Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.105
Filter
1.
Clin Respir J ; 18(4): e13742, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38664220

ABSTRACT

BACKGROUND: Allergic asthma is an important respiratory system problem characterized by airway inflammation, breathlessness, and bronchoconstriction. Allergic asthma and its outcomes are triggered by type 2 allergic immune responses. Tectorigenin is a methoxy-isoflavone with anti-inflammatory effects. In this study, we investigated the effects of tectorigenin on the pathophysiology of allergic asthma in an animal model. METHODS: Asthmatic mice were treated with tectorigenin. Then airway hyperresponsiveness (AHR), eosinophil percentage, levels of interleukin (IL)-33, IL-25, IL-13, IL-5, IL-4, total and ovalbumin (OVA)-specific immunoglobulin (Ig)E, and lung histopathology were evaluated. RESULT: Tectorigenin significantly (P 〈 0.05) reduced eosinophil infiltration (41 ± 7%) in the broncho-alveolar lavage fluid (BALF), serum IL-5 level (41 ± 5, pg/mL), and bronchial and vascular inflammation (scores of 1.3 ± 0.2 and 1.1 ± 0.3, respectively) but had no significant effects on AHR, serum levels of IL-33, -25, -13, and -4 (403 ± 24, 56 ± 7, 154 ± 11, and 89 ± 6 pg/mL, respectively), total and OVA-specific IgE (2684 ± 265 and 264 ± 19 ng/mL, respectively), goblet cell hyperplasia, and mucus production. CONCLUSION: Tectorigenin could control inflammation and the secretion of inflammatory mediators of asthma, so it can be regarded as a potential antiasthma treatment with the ability to control eosinophilia-related problems.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Asthma , Disease Models, Animal , Isoflavones , Mice, Inbred BALB C , Ovalbumin , Animals , Asthma/drug therapy , Asthma/chemically induced , Asthma/metabolism , Asthma/immunology , Asthma/pathology , Mice , Ovalbumin/toxicity , Ovalbumin/adverse effects , Isoflavones/pharmacology , Isoflavones/therapeutic use , Antioxidants/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Immunoglobulin E/blood , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Female , Lung/pathology , Lung/drug effects , Lung/metabolism , Lung/immunology , Cytokines/metabolism
2.
JCI Insight ; 9(8)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38502186

ABSTRACT

BACKGROUNDSurvivors of pneumonia, including SARS-CoV-2 pneumonia, are at increased risk for cognitive dysfunction and dementia. In rodent models, cognitive dysfunction following pneumonia has been linked to the systemic release of lung-derived pro-inflammatory cytokines. Microglia are poised to respond to inflammatory signals from the circulation, and their dysfunction has been linked to cognitive impairment in murine models of dementia and in humans.METHODSWe measured levels of 55 cytokines and chemokines in bronchoalveolar lavage fluid and plasma from 341 patients with respiratory failure and 13 healthy controls, including 93 unvaccinated patients with COVID-19 and 203 patients with other causes of pneumonia. We used flow cytometry to sort neuroimmune cells from postmortem brain tissue from 5 patients who died from COVID-19 and 3 patients who died from other causes for single-cell RNA-sequencing.RESULTSMicroglia from patients with COVID-19 exhibited a transcriptomic signature suggestive of their activation by circulating pro-inflammatory cytokines. Peak levels of pro-inflammatory cytokines were similar in patients with pneumonia irrespective of etiology, but cumulative cytokine exposure was higher in patients with COVID-19. Treatment with corticosteroids reduced expression of COVID-19-specific cytokines.CONCLUSIONProlonged lung inflammation results in sustained elevations in circulating cytokines in patients with SARS-CoV-2 pneumonia compared with those with pneumonia secondary to other pathogens. Microglia from patients with COVID-19 exhibit transcriptional responses to inflammatory cytokines. These findings support data from rodent models causally linking systemic inflammation with cognitive dysfunction in pneumonia and support further investigation into the role of microglia in pneumonia-related cognitive dysfunction.FUNDINGSCRIPT U19AI135964, UL1TR001422, P01AG049665, P01HL154998, R01HL149883, R01LM013337, R01HL153122, R01HL147290, R01HL147575, R01HL158139, R01ES034350, R01ES027574, I01CX001777, U01TR003528, R21AG075423, T32AG020506, F31AG071225, T32HL076139.


Subject(s)
COVID-19 , Cytokines , Microglia , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/complications , Microglia/metabolism , Microglia/immunology , Cytokines/metabolism , Cytokines/blood , Male , Female , Middle Aged , Aged , Lung/immunology , Lung/pathology , Lung/virology , Lung/metabolism , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/cytology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Adult , Brain/metabolism , Brain/immunology , Brain/pathology
3.
Sci Data ; 9(1): 269, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35660753

ABSTRACT

Pulmonary hemorrhage (P-Hem) occurs among multiple species and can have various causes. Cytology of bronchoalveolar lavage fluid (BALF) using a 5-tier scoring system of alveolar macrophages based on their hemosiderin content is considered the most sensitive diagnostic method. We introduce a novel, fully annotated multi-species P-Hem dataset, which consists of 74 cytology whole slide images (WSIs) with equine, feline and human samples. To create this high-quality and high-quantity dataset, we developed an annotation pipeline combining human expertise with deep learning and data visualisation techniques. We applied a deep learning-based object detection approach trained on 17 expertly annotated equine WSIs, to the remaining 39 equine, 12 human and 7 feline WSIs. The resulting annotations were semi-automatically screened for errors on multiple types of specialised annotation maps and finally reviewed by a trained pathologist. Our dataset contains a total of 297,383 hemosiderophages classified into five grades. It is one of the largest publicly available WSIs datasets with respect to the number of annotations, the scanned area and the number of species covered.


Subject(s)
Bronchoalveolar Lavage Fluid , Macrophages, Alveolar , Animals , Bronchoalveolar Lavage Fluid/cytology , Cats , Hemosiderin , Horses , Humans , Species Specificity
4.
Respir Res ; 23(1): 44, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35241086

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is among the top 5 causes of mortality in the world and can develop as a consequence of genetic and/or environmental factors. Current efforts are focused on identifying early life insults and how these contribute to COPD development. In line with this, our study focuses on the influence of early life nicotine exposure and its potential impact on (a) lung pulmonary functions, and (b) elastase-induced emphysema in adulthood. METHODS: To address this hypothesis, we developed a model of 2 hits, delivered at different time points: mouse pups were first exposed to nicotine/placebo in utero and during lactation, and then subsequently received elastase/placebo at the age of 11 weeks. The effect of nicotine pretreatment and elastase instillation was assessed by (a) measurement of pulmonary function at post-elastase day (ped) 21, and (b) transcriptomic profiling at ped3 and 21, and complementary protein determination. Statistical significance was determined by 3- and 2-way ANOVA for pulmonary functions, and RNAseq results were analyzed using the R project. RESULTS: We did not observe any impact of nicotine pre- and early post-natal exposure compared to control samples on lung pulmonary functions in adulthood, as measured by FLEXIVENT technology. After elastase instillation, substantial lung damage was detected by x-ray tomography and was accompanied by loss in body weight at ped3 as well as an increase in cell numbers, inflammatory markers in BAL and lung volume at ped21. Lung functions showed a decrease in elastance and an increase in deep inflation volume and pressure volume (pv) loop area in animals with emphysema at ped21. Nicotine had no effect on elastance and deep inflation volume, but did affect the pv loop area in animals with emphysema at ped21. Extensive transcriptomic changes were induced by elastase at ped3 both in the nicotine-pretreated and the control samples, with several pathways common to both groups, such as for cell cycle, DNA adhesion and DNA damage. Nicotine pretreatment affected the number of lymphocytes present in BAL after elastase instillation and some of the complement pathway related proteins, arguing for a slight modification of the immune response, as well as changes related to general body metabolism. The majority of elastase-induced transcriptomic changes detected at ped3 had disappeared at ped21. In addition, transcriptomic profiling singled out a common gene pool that was independently activated by nicotine and elastase. CONCLUSIONS: Our study reports a broad spectrum of transient transcriptomic changes in mouse emphysema and identifies nicotine as influencing the emphysema-associated immune system response.


Subject(s)
Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Life Expectancy , Nicotine/adverse effects , Pulmonary Emphysema/genetics , RNA/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Disease Models, Animal , H-2 Antigens , Histocompatibility Antigens Class I/biosynthesis , Mice , Mice, Inbred C57BL , Pancreatic Elastase/toxicity , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/metabolism
5.
Regul Toxicol Pharmacol ; 130: 105129, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35124138

ABSTRACT

Lung cancer following inhalation in rodents is a major concern regarding exposure to cobalt substances. However, little information is available on adverse effects and toxicity following long-term inhalation exposure to poorly soluble cobalt substances with low bioavailability. Thus, the present study focused on pulmonary effects of the poorly soluble tricobalt tetraoxide (5, 20, 80 mg/m³) in a 28-day inhalation exposure study. Lung weights increased with increasing exposures. Bronchoalveolar lavage fluid analysis and histopathology revealed lung tissue inflammation at the mid-dose with increasing severity in the high-dose group and post-exposure persistency. Markers for cellular damage and cell proliferation were statistically significantly increased. No increase in 8-OH-dG lesions was observed, indicating an absence of oxidative DNA lesions. The primary effect of inhaled Co3O4 particles is inflammation of the respiratory tract strongly resembling responses of inhaled "inert dust" substances, with a NOAEC of 5 mg/m³ under the conditions of this test.


Subject(s)
Cobalt/toxicity , Lung/drug effects , Oxides/toxicity , Pneumonia/pathology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Inhalation Exposure , Male , Particle Size , Random Allocation , Rats , Toxicity Tests
6.
Front Immunol ; 13: 810338, 2022.
Article in English | MEDLINE | ID: mdl-35185901

ABSTRACT

Background: Idiopathic pulmonary fibrosis (IPF) is one of interstitial lung diseases (ILDs) with poor prognosis. S100 calcium binding protein A12 (S100A12) has been reported as a prognostic serum biomarker in the IPF, but its correlation with IPF remains unclear in the lung tissue and bronchoalveolar lavage fluids (BALF). Methods: Datasets were collected from the Gene Expression Omnibus (GEO) database. Person correlation coefficient, Kaplan-Meier analysis, Cox regression analysis, functional enrichment analysis and so on were used. And single cell RNA-sequencing (scRNA-seq) analysis was also used to explore the role of S100A12 and related genes in the IPF. Results: S100A12 was mainly and highly expressed in the monocytes, and its expression was downregulated in the lung of patients with IPF according to scRNA-seq and the transcriptome analysis. However, S100A12 expression was upregulated both in blood and BALF of patients with IPF. In addition, 10 genes were found to interact with S100A12 according to protein-protein interaction (PPI) network, and the first four transcription factors (TF) targeted these genes were found according to hTFtarget database. Two most significant co-expression genes of S100A12 were S100A8 and S100A9. The 3 genes were significantly negatively associated with lung function and positively associated with the St. George's Respiratory Questionnaire (SGRQ) scores in the lung of patients with IPF. And, high expression of the 3 genes was associated with higher mortality in the BALF, and shorter transplant-free survival (TFS) and progression-free survival (PFS) time in the blood. Prognostic predictive value of S100A12 was more superior to S100A8 and S100A9 in patients with IPF, and the composited variable [S100A12 + GAP index (gender, age, and physiological index)] may be a more effective predictive index. Conclusion: These results imply that S100A12 might be an efficient disease severity and prognostic biomarker in patients with IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , S100A12 Protein/metabolism , Severity of Illness Index , Aged , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/cytology , Databases, Factual , Female , Gene Expression Profiling , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/mortality , Male , Middle Aged , Prognosis , RNA-Seq , S100A12 Protein/genetics , Survival Analysis
7.
J Ethnopharmacol ; 290: 115086, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35157952

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Chestnut flowers were one of the by-products during chestnut industrial processing. Chestnut (Castanea mollissima Blume) flower is rich in flavonoids and has been used as a traditional medicine to treat a variety of diseases including respiratory disorders for a long history. AIM OF THE STUDY: The present study aims to investigate the potential anti-inflammatory effect of flavonoids from chestnut flower (FCF) in lipopolysaccharide (LPS)-treated RAW 264.7 cells and stimulated acute lung injury (ALI) in mice. MATERIALS AND METHODS: HPLC-ESI-MS/MS was applied to identify flavonoids from Chestnut flower. The ROS content in cells and lung tissue was measured by flow cytometry. The malondialdehyde (MDA) content, superoxide dismutase (SOD) activity and glutathione (GSH) content in cells and bronchoalveolar lavage fluid (BALF) was analyzed by photometry. Furthermore, the level of pro-inflammatory factors was analyzed by ELISA, and the expression of inflammatory gene mRNA by fluorescence quantitative PCR. H&E staining was used to evaluate the degree of lung tissue injury in mice. MPO activity was used to measure the degree of neutrophil infiltration. Total protein content was detected by BCA method. RESULTS: A total of forty-nine flavonoids compounds were tentatively identified in FCF by mass spectrometry analysis. The results of cell experiment suggested that FCF could alleviate oxidative injury via increasing SOD activity and GSH content, as well as inhibiting the production of intracellular ROS and MDA. FCF exerted its protective effect by suppressing the expression of both inducible nitric oxide synthase (iNOS) and cycooxygenase 2 (COX-2) to inhibit the synthesis of pro-inflammatory factors and cytokines, including NO, PGE2, TNF-α, IL-6 and IL-1ß. Besides, FCF treatment could alleviate the thickening of alveolar wall and pulmonary congestion in LPS-treated ALI mice, and significantly inhibit the activity of myeloperoxidas (MPO) and the expression of cytokines in BALF. CONCLUSIONS: FCF could ameliorate inflammation and oxidative stress in LPS-treated inflammation, resulting in an overall improvement in both macroscopic and histological parameters.


Subject(s)
Acute Lung Injury/pathology , Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Plant Extracts/pharmacology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cytokines/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Flowers , Glutathione/drug effects , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Lung/drug effects , Macrophages/drug effects , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/drug effects , Oxidative Stress/drug effects , RAW 264.7 Cells , Random Allocation , Superoxide Dismutase/drug effects , Tandem Mass Spectrometry
8.
JCI Insight ; 7(4)2022 02 22.
Article in English | MEDLINE | ID: mdl-35191395

ABSTRACT

The intensity and longevity of inflammatory responses to inhaled allergens is determined largely by the balance between effector and regulatory immune responses, but the mechanisms that determine the relative magnitudes of these opposing forces remain poorly understood. We have found that the type of adjuvant used during allergic sensitization has a profound effect on both the nature and longevity of the pulmonary inflammation triggered by subsequent reexposure to that same provoking allergen. TLR ligand adjuvants and house dust extracts primed immune responses characterized by a mixed neutrophilic and eosinophilic inflammation that was suppressed by multiple daily allergen challenges. During TLR ligand-mediated allergic sensitization, mice displayed transient airway neutrophilia, which triggered the release of TGF-ß into the airway. This neutrophil-dependent production of TGF-ß during sensitization had a delayed, suppressive effect on eosinophilic responses to subsequent allergen challenge. Neutrophil depletion during sensitization did not affect numbers of Foxp3+ Tregs but increased proportions of Gata3+CD4+ T cells, which, upon their transfer to recipient mice, triggered stronger eosinophilic inflammation. Thus, a neutrophil/TGF-ß axis acts during TLR-mediated allergic sensitization to fine-tune the phenotype of developing allergen-specific CD4+ T cells and limit their pathogenicity, suggesting a novel immunotherapeutic approach to control eosinophilia in asthma.


Subject(s)
Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , Neutrophils/metabolism , Respiratory Hypersensitivity/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Neutrophils/pathology , Respiratory Hypersensitivity/pathology , Transforming Growth Factor beta/metabolism
9.
Parasit Vectors ; 15(1): 16, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991711

ABSTRACT

BACKGROUND: There is ample evidence demonstrating a reverse relationship between helminth infection and immune-mediated diseases. Accordingly, several studies have shown that Echinococcus granulosus infection and hydatid cyst compounds are able to suppress immune responses in allergic airway inflammation. Previous studies have documented the ability of hydatid cysts to suppress aberrant Th2 immune response in a mouse model of allergic asthma. However, there is a paucity of research on the effects of protoscoleces on allergic asthma. Thus, this study was designed to evaluate the effects of somatic antigens of protoscoleces in a murine model of allergic airway inflammation. METHODS: Ovalbumin (OVA)/aluminum hydroxide (alum) was injected intraperitoneally to sensitize BALB/c mice over a period of 0 to 7 days, followed by challenge with 1% OVA. The treatment group received somatic antigens of protoscoleces emulsified with PBS on these days in each sensitization before being challenged with 1% OVA on days 14, 15, and 16. The effects of somatic antigens of protoscoleces on allergic airway inflammation were evaluated by examining histopathological changes, the recruitment of inflammatory cells in the bronchoalveolar lavage, cytokine production in the homogenized lung tissue (IL-4, IL-5, IL-10, IL-17, and IFN-γ), and total antioxidant capacity in serum. RESULTS: Overall, administration of somatic antigens of protoscoleces exacerbated allergic airway inflammation via increased Th2 cytokine levels in the lung homogenate, recruitment of eosinophils into bronchoalveolar lavage fluid, and pathological changes. In addition, total antioxidant capacity and IFN-γ levels declined following the administration of somatic antigens. CONCLUSIONS: The results revealed that the co-administration of somatic products of protoscoleces with OVA/alum contributed to the exacerbation of allergic airway inflammation in BALB/c mice. Currently, the main cause of allergic-type inflammation exacerbation is unknown, and further research is needed to understand the mechanism of these interactions.


Subject(s)
Antigens, Helminth/immunology , Asthma/pathology , Echinococcosis, Pulmonary/immunology , Echinococcus granulosus/immunology , Animals , Antioxidants/analysis , Asthma/complications , Asthma/immunology , Bronchoalveolar Lavage Fluid/cytology , Cytokines/analysis , Disease Models, Animal , Echinococcosis, Pulmonary/complications , Echinococcosis, Pulmonary/pathology , Female , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Sheep , Specific Pathogen-Free Organisms
10.
Arch Pharm Res ; 45(1): 51-62, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34984603

ABSTRACT

Asian sand dust (ASD), which mainly originates in China and Mongolia in the spring and blows into Korea, can exacerbate respiratory and immunological diseases. This study aims to observe effects of co-exposure to ASD on ovalbumin (OVA)-induced asthmatic lung inflammation and of treatment with a phosphodiesterase 7 (PDE7) inhibitor in a mouse model. The challenge with OVA increased airway hyperresponsiveness (AHR) and inflammatory cell infiltration into the lung tissue. Interleukin (IL)-13, tumor necrosis factor-alpha, monocyte-protein-1, mucin, and antigen-specific IgE and IgG1 production increased in mouse serum. The co-exposure of ASD significantly exacerbated these effects in this asthma model. Notably, the administration of a PDE7 inhibitor, BRL-50481 (BRL), significantly reduced AHR, infiltration of inflammatory cells into the lungs, and the levels of type 2 T helper cell-related cytokines, antigen-specific immunoglobulins, and mucin. Thus, the administration of BRL ameliorated OVA-induced allergic asthmatic responses exacerbated by co-exposure to ASD. This study suggests that PDE7 inhibition can be a therapeutic strategy for inflammatory lung diseases and asthma via the regulation of T lymphocytes and reduction of IL-13, and, consequently, mucin production.


Subject(s)
Anti-Inflammatory Agents , Asthma , Cyclic Nucleotide Phosphodiesterases, Type 7 , Pneumonia , Animals , Mice , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Asthma/etiology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Cytokines/analysis , Disease Models, Animal , Dust , Fluorescent Antibody Technique , Inhalation Exposure/adverse effects , Lung/pathology , Mice, Inbred BALB C , Ovalbumin/adverse effects , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/pathology , Sand
11.
Int Immunopharmacol ; 104: 108509, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34998035

ABSTRACT

The present study aims to investigate the effects of CCR3 gene knockout in bone marrow cells (CCR3-KO) on the mouse model of combined allergic rhinitis and asthma syndrome (CARAS). It was found that CCR3-KO significantly reduced eosinophil (EOS) migration into the nasal (NALF) and bronchoalveolar (BALF) cavities of mice, and decreased Th2 cytokines (such as, IL-4, IL-5 and IL-13) levels in nasal mucosa and lung tissues. In addition, histological analysis showed that the damage degree of nasal mucosa structure in ovalbumin (OVA) modulated CCR3-KO mice was significantly less than that in OVA modulated Wild type (WT) mice, with reduced inflammatory cell infiltration and nasal mucus secretion. The infiltration of inflammatory cells in lung tissue was significantly reduced, and the proliferation of lung smooth muscle layer and extracellular matrix (ECM) production were decreased. Symptom analysis showed that CCR3-KO can reduced allergic rhinitis (AR) signals as nose scratching and sneezing. It was also found CCR3-KO reduce OVA-induced weight loss. The results showed that CCR3-KO could reduce the symptoms of allergic inflammation in CARAS mice by reducing airway inflammatory cell infiltration and down-regulating the expression of Th2 cytokines, and CCR3 gene could be used as a target gene for the treatment of CARAS.


Subject(s)
Asthma/genetics , Receptors, CCR3/genetics , Rhinitis, Allergic/genetics , Allergens/immunology , Animals , Asthma/metabolism , Asthma/pathology , Bone Marrow Cells , Bronchoalveolar Lavage Fluid/cytology , Cytokines/genetics , Eosinophils/immunology , Immunoglobulin E/blood , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Nasal Lavage Fluid/cytology , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Ovalbumin/immunology , Receptors, CCR3/metabolism , Rhinitis, Allergic/metabolism , Rhinitis, Allergic/pathology , Syndrome , Th2 Cells
12.
PLoS One ; 17(1): e0261242, 2022.
Article in English | MEDLINE | ID: mdl-35007307

ABSTRACT

Hyperactive and damaging inflammation is a hallmark of severe rather than mild Coronavirus disease 2019 (COVID-19). To uncover key inflammatory differentiators between severe and mild COVID-19, we applied an unbiased single-cell transcriptomic analysis. We integrated two single-cell RNA-seq datasets with COVID-19 patient samples, one that sequenced bronchoalveolar lavage (BAL) cells and one that sequenced peripheral blood mononuclear cells (PBMCs). The combined cell population was then analyzed with a focus on genes associated with disease severity. The immunomodulatory long non-coding RNAs (lncRNAs) NEAT1 and MALAT1 were highly differentially expressed between mild and severe patients in multiple cell types. Within those same cell types, the concurrent detection of other severity-associated genes involved in cellular stress response and apoptosis regulation suggests that the pro-inflammatory functions of these lncRNAs may foster cell stress and damage. Thus, NEAT1 and MALAT1 are potential components of immune dysregulation in COVID-19 that may provide targets for severity related diagnostic measures or therapy.


Subject(s)
COVID-19/genetics , Gene Expression Profiling/methods , Leukocytes, Mononuclear/metabolism , RNA, Long Noncoding/genetics , Single-Cell Analysis/methods , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/virology , COVID-19/virology , Cells, Cultured , Cluster Analysis , Gene Ontology , Humans , Inflammation/genetics , Inflammation/virology , Leukocytes, Mononuclear/virology , RNA-Seq/methods , SARS-CoV-2/physiology , Severity of Illness Index
13.
Comput Math Methods Med ; 2022: 1452116, 2022.
Article in English | MEDLINE | ID: mdl-35047052

ABSTRACT

OBJECTIVES: This study sought to examine whether ligustrazine was capable of inhibiting phosphodiesterase (PDE) activity and improving lung function in a rat model of asthma. METHODS: Rats were initially sensitized using ovalbumin (OVA) and then were challenged daily with aerosolized OVA beginning 14 days later (30 min/day) to generate a rat model of asthma. Changes in airway function following methacholine (MCh) injection were evaluated by monitoring lung resistance (R L) and dynamic lung compliance (C dyn) values using an AniRes2005 analytic system. In addition, serum IgE was measured via ELISA, while PDE expression was evaluated via qPCR and western blotting. Key Findings. Ligustrazine significantly impaired allergen-induced lung hyperresponsivity and inflammation in this asthma model system. Ligustrazine treatment was also associated with reduced expression of PDEs including PDE4 in the lungs of these rats. CONCLUSIONS: Ligustrazine suppresses airway inflammation and bronchial hyperresponsivity in this rat model system, and these changes are associated with decreased PDE expression at the protein and mRNA levels.


Subject(s)
Asthma/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Pyrazines/pharmacology , Airway Resistance/drug effects , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/immunology , Asthma/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Computational Biology , Disease Models, Animal , Immunoglobulin E/blood , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Ovalbumin/administration & dosage , Ovalbumin/immunology , Phosphoric Diester Hydrolases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/physiopathology
14.
Respir Res ; 23(1): 11, 2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35042537

ABSTRACT

BACKGROUND: Airway mucus hypersecretion is one of the important pathological features of chronic obstructive pulmonary disease (COPD). MUC5B is the main mucin expressed in the airways of COPD patients and has been indicated to play an important role in airway defense. However, the specific biological function of MUC5B in COPD and the possible mechanism are not clear. METHODS: We established a COPD model with 24-week-old MUC5B-/- mice exposed to cigarette smoke and tested our hypothesis through lung function tests, HE and PAS staining, immunohistochemistry (IHC), western blot, q-PCR and ELISA. RESULTS: Compared with MUC5B+/+ mice, MUC5B-/- mice had worse general condition and lung function, increased inflammatory infiltration, reduced goblet cell differentiation as indicated by decreased PAS staining (PAS grade: 1.8 ± 0.24 vs. 0.6 ± 0.16), reduced MUC5AC expression (ELISA: 0.30 ± 0.01 vs. 0.17 ± 0.01 mg/ml, q-PCR: 9.4 ± 1.7 vs. 4.1 ± 0.1 fold, IHC score: 3.1 ± 0.9 vs. 1.6 ± 0.7), increased macrophage secretion of inflammatory factors (TNF-α and IL-6) and expression of downstream pathway factors (ERK1/2 and NF-κB), decreased expression of SPDEF and STAT6, and increased expression of FOXA2. CONCLUSION: The protective effect of MUC5B in the development of COPD was mediated by the promotion of goblet cell differentiation and the inhibition of inflammation. The role of MUC5B in regulating inflammation was related to macrophage function, and goblet cell differentiation was promoted by the induced expression of STAT6 and SPDEF. This study describes a mechanism of mucus hypersecretion and identifies MUC5B as a new target for the treatment of mucus hypersecretion.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Lung/pathology , Mucin-5B/genetics , Pulmonary Disease, Chronic Obstructive/genetics , RNA/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Differentiation , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-5B/biosynthesis , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology
15.
J Immunol ; 208(3): 745-752, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35031577

ABSTRACT

Cystic fibrosis (CF) is an inherited life-threatening disease accompanied by repeated lung infections and multiorgan inflammation that affects tens of thousands of people worldwide. The causative gene, cystic fibrosis transmembrane conductance regulator (CFTR), is mutated in CF patients. CFTR functions in epithelial cells have traditionally been thought to cause the disease symptoms. Recent work has shown an additional defect: monocytes from CF patients show a deficiency in integrin activation and adhesion. Because monocytes play critical roles in controlling infections, defective monocyte function may contribute to CF progression. In this study, we demonstrate that monocytes from CFTRΔF508 mice (CF mice) show defective adhesion under flow. Transplanting CF mice with wild-type (WT) bone marrow after sublethal irradiation replaced most (60-80%) CF monocytes with WT monocytes, significantly improved survival, and reduced inflammation. WT/CF mixed bone marrow chimeras directly demonstrated defective CF monocyte recruitment to the bronchoalveolar lavage and the intestinal lamina propria in vivo. WT mice reconstituted with CF bone marrow also show lethality, suggesting that the CF defect in monocytes is not only necessary but also sufficient to cause disease. We also show that monocyte-specific knockout of CFTR retards weight gains and exacerbates dextran sulfate sodium-induced colitis. Our findings show that providing WT monocytes by bone marrow transfer rescues mortality in CF mice, suggesting that similar approaches may mitigate disease in CF patients.


Subject(s)
Cell Adhesion/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Monocytes/immunology , Monocytes/transplantation , Animals , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/cytology , Colitis/pathology , Cystic Fibrosis/pathology , Integrins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL
16.
Nat Immunol ; 23(1): 62-74, 2022 01.
Article in English | MEDLINE | ID: mdl-34764490

ABSTRACT

The molecular mechanisms governing orderly shutdown and retraction of CD4+ type 1 helper T (TH1) cell responses remain poorly understood. Here we show that complement triggers contraction of TH1 responses by inducing intrinsic expression of the vitamin D (VitD) receptor and the VitD-activating enzyme CYP27B1, permitting T cells to both activate and respond to VitD. VitD then initiated the transition from pro-inflammatory interferon-γ+ TH1 cells to suppressive interleukin-10+ cells. This process was primed by dynamic changes in the epigenetic landscape of CD4+ T cells, generating super-enhancers and recruiting several transcription factors, notably c-JUN, STAT3 and BACH2, which together with VitD receptor shaped the transcriptional response to VitD. Accordingly, VitD did not induce interleukin-10 expression in cells with dysfunctional BACH2 or STAT3. Bronchoalveolar lavage fluid CD4+ T cells of patients with COVID-19 were TH1-skewed and showed de-repression of genes downregulated by VitD, from either lack of substrate (VitD deficiency) and/or abnormal regulation of this system.


Subject(s)
Interferon-gamma/immunology , Interleukin-10/immunology , SARS-CoV-2/immunology , Th1 Cells/immunology , Vitamin D/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Bronchoalveolar Lavage Fluid/cytology , COVID-19/immunology , COVID-19/pathology , Complement C3a/immunology , Complement C3b/immunology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lymphocyte Activation/immunology , Receptors, Calcitriol/metabolism , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Transcription, Genetic/genetics
17.
JCI Insight ; 7(1)2022 01 11.
Article in English | MEDLINE | ID: mdl-34793331

ABSTRACT

Neutrophils are recognized as important circulating effector cells in the pathophysiology of severe coronavirus disease 2019 (COVID-19). However, their role within the inflamed lungs is incompletely understood. Here, we collected bronchoalveolar lavage (BAL) fluids and parallel blood samples of critically ill COVID-19 patients requiring invasive mechanical ventilation and compared BAL fluid parameters with those of mechanically ventilated patients with influenza, as a non-COVID-19 viral pneumonia cohort. Compared with those of patients with influenza, BAL fluids of patients with COVID-19 contained increased numbers of hyperactivated degranulating neutrophils and elevated concentrations of the cytokines IL-1ß, IL-1RA, IL-17A, TNF-α, and G-CSF; the chemokines CCL7, CXCL1, CXCL8, CXCL11, and CXCL12α; and the protease inhibitors elafin, secretory leukocyte protease inhibitor, and tissue inhibitor of metalloproteinases 1. In contrast, α-1 antitrypsin levels and net proteolytic activity were comparable in COVID-19 and influenza BAL fluids. During antibiotic treatment for bacterial coinfections, increased BAL fluid levels of several activating and chemotactic factors for monocytes, lymphocytes, and NK cells were detected in patients with COVID-19 whereas concentrations tended to decrease in patients with influenza, highlighting the persistent immunological response to coinfections in COVID-19. Finally, the high proteolytic activity in COVID-19 lungs suggests considering protease inhibitors as a treatment option.


Subject(s)
Bacterial Infections , Bronchoalveolar Lavage Fluid , COVID-19 , Coinfection , Influenza, Human , Adult , Aged , Bacterial Infections/complications , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/pathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , COVID-19/complications , COVID-19/diagnosis , COVID-19/immunology , COVID-19/pathology , Coinfection/immunology , Coinfection/metabolism , Coinfection/pathology , Cytokines/analysis , Female , Humans , Inflammation , Influenza, Human/complications , Influenza, Human/diagnosis , Influenza, Human/immunology , Influenza, Human/pathology , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Middle Aged
18.
Int J Biol Macromol ; 195: 246-254, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34863838

ABSTRACT

Purified acid polysaccharides PSAP-1 and PSAP-2 with apparent molecular weights of 64.6 and 38.9 kDa, respectively, were isolated from Pyrus sinkiangensis Yu. through combined techniques of ion-exchange and gel permeation chromatography. Both polysaccharides were composed of predominant amounts of GalA and small amounts of Ara, Rha, and Gal. They are deduced to be native pectin-type polysaccharides containing the HG backbone consisting of α-1,4-GalAp and methyl-esterified α-1,4-GalAp residues by IR, GC-MS and NMR spectra analyses. The immunoregulatory activity test showed that PSAP-1 and PSAP-2 could increase the cell viability and the release of NO, IL-6, and TNF-α on the RAW264.7 macrophage. It indicated that PSAP-1 and PSAP-2 could increase macrophage-mediated immunostimulatory activity. The airway inflammation test of antiasthmatic mice showed that PSAP-1 could decrease the contents of IL-4, IL-5, and IL-13 and the number of inflammatory cells in BALF and improve the pathological changes in lung tissue. PSAP-1 could also decrease the amount of mucus secreted by goblet cells and the expression levels of NF-κB p65, IκBα, IKK, ERK, JNK, P38, and Muc5ac mRNA and increase the expression levels of TLR2 and TLR4 mRNA in lung tissues. This suggested that PSAP-1 may resist airway inflammation in mice. PSAP-1 and PSAP-2 had potential clinical application value.


Subject(s)
Adjuvants, Immunologic/pharmacology , Polysaccharides/isolation & purification , Pyrus/metabolism , Adjuvants, Immunologic/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Survival , Chromatography, Ion Exchange/methods , Cytokines/metabolism , Inflammation/metabolism , Lipopolysaccharides/metabolism , Lung/metabolism , Macrophages/metabolism , Mice , Mucus/metabolism , NF-KappaB Inhibitor alpha/metabolism , Pectins/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , RAW 264.7 Cells , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 322(1): L116-L128, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34850640

ABSTRACT

Obesity impairs host defense against Klebsiella pneumoniae, but responsible mechanisms are incompletely understood. To determine the impact of diet-induced obesity on pulmonary host defense against K. pneumoniae, we fed 6-wk-old male C57BL/6j mice a normal diet (ND) or high-fat diet (HFD) (13% vs. 60% fat, respectively) for 16 wk. Mice were intratracheally infected with Klebsiella, assayed at 24 or 48 h for bacterial colony-forming units, lung cytokines, and leukocytes from alveolar spaces, lung parenchyma, and gonadal adipose tissue were assessed using flow cytometry. Neutrophils from uninfected mice were cultured with and without 2-deoxy-d-glucose (2-DG) and assessed for phagocytosis, killing, reactive oxygen intermediates (ROI), transport of 2-DG, and glucose transporter (GLUT1-4) transcripts, and protein expression of GLUT1 and GLUT3. HFD mice had higher lung and splenic bacterial burdens. In HFD mice, baseline lung homogenate concentrations of IL-1ß, IL-6, IL-17, IFN-γ, CXCL2, and TNF-α were reduced relative to ND mice, but following infection were greater for IL-6, CCL2, CXCL2, and IL-1ß (24 h only). Despite equivalent lung homogenate leukocytes, HFD mice had fewer intraalveolar neutrophils. HFD neutrophils exhibited decreased Klebsiella phagocytosis and killing and reduced ROI to heat-killed Klebsiella in vitro. 2-DG transport was lower in HFD neutrophils, with reduced GLUT1 and GLUT3 transcripts and protein (GLUT3 only). Blocking glycolysis with 2-DG impaired bacterial killing and ROI production in neutrophils from mice fed ND but not HFD. Diet-induced obesity impairs pulmonary Klebsiella clearance and augments blood dissemination by reducing neutrophil killing and ROI due to impaired glucose transport.


Subject(s)
Diet , Glucose/metabolism , Host-Pathogen Interactions , Klebsiella Infections/microbiology , Klebsiella pneumoniae/physiology , Neutrophils/metabolism , Obesity/microbiology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Bacterial Load/drug effects , Biological Transport/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Bone Marrow/pathology , Bronchoalveolar Lavage Fluid/cytology , Cytokines/metabolism , Deoxyglucose/pharmacology , Diet, High-Fat , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/drug effects , Host-Pathogen Interactions/drug effects , Klebsiella Infections/blood , Klebsiella Infections/complications , Klebsiella pneumoniae/drug effects , Leukocyte Count , Lung/microbiology , Lung/pathology , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Obesity/blood , Obesity/complications , Phagocytosis/drug effects , Pneumonia/microbiology , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/microbiology
20.
Shock ; 57(3): 408-416, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34387224

ABSTRACT

ABSTRACT: Extracellular vesicles (EVs) have now been recognized as important mediators of cellular communication during injury and repair. We previously found that plasma EVs isolated from ex vivo perfused human lungs injured with Escherichia coli bacterial pneumonia were inflammatory, and exogenous administration of high molecular weight (HMW) hyaluronic acid (HA) as therapy bound to these EVs, decreasing inflammation and injury. In the current study, we studied the role of EVs released during severe Pseudomonas aeruginosa (PA) pneumonia in mice and determined whether intravenous administration of exogenous HMW HA would have therapeutic effects against the bacterial pneumonia. EVs were collected from the bronchoalveolar lavage fluid (BALF) of mice infected with PA103 by ultracentrifugation and analyzed by NanoSight and flow cytometry. In a cytotoxicity assay, administration of EVs released from infected mice (I-EVs) decreased the viability of A549 cells compared to EV isolated from sham control mice (C-EVs). Either exogenous HMW HA or an anti-CD44 antibody, when co-incubated with I-EVs, significantly improved the viability of the A549 cells. In mice with PA103 pneumonia, administration of HMW HA improved pulmonary edema and bacterial count in the lungs and decreased TNF-α and caspase-3 levels in the supernatant of lung homogenates. In conclusion, EVs isolated from BALF of mice with P. aeruginosa pneumonia were cytotoxic and inflammatory, and intravenous HMW HA administration was protective against P. aeruginosa pneumonia.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Extracellular Vesicles/drug effects , Hyaluronic Acid/therapeutic use , Pneumonia, Bacterial/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa , Animals , Bronchoalveolar Lavage Fluid/cytology , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Pneumonia, Bacterial/etiology , Pseudomonas Infections/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...