Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
J Inflamm (Lond) ; 21(1): 14, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689261

ABSTRACT

BACKGROUND: The DNA-dependent protein kinase (DNA-PK) complex comprises a catalytic (PRKDC) and two requisite DNA-binding (Ku70/Ku80) subunits. The role of the complex in repairing double-stranded DNA breaks (DSBs) is established, but its role in inflammation, as a complex or individual subunits, remains elusive. While only ~ 1% of PRKDC is necessary for DNA repair, we reported that partial inhibition blocks asthma in mice without causing SCID. METHODS: We investigated the central role of PRKDC in inflammation and its potential association with DNA repair. We also elucidated the relationship between inflammatory cytokines (e.g., TNF-α) and PRKDC by analyzing its connections to inflammatory kinases. Human cell lines, primary human endothelial cells, and mouse fibroblasts were used to conduct the in vitro studies. For animal studies, LPS- and oxazolone-induced mouse models of acute lung injury (ALI) and delayed-type hypersensitivity (DHT) were used. Wild-type, PRKDC+/-, or Ku70+/- mice used in this study. RESULTS: A ~ 50% reduction in PRKDC markedly blocked TNF-α-induced expression of inflammatory factors (e.g., ICAM-1/VCAM-1). PRKDC regulates Th1-mediated inflammation, such as DHT and ALI, and its role is highly sensitive to inhibition achieved by gene heterozygosity or pharmacologically. In endothelial or epithelial cells, TNF-α promoted rapid PRKDC phosphorylation in a fashion resembling that induced by, but independent of, DSBs. Ku70 heterozygosity exerted little to no effect on ALI in mice, and whatever effect it had was associated with a specific increase in MCP-1 in the lungs and systemically. While Ku70 knockout blocked VP-16-induced PRKDC phosphorylation, it did not prevent TNF-α - induced phosphorylation of the kinase, suggesting Ku70 dispensability. Immunoprecipitation studies revealed that PRKDC transiently interacts with p38MAPK. Inhibition of p38MAPK blocked TNF-α-induced PRKDC phosphorylation. Direct phosphorylation of PRKDC by p38MAPK was demonstrated using a cell-free system. CONCLUSIONS: This study presents compelling evidence that PRKDC functions independently of the DNA-PK complex, emphasizing its central role in Th1-mediated inflammation. The distinct functionality of PRKDC as an individual enzyme, its remarkable sensitivity to inhibition, and its phosphorylation by p38MAPK offer promising therapeutic opportunities to mitigate inflammation while sparing DNA repair processes. These findings expand our understanding of PRKDC biology and open new avenues for targeted anti-inflammatory interventions.

2.
Respir Med ; 180: 106354, 2021.
Article in English | MEDLINE | ID: mdl-33721696

ABSTRACT

BACKGROUND AND OBJECTIVE: We tested whether the prostacyclin analog inhaled iloprost modulates dead space, dynamic hyperinflation (DH), and systemic inflammation/oxidative stress during maximal exercise in subjects with chronic obstructive pulmonary disease (COPD) who were not selected based on pulmonary hypertension (PH). METHODS: Twenty-four COPD patients with moderate-severe obstruction (age 59 ± 7 years, FEV1 53 ± 13% predicted) participated in a randomized, double-blind, placebo-controlled crossover trial. Each subject received a single nebulized dose of 5.0 µg iloprost or placebo on non-consecutive days followed by maximal cardiopulmonary exercise tests. The primary outcome was DH quantified by end-expiratory lung volume/total lung capacity ratio (EELV/TLC) at metabolic isotime. RESULTS: Inhaled iloprost was well-tolerated and reduced submaximal alveolar dead-space fraction but did not significantly reduce DH (0.70 ± 0.09 vs 0.69 ± 0.07 following placebo and iloprost, respectively, p = 0.38). Maximal exercise time (9.1 ± 2.3 vs 9.3 ± 2.2 min, p = 0.31) and peak oxygen uptake (17.4 ± 6.3 vs 17.9 ± 6.9 mL/kg/min, p = 0.30) were not significantly different following placebo versus iloprost. CONCLUSIONS: A single dose of inhaled iloprost was safe and reduced alveolar dead space fraction; however, it was not efficacious in modulating DH or improving exercise capacity in COPD patients who were not selected for the presence of PH.


