Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.982
Filter
1.
Cell Mol Biol (Noisy-le-grand) ; 70(8): 170-174, 2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39262245

ABSTRACT

Asthma is a chronic airway inflammatory disease. The excessive proliferation of airway smooth muscle cells (ASMCs) is associated with airway remodeling. Ze-Qi-Tang (ZQT) is a popular traditional Chinese medicine preparation and has been confirmed to have therapeutic effects on lung diseases. This study is aimed to probe the biological function of ZQT in asthma. RT-qPCR and ELISA were utilized for testing the mRNA levels and concentrations of pro-inflammatory factors. Colony formation and transwell assay were applied to test cell viability and migration. The mouse model with asthma was established by ovalbumin (OVA) induction. Western blot was utilized for detecting the activation of PI3K/AKT/NF-κB pathway. We found that the concentrations of proinflammatory factors in cells induced by PDGF-BB could been suppressed by ZQT. ZQT-H treatment notably repressed cell viability and proliferation. Furthermore, we proved the suppressive effect of ZQT on airway inflammation in asthma mice. Additionally, we discovered that ZQT could suppress the PI3K/AKT/NF-κB pathway in PDGF-BB-induced ASMCs. To sum up, ZQT reduced airway inflammation and remodeling in mice with asthma via inactivating PI3K/AKT/NF-κB pathway.


Subject(s)
Asthma , Cell Proliferation , Drugs, Chinese Herbal , Inflammation , NF-kappa B , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Proto-Oncogene Proteins c-akt/metabolism , NF-kappa B/metabolism , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Mice , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Cell Proliferation/drug effects , Medicine, Chinese Traditional/methods , Mice, Inbred BALB C , Disease Models, Animal , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Cell Survival/drug effects , Becaplermin/metabolism , Cell Movement/drug effects , Ovalbumin
2.
Int J Mol Sci ; 25(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39273478

ABSTRACT

The prevalence of obesity-induced asthma increases in women after menopause. We hypothesized that the increase in obese asthma in middle-aged women results from estrogen loss. In particular, we focused on the acute action of estrogen through the G protein-coupled estrogen receptor 1 (GPER), previously known as GPR30. We investigated whether GPER activation ameliorates obesity-induced asthma with a high-fat diet (HFD) using G-1, the GPER agonist, and G-36, the GPER antagonist. Administration of G-1 (0.5 mg/kg) suppressed HFD-induced airway hypersensitivity (AHR), and increased immune cell infiltration, whereas G-36 co-treatment blocked it. Histological analysis showed that G-1 treatment inhibited HFD-induced inflammation, fibrosis, and mucus hypersecretion in a GPER-dependent manner. G-1 inhibited the HFD-induced rise in the mRNA levels of pro-inflammatory cytokines in the gonadal white adipose tissue and lungs, whereas G-36 co-treatment reversed this effect. G-1 increased anti-inflammatory M2 macrophages and inhibited the HFD-induced rise in pro-inflammatory M1 macrophages in the lungs. In addition, G-1 treatment reversed the HFD-induced increase in leptin expression and decrease in adiponectin expression in the lungs and gonadal white adipose tissue. The results suggest that activation of GPER could be a therapeutic option for obesity-induced asthma.


Subject(s)
Asthma , Diet, High-Fat , Macrophages , Obesity , Receptors, Estrogen , Receptors, G-Protein-Coupled , Animals , Receptors, G-Protein-Coupled/metabolism , Obesity/complications , Obesity/metabolism , Obesity/drug therapy , Asthma/metabolism , Asthma/drug therapy , Macrophages/metabolism , Macrophages/drug effects , Mice , Receptors, Estrogen/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Female , Quinolines/pharmacology , Lung/pathology , Lung/metabolism , Lung/drug effects , Cytokines/metabolism
3.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273095

ABSTRACT

Respiratory diseases, including chronic obstructive pulmonary disease (COPD), asthma, lung cancer, and coronavirus pneumonia, present a major global health challenge. Current diagnostic and therapeutic options for these diseases are limited, necessitating the urgent development of novel biomarkers and therapeutic strategies. In recent years, microRNAs (miRNAs) within extracellular vesicles (EVs) have received considerable attention due to their crucial role in intercellular communication and disease progression. EVs are membrane-bound structures released by cells into the extracellular environment, encapsulating a variety of biomolecules such as DNA, RNA, lipids, and proteins. Specifically, miRNAs within EVs, known as EV-miRNAs, facilitate intercellular communication by regulating gene expression. The expression levels of these miRNAs can reflect distinct disease states and significantly influence immune cell function, chronic airway inflammation, airway remodeling, cell proliferation, angiogenesis, epithelial-mesenchymal transition, and other pathological processes. Consequently, EV-miRNAs have a profound impact on the onset, progression, and therapeutic responses of respiratory diseases, with great potential for disease management. Synthesizing the current understanding of EV-miRNAs in respiratory diseases such as COPD, asthma, lung cancer, and novel coronavirus pneumonia, this review aims to explore the potential of EV-miRNAs as biomarkers and therapeutic targets and examine their prospects in the diagnosis and treatment of these respiratory diseases.


Subject(s)
Biomarkers , COVID-19 , Extracellular Vesicles , MicroRNAs , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , COVID-19/genetics , Asthma/genetics , Asthma/metabolism , Asthma/therapy , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals , SARS-CoV-2
4.
Zhongguo Zhong Yao Za Zhi ; 49(15): 4148-4157, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39307747

ABSTRACT

This study aims to explore the active components and mechanism of Wuhu Decoction in treating respiratory syncytial virus(RSV)-induced asthma. Ultra-high performance liquid chromatography coupled with high-resolution mass spectrometry was used to determine the components of Wuhu Decoction in the blood. By utilizing databases, Kyoto Encyclopedia of Genes and Genomes(KEGG) enrichment analysis and Gene Ontology(GO) functional analysis were conducted to investigate the targets of the components of Wuhu Decoction in asthma. Furthermore, the information on target proteins, and metabolite-target-pathway was imported into the STRING database to construct a network interaction diagram to identify the core components and key pathways of Wuhu Decoction. In the in vivo experiment, an asthma model was established using RSV combined with ovalbumin(OVA) in mice. The intervention effect of Wuhu Decoction on RSV-induced asthma in mice was validated through lung function tests, hematoxylin-eosin(HE) staining, enzyme-linked immunosorbent assay(ELISA), Western blot, and immunohistochemistry. The results showed that the main components of Wuhu Decoction in the blood were flavonoids, phenylpropanoids, lignans, and terpenoids. The core components of Wuhu Decoction in treating pediatric asthma included(-)-epigallocatechin, kaempferol, isoliquiritigenin, diosmetin, betulinic acid, ursolic acid, daphnetin, aescin. The main pathways targeted by Wuhu Decoction were calcium signaling pathway, neuroactive ligand-receptor interaction, NOD-like receptor signaling pathway, T cell receptor signaling pathway, and Toll-like receptor signaling pathway. The results of in vivo experiments demonstrated that Wuhu Decoction could improve lung function indicators, down-regulate levels of interleukin-6(IL-6), interleukin-17(IL-17), and tumor necrosis factor-alpha(TNF-α), and reduce the expression of proteins such as NOD-like receptor pyrin domain-containing 3(NLRP3), mitogen-activated protein kinase 1(MAPK1), mitogen-activated protein kinase 14(MAPK14), and nuclear factor kappaB subunit 1(NFKB1) in lung tissue, thereby alleviating neutrophilic inflammation and pulmonary congestion. These findings indicate that Wuhu Decoction intervenes in virus-induced asthma through a synergistic effect on multiple components, targets, and pathways, and it can inhibit the activation of the NOD-like receptor signaling pathway, thereby alleviating airway inflammation and injury in asthmatic mice.


Subject(s)
Asthma , Drugs, Chinese Herbal , Mice, Inbred BALB C , Network Pharmacology , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Asthma/drug therapy , Asthma/metabolism , Mice , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Humans , Female , Lung/drug effects , Lung/metabolism
5.
J Biochem Mol Toxicol ; 38(10): e23836, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39308040

ABSTRACT

This study examines the anti-inflammatory activity of cynaropicrin against lipopolysaccharide (LPS) in vitro and ovalbumin (OVA)-challenged asthma in mice. Cynaropicrin's antimicrobial effects were tested on Escherichia coli (E. coli) and Streptococcus pyogenes (S. pyogenes) using the disc diffusion technique. Cytotoxicity was assessed with an (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) assay. The anti-inflammatory property was evaluated in LPS-induced RAW264.7 cells, while OVA-challenged asthmatic mice were treated with 10 mg/kg of cynaropicrin. Key inflammatory and antioxidant markers were quantified, and lung histology was examined to confirm therapeutic roles. The antimicrobial studies proved that cynaropicrin effectively inhibited the growth of E. coli and S. pyogenes. Cynaropicrin displayed no cytotoxicity on RAW264.7 cells. Furthermore, it significantly inhibited inflammatory cytokine synthesis upon LPS induction. Cynaropicrin treatment decreased the inflammatory cell counts and also suppressed specific allergic markers in OVA-challenged mice. It also decreased nitric oxide and myeloperoxidase levels and reduced pulmonary edema. Cynaropicrin increased antioxidant levels and decreased proinflammatory cytokines in the asthmatic mice. Lung histological examination confirms the ameliorative potency of cynaropicrin against OVA-induced asthmatic pulmonary inflammation in mice. Our findings suggest cynaropicrin possesses significant ameliorative potency against allergen-induced pulmonary inflammation.


Subject(s)
Asthma , Cytokines , Lipopolysaccharides , Ovalbumin , Animals , Mice , Asthma/drug therapy , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Lipopolysaccharides/toxicity , RAW 264.7 Cells , Cytokines/metabolism , Sesquiterpenes/pharmacology , Mice, Inbred BALB C , Escherichia coli , Streptococcus pyogenes , Anti-Inflammatory Agents/pharmacology , Male , Female , Lactones
6.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273372

ABSTRACT

The mechanisms underlying severe allergic asthma are complex and unknown, meaning it is a challenge to provide the most appropriate treatment. This study aimed to identify novel biomarkers for stratifying allergic asthmatic patients according to severity, and to uncover the biological mechanisms that lead to the development of the severe uncontrolled phenotype. By using miRNA PCR panels, we analyzed the expression of 752 miRNAs in serum samples from control subjects (n = 15) and mild (n = 11) and severe uncontrolled (n = 10) allergic asthmatic patients. We identified 40 differentially expressed miRNAs between severe uncontrolled and mild allergic asthmatic patients. Functional enrichment analysis revealed signatures related to inflammation, angiogenesis, lipid metabolism and mRNA regulation. A random forest classifier trained with DE miRNAs achieved a high accuracy of 97% for severe uncontrolled patient stratification. Validation of the identified biomarkers was performed on a subset of allergic asthmatic patients from the CAMP cohort at Brigham and Women's Hospital, Harvard Medical School. Four of these miRNAs (hsa-miR-99b-5p, hsa-miR-451a, hsa-miR-326 and hsa-miR-505-3p) were validated, pointing towards their potential as biomarkers for stratifying allergic asthmatic patients by severity and providing insights into severe uncontrolled asthma molecular pathways.


Subject(s)
Asthma , Biomarkers , Inflammation , Lipid Metabolism , MicroRNAs , Severity of Illness Index , Humans , Asthma/genetics , Asthma/blood , Asthma/metabolism , MicroRNAs/genetics , MicroRNAs/blood , Female , Male , Lipid Metabolism/genetics , Adult , Biomarkers/blood , Inflammation/genetics , Inflammation/blood , Inflammation/metabolism , Middle Aged , Gene Expression Profiling , Gene Expression Regulation
7.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273399

ABSTRACT

Type 2 airway inflammation (T2AI), driven by type 2 innate lymphoid and CD4+ T helper 2 cells, leads to various diseases and conditions, such as chronic rhinosinusitis with nasal polyps, allergic rhinitis, and asthma. Emerging evidence suggests the involvement of extracellular vesicles (EVs) in these diseases. In this review, we describe the immunological T2AI pathogenic mechanisms, outline EV characteristics, and highlight their applications in the diagnosis and treatment of T2AI. An extensive literature search was conducted using appropriate strategies to identify relevant articles from various online databases. EVs in various biological samples showed disease-specific characteristics for chronic rhinosinusitis with nasal polyps, allergic rhinitis, and asthma, with some demonstrating therapeutic effects against these conditions. However, most studies have been limited to in vitro and animal models, highlighting the need for further clinical research on the diagnostic and therapeutic applications of EVs.


Subject(s)
Extracellular Vesicles , Th2 Cells , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Humans , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Asthma/therapy , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Sinusitis/immunology , Sinusitis/metabolism , Sinusitis/pathology , Sinusitis/therapy , Rhinitis, Allergic/immunology , Rhinitis, Allergic/metabolism , Rhinitis, Allergic/therapy , Nasal Polyps/immunology , Nasal Polyps/therapy , Nasal Polyps/metabolism , Nasal Polyps/pathology , Rhinitis/immunology , Rhinitis/therapy , Rhinitis/metabolism , Rhinitis/pathology
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(7): 577-584, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39179401

ABSTRACT

Objective To explore the regulatory mechanism of interleukin-33 (IL-33) on the inflammatory response in asthmatic mice. Methods Using 10 µg/mL of lipopolysaccharide (LPS) to establish a cellular inflammation model of mouse bone marrow mesenchymal stem cells (BMMSCs), the cells were divided into three groups: small interfering RNA of IL-33(si-IL-33) group, IL-33 overexpression (IL-33-OE) group, and model group, based on the transfection status of si-IL-33 plasmid and IL-33-OE plasmid. Real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) was used to detect mRNA expression of IL-33, nucleotide binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), IL-1ß and IL-6. Fluo-3 AM was used to measure calcium ion content, and JC-1 mitochondrial membrane potential detection kit was performed to detect mitochondrial membrane potential changes. An asthma mouse model was established by intraperitoneal injection of sensitizers and activators. According to different treatment plans, the asthmatic mice were divided into si-IL-33 group, IL-33-OE group and model group, with 5 mice in each group. ELISA was used to detect the levels of IL-1ß, IL-6 and NLRP3 in mouse serum, while HE staining and Masson staining were performed to assess lung tissue lesions. Results Compared with the model group, the mRNA expression levels of IL-33, NLRP3, IL-1ß and IL-6 in the si-IL-33 group were all reduced, while those in the IL-33-OE group were increased. The calcium ion fluorescence was decreased in BMMSCs, while it was increased in the IL-33-OE group. In the si-IL-33 group, JC-1 existed in a polymer form in mitochondria, showing bright red fluorescence and weak green fluorescence, indicating stable mitochondria and normal mitochondrial function. After treating with IL-33-OE plasmid to reduce the mitochondrial membrane potentia, JC-1 cannot exist in polymer form within the mitochondrial matrix. At this point, the red fluorescence intensity inside the mitochondria decreases significantly, while the green fluorescence in the cytoplasm increases significantly. The levels of IL-1ß, IL-6, and NLRP3 in the serum of mice in the si-IL-33 group were significantly lower, while those in the IL-33-OE group were significantly higher. In the si-IL-33 group, almost no inflammatory cell infiltration was observed, indicating a relief of inflammation and normal epithelial cell morphology. Additionally, the fibrotic portion of the lung tissue in the si-IL-33 group tended to be normal. The total wall area of bronchus (WAt)/basement membrane perimeter (Pbm) and wall area of bronchial smooth muscle (WAm)/Pbm were reduced in the si-IL-33 group compared with the model group, while they were increased in the IL-33-OE group. Conclusion Knockdown of IL-33 inhibits the inflammatory response in asthmatic mice by downregulating the expression of NLRP3, IL-1ß and IL-6.


Subject(s)
Asthma , Interleukin-33 , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Interleukin-33/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Asthma/therapy , Mice , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Inflammation/genetics , Inflammation/metabolism , Female , Membrane Potential, Mitochondrial , Interleukin-6/genetics , Interleukin-6/metabolism , Gene Knockdown Techniques , Mesenchymal Stem Cells/metabolism
9.
J Innate Immun ; 16(1): 397-412, 2024.
Article in English | MEDLINE | ID: mdl-39134014

ABSTRACT

INTRODUCTION: MDM2 is known as the primary negative regulator of p53, and MDM2 promotes lung cancer fibrosis and lung injury through p53-dependent and p53-independent pathways. However, the mechanism by which MDM2 influences the pathogenesis of asthma is unknown. In this study, we investigated the function of MDM2 in lung epithelial cells in type 2 lung inflammation. METHODS: We used type II alveolar epithelial cell-specific heterozygous knockout of Mdm2 mice to validate its function. Then papain-induced asthma model was established, and changes in inflammation were observed by measuring immunohistochemistry and flow cytometry analysis. RESULTS: In this study, we knockdown the mouse Mdm2 gene in type 2 alveolar epithelial cells. We demonstrated that heterozygous Mdm2 gene-deleted mice were highly susceptible to protease allergen papain-induced pulmonary inflammation characterized by increased ILC2 numbers, IL-5 and IL-13 cytokine levels, and lung pathology. A mechanistic study showed that following the decreased expression of Mdm2 in lung epithelial cells and A549 cell line, p53 was overactivated, and the expression of its downstream genes p21, Puma, and Noxa was elevated, which resulted in apoptosis. After Mdm2 knockdown, the mRNA expression of inflammation-related gene IL-25, HMGB1, and TNF-α were increased, which further amplified the downstream ILC2 response and lung inflammation. CONCLUSION: These results indicate that Mdm2 maintains the homeostasis of lung epithelial cells by targeting P53 and regulates the function of lung epithelial cells under type 2 lung inflammation.


Subject(s)
Asthma , Homeostasis , Mice, Knockout , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , Animals , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice , Humans , Asthma/immunology , Asthma/metabolism , Asthma/chemically induced , Asthma/genetics , A549 Cells , Disease Models, Animal , Apoptosis , Epithelial Cells/metabolism , Alveolar Epithelial Cells/metabolism , Papain , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism
10.
FASEB J ; 38(15): e23846, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39093041

ABSTRACT

Epithelial cells play a crucial role in asthma, contributing to chronic inflammation and airway hyperresponsiveness. m6A modification, which involves key proteins such as the demethylase fat mass and obesity-associated protein (FTO), is crucial in the regulation of various diseases, including asthma. However, the role of FTO in epithelial cells and the development of asthma remains unclear. In this study, we investigated the demethylase activity of FTO using a small-molecule inhibitor FB23 in epithelial cells and allergic inflammation in vivo and in vitro. We examined the FTO-regulated transcriptome-wide m6A profiling by methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-seq under FB23 treatment and allergic inflammation conditions. Immunofluorescence staining was performed to assess the tissue-specific expression of FTO in asthmatic bronchial mucosa. We demonstrated that FB23 alleviated allergic inflammation in IL-4/IL-13-treated epithelial cells and house dust mite (HDM)-induced allergic airway inflammation mouse model. The demethylase activity of FTO contributed to the regulation of TNF-α signaling via NF-κB and epithelial-mesenchymal transition-related pathways under allergic inflammation conditions in epithelial cells. FTO was expressed in epithelial, submucosal gland, and smooth muscle cells in human bronchial mucosa. In conclusion, FB23-induced inhibition of FTO alleviates allergic inflammation in epithelial cells and HDM-induced mice, potentially through diverse cellular processes and epithelial-mesenchymal transition signaling pathways, suggesting that FTO is a potential therapeutic target in asthma management.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Asthma , Inflammation , Animals , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Mice , Asthma/metabolism , Asthma/genetics , Inflammation/metabolism , Humans , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Epithelial Cells/metabolism , Mice, Inbred BALB C , Female , Hypersensitivity/metabolism , Hypersensitivity/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Mice, Inbred C57BL
11.
PLoS One ; 19(8): e0309868, 2024.
Article in English | MEDLINE | ID: mdl-39213301

ABSTRACT

Dendritic cells (DCs) are pivotal in regulating allergic asthma. Our research has shown that the absence of Sema3E worsens asthma symptoms in acute and chronic asthma models. However, the specific role of PlexinD1 in these processes, particularly in DCs, remains unclear. This study investigates the role of PlexinD1 in CD11c+ DCs using a house dust mite (HDM) model of asthma. We generated CD11c+ DC-specific PlexinD1 knockout (CD11cPLXND1 KO) mice and subjected them, alongside wild-type controls (PLXND1fl/fl), to an HDM allergen protocol. Airway hyperresponsiveness (AHR) was measured using FlexiVent, and immune cell populations were analyzed via flow cytometry. Cytokine levels and immunoglobulin concentrations were assessed using mesoscale and ELISA, while collagen deposition and mucus production were examined through Sirius-red and periodic acid Schiff (PAS) staining respectively. Our results indicate that CD11cPLXND1 KO mice exhibit significantly exacerbated AHR, characterized by increased airway resistance and tissue elastance. Enhanced mucus production and collagen gene expression were observed in these mice compared to wild-type counterparts. Flow cytometry revealed higher CD11c+ MHCIIhigh CD11b+ cell recruitment into the lungs, and elevated total and HDM-specific serum IgE levels in CD11cPLXND1 KO mice. Mechanistically, co-cultures of B cells with DCs from CD11cPLXND1 KO mice showed significantly increased IgE production compared to wild-type mice.These findings highlight the critical regulatory role of the plexinD1 signaling pathway in CD11c+ DCs in modulating asthma features.


Subject(s)
Asthma , CD11c Antigen , Dendritic Cells , Disease Models, Animal , Immunoglobulin E , Mice, Knockout , Mucus , Animals , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunoglobulin E/blood , Immunoglobulin E/immunology , Mice , CD11c Antigen/metabolism , Mucus/metabolism , Pyroglyphidae/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Allergens/immunology , Mice, Inbred C57BL , Membrane Glycoproteins , Intracellular Signaling Peptides and Proteins
12.
Int J Mol Sci ; 25(16)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39201796

ABSTRACT

Steroid-resistant asthma (SRA), resisting glucocorticoids such as dexamethasone (DEX), is a bottleneck in the treatment of asthma. It is characterized by a predominantly neutrophilic inflammatory subtype and is prone to developing into severe refractory asthma and fatal asthma. Currently, there is a lack of universally effective treatments for SRA. Moreover, since cold stimulation does increase the risk of asthma development and exacerbate asthma symptoms, the treatment of cold-stimulated SRA (CSRA) will face greater challenges. To find effective new methods to ameliorate CSRA, this study established a CSRA mouse model of allergic airway inflammation mimicking human asthma for the first time and evaluated the alleviating effects of 80% ethanol extract of mountain-cultivated ginseng (MCG) based on multi-omics analysis. The results indicate that cold stimulation indeed exacerbated the SRA-related symptoms in mice; the DEX individual treatment did not show a satisfactory effect; while the combination treatment of DEX and MCG could dose-dependently significantly enhance the lung function; reduce neutrophil aggregation; decrease the levels of LPS, IFN-γ, IL-1ß, CXCL8, and IL-17; increase the level of IL-10; alleviate the inflammatory infiltration; and decrease the mucus secretion and the expression of MUC5AC. Moreover, the combination of DEX and high-dose (200 mg/kg) MCG could significantly increase the levels of tight junction proteins (TJs), regulate the disordered intestinal flora, increase the content of short-chain fatty acids (SCFAs), and regulate the abnormal gene profile and metabolic profile. Multi-omics integrated analysis showed that 7 gut microbes, 34 genes, 6 metabolites, and the involved 15 metabolic/signaling pathways were closely related to the pharmacological effects of combination therapy. In conclusion, integrated multi-omics profiling highlighted the benefits of MCG for CSRA mice by modulating the interactions of microbiota, genes, and metabolites. MCG shows great potential as a functional food in the adjuvant treatment of CSRA.


Subject(s)
Asthma , Dexamethasone , Panax , Plant Extracts , Animals , Asthma/drug therapy , Asthma/microbiology , Asthma/metabolism , Panax/chemistry , Mice , Dexamethasone/pharmacology , Plant Extracts/pharmacology , Cold Temperature , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Metabolomics/methods , Microbiota/drug effects , Mice, Inbred BALB C , Lung/drug effects , Lung/metabolism , Lung/microbiology , Lung/pathology , Mucin 5AC/metabolism , Mucin 5AC/genetics , Cytokines/metabolism , Drug Resistance/genetics , Female , Multiomics
13.
Mol Med ; 30(1): 120, 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39129025

ABSTRACT

BACKGROUND: Asthma is a heterogeneous, inflammatory disease with several phenotypes and endotypes. Severe asthmatics often exhibit mixed granulocytosis with reduced corticosteroid sensitivity. Bronchom is a newly developed Ayurvedic prescription medicine, indicated for the treatment of obstructive airway disorders. The purpose of the present study was to evaluate the in-vivo efficacy of Bronchom in mouse model of mixed granulocytic asthma with steroidal recalcitrance. METHODS: High-performance thin layer chromatography (HPTLC) and Ultra-high performance liquid chromatography (UHPLC) were employed to identify and quantitate the phytometabolites present in Bronchom. The preclinical effectiveness of Bronchom was assessed in house dust mite (HDM) and Complete Freund's adjuvant (CFA)-induced mixed granulocytic asthma model in mice. High dose of dexamethasone was tested parallelly. Specific-pathogen-free C57BL/6 mice were immunized with HDM and CFA and nineteen days later, they were intranasally challenged with HDM for four consecutive days. Then the mice were challenged with nebulized methacholine to evaluate airway hyperresponsiveness (AHR). Inflammatory cell influx was enumerated in the bronchoalveolar lavage fluid (BALF) followed by lung histology. Additionally, the concentrations of Th2 and pro-inflammatory cytokines was assessed in the BALF by multiplexed immune assay. The mRNA expression of pro-inflammatory cytokines and Mucin 5AC (MUC5AC) was also evaluated in the lung. RESULTS: HPTLC fingerprinting and UHPLC quantification of Bronchom revealed the presence of bioactive phytometabolites, namely, rosmarinic acid, gallic acid, methyl gallate, piperine, eugenol and glycyrrhizin. Bronchom effectively reduced AHR driven by HDM-CFA and the influx of total leukocytes, eosinophils and neutrophils in the BALF. In addition, Bronchom inhibited the infiltration of inflammatory cells in the lung as well as goblet cell metaplasia. Further, it also suppressed the elevated levels of Th2 cytokines and pro-inflammatory cytokines in the BALF. Similarly, Bronchom also regulated the mRNA expression of pro-inflammatory cytokines as well as MUC5AC in mice lungs. Reduced effectiveness of a high dose of the steroid, dexamethasone was observed in the model. CONCLUSIONS: We have demonstrated for the first time the robust pharmacological effects of an herbo-mineral medicine in an animal model of mixed granulocytic asthma induced by HDM and CFA. The outcomes suggest the potential utility of Bronchom in severe asthmatics with a mixed granulocytic phenotype.


Subject(s)
Airway Remodeling , Asthma , Disease Models, Animal , Animals , Asthma/drug therapy , Asthma/immunology , Asthma/metabolism , Mice , Airway Remodeling/drug effects , Adrenal Cortex Hormones/therapeutic use , Adrenal Cortex Hormones/pharmacology , Cytokines/metabolism , Medicine, Ayurvedic , Bronchoalveolar Lavage Fluid , Female , Mice, Inbred C57BL , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Plant Extracts/pharmacology , Lung/drug effects , Lung/pathology , Inflammation/drug therapy , Respiratory Hypersensitivity/drug therapy , Pyroglyphidae/immunology
14.
Int Immunopharmacol ; 141: 112995, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39191121

ABSTRACT

Zymogen granule 16 (ZG16) is a secretory glycoprotein found in zymogen granules, which also plays an important role in colorectal inflammation and cancer. Herein, a ZG16 gene knock-out (ZG16-/-) mouse line was established and we found that ZG16 deletion damaged the intestinal mucosal barrier and gut microbiota, which resulted in low-level inflammation and further promoted the development of ulcerative colitis and inflammation-related colorectal cancer. Meanwhile, a metabolomics analysis on mouse feces showed that the metabolites significantly differed between ZG16-/- and WT mice, which were important mediators of host-microbiota communication and may impact the pulmonary inflammation of mice. Indeed, ZG16-/- mice showed more severe inflammation in a bronchial asthma model. Taken together, the results demonstrate that ZG16 plays a pivotal role in inhibiting inflammation and regulating immune responses in colorectum and lung of experimental animals, which may provide a better understanding of the underlying mechanism of human inflammatory diseases associated with ZG16.


Subject(s)
Gastrointestinal Microbiome , Intestinal Mucosa , Mice, Inbred C57BL , Mice, Knockout , Animals , Gastrointestinal Microbiome/immunology , Mice , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Asthma/immunology , Asthma/metabolism , Asthma/microbiology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Humans , Lung/immunology , Lung/pathology , Lung/metabolism , Disease Models, Animal , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Male , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Bacteria/metabolism , Bacteria/immunology , Glycoproteins/metabolism , Glycoproteins/genetics
15.
Int Immunopharmacol ; 141: 113018, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39216235

ABSTRACT

Turmeric (Curcuma longa L.) extract (CLE) has been shown to elicit several pharmacological properties and is widely used in Asian traditional medicine. Herein, we assessed the impact of CLE on airway inflammation in BALB/c mice and A549 cells to clarify the underlying mechanism. An asthmatic mouse model was established by administering ovalbumin (OVA). CLE (100 or 300 mg/kg/day) was orally administered daily from days 18 to 23, with dexamethasone (3 mg/kg/day) used as the positive control. Human airway epithelial cells, A549, were stimulated using recombinant tumor necrosis factor-α. The CLE100 and CLE400 groups exhibited a significant downregulation in eosinophil counts, cytokine levels, and immunoglobulin-E levels. Moreover, CLE administration dose-dependently suppressed oxidative stress and airway inflammation in the lung tissue. CLE administration inhibited the phosphorylation of mitogen-activated protein kinases (MAPKs) and the expression and activity of matrix metalloproteinase (MMP)-9. In vitro, CLE treatment reduced mRNA levels of proinflammatory cytokines, MAPK phosphorylation, and the expression and activity of MMP-2 and MMP-9. Additionally, 50 µg/mL CLE and 2.5 µg/mL curcumin showed similar anti-inflammatory effects. Collectively, our findings revealed that CLE could suppress airway inflammation in asthmatic mice and A549 cells via oxidative stress-driven MAPK/MMPs signaling, suggesting that CLE could be developed as a potential treatment option for patients with asthma.


Subject(s)
Anti-Inflammatory Agents , Asthma , Curcuma , Matrix Metalloproteinase 9 , Mice, Inbred BALB C , Oxidative Stress , Plant Extracts , Animals , Humans , Oxidative Stress/drug effects , Asthma/drug therapy , Asthma/metabolism , Asthma/immunology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Curcuma/chemistry , A549 Cells , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Mice , Cytokines/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ovalbumin/immunology , Disease Models, Animal , Female , Lung/drug effects , Lung/pathology , Lung/metabolism , Lung/immunology , Immunoglobulin E/blood , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , MAP Kinase Signaling System/drug effects
16.
Proc Natl Acad Sci U S A ; 121(34): e2401251121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39136993

ABSTRACT

Integrin activation resulting in enhanced adhesion to the extracellular matrix plays a key role in fundamental cellular processes. Although integrin activation has been extensively studied in circulating cells such as leukocytes and platelets, much less is known about the regulation and functional impact of integrin activation in adherent cells such as smooth muscle. Here, we show that two different asthmagenic cytokines, IL-13 and IL-17A, activate type I and IL-17 cytokine receptor families, respectively, to enhance adhesion of airway smooth muscle. These cytokines also induce activation of ß1 integrins detected by the conformation-specific antibody HUTS-4. Moreover, HUTS-4 binding is increased in the smooth muscle of patients with asthma compared to nonsmokers without lung disease, suggesting a disease-relevant role for integrin activation in smooth muscle. Indeed, integrin activation induced by the ß1-activating antibody TS2/16, the divalent cation manganese, or the synthetic peptide ß1-CHAMP that forces an extended-open integrin conformation dramatically enhances force transmission in smooth muscle cells and airway rings even in the absence of cytokines. We demonstrate that cytokine-induced activation of ß1 integrins is regulated by a common pathway of NF-κB-mediated induction of RhoA and its effector Rho kinase, which in turn stimulates PIP5K1γ-mediated synthesis of PIP2 at focal adhesions, resulting in ß1 integrin activation. Taken together, these data identify a pathway by which type I and IL-17 cytokine receptor family stimulation induces functionally relevant ß1 integrin activation in adherent smooth muscle and help to explain the exaggerated force transmission that characterizes chronic airway diseases such as asthma.


Subject(s)
Asthma , Integrin beta1 , Interleukin-13 , Interleukin-17 , Muscle, Smooth , NF-kappa B , rho-Associated Kinases , Humans , Integrin beta1/metabolism , Interleukin-17/metabolism , Muscle, Smooth/metabolism , NF-kappa B/metabolism , rho-Associated Kinases/metabolism , Interleukin-13/metabolism , Asthma/metabolism , Signal Transduction , Cell Adhesion , Myocytes, Smooth Muscle/metabolism , Animals
17.
Cell Death Dis ; 15(8): 637, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214980

ABSTRACT

Eosinophils, traditionally associated with allergic phenomena, play a pivotal role in inflammatory responses. Despite accumulating evidence suggesting their pro-inflammatory function upon activation, the underlying mechanisms governing eosinophil activation remain incompletely characterized. In this study, we investigate the local activation of pulmonary and colon eosinophils within the inflammatory microenvironment. Leveraging transcriptional sequencing, we identify TGF-ß as a putative regulator of eosinophil activation, leading to the secretion of granule proteins, including peroxidase. Genetic deletion of TGF-ß receptors on eosinophils resulted in the inhibition of peroxidase synthesis, affirming the significance of TGF-ß signaling in eosinophil activation. Using models of HDM-induced asthma and DSS-induced colitis, we demonstrate the indispensability of TGF-ß-driven eosinophil activation in both disease contexts. Notably, while TGF-ß signaling did not significantly influence asthmatic inflammation, its knockout conferred protection against experimental colitis. This study delineates a distinct pattern of eosinophil activation within inflammatory responses, highlighting the pivotal role of TGF-ß signaling in regulating eosinophil behavior. These findings deepen our comprehension of eosinophil-related pathophysiology and may pave the way for targeted therapeutic approaches in allergic and inflammatory diseases.


Subject(s)
Asthma , Colitis , Eosinophils , Inflammation , Signal Transduction , Transforming Growth Factor beta , Animals , Eosinophils/metabolism , Transforming Growth Factor beta/metabolism , Mice , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/genetics , Asthma/metabolism , Asthma/pathology , Asthma/genetics , Asthma/immunology , Mice, Inbred C57BL , Mice, Knockout , Humans
18.
Eur J Pharmacol ; 980: 176867, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39111683

ABSTRACT

BACKGROUND: MARCKS (myristoylated alanine-rich C kinase substrates) serves as a substrate for protein kinase C, residing in the plasma membrane while acts as an actin filament crosslinking protein. This investigation aims to elucidate phosphorylated MARCKS (p-MARCKS) levels and activity in allergic asthma patients and explore the therapeutic potential of peptide inhibitors targeting p-MARCKS in an acute mouse model of allergic asthma. METHODS: Immunohistochemistry and histology staining were employed on lung tissue slides to evaluate p-MARCKS expression and allergic asthma symptoms. Airway resistance was measured using invasive whole-body plethysmography. Flow cytometry detected lung dendritic cell migration, and migration/maturation assays were conducted on isolated murine bone marrow-derived dendritic cells (BM-DCs). RESULTS: Elevated p-MARCKS expression was observed in both human asthmatic tissues and animal models immunized with ovalbumin or Alternaria alternata. Remarkably, asthmatic individuals showed elevated high p-MARCKS expression in lung tissues. Intraperitoneal injection of the peptide MPS, targeting the MARCKS phosphorylation site domain, before allergen challenged, effectively suppressed MARCKS phosphorylation in murine lung tissues. MPS inhibited both in vivo and in vitro migration and maturation of dendritic cells (BM-DCs) and reduced Th2-related lymphocyte activation in bronchoalveolar lavage fluid (BALF). MPS pretreatment additionally suppressed all symptoms associated with allergic airway asthma, including a reduction in inflammatory cell influx, airway mucous cell metaplasia, and airway hyperreactivity. CONCLUSION: These findings suggest that phosphorylated MARCKS occurs in asthmatic lung tissue, and the inhibition of MARCKS phosphorylation by the MPS peptide reduces dendritic cell migration and Th2-related lymphocytes in the lungs in a murine model of acute asthma.


Subject(s)
Asthma , Cell Movement , Dendritic Cells , Myristoylated Alanine-Rich C Kinase Substrate , Animals , Female , Humans , Male , Mice , Acute Disease , Asthma/immunology , Asthma/drug therapy , Asthma/pathology , Asthma/metabolism , Cell Movement/drug effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Lung/pathology , Lung/immunology , Lung/drug effects , Mice, Inbred BALB C , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Peptides/pharmacology , Phosphorylation
19.
Autoimmunity ; 57(1): 2387100, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39097915

ABSTRACT

Childhood asthma, a common chronic childhood disease, leads to high mortality and morbidity in the world. Airway smooth muscle cells (ASMCs) is a group of multifunctional cells that has been found to be correlated with the pathogenesis of asthma. Astragaloside IV (AS-IV) is a compound extracted from Astragalus membranaceus, which has the anti-asthmatic effect. However, the role of molecular mechanisms regulated by AS-IV in the biological processes of ASMCs in asthma remains unclear. Our current study aims to investigate the downstream molecular mechanism of AS-IV in modulating the aberrant proliferation and pyroptosis of ASMCs in asthma. At first, we determined that the viability of ASMCs could be efficiently suppressed by AS-IV treatment (200 µM). Moreover, AS-IV promoted the pyroptosis and suppressed PDGF-BB-induced aberrant proliferation. Through mechanism investigation, we confirmed that AS-IV could suppress high mobility group box 1 (HMGB1) expression and prevent it from entering the cytoplasm. Subsequently, AS-IV blocked the interaction between HMGB1 and advanced glycosylation end product-specific receptor (RAGE) to inactivate NF-κB pathway. Finally, in vivo experiments demonstrated that AS-IV treatment can alleviate the lung inflammation in asthma mice. Collectively, AS-IV alleviates asthma and suppresses the pyroptosis of AMSCs through blocking HMGB1/RAGE axis to inactivate NF-κB pathway.


Subject(s)
Asthma , HMGB1 Protein , Myocytes, Smooth Muscle , NF-kappa B , Pyroptosis , Receptor for Advanced Glycation End Products , Saponins , Signal Transduction , Triterpenes , Saponins/pharmacology , Pyroptosis/drug effects , HMGB1 Protein/metabolism , Animals , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , NF-kappa B/metabolism , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Triterpenes/pharmacology , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , Humans , Disease Models, Animal
20.
Respir Res ; 25(1): 295, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095798

ABSTRACT

Shortening of airway smooth muscle and bronchoconstriction are pathognomonic for asthma. Airway shortening occurs through calcium-dependent activation of myosin light chain kinase, and RhoA-dependent calcium sensitization, which inhibits myosin light chain phosphatase. The mechanism through which pro-contractile stimuli activate calcium sensitization is poorly understood. Our review of the literature suggests that pro-contractile G protein coupled receptors likely signal through G12/13 to activate RhoA and mediate calcium sensitization. This hypothesis is consistent with the effects of pro-contractile agonists on RhoA and Rho kinase activation, actin polymerization and myosin light chain phosphorylation. Recognizing the likely role of G12/13 signaling in the pathophysiology of asthma rationalizes the effects of pro-contractile stimuli on airway hyperresponsiveness, immune activation and airway remodeling, and suggests new approaches for asthma treatment.


Subject(s)
Asthma , Signal Transduction , Asthma/metabolism , Asthma/physiopathology , Asthma/drug therapy , Humans , Signal Transduction/physiology , Animals , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , Muscle, Smooth/drug effects , Airway Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL