Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.853
Filter
1.
Int J Chron Obstruct Pulmon Dis ; 19: 1905-1920, 2024.
Article in English | MEDLINE | ID: mdl-39206144

ABSTRACT

Purpose: The main objective of this study was to explore the mechanism of effective component compatibility of Bufei Yishen formula III (ECC-BYF III) in inhibiting mitochondrial oxidative stress in a rat model of chronic obstructive pulmonary disease (COPD). Methods: A549 cells exposed to cigarette smoke extract (CSE) were used to establish a model of mitochondrial oxidative damage. The cells were treated with the plasmid encoding Pkm2 and the enzymes and proteins involved in oxidative stress and mitochondrial function were measured. A rat model of COPD was established using CS and bacteria. Two different treatments were established, ECC-BYF III (5.5 mg/kg/d) and N-acetylcysteine (54 mg/kg/day). Animals were tested for pulmonary function (Vt, PEF, FVC, FEV0.1s and Cdyn) after eight weeks of therapy and were sacrificed. Pulmonary H&E staining was performed, and the total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), total antioxidant capacity (T-AOC), and malondialdehyde (MDA) content were measured. The mitochondrial function was also examined. Furthermore, the Pkm2/Nrf2 signaling pathway was evaluated. Results: Overexpression of Pkm2 dramatically ameliorated the CS-induced mitochondrial oxidative damage. Further studies indicated that ECC-BYF III significantly improved mitochondrial function and inhibited oxidative stress in the lung tissues of COPD rats. Moreover, it can upregulate mitochondrial respiratory chain enzyme activity. ECC-BYF III also decreased the MDA content and increased T-SOD, GSH-Px, and T-AOC expression to facilitate oxidative homeostasis. Finally, our results indicated that the Pkm2/Nrf2 pathway is regulated by ECC-BYF III in A549 cells and lung tissue. Conclusion: These results indicate that ECC-BYF III exerts a strong effective therapeutic effect against cigarette smoke combined with bacteria-induced COPD in rats by activating the Pkm2/Nrf2 signaling pathway and restoring mitochondrial oxidative stress. Although more in vivo animal model research is needed to confirm these findings, this study contributes new data to support the conventional usage of ECC-BYF III.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal , Lung , Mitochondria , NF-E2-Related Factor 2 , Oxidative Stress , Pulmonary Disease, Chronic Obstructive , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Humans , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Lung/enzymology , Drugs, Chinese Herbal/pharmacology , A549 Cells , Male , Thyroid Hormone-Binding Proteins , Pyruvate Kinase/metabolism , Antioxidants/pharmacology , Rats, Sprague-Dawley , Carrier Proteins/metabolism , Thyroid Hormones/metabolism , Smoke/adverse effects , Membrane Proteins/metabolism , Membrane Proteins/genetics
2.
Arterioscler Thromb Vasc Biol ; 44(8): 1784-1798, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38868942

ABSTRACT

BACKGROUND: The chromatin-remodeling enzymes BRG1 (brahma-related gene 1) and CHD4 (chromodomain helicase DNA-binding protein 4) independently regulate the transcription of genes critical for vascular development, but their coordinated impact on vessels in late-stage embryos has not been explored. METHODS: In this study, we genetically deleted endothelial Brg1 and Chd4 in mixed background mice (Brg1fl/fl;Chd4fl/fl;VE-Cadherin-Cre), and littermates that were negative for Cre recombinase were used as controls. Tissues were analyzed by immunostaining, immunoblot, and flow cytometry. Quantitative reverse transcription polymerase chain reaction was used to determine gene expression, and chromatin immunoprecipitation revealed gene targets of BRG1 and CHD4 in cultured endothelial cells. RESULTS: We found Brg1/Chd4 double mutants grew normally but died soon after birth with small and compact lungs. Despite having normal cellular composition, distal air sacs of the mutant lungs displayed diminished ECM (extracellular matrix) components and TGFß (transforming growth factor-ß) signaling, which typically promotes ECM synthesis. Transcripts for collagen- and elastin-related genes and the TGFß ligand Tgfb1 were decreased in mutant lung endothelial cells, but genetic deletion of endothelial Tgfb1 failed to recapitulate the small lungs and ECM defects seen in Brg1/Chd4 mutants. We instead found several ECM genes to be direct targets of BRG1 and CHD4 in cultured endothelial cells. CONCLUSIONS: Collectively, our data highlight essential roles for endothelial chromatin-remodeling enzymes in promoting ECM deposition in the distal lung tissue during the saccular stage of embryonic lung development.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases , Endothelial Cells , Gene Expression Regulation, Developmental , Lung , Nuclear Proteins , Transcription Factors , Animals , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/deficiency , Lung/embryology , Lung/metabolism , Lung/enzymology , Transcription Factors/metabolism , Transcription Factors/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/enzymology , Mice, Knockout , Signal Transduction , Extracellular Matrix/metabolism , Mice , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice, Inbred C57BL , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Phenotype , Humans , Organogenesis
3.
Res Vet Sci ; 175: 105314, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38823354