Subject(s)
Exercise/physiology , Iloprost/administration & dosage , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Administration, Inhalation , Aged , Cross-Over Studies , Double-Blind Method , Exercise Test/methods , Female , Humans , Inflammation , Lung Volume Measurements , Male , Middle Aged , Oxidative Stress/drug effects , Oxygen Consumption , Pulmonary Disease, Chronic Obstructive/metabolism , Total Lung Capacity
3.
Respirology ; 24(7): 675-683, 2019 07.
Article in English | MEDLINE | ID: mdl-30747487

ABSTRACT

BACKGROUND AND OBJECTIVE: Endothelial microparticles (EMP) are submicron vesicles released from endothelial cells. We aimed to determine the utility of EMP as biomarkers of pulmonary arterial hypertension (PAH) in systemic sclerosis (SSc) patients and the pathogenic role of microparticles (MP) in vascular inflammation. METHODS: Levels of EMP (CD144+, CD31+, CD62E+ and CD143+) were compared between three groups (10 SSc patients with PAH, 10 SSc patients without pulmonary hypertension (no-PH) and 10 healthy age- and sex-matched controls). Human pulmonary artery endothelial cells (HPAEC) were exposed in vitro to MP obtained from SSc patients or healthy controls, and levels of cytokines and inflammatory adhesion molecules were compared. RESULTS: CD144+ EMP were significantly higher in the SSc-PAH group compared to either the SSc-no PH or healthy controls (diagnostic accuracy 80%, P = 0.02). Compared to controls, SSc patients had higher CD31+/CD62E+ ratios, indicating larger contributions of apoptosis to EMP release (P = 0.04). Patients with limited SSc had significantly higher levels of CD143+ EMP compared to those with diffuse subtype (P = 0.008). When HPAEC were exposed to MP from SSc patients, there was a significant increase in inflammatory cytokines and adhesion molecules. Interestingly, exposure to healthy control MP caused a reduction in inflammatory markers. CONCLUSION: EMP (particularly CD144+) are promising biomarkers of PAH in SSc but require further study. MP isolated from SSc patients induced an increase in endothelial cell inflammation and may be an important pathogenic factor in SSc.


Subject(s)
Cell-Derived Microparticles , Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Scleroderma, Systemic/metabolism , Biomarkers/metabolism , Cytokines/metabolism , Female , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Male , Middle Aged , Pilot Projects , Pulmonary Artery/pathology , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology
4.
J Transl Med ; 16(1): 243, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30172259

ABSTRACT

BACKGROUND: The efficacy of heparins and low-MW-heparins (LMWH) against human asthma has been known for decades. However, the clinical utility of these compounds has been hampered by their anticoagulant properties. Much effort has been put into harnessing the anti-inflammatory properties of LMWH but none have been used as therapy for asthma. Sulfated-non-anticoagulant heparin (S-NACH) is an ultra-LMWH with no systemic anticoagulant effects. OBJECTIVE: The present study explored the potential of S-NACH in blocking allergic asthma and examined the potential mechanism by which it exerts its effects. METHODS: Acute and chronic ovalbumin-based mouse models of asthma, splenocytes, and a lung epithelial cell line were used. Mice were challenged with aerosolized ovalbumin and administered S-NACH or saline 30 min after each ovalbumin challenge. RESULTS: Sulfated-non-anticoagulant heparin administration in mice promoted a robust reduction in airway eosinophilia, mucus production, and airway hyperresponsiveness even after chronic repeated challenges with ovalbumin. Such effects were linked to suppression of Th2 cytokines IL-4/IL-5/IL-13/GM-CSF and ovalbumin-specific IgE without any effect on IFN-γ. S-NACH also reduced lung fibrosis in mice that were chronically-exposed to ovalbumin. These protective effects of S-NACH may be attributed to modulation of the IL-4/JAK1 signal transduction pathway through an inhibition of STAT6 phosphorylation and a subsequent inhibition of GATA-3 and inducible NO synthase expression. The effect of the drug on STAT6 phosphorylation coincided with a reduction in JAK1 phosphorylation upon IL-4 treatment. The protective effects of S-NACH treatment was associated with reduction of the basal expression of the two isoforms of arginase ARG1 and ARG2 in lung epithelial cells. CONCLUSIONS: Our study demonstrates that S-NACH constitutes an opportunity to benefit from the well-known anti-asthma properties of heparins/LMWH while bypassing the risk of bleeding. Our results show, for the first time, that such anti-asthma effects may be associated with reduction of the IL-4/JAK1/STAT6 pathway.


Subject(s)
Asthma/drug therapy , Heparin/adverse effects , Interleukin-4/metabolism , Janus Kinase 1/metabolism , STAT6 Transcription Factor/metabolism , Th2 Cells/metabolism , A549 Cells , Animals , Anti-Inflammatory Agents/chemistry , Anticoagulants/chemistry , Asthma/complications , Cell Line , Cell Separation , Disease Models, Animal , Flow Cytometry , Hemorrhage/drug therapy , Humans , Hypersensitivity/complications , Hypersensitivity/drug therapy , Male , Mice , Mice, Inbred C57BL , Ovalbumin/chemistry , Signal Transduction , Spleen/cytology
5.
Mediators Inflamm ; 2016: 1984703, 2016.
Article in English | MEDLINE | ID: mdl-27524861

ABSTRACT

Although expression of inducible NO synthase (iNOS) in the lungs of asthmatics and associated nitrosative damage are established, iNOS failed as a therapeutic target for blocking airway hyperresponsiveness (AHR) and inflammation in asthmatics. This dichotomy calls for better strategies with which the enzyme is adequately targeted. Here, we confirm iNOS expression in the asthmatic lung with concomitant protein nitration and poly(ADP-ribose) polymerase (PARP) activation. We show, for the first time, that iNOS is highly expressed in peripheral blood mononuclear cells (PBMCs) of asthmatics with uncontrolled disease, which did not correspond to protein nitration. Selective iNOS inhibition with L-NIL protected against AHR upon acute, but not chronic, exposure to ovalbumin or house dust mite (HDM) in mice. Supplementation of NO by nitrite administration significantly blocked AHR in chronically HDM-exposed mice that were treated with L-NIL. Protection against chronic HDM exposure-induced AHR by olaparib-mediated PARP inhibition may be associated with the partial but not the complete blockade of iNOS expression. Indeed, L-NIL administration prevented olaparib-mediated protection against AHR in chronically HDM-exposed mice. Our study suggests that the amount of iNOS and NO are critical determinants in the modulation of AHR by selective iNOS inhibitors and renews the potential of iNOS as a therapeutic target for asthma.


Subject(s)
Allergens/immunology , Asthma/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Respiratory Hypersensitivity/metabolism , Animals , Asthma/immunology , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II/genetics , Nitrites/pharmacology , Nitrogen Oxides/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/immunology , Signal Transduction/drug effects
6.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L630-8, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26851260

ABSTRACT

Pulmonary endothelial prostacyclin appears to be involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). The effect of treatment with a prostacyclin analog in animal models of previously established COPD is unknown. We evaluated the short- and long-term effect of iloprost on inflammation and airway hyperresponsiveness (AHR) in a murine model of COPD. Nineteen mice were exposed to LPS/elastase, followed by either three doses of intranasal iloprost or saline. In the long-term treatment experiment, 18 mice were exposed to LPS/elastase and then received 6 wk of iloprost or were left untreated as controls. In the short-term experiment, iloprost did not change AHR but significantly reduced serum IL-5 and IFN-γ. Long-term treatment with iloprost for both 2 and 6 wk significantly improved AHR. After 6 wk of iloprost, there was a reduction in bronchoalveolar lavage (BALF) neutrophils, serum IL-1ß (30.0 ± 9.2 vs. 64.8 ± 7.4 pg/ml, P = 0.045), IL-2 (36.5 ± 10.6 vs. 83.8 ± 0.4 pg/ml, P = 0.01), IL-10 (75.7 ± 9.3 vs. 96.5 ± 3.5 pg/ml, P = 0.02), and nitrite (15.1 ± 5.4 vs. 30.5 ± 10.7 µmol, P = 0.01). Smooth muscle actin (SMA) in the lung homogenate was also significantly reduced after iloprost treatment (P = 0.02), and SMA thickness was reduced in the small and medium blood vessels after iloprost (P < 0.001). In summary, short- and long-term treatment with intranasal iloprost significantly reduced systemic inflammation in an LPS/elastase COPD model. Long-term iloprost treatment also reduced AHR, serum nitrite, SMA, and BALF neutrophilia. These data encourage future investigations of prostanoid therapy as a novel treatment for COPD patients.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Iloprost/administration & dosage , Pulmonary Disease, Chronic Obstructive/drug therapy , Administration, Intranasal , Animals , Cytokines/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Neutrophil Infiltration , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology
7.
Clin Sci (Lond) ; 129(11): 951-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26205779

ABSTRACT

Our laboratory established a role for poly(ADP-ribose)polymerase (PARP) in asthma. To increase the clinical significance of our studies, it is imperative to demonstrate that PARP is actually activated in human asthma, to examine whether a PARP inhibitor approved for human testing such as olaparib blocks already-established chronic asthma traits in response to house dust mite (HDM), a true human allergen, in mice and to examine whether the drug modulates human cluster of differentiation type 4 (CD4(+)) T-cell function. To conduct the study, human lung specimens and peripheral blood mononuclear cells (PBMCs) and a HDM-based mouse asthma model were used. Our results show that PARP is activated in PBMCs and lung tissues of asthmatics. PARP inhibition by olaparib or gene knockout blocked established asthma-like traits in mice chronically exposed to HDM including airway eosinophilia and hyper-responsiveness. These effects were linked to a marked reduction in T helper 2 (Th2) cytokine production without a prominent effect on interferon (IFN)-γ or interleukin (IL)-10. PARP inhibition prevented HDM-induced increase in overall cellularity, weight and CD4(+) T-cell population in spleens of treated mice whereas it increased the T-regulatory cell population. In CD3/CD28-stimulated human CD4 (+)T-cells, olaparib treatment reduced Th2 cytokine production potentially by modulating GATA binding protein-3 (gata-3)/IL-4 expression while moderately affecting T-cell proliferation. PARP inhibition inconsistently increased IL-17 in HDM-exposed mice and CD3/CD28-stimulated CD4(+) T cells without a concomitant increase in factors that can be influenced by IL-17. In the present study, we provide evidence for the first time that PARP-1 is activated in human asthma and that its inhibition is effective in blocking established asthma in mice.


Subject(s)
Anti-Allergic Agents/pharmacology , Anti-Asthmatic Agents/pharmacology , Antigens, Dermatophagoides , Asthma/prevention & control , Lung/drug effects , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Asthma/enzymology , Asthma/immunology , Asthma/physiopathology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Humans , Inflammation Mediators/metabolism , Lung/enzymology , Lung/immunology , Lung/physiopathology , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/drug effects , Th2 Cells/enzymology , Th2 Cells/immunology
8.
J Transl Med ; 13: 225, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26169874

ABSTRACT

BACKGROUND: An important portion of asthmatics do not respond to current therapies. Thus, the need for new therapeutic drugs is urgent. We have demonstrated a critical role for PARP in experimental asthma. Olaparib, a PARP inhibitor, was recently introduced in clinical trials against cancer. The objective of the present study was to examine the efficacy of olaparib in blocking established allergic airway inflammation and hyperresponsiveness similar to those observed in human asthma in animal models of the disease. METHODS: We used ovalbumin (OVA)-based mouse models of asthma and primary CD4(+) T cells. C57BL/6J WT or PARP-1(-/-) mice were subjected to OVA sensitization followed by a single or multiple challenges to aerosolized OVA or left unchallenged. WT mice were administered, i.p., 1 mg/kg, 5 or 10 mg/kg of olaparib or saline 30 min after each OVA challenge. RESULTS: Administration of olaparib in mice 30 min post-challenge promoted a robust reduction in airway eosinophilia, mucus production and hyperresponsiveness even after repeated challenges with ovalbumin. The protective effects of olaparib were linked to a suppression of Th2 cytokines eotaxin, IL-4, IL-5, IL-6, IL-13, and M-CSF, and ovalbumin-specific IgE with an increase in the Th1 cytokine IFN-γ. These traits were associated with a decrease in splenic CD4(+) T cells and concomitant increase in T-regulatory cells. The aforementioned traits conferred by olaparib administration were consistent with those observed in OVA-challenged PARP-1(-/-) mice. Adoptive transfer of Th2-skewed OT-II-WT CD4(+) T cells reversed the Th2 cytokines IL-4, IL-5, and IL-10, the chemokine GM-CSF, the Th1 cytokines IL-2 and IFN-γ, and ovalbumin-specific IgE production in ovalbumin-challenged PARP-1(-/-)mice suggesting a role for PARP-1 in CD4(+) T but not B cells. In ex vivo studies, PARP inhibition by olaparib or PARP-1 gene knockout markedly reduced CD3/CD28-stimulated gata-3 and il4 expression in Th2-skewed CD4(+) T cells while causing a moderate elevation in t-bet and ifn-γ expression in Th1-skewed CD4(+) T cells. CONCLUSIONS: Our findings show the potential of PARP inhibition as a viable therapeutic strategy and olaparib as a likely candidate to be tested in human asthma clinical trials.


Subject(s)
Asthma/drug therapy , Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , Gene Knockout Techniques , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Adoptive Transfer , Animals , Antigens, CD/metabolism , Asthma/complications , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/immunology , CD4-Positive T-Lymphocytes/drug effects , Cytokines/metabolism , Disease Models, Animal , Eosinophilia/complications , Eosinophilia/drug therapy , Eosinophilia/immunology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Humans , Immunoglobulin E/biosynthesis , Mice, Inbred C57BL , Mucus/metabolism , Ovalbumin/immunology , Phthalazines/pharmacology , Piperazines/pharmacology , Spleen/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism
9.
J Allergy Clin Immunol ; 135(2): 425-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25441643

ABSTRACT

BACKGROUND: We reported that DNA-dependent protein kinase (DNA-PK) is critical for the expression of nuclear factor κB-dependent genes in TNF-α-treated glioblastoma cells, suggesting an involvement in inflammatory diseases. OBJECTIVE: We sought to investigate the role of DNA-PK in asthma. METHODS: Cell culture and ovalbumin (OVA)- or house dust mite-based murine asthma models were used in this study. RESULTS: DNA-PK was essential for monocyte adhesion to TNF-α-treated endothelial cells. Administration of the DNA-PK inhibitor NU7441 reduced airway eosinophilia, mucus hypersecretion, airway hyperresponsiveness, and OVA-specific IgE production in mice prechallenged with OVA. Such effects correlated with a marked reduction in lung vascular cell adhesion molecule 1 expression and production of several cytokines, including IL-4, IL-5, IL-13, eotaxin, IL-2, and IL-12 and the chemokines monocyte chemoattractant protein 1 and keratinocyte-derived chemokine, with a negligible effect on IL-10/IFN-γ production. DNA-PK inhibition by gene heterozygosity of the 450-kDa catalytic subunit of the kinase (DNA-PKcs(+/-)) also prevented manifestation of asthma-like traits. These results were confirmed in a chronic model of asthma by using house dust mite, a human allergen. Remarkably, such protection occurred without causing severe combined immunodeficiency. Adoptive transfer of TH2-skewed OT-II wild-type CD4(+) T cells reversed IgE and TH2 cytokine production but not airway hyperresponsiveness in OVA-challenged DNA-PKcs(+/-) mice. DNA-PK inhibition reduced IL-4, IL-5, IL-13, eotaxin, IL-8, and monocyte chemoattractant protein 1 production without affecting IL-2, IL-12, IFN-γ, and interferon-inducible protein 10 production in CD3/CD28-stimulated human CD4(+) T cells, potentially by blocking expression of Gata3. These effects occurred without significant reductions in T-cell proliferation. In mouse CD4(+) T cells in vitro DNA-PK inhibition severely blocked CD3/CD28-induced Gata3 and T-bet expression in CD4(+) T cells and prevented differentiation of TH1 and TH2 cells under respective TH1- and TH2-skewing conditions. CONCLUSION: Our results suggest DNA-PK as a novel determinant of asthma and a potential target for the treatment of the disease.


Subject(s)
Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , DNA-Activated Protein Kinase/antagonists & inhibitors , Respiratory Mucosa/immunology , Adoptive Transfer , Allergens/immunology , Animals , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Cell Adhesion , Cytokines/metabolism , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Disease Models, Animal , Eosinophils/immunology , Eosinophils/metabolism , Epithelial Cells/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression , Genetic Heterogeneity , Humans , Immunoglobulin E/immunology , Lymphocyte Activation , Male , Mice , Mice, Knockout , Organ Size , Ovalbumin/adverse effects , Ovalbumin/immunology , Phenotype , Plasma Cells/immunology , Plasma Cells/metabolism , Pyroglyphidae/immunology , Receptors, Antigen, T-Cell/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Severe Combined Immunodeficiency , Spleen/anatomy & histology , Spleen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
10.
J Biol Chem ; 288(3): 1458-68, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23184953

ABSTRACT

Minocycline protects against asthma independently of its antibiotic function and was recently reported as a potent poly(ADP-ribose) polymerase (PARP) inhibitor. In an animal model of asthma, a single administration of minocycline conferred excellent protection against ovalbumin-induced airway eosinophilia, mucus hypersecretion, and Th2 cytokine production (IL-4/IL-5/IL-12(p70)/IL-13/GM-CSF) and a partial protection against airway hyperresponsiveness. These effects correlated with pronounced reduction in lung and sera allergen-specific IgE. A reduction in poly(ADP-ribose) immunoreactivity in the lungs of minocycline-treated/ovalbumin-challenged mice correlated with decreased oxidative DNA damage. The effect of minocycline on PARP may be indirect, as the drug failed to efficiently block direct PARP activation in lungs of N-methyl-N'-nitro-N-nitroso-guanidine-treated mice or H(2)O(2)-treated cells. Minocycline blocked allergen-specific IgE production in B cells potentially by modulating T cell receptor (TCR)-linked IL-4 production at the mRNA level but not through a modulation of the IL-4-JAK-STAT-6 axis, IL-2 production, or NFAT1 activation. Restoration of IL-4, ex vivo, rescued IgE production by minocycline-treated/ovalbumin-stimulated B cells. IL-4 blockade correlated with a preferential inhibition of the NF-κB activation arm of TCR but not GSK3, Src, p38 MAPK, or ERK1/2. Interestingly, the drug promoted a slightly higher Src and ERK1/2 phosphorylation. Inhibition of NF-κB was linked to a complete blockade of TCR-stimulated GATA-3 expression, a pivotal transcription factor for IL-4 expression. Minocycline also reduced TNF-α-mediated NF-κB activation and expression of dependent genes. These results show a potentially broad effect of minocycline but that it may block IgE production in part by modulating TCR function, particularly by inhibiting the signaling pathway, leading to NF-κB activation, GATA-3 expression, and subsequent IL-4 production.


Subject(s)
Asthma/drug therapy , GATA3 Transcription Factor/genetics , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Interleukin-4/genetics , Minocycline/therapeutic use , NF-kappa B/genetics , Receptors, Antigen, T-Cell/genetics , Animals , Asthma/complications , Asthma/genetics , Asthma/immunology , GATA3 Transcription Factor/agonists , GATA3 Transcription Factor/immunology , Gene Expression Regulation/drug effects , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunologic Factors/pharmacology , Inflammation/complications , Inflammation/genetics , Inflammation/immunology , Interleukin-4/antagonists & inhibitors , Interleukin-4/immunology , Male , Mice , Mice, Inbred C57BL , Minocycline/pharmacology , NF-kappa B/agonists , NF-kappa B/immunology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/immunology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL