Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
FASEB J ; 35(5): e21525, 2021 05.
Article in English | MEDLINE | ID: mdl-33817836

ABSTRACT

Glycolysis is a well-known process by which metabolically active cells, such as tumor or immune cells meet their high metabolic demands. Previously, our laboratory has demonstrated that in airway epithelial cells, the pleiotropic cytokine, interleukin-1 beta (IL1B) induces glycolysis and that this contributes to allergic airway inflammation and remodeling. Activation of glycolysis is known to increase NADPH reducing equivalents generated from the pentose phosphate pathway, linking metabolic reprogramming with redox homeostasis. In addition, numerous glycolytic enzymes are known to be redox regulated. However, whether and how redox chemistry regulates metabolic reprogramming more generally remains unclear. In this study, we employed a multi-omics approach in primary mouse airway basal cells to evaluate the role of protein redox biochemistry, specifically protein glutathionylation, in mediating metabolic reprogramming. Our findings demonstrate that IL1B induces glutathionylation of multiple proteins involved in metabolic regulation, notably in the glycolysis pathway. Cells lacking Glutaredoxin-1 (Glrx), the enzyme responsible for reversing glutathionylation, show modulation of multiple metabolic pathways including an enhanced IL1B-induced glycolytic response. This was accompanied by increased secretion of thymic stromal lymphopoietin (TSLP), a cytokine important in asthma pathogenesis. Targeted inhibition of glycolysis prevented TSLP release, confirming the functional relevance of enhanced glycolysis in cells stimulated with IL1B. Collectively, data herein point to an intriguing link between glutathionylation chemistry and glycolytic reprogramming in epithelial cells and suggest that glutathionylation chemistry may represent a therapeutic target in pulmonary pathologies with perturbations in the glycolysis pathway.


Subject(s)
Cellular Reprogramming , Glutaredoxins/physiology , Glutathione/metabolism , Glycolysis , Inflammation/immunology , Interleukin-1beta/pharmacology , Lung/immunology , Animals , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Lung/cytology , Lung/drug effects , Lung/metabolism , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction
2.
J Immunol ; 204(4): 763-774, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31924651

ABSTRACT

Asthma is a chronic disorder characterized by inflammation, mucus metaplasia, airway remodeling, and hyperresponsiveness. We recently showed that IL-1-induced glycolytic reprogramming contributes to allergic airway disease using a murine house dust mite model. Moreover, levels of pyruvate kinase M2 (PKM2) were increased in this model as well as in nasal epithelial cells from asthmatics as compared with healthy controls. Although the tetramer form of PKM2 converts phosphoenolpyruvate to pyruvate, the dimeric form of PKM2 has alternative, nonglycolysis functions as a transcriptional coactivator to enhance the transcription of several proinflammatory cytokines. In the current study, we examined the impact of PKM2 on the pathogenesis of house dust mite-induced allergic airways disease in C57BL/6NJ mice. We report, in this study, that activation of PKM2, using the small molecule activator, TEPP46, augmented PKM activity in lung tissues and attenuated airway eosinophils, mucus metaplasia, and subepithelial collagen. TEPP46 attenuated IL-1ß-mediated airway inflammation and expression of proinflammatory mediators. Exposure to TEPP46 strongly decreased the IL-1ß-mediated increases in thymic stromal lymphopoietin (TSLP) and GM-CSF in primary tracheal epithelial cells isolated from C57BL/6NJ mice. We also demonstrate that IL-1ß-mediated increases in nuclear phospho-STAT3 were decreased by TEPP46. Finally, STAT3 inhibition attenuated the IL-1ß-induced release of TSLP and GM-CSF, suggesting that the ability of PKM2 to phosphorylate STAT3 contributes to its proinflammatory function. Collectively, these results demonstrate that the glycolysis-inactive form of PKM2 plays a crucial role in the pathogenesis of allergic airways disease by increasing IL-1ß-induced proinflammatory signaling, in part, through phosphorylation of STAT3.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Pneumonia/immunology , Pyruvate Kinase/immunology , Signal Transduction/immunology , Airway Remodeling/physiology , Animals , Asthma/metabolism , Female , Hypersensitivity/metabolism , Male , Mice , Mice, Inbred C57BL , Pneumonia/metabolism , Pyroglyphidae/immunology , Pyruvate Kinase/metabolism
3.
Am J Respir Cell Mol Biol ; 64(6): 709-721, 2021 06.
Article in English | MEDLINE | ID: mdl-33662229

ABSTRACT

Obesity is a risk factor for the development of asthma and represents a difficult-to-treat disease phenotype. Aerobic glycolysis is emerging as a key feature of asthma, and changes in glucose metabolism are linked to leukocyte activation and adaptation to oxidative stress. Dysregulation of PKM2 (pyruvate kinase M2), the enzyme that catalyzes the last step of glycolysis, contributes to house dust mite (HDM)-induced airway inflammation and remodeling in lean mice. It remains unclear whether glycolytic reprogramming and dysregulation of PKM2 also contribute to obese asthma. The goal of the present study was to elucidate the functional role of PKM2 in a murine model of obese allergic asthma. We evaluated the small molecule activator of PKM2, TEPP46, and assessed the role of PKM2 using conditional ablation of the Pkm2 allele from airway epithelial cells. In obese C57BL/6NJ mice, parameters indicative of glycolytic reprogramming remained unchanged in the absence of stimulation with HDM. Obese mice that were subjected to HDM showed evidence of glycolytic reprogramming, and treatment with TEPP46 diminished airway inflammation, whereas parameters of airway remodeling were unaffected. Epithelial ablation of Pkm2 decreased central airway resistance in both lean and obese allergic mice in addition to decreasing inflammatory cytokines in the lung tissue. Lastly, we highlight a novel role for PKM2 in the regulation of glutathione-dependent protein oxidation in the lung tissue of obese allergic mice via a putative IFN-γ-glutaredoxin1 pathway. Overall, targeting metabolism and protein oxidation may be a novel treatment strategy for obese allergic asthma.


Subject(s)
Asthma/enzymology , Asthma/pathology , Hypersensitivity/enzymology , Hypersensitivity/pathology , Inflammation/enzymology , Inflammation/pathology , Pyruvate Kinase/metabolism , Animals , Asthma/complications , Asthma/parasitology , Bronchial Hyperreactivity/complications , Diet, High-Fat , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glutathione/metabolism , Glycolysis , Homeostasis/drug effects , Hypersensitivity/complications , Hypersensitivity/parasitology , Inflammation Mediators/metabolism , Lung/enzymology , Lung/pathology , Mice, Inbred C57BL , Mice, Obese , Models, Biological , Pyridazines/administration & dosage , Pyridazines/pharmacology , Pyroglyphidae , Pyrroles/administration & dosage , Pyrroles/pharmacology
4.
Histochem Cell Biol ; 155(2): 301-308, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33459870

ABSTRACT

The lung is comprised of more than 40 distinct cell types that support a complex 3-dimensional (3D) architecture that is required for efficient lung function. Loss of this proper architecture can accommodate and promote lung disease, highlighting researchers' growing need to analyze lung structures in detail. Additionally, in vivo cellular and molecular response to chemical and physical signals, along with the recapitulation of gene-expression patterns, can be lost during the transition from complex 3D tissues to 2D cell culture systems. Therefore, technologies that allow for the investigation of lung function under normal and disease states utilizing the entirety of the lung architecture are required to generate a complete understanding of these processes. Airway cell-derived organoids that can recapitulate lung structure and function ex vivo while being amenable to experimental manipulation, have provided a new and exciting model system to investigate lung biology. In this perspective, we discuss emerging technologies for culturing lung-derived organoids, techniques to visualize organoids using high-resolution microscopy and the resulting information extracted from organoids supporting research focused on lung function and diseases.


Subject(s)
Cell Culture Techniques , Imaging, Three-Dimensional , Lung/cytology , Organoids/cytology , Animals , Humans , Lung/metabolism , Microscopy, Fluorescence , Organoids/metabolism
5.
Am J Physiol Cell Physiol ; 318(2): C304-C327, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31693398

ABSTRACT

Glutathione is a major redox buffer, reaching millimolar concentrations within cells and high micromolar concentrations in airways. While glutathione has been traditionally known as an antioxidant defense mechanism that protects the lung tissue from oxidative stress, glutathione more recently has become recognized for its ability to become covalently conjugated to reactive cysteines within proteins, a modification known as S-glutathionylation (or S-glutathiolation or protein mixed disulfide). S-glutathionylation has the potential to change the structure and function of the target protein, owing to its size (the addition of three amino acids) and charge (glutamic acid). S-glutathionylation also protects proteins from irreversible oxidation, allowing them to be enzymatically regenerated. Numerous enzymes have been identified to catalyze the glutathionylation/deglutathionylation reactions, including glutathione S-transferases and glutaredoxins. Although protein S-glutathionylation has been implicated in numerous biological processes, S-glutathionylated proteomes have largely remained unknown. In this paper, we focus on the pathways that regulate GSH homeostasis, S-glutathionylated proteins, and glutaredoxins, and we review methods required toward identification of glutathionylated proteomes. Finally, we present the latest findings on the role of glutathionylation/glutaredoxins in various lung diseases: idiopathic pulmonary fibrosis, asthma, and chronic obstructive pulmonary disease.


Subject(s)
Glutaredoxins/metabolism , Glutathione/metabolism , Lung Diseases/metabolism , Lung/metabolism , Amino Acid Sequence , Animals , Antioxidants/metabolism , Cysteine/metabolism , Disulfides/metabolism , Humans , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Oxidative Stress/physiology
6.
Am J Physiol Endocrinol Metab ; 318(1): E22-E32, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31689144

ABSTRACT

Factors secreted from tumors/tumor cells are hypothesized to cause skeletal muscle wasting in cancer patients. We examined whether cancer cells secrete factors to promote atrophy by evaluating the effects of conditioned media (CM) from murine lung cancer cells and primary cultures of human lung tumor cells on cultured myotubes. We evaluated murine Lewis lung carcinoma (LLC) and KRASG12D cells, and primary cell lines derived from tumor biopsies from patients with lung cancer (hTCM; n = 6). In all experiments, serum content was matched across treatment groups. We hypothesized that CM from murine and human tumor cells would reduce myotube myosin content, decrease mitochondrial content, and increase mitochondrial reactive oxygen species (ROS) production. Treatment of myotubes differentiated for 7 days with CM from LLC and KRASG12D cells did not alter any of these variables. Effects of murine tumor cell CM were observed when myotubes differentiated for 4 days were treated with tumor cell CM and compared with undiluted differentiation media. However, these effects were not apparent if tumor cell CM treatments were compared with control cell CM or dilution controls. Finally, CM from human lung tumor primary cell lines did not modify myosin content or mitochondrial content or ROS production compared with either undiluted differentiated media, control cell CM, or dilution controls. Our results do not support the hypothesis that factors released from cultured lung cancer/tumor cells promote myotube wasting or mitochondrial abnormalities, but we cannot dismiss the possibility that these cells could secrete such factors in vivo within the native tumor microenvironment.


Subject(s)
Cachexia/metabolism , Carcinoma, Lewis Lung/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Culture Media, Conditioned/pharmacology , Lung Neoplasms/metabolism , Mitochondria, Muscle/drug effects , Muscle Fibers, Skeletal/drug effects , Myosins/metabolism , Adenocarcinoma/metabolism , Aged , Aged, 80 and over , Animals , Cachexia/etiology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Female , Humans , Male , Mice , Middle Aged , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal , Neoplasms/complications , Neoplasms/metabolism , Primary Cell Culture , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
7.
J Allergy Clin Immunol ; 142(2): 435-450.e10, 2018 08.
Article in English | MEDLINE | ID: mdl-29108965

ABSTRACT

BACKGROUND: Emerging studies suggest that enhanced glycolysis accompanies inflammatory responses. Virtually nothing is known about the relevance of glycolysis in patients with allergic asthma. OBJECTIVES: We sought to determine whether glycolysis is altered in patients with allergic asthma and to address its importance in the pathogenesis of allergic asthma. METHODS: We examined alterations in glycolysis in sputum samples from asthmatic patients and primary human nasal cells and used murine models of allergic asthma, as well as primary mouse tracheal epithelial cells, to evaluate the relevance of glycolysis. RESULTS: In a murine model of allergic asthma, glycolysis was induced in the lungs in an IL-1-dependent manner. Furthermore, administration of IL-1ß into the airways stimulated lactate production and expression of glycolytic enzymes, with notable expression of lactate dehydrogenase A occurring in the airway epithelium. Indeed, exposure of mouse tracheal epithelial cells to IL-1ß or IL-1α resulted in increased glycolytic flux, glucose use, expression of glycolysis genes, and lactate production. Enhanced glycolysis was required for IL-1ß- or IL-1α-mediated proinflammatory responses and the stimulatory effects of IL-1ß on house dust mite (HDM)-induced release of thymic stromal lymphopoietin and GM-CSF from tracheal epithelial cells. Inhibitor of κB kinase ε was downstream of HDM or IL-1ß and required for HDM-induced glycolysis and pathogenesis of allergic airways disease. Small interfering RNA ablation of lactate dehydrogenase A attenuated HDM-induced increases in lactate levels and attenuated HDM-induced disease. Primary nasal epithelial cells from asthmatic patients intrinsically produced more lactate compared with cells from healthy subjects. Lactate content was significantly higher in sputum supernatants from asthmatic patients, notably those with greater than 61% neutrophils. A positive correlation was observed between sputum lactate and IL-1ß levels, and lactate content correlated negatively with lung function. CONCLUSIONS: Collectively, these findings demonstrate that IL-1ß/inhibitory κB kinase ε signaling plays an important role in HDM-induced glycolysis and pathogenesis of allergic airways disease.