ABSTRACT

Over the course of the last twenty years, there has been a growing recognition of the pig's potential as a valuable model for studying human drug metabolism. This study aimed to investigate the expression, enzymatic activity, inhibitory susceptibility, and cellular localization of carboxylesterases (CES) in porcine lung tissue not yet explored. Our results showed that CESs hydrolysis activity followed Michaelis-Menten kinetics in both cytosolic and microsomal fractions of porcine lung tissues (N = 8), with comparable hydrolysis rates for tested substrates, namely 4-nitrophenyl acetate (pNPA), 4-methylumbelliferyl acetate (4-MUA), and fluorescein diacetate (FD). We also determined the CESs hydrolysis activity in a representative sample of the porcine liver that, as expected, displayed higher activity than the lung ones. The study demonstrated variable levels of enzyme activities and interindividual variability in both porcine lung fractions. Inhibition studies used to assess the CESs' involvement in the hydrolysis of pNPA, 4-MUA, and FD suggested that CESs may be the enzymes primarily involved in the metabolism of ester compounds in the pig lung tissue. Overall, this study provides insight into the distribution and diversity of CES isoforms involved in substrate hydrolysis across different cellular fractions (cytosol and microsomes) in porcine lungs.


Subject(s)
Carboxylic Ester Hydrolases , Lung , Animals , Lung/enzymology , Lung/metabolism , Swine , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/genetics , Microsomes/enzymology , Nitrophenols/metabolism , Umbelliferones/metabolism , Fluoresceins , Hydrolysis , Cytosol/enzymology , Liver/enzymology
4.
Int J Rheum Dis ; 27(5): e15164, 2024 May.
Article in English | MEDLINE | ID: mdl-38706209

ABSTRACT

BACKGROUND: JAK inhibitors are well known for the treatment of rheumatoid arthritis (RA), but whether they can be used to treat pulmonary fibrosis, a common extra-articular disease of RA, remains to be clarified. METHODS: A jak2 inhibitor, CEP33779 (CEP), was administered to a rat model of RA-associated interstitial lung disease to observe the degree of improvement in both joint swelling and pulmonary fibrosis. HFL1 cells were stimulated with TGF-ß1 to observe the expression of p-JAK2. Then, different concentrations of related gene inhibitors (JAK2, TGFß-R1/2, and p-STAT3) or silencers (STAT3, JAK2) were administered to HFL1 cells, and the expression levels of related proteins were detected to explore the underlying mechanisms of action. RESULTS: CEP not only reduced the degree of joint swelling and inflammation in rats but also improved lung function, inhibited the pro-inflammatory factors IL-1ß and IL-6, reduced lung inflammation and collagen deposition, and alleviated lung fibrosis. CEP decreased the expression levels of TGFß-R2, p-SMAD, p-STAT3, and ECM proteins in rat lung tissues. TGF-ß1 induced HFL1 cells to highly express p-JAK2, with the most pronounced expression at 48 h. The levels of p-STAT3, p-SMAD3, and ECM-related proteins were significantly reduced after inhibition of either JAK2 or STAT3. CONCLUSION: JAK2 inhibitors may be an important and novel immunotherapeutic drug that can improve RA symptoms while also delaying or blocking the development of associated pulmonary fibrotic disease. The mechanism may be related to the downregulation of p-STAT3 protein via inhibition of the JAK2/STAT signaling pathway, which affects the phosphorylation of SMAD3.


Subject(s)
Isoquinolines , Janus Kinase Inhibitors , Lung , Pulmonary Fibrosis , Pyridines , Pyrroles , Signal Transduction , Smad3 Protein , Animals , Humans , Male , Rats , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Cell Line , Disease Models, Animal , Down-Regulation , Janus Kinase 2/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/enzymology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad3 Protein/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors
5.
Respir Res ; 25(1): 175, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654248

ABSTRACT

BACKGROUND: Two isoforms of Phosphoinositide 3-kinase (PI3K), p110γ and p110δ, are predominantly expressed in leukocytes and represent attractive therapeutic targets for the treatment of allergic asthma. The study aim was to assess the impact of administration of an inhaled PI3Kγδ inhibitor (AZD8154) in a rat model of asthma. METHODS: Firstly, we checked that the tool compound, AZD8154, inhibited rat PI3K γ & δ kinases using rat cell-based assays. Subsequently, a time-course study was conducted in a rat model of asthma to assess PI3K activity in the lung and how it is temporally associated with other key transcription pathways and asthma like features of the model. Finally, the impact on lung dosed AZD8154 on target engagement, pathway specificity, airway inflammation and lung function changes was assessed. RESULTS: Data showed that AZD8154 could inhibit rat PI3K γ & δ isoforms and, in a rat model of allergic asthma the PI3K pathway was activated in the lung. Intratracheal administration of AZD8154 caused a dose related suppression PI3K pathway activation (reduction in pAkt) and unlike after budesonide treatment, STAT and NF-κB pathways were not affected by AZD8154. The suppression of the PI3K pathway led to a marked inhibition of airway inflammation and reduction in changes in lung function. CONCLUSION: These data show that a dual PI3Kγδ inhibitor suppress key features of disease in a rat model of asthma to a similar degree as budesonide and indicate that dual PI3Kγδ inhibition may be an effective treatment for people suffering from allergic asthma.


Subject(s)
Asthma , Disease Models, Animal , Animals , Asthma/drug therapy , Asthma/metabolism , Rats , Male , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Rats, Sprague-Dawley , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/enzymology , Dose-Response Relationship, Drug , Protein Kinase Inhibitors/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Anti-Asthmatic Agents/pharmacology , Ovalbumin/toxicity
6.
Biomed Pharmacother ; 174: 116480, 2024 May.
Article in English | MEDLINE | ID: mdl-38547765

ABSTRACT

Sepsis is caused by an inadequate or dysregulated host response to infection. Enzymes causing cellular degradation are matrix metalloproteinases (MMPs). Lipopolysaccharide (LPS) is used in models of sepsis in laboratory settings The aim of the study was to measure MMP 2 and 12 concentrations in spleen and lungs in rats in which septic shock was induced by LPS. The experiment was carried out on 40 male Wistar rats (5 groups of 8): 0. controls 1. administered LPS 2. administered bestatin 3. LPS and bestatin 4.bestatin and after 6 hours LPS Animals were decapitated. Lungs and spleens were collected. Concentrations of MMP-2 and MMP-12 were determined using immunoenzymatic methods. Mean (±SD) MMP-2 in the controls was 43.57 ± 20.53 ng/ml in the lungs and 1.7 ± 0.72 ng/ml in the spleen; Group 1: 31.28 ± 13.13 ng/ml, 0.83 ± 0.8 ng/ml; Group 2: 44.24 ± 22.75 ng /ml, 1.01 ± 0.32 ng/ml; Group 3: 35.94 ± 15.13 ng/ml, 0.41 ± 0.03 ng/ml; Group 4:79.42 ± 44.70 ng/ml, 0.45 ± 0.15, respectively. Mean MMP-12 in controls was 19.79 ± 10.01 ng/ml in lungs and 41.13 ± 15.99 ng/ml in the spleen; Group 1:27.97 ± 15.1 ng/ml; 40.44 ± 11.2 ng/ml; Group 2: 37.93 ± 25.38 ng/ml 41.05 ± 18.08 ng/ml; Group 3: 40.59 ± 11.46 ng/ml, 35.16 ± 12.89 ng/ml; Group 4: 39.4 ± 17.83 ng/ml, 42.04 ± 12.35 ng/ml, respectively. CONCLUSIONS: 1. Bestatin reduces MMP 2 and 12 levels in spleen and lungs. 2. Treatment with bestatin minimizes the effect of LPS.


Subject(s)
Disease Models, Animal , Leucine , Leucine/analogs & derivatives , Lipopolysaccharides , Lung , Matrix Metalloproteinase 12 , Matrix Metalloproteinase 2 , Rats, Wistar , Sepsis , Spleen , Animals , Spleen/drug effects , Spleen/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Lung/drug effects , Lung/enzymology , Lung/pathology , Lung/metabolism , Sepsis/drug therapy , Sepsis/chemically induced , Matrix Metalloproteinase 12/metabolism , Rats , Leucine/pharmacology , Leucine/therapeutic use , Matrix Metalloproteinase Inhibitors/pharmacology
7.
Exp Mol Med ; 56(2): 478-490, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38413821

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung remodeling and the excessive accumulation of extracellular matrix (ECM) proteins. In a previous study, we found that the levels of ornithine aminotransferase (OAT), a principal enzyme in the proline metabolism pathway, were increased in the lungs of patients with IPF. However, the precise role played by OAT in the pathogenesis of IPF is not yet clear. The mechanism by which OAT affects fibrogenesis was assessed in vitro using OAT-overexpressing and OAT-knockdown lung fibroblasts. The therapeutic effects of OAT inhibition were assessed in the lungs of bleomycin-treated mice. OAT expression was increased in fibrotic areas, principally in interstitial fibroblasts, of lungs affected by IPF. OAT levels in the bronchoalveolar lavage fluid of IPF patients were inversely correlated with lung function. The survival rate was significantly lower in the group with an OAT level >75.659 ng/mL than in the group with an OAT level ≤75.659 ng/mL (HR, 29.53; p = 0.0008). OAT overexpression and knockdown increased and decreased ECM component production by lung fibroblasts, respectively. OAT knockdown also inhibited transforming growth factor-ß1 (TGF)-ß1 activity and TGF-ß1 pathway signaling. OAT overexpression increased the generation of mitochondrial reactive oxygen species (ROS) by activating proline dehydrogenase. The OAT inhibitor L-canaline significantly attenuated bleomycin-induced lung injury and fibrosis. In conclusion, increased OAT levels in lungs affected by IPF contribute to the progression of fibrosis by promoting excessive mitochondrial ROS production, which in turn activates TGF-ß1 signaling. OAT may be a useful target for treating patients with fibrotic lung diseases, including IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Transforming Growth Factor beta1 , Animals , Humans , Mice , Bleomycin , Extracellular Matrix Proteins , Fibrosis , Lung/enzymology , Ornithine-Oxo-Acid Transaminase , Reactive Oxygen Species
8.
Proc Natl Acad Sci U S A ; 119(24): e2201707119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35671428

ABSTRACT

A number of inflammatory lung diseases, including chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and pneumonia, are modulated by WNT/ß-catenin signaling. However, the underlying molecular mechanisms remain unclear. Here, starting with a forward genetic screen in mouse, we identify the WNT coreceptor Related to receptor tyrosine kinase (RYK) acting in mesenchymal tissues as a cell survival and antiinflammatory modulator. Ryk mutant mice exhibit lung hypoplasia and inflammation as well as alveolar simplification due to defective secondary septation, and deletion of Ryk specifically in mesenchymal cells also leads to these phenotypes. By analyzing the transcriptome of wild-type and mutant lungs, we observed the up-regulation of proapoptotic and inflammatory genes whose expression can be repressed by WNT/RYK signaling in vitro. Moreover, mesenchymal Ryk deletion at postnatal and adult stages can also lead to lung inflammation, thus indicating a continued role for WNT/RYK signaling in homeostasis. Our results indicate that RYK signaling through ß-catenin and Nuclear Factor kappa B (NF-κB) is part of a safeguard mechanism against mesenchymal cell death, excessive inflammatory cytokine production, and inflammatory cell recruitment and accumulation. Notably, RYK expression is down-regulated in the stromal cells of pneumonitis patient lungs. Altogether, our data reveal that RYK signaling plays critical roles as an antiinflammatory modulator during lung development and homeostasis and provide an animal model to further investigate the etiology of, and therapeutic approaches to, inflammatory lung diseases.


Subject(s)
Pneumonia , Receptor Protein-Tyrosine Kinases , Wnt Signaling Pathway , beta Catenin , Animals , Humans , Lung/enzymology , Lung/growth & development , Mesoderm/metabolism , Mice , NF-kappa B/metabolism , Pneumonia/enzymology , Pneumonia/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Stromal Cells/metabolism , beta Catenin/genetics , beta Catenin/metabolism
9.
J Pharm Biomed Anal ; 211: 114518, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35124452

ABSTRACT

The inhalation of peptides comes with the advantage of directly targeting the lung as tissue of interest. However, peptides are often rapidly metabolized in lung tissue through proteolytic cleavage. We have developed an assay workflow to obtain half-life and metabolite ID data for peptides incubated with four proteases abundant in lungs of asthma and COPD patients. The assay system has been validated using 28 structurally diverse linear and cyclic peptides with a molecular weight between 708 and 5808 Da. Experimental conditions for incubation, sample preparation, chromatography, data acquisition and analysis are compatible with the required throughput in early stage peptide projects. Together with co-crystal structures and Ala scans, we are using the described assay workflow to guide the first chemical modifications of peptide hits in early respiratory drug discovery projects.


Subject(s)
Peptide Hydrolases , Peptides , Administration, Inhalation , Asthma/drug therapy , Asthma/enzymology , High-Throughput Screening Assays , Humans , Lung/enzymology , Peptide Hydrolases/metabolism , Peptides/administration & dosage , Peptides/chemistry , Peptides/pharmacokinetics , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/enzymology
10.
Respir Res ; 23(1): 20, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35130915

ABSTRACT

BACKGROUND: Tyrosine kinase activation plays an important role in the progression of pulmonary fibrosis. In this study, we analyzed the expression of 612 kinase-coding and cancer-related genes using next-generation sequencing to identify potential therapeutic targets for idiopathic pulmonary fibrosis (IPF). METHODS: Thirteen samples from five patients with IPF (Cases 1-5) and eight samples from four patients without IPF (control) were included in this study. Six of the thirteen samples were obtained from different lung segments of a single patient who underwent bilateral pneumonectomy. Gene expression analysis of IPF lung tissue samples (n = 13) and control samples (n = 8) was performed using SureSelect RNA Human Kinome Kit. The expression of the selected genes was further confirmed at the protein level by immunohistochemistry (IHC). RESULTS: Gene expression analysis revealed a correlation between the gene expression signatures and the degree of fibrosis, as assessed by Ashcroft score. In addition, the expression analysis indicated a stronger heterogeneity among the IPF lung samples than among the control lung samples. In the integrated analysis of the 21 samples, DCLK1 and STK33 were found to be upregulated in IPF lung samples compared to control lung samples. However, the top most upregulated genes were distinct in individual cases. DCLK1, PDK4, and ERBB4 were upregulated in IPF case 1, whereas STK33, PIM2, and SYK were upregulated in IPF case 2. IHC revealed that these proteins were expressed in the epithelial layer of the fibrotic lesions. CONCLUSIONS: We performed a comprehensive kinase expression analysis to explore the potential therapeutic targets for IPF. We found that DCLK1 and STK33 may serve as potential candidate targets for molecular targeted therapy of IPF. In addition, PDK4, ERBB4, PIM2, and SYK might also serve as personalized therapeutic targets of IPF. Additional large-scale studies are warranted to develop personalized therapies for patients with IPF.


Subject(s)
Gene Expression Regulation , Idiopathic Pulmonary Fibrosis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lung/enzymology , Phosphotransferases/genetics , RNA/genetics , Aged , Female , Follow-Up Studies , High-Throughput Nucleotide Sequencing/methods , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Immunohistochemistry , Male , Middle Aged , Phosphotransferases/biosynthesis , Retrospective Studies
11.
Nat Commun ; 13(1): 318, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35031603

ABSTRACT

Lung emphysema and chronic bronchitis are the two most common causes of chronic obstructive pulmonary disease. Excess macrophage elastase MMP-12, which is predominantly secreted from alveolar macrophages, is known to mediate the development of lung injury and emphysema. Here, we discovered the endolysosomal cation channel mucolipin 3 (TRPML3) as a regulator of MMP-12 reuptake from broncho-alveolar fluid, driving in two independently generated Trpml3-/- mouse models enlarged lung injury, which is further exacerbated after elastase or tobacco smoke treatment. Mechanistically, using a Trpml3IRES-Cre/eR26-τGFP reporter mouse model, transcriptomics, and endolysosomal patch-clamp experiments, we show that in the lung TRPML3 is almost exclusively expressed in alveolar macrophages, where its loss leads to defects in early endosomal trafficking and endocytosis of MMP-12. Our findings suggest that TRPML3 represents a key regulator of MMP-12 clearance by alveolar macrophages and may serve as therapeutic target for emphysema and chronic obstructive pulmonary disease.


Subject(s)
Macrophages, Alveolar/enzymology , Matrix Metalloproteinase 12/metabolism , Pancreatic Elastase/metabolism , Pulmonary Emphysema/enzymology , Transient Receptor Potential Channels/deficiency , Animals , Disease Models, Animal , Endosomes/metabolism , Female , Humans , Lung/enzymology , Matrix Metalloproteinase 12/genetics , Mice , Mice, Knockout , Pancreatic Elastase/genetics , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Transient Receptor Potential Channels/genetics
12.
Am J Physiol Regul Integr Comp Physiol ; 322(2): R126-R135, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34984926

ABSTRACT

Many lung diseases are caused by an excessive inflammatory response, and inflammatory lung diseases are often modeled using lipopolysaccharide (LPS) in mice. Cyclooxygenase-2 (COX-2) encoded by the Ptgs2 gene is induced in response to inflammatory stimuli including LPS. The objective of this study was to test the hypothesis that mice deficient in COX-2 (Ptgs2-/-) will be protected from LPS-induced lung injury. Wild-type (WT; CD1 mice) and Ptgs2-/- mice (on a CD1 background) were treated with LPS or vehicle for 24 h. LPS treatment resulted in histological evidence of lung injury, which was attenuated in the Ptgs2-/- mice. LPS treatment increased the mRNA levels for tumor necrosis factor-α, interleukin-10, and monocyte chemoattractant protein-1 in the lungs of WT mice, and the LPS-induced increases in these levels were attenuated in the Ptgs2-/- mice. The protein levels of active caspase-3 and caspase-9 were lower in the LPS-treated lungs of Ptgs2-/- mice than in LPS-treated WT mice, as were the number of terminal deoxynucleotide transferase dUTP nick end labeling-positive cells in lung sections. LPS exposure resulted in a greater lung wet-to-dry weight ratio (W/D) in WT mice, suggestive of pulmonary edema, while in LPS-treated Ptgs2-/- mice, the W/D was not different from controls and less than in LPS-treated WT mice. These results demonstrate that COX-2 is involved in the inflammatory response to LPS and suggest that COX-2 not only acts as a downstream participant in the inflammatory response, but also acts as a regulator of the inflammatory response likely through a feed-forward mechanism following LPS stimulation.


Subject(s)
Acute Lung Injury/prevention & control , Apoptosis , Cyclooxygenase 2/deficiency , Lung/enzymology , Pneumonia/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2/genetics , Disease Models, Animal , Female , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopolysaccharides , Lung/pathology , Male , Mice, Knockout , Pneumonia/chemically induced , Pneumonia/enzymology , Pneumonia/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L204-L223, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34878944

ABSTRACT

During newborn lung injury, excessive activity of lysyl oxidases (LOXs) disrupts extracellular matrix (ECM) formation. Previous studies indicate that TGFß activation in the O2-injured mouse pup lung increases lysyl oxidase (LOX) expression. But how TGFß regulates this, and whether the LOXs generate excess pulmonary aldehydes are unknown. First, we determined that O2-mediated lung injury increases LOX protein expression in TGFß-stimulated pup lung interstitial fibroblasts. This regulation appeared to be direct; this is because TGFß treatment also increased LOX protein expression in isolated pup lung fibroblasts. Then using a fibroblast cell line, we determined that TGFß stimulates LOX expression at a transcriptional level via Smad2/3-dependent signaling. LOX is translated as a pro-protein that requires secretion and extracellular cleavage before assuming amine oxidase activity and, in some cells, reuptake with nuclear localization. We found that pro-LOX is processed in the newborn mouse pup lung. Also, O2-mediated injury was determined to increase pro-LOX secretion and nuclear LOX immunoreactivity particularly in areas populated with interstitial fibroblasts and exhibiting malformed ECM. Then, using molecular probes, we detected increased aldehyde levels in vivo in O2-injured pup lungs, which mapped to areas of increased pro-LOX secretion in lung sections. Increased activity of LOXs plays a critical role in the aldehyde generation; an inhibitor of LOXs prevented the elevation of aldehydes in the O2-injured pup lung. These results reveal new mechanisms of TGFß and LOX in newborn lung disease and suggest that aldehyde-reactive probes might have utility in sensing the activation of LOXs in vivo during lung injury.


Subject(s)
Aldehydes/metabolism , Lung Injury/metabolism , Lung/enzymology , Lung/pathology , Protein-Lysine 6-Oxidase/metabolism , Aldehydes/chemistry , Animals , Animals, Newborn , Embryo, Mammalian/pathology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic , Mice , Mice, Inbred C57BL , Molecular Probes/metabolism , NIH 3T3 Cells , Protein-Lysine 6-Oxidase/genetics , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Signal Transduction , Smad Proteins/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism
14.
Biosci Rep ; 42(1)2022 01 28.
Article in English | MEDLINE | ID: mdl-34750610

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a respiratory disease with high morbidity and mortality rates. Currently, there is no effective treatment to complement mechanical ventilation. Exosomes and microRNAs (miRNAs) are promising agents for the management of this disease. METHODS: Exosomes were isolated from mouse bone marrow stromal stem cells (BMSCs). The levels of two miRNAs, miR-542-3P and miR-150, in exosomes were determined using RT-PCR, and miR-150 was selected for further study. ALI model was established in mice using lipopolysaccharides, and then, they were treated with saline, exosomes, miRNA agomirs, or miRNA antagomirs. The concentrations of TNF-α, IL-6, and IL-1ß and the number of neutrophils and macrophages in the bronchoalveolar lavage fluid were measured. The wet/dry weight ratio of the lung tissue was calculated, and tissue pathology and apoptosis were observed using hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. CD34 and VE-cadherin expression was detected using immunofluorescence. Proteins associated with apoptosis and MAPK signaling were detected using Western blotting, and miR-150 expression in lung tissue was evaluated using RT-PCR. RESULTS: We successfully isolated BMSCs and exosomes and showed that the level of miR-150 was significantly higher than that of miR-542-3p. Exosomes and miR-150 reduced inflammation and lung edema while maintaining the integrity of the alveolar structure. They also mitigated microvascular endothelial cell injury by regulating the caspase-3, Bax/Bcl-2, and MAPK signaling. CONCLUSIONS: Exosomal miR-150 attenuates lipopolysaccharide-induced ALI through the MAPK pathway.


Subject(s)
Acute Lung Injury/prevention & control , Exosomes/transplantation , Lung/blood supply , Mesenchymal Stem Cell Transplantation , MicroRNAs/metabolism , Microvessels/enzymology , Mitogen-Activated Protein Kinases/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Exosomes/genetics , Exosomes/metabolism , Lipopolysaccharides , Lung/enzymology , Lung/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , Signal Transduction
15.
J Mol Cell Cardiol ; 164: 13-16, 2022 03.
Article in English | MEDLINE | ID: mdl-34774871

ABSTRACT

Aged males disproportionately succumb to increased COVID-19 severity, hospitalization, and mortality compared to females. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease, serine 2 (TMPRSS2) facilitate SARS-CoV-2 viral entry and may have sexually dimorphic regulation. As viral load dictates disease severity, we investigated the expression, protein levels, and activity of ACE2 and TMPRSS2. Our data reveal that aged males have elevated ACE2 in both mice and humans across organs. We report the first comparative study comprehensively investigating the impact of sex and age in murine and human levels of ACE2 and TMPRSS2, to begin to elucidate the sex bias in COVID-19 severity.


Subject(s)
Aging/metabolism , Angiotensin-Converting Enzyme 2/biosynthesis , COVID-19/epidemiology , Gene Expression Regulation, Enzymologic , Receptors, Virus/biosynthesis , SARS-CoV-2/physiology , Sex Characteristics , Aging/genetics , Angiotensin-Converting Enzyme 2/genetics , Animals , Disease Susceptibility , Female , Heart/virology , Humans , Intestine, Small/enzymology , Intestine, Small/virology , Kidney/enzymology , Kidney/virology , Lung/enzymology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardium/enzymology , Organ Specificity , Receptors, Virus/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Young Adult
16.
J Pathol ; 256(3): 256-261, 2022 03.
Article in English | MEDLINE | ID: mdl-34859884

ABSTRACT

COVID-19 is a pandemic with high morbidity and mortality. In an autopsy cohort of COVID-19 patients, we found extensive accumulation of the tryptophan degradation products 3-hydroxy-anthranilic acid and quinolinic acid in the lungs, heart, and brain. This was not related to the expression of the tryptophan-catabolizing indoleamine 2,3-dioxygenase (IDO)-1, but rather to that of its isoform IDO-2, which otherwise is expressed rarely. Bioavailability of tryptophan is an absolute requirement for proper cell functioning and synthesis of hormones, whereas its degradation products can cause cell death. Markers of apoptosis and severe cellular stress were associated with IDO-2 expression in large areas of lung and heart tissue, whereas affected areas in brain were more restricted. Analyses of tissue, cerebrospinal fluid, and sequential plasma samples indicate early initiation of the kynurenine/aryl-hydrocarbon receptor/IDO-2 axis as a positive feedback loop, potentially leading to severe COVID-19 pathology. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Brain/enzymology , COVID-19/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Lung/enzymology , Myocardium/enzymology , 3-Hydroxyanthranilic Acid/analysis , Adult , Aged , Apoptosis , Autopsy , Brain/pathology , COVID-19/mortality , COVID-19/pathology , COVID-19/virology , Humans , Kynurenine/analysis , Lung/pathology , Middle Aged , Myocardium/pathology , Prospective Studies , Quinolinic Acid/analysis , Severity of Illness Index , Tryptophan/analysis
17.
Cell Rep ; 37(6): 109921, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758300

ABSTRACT

Regulatory T (Treg) cells are critical for immunological tolerance and immune homeostasis. Treg cells strongly rely on mitochondrial metabolism and show a lower level of glycolysis. However, little is known about the role of lipid metabolism in the regulation of Treg cell homeostasis. Some members of the ACSL family of acyl-coenzyme A (CoA) synthases are expressed in T cells, but their function remains unclear. A combination of RNA-sequencing and proteome analyses shows that Acsbg1, a member of ACSL, is selectively expressed in Treg cells. We show that the genetic deletion of Acsbg1 not only causes mitochondrial dysfunction, but it also dampens other metabolic pathways. The extrinsic supplementation of Acsbg1-deficient Treg cells with oleoyl-CoA restores the phenotype of the Treg metabolic signature. Furthermore, this pathway in ST2+ effector Treg cells enhances immunosuppressive capacity in airway inflammation. Thus, Acsbg1 serves as a metabolic checkpoint governing Treg cell homeostasis and the resolution of lung inflammation.


Subject(s)
Coenzyme A Ligases/metabolism , Energy Metabolism , Lung/enzymology , Mitochondria/enzymology , Pneumonia/enzymology , T-Lymphocytes, Regulatory/enzymology , Animals , Coenzyme A Ligases/genetics , Disease Models, Animal , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Homeostasis , Interleukin-33 , Lung/immunology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , Organelle Biogenesis , Pneumonia/genetics , Pneumonia/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology
18.
Int J Mol Sci ; 22(22)2021 Nov 14.
Article in English | MEDLINE | ID: mdl-34830188

ABSTRACT

Cytochrome P450 2A13 is an omitted brother of CYP2A6 that has an important role in the drug metabolism of liver. Due to extrahepatic expression, it has gained less attention than CYP2A6, despite the fact that it plays a significant role in toxicant-induced pulmonary lesions and, therefore, lung cancer. The purpose of this mini-review is to summarize the basic knowledge about this enzyme in relation to the substrates, inhibitors, genetic polymorphisms, and transcriptional regulation that are known so far (September 2021).


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung/metabolism , Polymorphism, Genetic , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/metabolism , Humans , Lung/enzymology , Lung/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Methoxsalen/pharmacology , Substrate Specificity
19.
Toxicol Appl Pharmacol ; 432: 115757, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34673086

ABSTRACT

An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1ß, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-ß1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-ß1/SMAD pathways.


Subject(s)
HMGB1 Protein/metabolism , Lung/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Pneumonia/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/enzymology , Receptor for Advanced Glycation End Products/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Arsenites , Collagen/metabolism , Cytokines/metabolism , Disease Models, Animal , Lung/ultrastructure , Male , Phosphorylation , Pneumonia/chemically induced , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats, Wistar , Signal Transduction , Sodium Compounds
20.
Biomed Pharmacother ; 144: 112340, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34678729

ABSTRACT

Fibrosis is an outcome of tissue repair after different types of injuries. The homeostasis of extracellular matrix is broken, and excessive deposition occurs, affecting the normal function of tissues and organs, which could become prostrated in serious cases.Finding a suitable target to regulate the repair process and reduce the damage caused by fibrosis is a hot research topic at present. The TRIM family is number of one of the E3 ubiquitin ligase subfamilies and participates in various biological processes including intracellular signal transduction, apoptosis, autophagy, and immunity by regulating the ubiquitination of target proteins. For the past few years, the important role of TRIM in the occurrence and development of fibrosis has been gradually revealed. In this review, we focus on the recent emerging topics on TRIM proteins in the regulation of fibrosis, fibrosis-related cytokines and pathways.


Subject(s)
Extracellular Matrix/metabolism , Liver Cirrhosis/enzymology , Liver/enzymology , Lung/enzymology , Pulmonary Fibrosis/enzymology , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Antifibrotic Agents/therapeutic use , Cytokines/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibrosis , Humans , Inflammation Mediators/metabolism , Liver/drug effects , Liver/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Lung/drug effects , Lung/pathology , Protein Conformation , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Signal Transduction , Structure-Activity Relationship , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/chemistry , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL