representative disease of the upper respiratory tract where airway remodeling occurs, including epithelial damage, thickening of the basement membrane, fibrosis, goblet cell hyperplasia, subepithelial edema, and osteitis. CRS is divided into two phenotypes according to the presence or absence of nasal polyps: CRS with nasal polyp (CRSwNP) and CRS without nasal polyps (CRSsNP). Based on the underlying pathophysiologic mechanism, CRS is also classified as eosinophilic CRS and non-eosinophilic CRS, owing to Type 2 T helper (Th2)-based inflammation and Type 1 T helper (Th1)/Type 17 T helper (Th17) skewed immune response, respectively. Differences in tissue remodeling in CRS are suggested to be based on the clinical phenotype and endotypes; this is because fibrosis is prominent in CRSsNP, whereas edematous changes occur in CRSwNP, especially in the eosinophilic type. This review aims to summarize the latest information on the different mechanisms of airway remodeling in CRS according to distinct endotypes.


Subject(s)
Airway Remodeling/genetics , Inflammation/immunology , Nasal Polyps/immunology , Rhinitis/immunology , Airway Remodeling/immunology , Airway Remodeling/physiology , Chronic Disease/epidemiology , Fibrosis , Goblet Cells/classification , Goblet Cells/immunology , Humans , Inflammation/pathology , Nasal Polyps/genetics , Nasal Polyps/pathology , Rhinitis/genetics , Rhinitis/pathology , Sinusitis , Th1 Cells/classification , Th1 Cells/immunology , Th17 Cells/classification , Th17 Cells/immunology , Th2 Cells/classification , Th2 Cells/immunology
17.
EBioMedicine ; 64: 103186, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33478923

ABSTRACT

BACKGROUND: Substantial recent advances in the comprehension of the molecular and cellular mechanisms behind asthma have evidenced the importance of the lung immune environment for disease outcome, making modulation of local immune responses an attractive therapeutic target against this pathology. Live attenuated mycobacteria, such as the tuberculosis vaccine BCG, have been classically linked with a type 1 response, and proposed as possible modulators of the type 2 response usually associated with asthma. METHODS: In this study we used different acute and chronic murine models of asthma to investigate the therapeutic efficacy of intranasal delivery of the live tuberculosis vaccines BCG and MTBVAC by regulating the lung immune environment associated with airway hyperresponsiveness (AHR). FINDINGS: Intranasal administration of BCG, or the novel tuberculosis vaccine candidate MTBVAC, abrogated AHR-associated hallmarks, including eosinophilia and lung remodeling. This correlated with the re-polarization of allergen-induced M2 macrophages towards an M1 phenotype, as well as with the induction of a strong allergen-specific Th1 response. Importantly, vaccine treatment was effective in a scenario of established chronic asthma where a strong eosinophil infiltration was already present prior to immunization. We finally compared the nebulization efficiency of clinical formulations of MTBVAC and BCG using a standard commercial nebulizer for potential aerosol application. INTERPRETATION: Our results demonstrate that pulmonary live tuberculosis vaccines efficiently revert established asthma in mice. These data support the further exploration of this approach as potential therapy against asthma. FUNDING: Spanish Ministry of Science [grant numbers: BIO2014-5258P, RTI2018-097625-B-I00], Instituto de Salud Carlos III, Gobierno de Aragón/Fondo Social Europeo, University of Zaragoza [grant number: JIUZ-2018-BIO-01].


Subject(s)
Asthma/immunology , Asthma/therapy , Tuberculosis Vaccines/therapeutic use , Vaccines, Attenuated/therapeutic use , Administration, Intranasal , Airway Remodeling/immunology , Allergens/immunology , Animals , BCG Vaccine , Biomarkers , Cellular Microenvironment/immunology , Cytokines/metabolism , Disease Models, Animal , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Female , Immunization , Mice , Ovalbumin/immunology , Tuberculosis Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage
18.
Int Arch Allergy Immunol ; 182(4): 277-291, 2021.
Article in English | MEDLINE | ID: mdl-33147596

ABSTRACT

INTRODUCTION: The use of probiotics has been broadly popularized due to positive effects in the attenuation of aberrant immune responses such as asthma. Allergic asthma is a chronic respiratory disease characterized by airway inflammation and remodelling. OBJECTIVE: This study was aimed to evaluate the effect of oral administration of Lactococcus lactis NZ9000 on asthmatic airway inflammation and lung tissue remodelling in rats and its relation to the maintenance of an adequate intestinal barrier. METHODS: Wistar rats were ovalbumin (OVA) sensitized and challenged and orally treated with L. lactis. Lung inflammatory infiltrates and cytokines were measured, and remodelling was evaluated. Serum OVA-specific immunoglobulin (Ig) E levels were assessed. We also evaluated changes on intestinal environment and on systemic immune response. RESULTS: L. lactis diminished the infiltration of proinflammatory leucocytes, mainly eosinophils, in the bronchoalveolar compartment, decreased lung IL-4 and IL-5 expression, and reduced the level of serum allergen-specific IgE. Furthermore, L. lactis prevented eosinophil influx, collagen deposition, and goblet cell hyperplasia in lung tissue. In the intestine, L. lactis-treated asthmatic rats increased Peyer's patch and goblet cell quantity and mRNA expression of IgA, MUC-2, and claudin. Additionally, intestinal morphological alterations were normalized by L. lactis administration. Splenocyte proliferative response to OVA was abolished, and serum levels of transforming growth factor (TGF)-ß were increased by L. lactis treatment. CONCLUSIONS: These findings suggest that L. lactis is a potential candidate for asthma prevention, and the effect is mediated by the improvement of intestinal barrier function and systemic TGF-ß production.


Subject(s)
Airway Remodeling , Asthma/metabolism , Asthma/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lactococcus lactis/physiology , Probiotics/administration & dosage , Transforming Growth Factor beta/biosynthesis , Airway Remodeling/immunology , Animals , Asthma/etiology , Asthma/prevention & control , Cytokines/metabolism , Disease Models, Animal , Immunoglobulin E/blood , Immunoglobulin E/immunology , Inflammation Mediators/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Ovalbumin/immunology , Rats
19.
Front Immunol ; 12: 770305, 2021.
Article in English | MEDLINE | ID: mdl-35069543

ABSTRACT

HAS2 is a member of the gene family encoding the hyaluronan synthase 2, which can generate high-molecular-weight hyaluronan (HMW-HA). Our previous study identified HAS2 as a candidate gene for increased susceptibility to adult asthma. However, whether HAS2 dysfunction affects airway remodeling and steroid insensitivity is still limited. Therefore, this study aimed to clarify the Has2 dysfunction, triggering severe airway remodeling and steroid insensitivity in a murine model of asthma. Has2 heterozygous-deficient (Has2+/-) mice and their wild-type littermates have been evaluated in a model of chronic ovalbumin (OVA) sensitization and challenge. Mice present a higher sensitivity to OVA and higher IL-17 release as well as eosinophilic infiltration. RNA sequencing demonstrated the downregulation of EIF2 signaling pathways, TGF-ß signaling pathways, and heat shock proteins with Th17 bias in Has2+/--OVA mice. The combined treatment with anti-IL-17A antibody and dexamethasone reduces steroid insensitivity in Has2+/--OVA mice. Has2 attenuation worsens eosinophilic airway inflammation, airway remodeling, and steroid insensitivity. These data highlight that HAS2 and HMW-HA are important for controlling intractable eosinophilic airway inflammation and remodeling and could potentially be exploited for their therapeutic benefits in patients with asthma.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Drug Resistance/immunology , Hyaluronan Synthases/immunology , Airway Remodeling/drug effects , Airway Remodeling/genetics , Animals , Asthma/chemically induced , Asthma/genetics , Drug Resistance/genetics , Hyaluronan Synthases/genetics , Mice , Mice, Knockout , Ovalbumin/toxicity , Steroids/pharmacology
20.
J Allergy Clin Immunol ; 147(6): 2281-2294.e7, 2021 06.
Article in English | MEDLINE | ID: mdl-33249168

ABSTRACT

BACKGROUND: Asthma is a widespread, multifactorial chronic airway disease. The influence of regulatory B cells on airway hyperreactivity (AHR) and remodeling in asthma is poorly understood. OBJECTIVE: Our aim was to analyze the role of B cells in a house dust mite (HDM)-based murine asthma model. METHODS: The influence of B cells on lung function, tissue remodeling, and the immune response were analyzed by using wild-type and B-cell-deficient (µMT) mice and transfer of IL-10-proficient and IL-10-deficient B cells to µMT mice. RESULTS: After HDM-sensitization, both wild-type and µMT mice developed AHR, but the AHR was significantly stronger in µMT mice, as confirmed by 2 independent techniques: invasive lung function measurement in vivo and examination of precision-cut lung slices ex vivo. Moreover, airway remodeling was significantly increased in allergic µMT mice, as shown by enhanced collagen deposition in the airways, whereas the numbers of FoxP3+ and FoxP3- IL-10-secreting regulatory T cells were reduced. Adoptive transfer of IL-10-proficient but not IL-10-deficient B cells into µMT mice before HDM-sensitization attenuated AHR and lung remodeling. In contrast, FoxP3+ regulatory T cells were equally upregulated by transfer of IL-10-proficient and IL-10-deficient B cells. CONCLUSION: Our data in a murine asthma model illustrate a central role of regulatory B cells in the control of lung function and airway remodeling and may support future concepts for B-cell-targeted prevention and treatment strategies for allergic asthma.


Subject(s)
Airway Remodeling/immunology , Asthma/etiology , Asthma/metabolism , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Allergens/immunology , Animals , Asthma/pathology , Biomarkers , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Lymphocyte Activation , Mice , Pyroglyphidae/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...