Subject(s)
Asthma/metabolism , Hypersensitivity/metabolism , Interleukin-1beta/metabolism , Lung/metabolism , Nose/pathology , Respiratory Mucosa/metabolism , Sputum/metabolism , Animals , Antigens, Dermatophagoides/immunology , Cells, Cultured , Cohort Studies , Disease Models, Animal , Female , Glycolysis , Humans , I-kappa B Proteins/metabolism , Interleukin-1beta/genetics , Lactic Acid/metabolism , Lung/pathology , Male , Mice , Middle Aged , Neutrophils/pathology , Proto-Oncogene Proteins/metabolism , Pyroglyphidae , RNA, Small Interfering/genetics , Respiratory Mucosa/pathology , Signal Transduction
8.
J Cell Physiol ; 233(4): 3487-3497, 2018 04.
Article in English | MEDLINE | ID: mdl-28926105

ABSTRACT

The mammalian runt-related factor 1 (RUNX1) is a master transcription factor that regulates lineage specification of hematopoietic stem cells. RUNX1 translocations result in the development of myeloid leukemias. Recently, RUNX1 has been implicated as a tumor suppressor in other cancers. We postulated RUNX1 expression may be associated with lung adenocarcinoma etiology and/or progression. We evaluated the association of RUNX1 mRNA expression with overall survival data from The Cancer Genome Atlas (TCGA), a publically available database. Compared to high expression levels, Low RUNX1 levels from lung adenocarcinomas were associated with a worse overall survival (Hazard Ratio = 2.014 (1.042-3.730 95% confidence interval), log-rank p = 0.035) compared to those that expressed high RUNX1 levels. Further immunohistochemical examination of 85 surgical specimens resected at the University of Vermont Medical Center identified that low RUNX1 protein expression was associated with larger tumors (p = 0.038). Gene expression network analysis was performed on the same subset of TCGA cases that demonstrated differential survival by RUNX1 expression. This analysis, which reveals regulatory relationships, showed that reduced RUNX1 levels were closely linked to upregulation of the transcription factor E2F1. To interrogate this relationship, RUNX1 was depleted in a lung cancer cell line that expresses high levels of RUNX1. Loss of RUNX1 resulted in enhanced proliferation, migration, and invasion. RUNX1 depletion also resulted in increased mRNA expression of E2F1 and multiple E2F1 target genes. Our data implicate loss of RUNX1 as driver of lung adenocarcinoma aggression, potentially through deregulation of the E2F1 pathway.


Subject(s)
Adenocarcinoma/metabolism , Aggression/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation/physiology , Lung Neoplasms/metabolism , Adenocarcinoma of Lung/metabolism , Cell Proliferation/physiology , Gene Expression Profiling/methods , Hematopoietic Stem Cells/metabolism , Humans , Transcriptional Activation/physiology
9.
Am J Physiol Lung Cell Mol Physiol ; 314(6): L984-L997, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29469614

ABSTRACT

Epithelial cells have been suggested as potential drivers of lung fibrosis, although the epithelial-dependent pathways that promote fibrogenesis remain unknown. Extracellular matrix is increasingly recognized as an environment that can drive cellular responses in various pulmonary diseases. In this study, we demonstrate that transforming growth factor-ß1 (TGF-ß1)-stimulated mouse tracheal basal (MTB) cells produce provisional matrix proteins in vitro, which initiate mesenchymal changes in subsequently freshly plated MTB cells via Rho kinase- and c-Jun NH2-terminal kinase (JNK1)-dependent processes. Repopulation of decellularized lung scaffolds, derived from mice with bleomycin-induced fibrosis or from patients with idiopathic pulmonary fibrosis, with wild-type MTB cells resulted in a loss of epithelial gene expression and augmentation of mesenchymal gene expression compared with cells seeded into decellularized normal lungs. In contrast, Jnk1-/- basal cells seeded into fibrotic lung scaffolds retained a robust epithelial expression profile, failed to induce mesenchymal genes, and differentiated into club cell secretory protein-expressing cells. This new paradigm wherein TGF-ß1-induced extracellular matrix derived from MTB cells activates a JNK1-dependent mesenchymal program, which impedes subsequent normal epithelial cell homeostasis, provides a plausible scenario of chronic aberrant epithelial repair, thought to be critical in lung fibrogenesis. This study identifies JNK1 as a possible target for inhibition in settings wherein reepithelialization is desired.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Extracellular Matrix/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Pulmonary Fibrosis/metabolism , Respiratory Mucosa/pathology , Trachea/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Respiratory Mucosa/metabolism , Trachea/pathology , Transforming Growth Factor beta1/genetics
10.
Am J Respir Cell Mol Biol ; 56(3): 393-401, 2017 03.
Article in English | MEDLINE | ID: mdl-27875656

ABSTRACT

Lung tissue remodeling in chronic obstructive pulmonary disease (COPD) is characterized by airway wall thickening and/or emphysema. Although the bronchial and alveolar compartments are functionally independent entities, we recently showed comparable alterations in matrix composition comprised of decreased elastin content and increased collagen and hyaluronan contents of alveolar and small airway walls. Out of several animal models tested, surfactant protein C (SPC)-TNF-α mice showed remodeling in alveolar and airway walls similar to what we observed in patients with COPD. Epithelial cells are able to undergo a phenotypic shift, gaining mesenchymal properties, a process in which c-Jun N-terminal kinase (JNK) signaling is involved. Therefore, we hypothesized that TNF-α induces JNK-dependent epithelial plasticity, which contributes to lung matrix remodeling. To this end, the ability of TNF-α to induce a phenotypic shift was assessed in A549, BEAS2B, and primary bronchial epithelial cells, and phenotypic markers were studied in SPC-TNF-α mice. Phenotypic markers of mesenchymal cells were elevated both in vitro and in vivo, as shown by the expression of vimentin, plasminogen activator inhibitor-1, collagen, and matrix metalloproteinases. Concurrently, the expression of the epithelial markers, E-cadherin and keratin 7 and 18, was attenuated. A pharmacological inhibitor of JNK attenuated this phenotypic shift in vitro, demonstrating involvement of JNK signaling in this process. Interestingly, activation of JNK signaling was also clearly present in lungs of SPC-TNF-α mice and patients with COPD. Together, these data show a role for TNF-α in the induction of a phenotypic shift in vitro, resulting in increased collagen production and the expression of elastin-degrading matrix metalloproteinases, and provide evidence for involvement of the TNF-α-JNK axis in extracellular matrix remodeling.


Subject(s)
Extracellular Matrix/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Biomarkers/metabolism , Cell Hypoxia/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Matrix/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Lung/drug effects , Lung/metabolism , Mesoderm/metabolism , Mice , Phenotype , Phosphorylation/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Surfactant-Associated Protein C/metabolism , Signal Transduction/drug effects
12.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L866-75, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24610935

ABSTRACT

Chronic allergic asthma leads to airway remodeling and subepithelial fibrosis via mechanisms not fully understood. Airway remodeling is amplified by profibrotic mediators, such as transforming growth factor-ß1 (TGF-ß1), which plays a cardinal role in various models of fibrosis. We recently have identified a critical role for c-Jun-NH2-terminal-kinase (JNK) 1 in augmenting the profibrotic effects of TGF-ß1, linked to epithelial-to-mesenchymal transition of airway epithelial cells. To examine the role of JNK1 in house dust mite (HDM)-induced airway remodeling, we induced allergic airway inflammation in wild-type (WT) and JNK1-/- mice by intranasal administration of HDM extract. WT and JNK1-/- mice were sensitized with intranasal aspirations of HDM extract for 15 days over 3 wk. HDM caused similar increases in airway hyperresponsiveness, mucus metaplasia, and airway inflammation in WT and JNK1-/- mice. In addition, the profibrotic cytokine TGF-ß1 and phosphorylation of Smad3 were equally increased in WT and JNK1-/- mice. In contrast, increases in collagen content in lung tissue induced by HDM were significantly attenuated in JNK1-/- mice compared with WT controls. Furthermore HDM-induced increases of α-smooth muscle actin (α-SMA) protein and mRNA expression as well as the mesenchymal markers high-mobility group AT-hook 2 and collagen1A1 in WT mice were attenuated in JNK1-/- mice. The let-7 family of microRNAs has previously been linked to fibrosis. HDM exposure in WT mice and primary lung epithelial cells resulted in striking decreases in let-7g miRNA that were not observed in mice or primary lung epithelial cells lacking JNK1-/- mice. Overexpression of let-7g in lung epithelial cells reversed the HDM-induced increases in α-SMA. Collectively, these findings demonstrate an important requirement for JNK1 in promoting HDM-induced fibrotic airway remodeling.


Subject(s)
Airway Remodeling , Bronchial Hyperreactivity/pathology , Dermatophagoides pteronyssinus/pathogenicity , JNK Mitogen-Activated Protein Kinases/physiology , Pneumonia/pathology , Respiratory System/pathology , Animals , Blotting, Western , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/metabolism , Cytokines/genetics , Cytokines/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/etiology , Pneumonia/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Respiratory System/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 306(2): L196-206, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24285269

ABSTRACT

Lung mastocytosis and antigen-induced bronchoconstriction are common features in allergic asthmatics. It is therefore important that animal models of asthma show similar features of mast cell inflammation and reactivity to inhaled allergen. We hypothesized that house dust mite (HDM) would induce mastocytosis in the lung and that inhalation of HDM would trigger bronchoconstriction. Mice were sensitized with intranasal HDM extract, and the acute response to nebulized HDM or the mast cell degranulating compound 48/80 was measured with respiratory input impedance. Using the constant-phase model we calculated Newtonian resistance (Rn) reflecting the conducting airways, tissue dampening (G), and lung elastance (H). Bronchoalveolar lavage fluid was analyzed for mouse mast cell protease-1 (mMCP-1). Lung tissue was analyzed for cytokines, histamine, and α-smooth muscle actin (α-SMA), and histological slides were stained for mast cells. HDM significantly increased Rn but H and G remained unchanged. HDM significantly expanded mast cells compared with control mice; at the same time mMCP-1, α-SMA, Th2 cytokines, and histamine were significantly increased. Compound 48/80 inhalation caused bronchoconstriction and mMCP-1 elevation similarly to HDM inhalation. Bronchoconstriction was eliminated in mast cell-deficient mice. We found that antigen-induced acute bronchoconstriction has a distinct phenotype in mice. HDM sensitization caused lung mastocytosis, and we conclude that inhalation of HDM caused degranulation of mast cells leading to an acute bronchoconstriction without affecting the lung periphery and that mast cell-derived mediators are responsible for the development of the HDM-induced bronchoconstriction in this model.


Subject(s)
Antigens/immunology , Asthma/immunology , Bronchoconstriction/immunology , Mast Cells/immunology , Mastocytosis/immunology , Pyroglyphidae/immunology , Animals , Antigens/pharmacology , Asthma/physiopathology , Bronchoalveolar Lavage Fluid/immunology , Bronchoconstriction/drug effects , Cell Degranulation/drug effects , Cell Degranulation/immunology , Disease Models, Animal , Female , Male , Mast Cells/cytology , Mastocytosis/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , p-Methoxy-N-methylphenethylamine/pharmacology
14.
J Cell Biochem ; 114(9): 1962-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23554102

ABSTRACT

Glutathione has traditionally been considered as an antioxidant that protects cells against oxidative stress. Hence, the loss of reduced glutathione and formation of glutathione disulfide is considered a classical parameter of oxidative stress that is increased in diseases. Recent studies have emerged that demonstrate that glutathione plays a more direct role in biological and pathophysiological processes through covalent modification to reactive cysteines within proteins, a process known as S-glutathionylation. The formation of an S-glutathionylated moiety within the protein can lead to structural and functional modifications. Activation, inactivation, loss of function, and gain of function have all been attributed to S-glutathionylation. In pathophysiological settings, S-glutathionylation is tightly regulated. This perspective offers a concise overview of the emerging field of protein thiol redox modifications. We will also cover newly developed methodology to detect S-glutathionylation in situ, which will enable further discovery into the role of S-glutathionylation in biology and disease.


Subject(s)
Glutathione/metabolism , Animals , Biotin/metabolism , Glutaredoxins/metabolism , Humans , Oxidation-Reduction , Sulfhydryl Compounds/metabolism
15.
Sci Adv ; 9(37): eadi5192, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37703360

ABSTRACT

Glutathione (GSH) is a critical component of the cellular redox system that combats oxidative stress. The glutamate-cystine antiporter, system xC-, is a key player in GSH synthesis that allows for the uptake of cystine, the rate-limiting building block of GSH. It is unclear whether GSH or GSH-dependent protein oxidation [protein S-glutathionylation (PSSG)] regulates the activity of system xC-. We demonstrate that an environment of enhanced PSSG promotes GSH increases via a system xC--dependent mechanism. Absence of the deglutathionylase, glutaredoxin (GLRX), augmented SLC7A11 protein and led to significant increases of GSH content. S-glutathionylation of C23 or C204 of the deubiquitinase OTUB1 promoted interaction with the E2-conjugating enzyme UBCH5A, leading to diminished ubiquitination and proteasomal degradation of SLC7A11 and augmentation of GSH, effects that were reversed by GLRX. These findings demonstrate an intricate link between GLRX and GSH via S-glutathionylation of OTUB1 and system xC- and illuminate a previously unknown feed-forward regulatory mechanism whereby enhanced GSH protein oxidation augments cellular GSH.


Subject(s)
Cystine , Glutaredoxins , Biological Transport , Glutamic Acid , Glutathione
16.
Am J Respir Cell Mol Biol ; 47(3): 306-14, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22461429

ABSTRACT

Recent studies suggest the importance of the transition of airway epithelial cells (EMT) in pulmonary fibrosis, and also indicate a role for Wingless protein (Wnt)/ß-catenin signaling in idiopathic pulmonary fibrosis. We investigated the possible role of the Wnt signaling pathway in inducing EMT in lung epithelial cells, and sought to unravel the role of c-Jun-N-terminal-kinase-1 (JNK1). The exposure of C10 lung epithelial cells or primary mouse tracheal epithelial cells (MTECs) to Wnt3a resulted in increases in JNK phosphorylation and nuclear ß-catenin content. Because the role of ß-catenin as a transcriptional coactivator is well established, we investigated T-cell factor/lymphocyte-enhancement factor (TCF/LEF) transcriptional activity in C10 lung epithelial cells after the activation of Wnt. TCF/LEF transcriptional activity was enhanced after the activation of Wnt, and this increase in TCF/LEF transcriptional activity was diminished after the small interfering (si)RNA-mediated ablation of JNK. The activation of the Wnt pathway by Wnt3a, or the expression of either wild-type or constitutively active ß-catenin (S37A), led to the activation of an EMT transcriptome, manifested by the increased mRNA expression of CArG box-binding factor-A, fibroblast-specific protein (FSP)-1, α-smooth muscle actin (α-SMA), and vimentin, increases in the content of α-SMA and FSP1, and the concomitant loss of zona occludens-1. The siRNA-mediated ablation of ß-catenin substantially decreased Wnt3a-induced EMT. The siRNA ablation of JNK1 largely abolished Wnt3a, ß-catenin, and ß-catenin S37a-induced EMT. In MTECs lacking Jnk1, Wnt3a-induced increases in nuclear ß-catenin, EMT transcriptome, and the content of α-SMA or FSP1 were substantially diminished. These data show that the activation of the Wnt signaling pathway is capable of inducing an EMT program in lung epithelial cells through ß-catenin, and that this process is controlled by JNK1.


Subject(s)
Lung/metabolism , Mesoderm/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Wnt Proteins/physiology , beta Catenin/physiology , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Lung/cytology , Mice , Phosphorylation , RNA, Small Interfering , Transcription, Genetic
17.
Am J Respir Cell Mol Biol ; 47(4): 497-508, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22652196

ABSTRACT

The transcription factor NF-κB has been causally linked to inflammatory lung diseases. Recent studies have unraveled the complexity of NF-κB activation by identifying two parallel activation pathways: the classical NF-κB pathway, which is controlled by IκB kinase complex-ß (IKKß) and RelA/p50, and the alternative pathway, which is controlled by IKKα and RelB/p52. The alternative pathway regulates adaptive immune responses and lymphoid development, yet its role in the regulation of innate immune responses remains largely unknown. In this study, we determined the relevance of the alternative NF-κB pathway in proinflammatory responses in lung epithelial cells. The exposure of C10 murine alveolar lung epithelial cells to diverse stimuli, or primary murine tracheal epithelial cells to LPS, resulted in the activation of both NF-κB pathways, based on the nuclear translocation of RelA, p50, RelB, and p52. Increases in the nuclear content of RelA occurred rapidly, but transiently, whereas increases in nuclear RelB content were protracted. The small interfering (si) RNA-mediated knockdown of IKKα, RelA, or RelB resulted in decreases of multiple LPS-induced proinflammatory cytokines. Surprisingly, the siRNA ablation of IKKα or RelB led to marked increases in the production of IL-6 in response to LPS. The simultaneous expression of constitutively active (CA)-IKKα and CA-IKKß caused synergistic increases in proinflammatory mediators. Lastly, the disruption of the IKK signalsome inhibited the activation of both NF-κB pathways. These results demonstrate that the coordinated activation of both NF-κB pathways regulates the magnitude and nature of proinflammatory responses in lung epithelial cells.


Subject(s)
Inflammation Mediators/metabolism , NF-kappa B/metabolism , Respiratory Mucosa/metabolism , Signal Transduction , Animals , Anoctamins , Cells, Cultured , Chloride Channels , Cytokines/genetics , Cytokines/metabolism , Cytokines/physiology , Gene Expression , Gene Knockdown Techniques , Histones/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Primary Cell Culture , RNA Interference , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Trachea/pathology
18.
Am J Physiol Lung Cell Mol Physiol ; 303(6): L528-38, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22752969

ABSTRACT

Protein-S-glutathionylation (PSSG) is an oxidative modification of reactive cysteines that has emerged as an important player in pathophysiological processes. Under physiological conditions, the thiol transferase, glutaredoxin-1 (Glrx1) catalyses deglutathionylation. Although we previously demonstrated that Glrx1 expression is increased in mice with allergic inflammation, the impact of Glrx1/PSSG in the development of allergic airways disease remains unknown. In the present study we examined the impact of genetic ablation of Glrx1 in the pathogenesis of allergic inflammation and airway hyperresponsiveness (AHR) in mice. Glrx1(-/-) or WT mice were subjected to the antigen, ovalbumin (OVA), and parameters of allergic airways disease were evaluated 48 h after three challenges, and 48 h or 7 days after six challenges with aerosolized antigen. Although no clear increases in PSSG were observed in WT mice in response to OVA, marked increases were detected in lung tissue of mice lacking Glrx1 48 h following six antigen challenges. Inflammation and expression of proinflammatory mediators were decreased in Glrx1(-/-) mice, dependent on the time of analysis. WT and Glrx1(-/-) mice demonstrated comparable increases in AHR 48 h after three or six challenges with OVA. However, 7 days postcessation of six challenges, parameters of AHR in Glrx1(-/-) mice were resolved to control levels, accompanied by marked decreases in mucus metaplasia and expression of Muc5AC and GOB5. These results demonstrate that the Glrx1/S-glutathionylation redox status in mice is a critical regulator of AHR, suggesting that avenues to increase S-glutathionylation of specific target proteins may be beneficial to attenuate AHR.


Subject(s)
Bronchial Hyperreactivity/immunology , Glutaredoxins/genetics , Lung/pathology , Mucus , Animals , Glutaredoxins/deficiency , Glutathione/metabolism , Lung Diseases/pathology , Metaplasia/pathology , Mice , Ovalbumin/immunology , Pneumonia/etiology , Proteins/metabolism
19.
Am J Physiol Cell Physiol ; 301(5): C995-C1007, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21832246

ABSTRACT

Skeletal muscle atrophy commonly occurs in acute and chronic disease. The expression of the muscle-specific E3 ligases atrogin-1 (MAFbx) and muscle RING finger 1 (MuRF1) is induced by atrophy stimuli such as glucocorticoids or absence of IGF-I/insulin and subsequent Akt signaling. We investigated whether glycogen synthase kinase-3ß (GSK-3ß), a downstream molecule in IGF-I/Akt signaling, is required for basal and atrophy stimulus-induced expression of atrogin-1 and MuRF1, and myofibrillar protein loss in C(2)C(12) skeletal myotubes. Abrogation of basal IGF-I signaling, using LY294002, resulted in a prominent induction of atrogin-1 and MuRF1 mRNA and was accompanied by a loss of myosin heavy chain fast (MyHC-f) and myosin light chains 1 (MyLC-1) and -3 (MyLC-3). The synthetic glucocorticoid dexamethasone (Dex) also induced the expression of both atrogenes and likewise resulted in the loss of myosin protein abundance. Genetic ablation of GSK-3ß using small interfering RNA resulted in specific sparing of MyHC-f, MyLC-1, and MyLC-3 protein levels after Dex treatment or impaired IGF-I/Akt signaling. Interestingly, loss of endogenous GSK-3ß suppressed both basal and atrophy stimulus-induced atrogin-1 and MuRF1 expression, whereas pharmacological GSK-3ß inhibition, using CHIR99021 or LiCl, only reduced atrogin-1 mRNA levels in response to LY294002 or Dex. In conclusion, our data reveal that myotube atrophy and myofibrillar protein loss are GSK-3ß dependent, and demonstrate for the first time that basal and atrophy stimulus-induced atrogin-1 mRNA expression requires GSK-3ß enzymatic activity, whereas MuRF1 expression depends solely on the physical presence of GSK-3ß.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Muscle, Skeletal/enzymology , Muscular Atrophy/enzymology , Myoblasts/enzymology , Animals , Cell Line , Chromones/pharmacology , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Insulin-Like Growth Factor I/antagonists & inhibitors , Lithium Chloride/pharmacology , Mice , Morpholines/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/drug effects , Muscular Atrophy/drug therapy , Myoblasts/drug effects , Myoblasts/physiology , Myosin Heavy Chains/biosynthesis , Myosin Light Chains/biosynthesis , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , Signal Transduction/drug effects , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/biosynthesis
20.
Redox Biol ; 47: 102160, 2021 11.
Article in English | MEDLINE | ID: mdl-34624602

ABSTRACT

BACKGROUND: Interleukin-1-dependent increases in glycolysis promote allergic airways disease in mice and disruption of pyruvate kinase M2 (PKM2) activity is critical herein. Glutathione-S-transferase P (GSTP) has been implicated in asthma pathogenesis and regulates the oxidation state of proteins via S-glutathionylation. We addressed whether GSTP-dependent S-glutathionylation promotes allergic airways disease by promoting glycolytic reprogramming and whether it involves the disruption of PKM2. METHODS: We used house dust mite (HDM) or interleukin-1ß in C57BL6/NJ WT or mice that lack GSTP. Airway basal cells were stimulated with interleukin-1ß and the selective GSTP inhibitor, TLK199. GSTP and PKM2 were evaluated in sputum samples of asthmatics and healthy controls and incorporated analysis of the U-BIOPRED severe asthma cohort database. RESULTS: Ablation of Gstp decreased total S-glutathionylation and attenuated HDM-induced allergic airways disease and interleukin-1ß-mediated inflammation. Gstp deletion or inhibition by TLK199 decreased the interleukin-1ß-stimulated secretion of pro-inflammatory mediators and lactate by epithelial cells. 13C-glucose metabolomics showed decreased glycolysis flux at the pyruvate kinase step in response to TLK199. GSTP and PKM2 levels were increased in BAL of HDM-exposed mice as well as in sputum of asthmatics compared to controls. Sputum proteomics and transcriptomics revealed strong correlations between GSTP, PKM2, and the glycolysis pathway in asthma. CONCLUSIONS: GSTP contributes to the pathogenesis of allergic airways disease in association with enhanced glycolysis and oxidative disruption of PKM2. Our findings also suggest a PKM2-GSTP-glycolysis signature in asthma that is associated with severe disease.


Subject(s)
Asthma , Carrier Proteins/metabolism , Glutathione S-Transferase pi/metabolism , Membrane Proteins/metabolism , Pyruvate Kinase , Thyroid Hormones/metabolism , Animals , Carrier Proteins/genetics , Glutathione/metabolism , Glutathione S-Transferase pi/genetics , Glutathione Transferase , Glycolysis , Humans , Lung/metabolism , Membrane Proteins/genetics , Mice , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL