Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
COPD ; 21(1): 2342797, 2024 12.
Article in English | MEDLINE | ID: mdl-38712759

ABSTRACT

Objective: To investigate the effects of cigarette smoke (CS) on Serine/Threonine Kinase 11 (STK11) and to determine STK11's role in CS-induced airway epithelial cell cytotoxicity.Methods: STK11 expression levels in the lung tissues of smokers with or without COPD and mice exposed to CS or room air (RA) were determined by immunoblotting and RT-PCR. BEAS-2Bs-human bronchial airway epithelial cells were exposed to CS extract (CSE), and the changes in STK11 expression levels were determined by immunoblotting and RT-PCR. BEAS-2B cells were transfected with STK11-specific siRNA or STK11 expression plasmid, and the effects of CSE on airway epithelial cell cytotoxicity were measured. To determine the specific STK11 degradation-proteolytic pathway, BEAS-2Bs were treated with cycloheximide alone or combined with MG132 or leupeptin. Finally, to identify the F-box protein mediating the STK11 degradation, a screening assay was performed using transfection with a panel of FBXL E3 ligase subunits.Results: STK11 protein levels were significantly decreased in the lung tissues of smokers with COPD relative to smokers without COPD. STK11 protein levels were also significantly decreased in mouse lung tissues exposed to CS compared to RA. Exposure to CSE shortened the STK11 mRNA and protein half-life to 4 h in BEAS-2B cells. STK11 protein overexpression attenuated the CSE-induced cytotoxicity; in contrast, its knockdown augmented CSE-induced cytotoxicity. FBXL19 mediates CSE-induced STK11 protein degradation via the ubiquitin-proteasome pathway in cultured BEAS-2B cells. FBXL19 overexpression led to accelerated STK11 ubiquitination and degradation in a dose-dependent manner.Conclusions: Our results suggest that CSE enhances the degradation of STK11 protein in airway epithelial cells via the FBXL19-mediated ubiquitin-proteasomal pathway, leading to augmented cell death.HIGHLIGHTSLung tissues of COPD-smokers exhibited a decreased STK11 RNA and protein expression.STK11 overexpression attenuates CS-induced airway epithelial cell cytotoxicity.STK11 depletion augments CS-induced airway epithelial cell cytotoxicity.CS diminishes STK11 via FBXL19-mediated ubiquitin-proteasome degradation.


Subject(s)
AMP-Activated Protein Kinases , Epithelial Cells , F-Box Proteins , Protein Serine-Threonine Kinases , Smoke , Animals , Humans , Male , Mice , AMP-Activated Protein Kinase Kinases , Cell Line , Cigarette Smoking/adverse effects , Cycloheximide/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , F-Box Proteins/metabolism , F-Box Proteins/genetics , Leupeptins/pharmacology , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteolysis/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , RNA, Small Interfering , Smoke/adverse effects
2.
Arch Microbiol ; 205(1): 41, 2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36571636

ABSTRACT

For the sustainable farming of disease-free and healthy shrimps, antimicrobial use is frequent nowadays in shrimp-cultured system. Considering the serious impact of global antimicrobial resistance (AMR), the present study was focused to investigate the prevalence of antimicrobial-resistant vibrios among infected shrimps (Penaeus vannamei) from two brackish water-cultured farms. Diverse species of vibrios viz. V. alginolyticus, V. parahaemolyticus, V. cholerae, V. mimicus, and V. fluvialis along with Aeromonas hydrophila, A. salmonicida and Shewanella algae were recovered from the shrimps on TCBS medium. Shannon-Wiener diversity index and H' (loge) were 1.506 and 1.69 for the isolates from farm 1 and farm 2, respectively. V. alginolyticus was found to be the most resistant isolate by showing multiple antibiotic resistance (MAR) index of 0.60 followed by V. mimicus (0.54) and V. parahaemolyticus (0.42). Among the 35 antibiotics of 15 different classes tested, tetracyclines, beta-lactams and cephalosporins were found as the most resistant antibiotic classes. All the isolates possessed a MAR index > 0.2 and the majority exhibited minimum inhibitory concentration (MIC) > 256 mcg/ml, thereby indicating the excess exposure of antibiotics in the systems. An enhanced altered resistance phenotype and a significant shift in the MAR index were noticed after plasmid curing. Public health is further concerning because plasmid-borne AMR is evident among the isolates and the studied shrimp samples are significant in the food industry. This baseline information will help the authorities to curb antimicrobial use and pave the way for establishing new alternative strategies by undertaking a multidimensional "One-Health" approach.


Subject(s)
Anti-Infective Agents , Penaeidae , Vibrio cholerae , Vibrio parahaemolyticus , Vibrio , Animals , Anti-Bacterial Agents/pharmacology
3.
Int J Biol Macromol ; 176: 244-252, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33548322

ABSTRACT

Polyhydroxyalkanoates (PHAs) are natural polyesters produced by microorganisms as a source of intracellular energy reserves. These polymers have been extensively studied for tissue engineering and drug delivery applications due to their desirable material properties. Solvent-cast film of poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV), produced by Bacillus cereus MCCB 281 was characterized to study the surface morphology, roughness, thermal and mechanical properties. PHBV films were slightly hydrophilic with an average surface roughness of 43.66 nm. In vitro cell viability and proliferation studies on PHBV film surface investigated using L929 fibroblasts showed good cell attachment and proliferation. Hemocompatibility of PHBV evaluated by hemolysis assay, in vitro platelet adhesion and coagulation assays demonstrated good blood compatibility for use as blood contact graft materials. Therefore, PHBV produced from the marine bacterium favoured cellular growth of L929 fibroblasts indicating its potential to be used as a biomaterial substrate for cell adhesion in tissue engineering and medical applications.


Subject(s)
Bacillus cereus/chemistry , Blood Platelets/metabolism , Fibroblasts/metabolism , Materials Testing , Membranes, Artificial , Platelet Adhesiveness , Polyhydroxyalkanoates/chemistry , Animals , Blood Platelets/cytology , Cell Line , Fibroblasts/cytology , Humans , Mice
4.
Arch Biochem Biophys ; 695: 108620, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33038311

ABSTRACT

Potential health benefits of consuming tea are thought to include anti-inflammatory actions of its constituent flavonoids including catechins, which are well-recognized antioxidants. We analyzed and discovered a novel mechanism by which epigallocatechin gallate (EGCG), the most abundant polyphenol in tea and a putative health-promoting constituent, inhibits activation of the nuclear transcription factor NF-κB, which mediates inflammatory responses to cytokines and other agents. We found that EGCG inhibits NF-κB-p65 transcriptional activity, by preventing NF-κB-p65 binding to κBs in normal human bronchial epithelial cells. We also analyzed the chemical mechanism by which EGCG binds directly to NF-κB-p65, and found that it involves covalent reaction via enones within EGCG ring structures, as the oxidizer diamide, which prevents 1, 4-addition reactions, blocked adduct-forming reaction of biotinylated EGCG with NF-κB-p65. Such blockade was inhibited by competing unlabeled EGCG. Furthermore, such covalent binding reflected irreversible reaction of EGCG with sulfhydryls of NF-κB-p65, as it was inhibited by glutathione but not reversible by it. We identified the reactive sulfhydryl moiety as that of cysteine, as S-carboxymethylation to block cysteine sulfhydryls prevented NF-κB-p65-Cys-alkylation reaction with EGCG. We also tested if EGCG can inhibit NF-κB-p65 binding to DNA within the nucleus, after its phosphorylation and translocation (activation). EGCG did not alter intranuclear phosphorylation levels of NF-κB-p65, but strongly repressed DNA-binding ability of activated NF-κB-p65, indicating that EGCG inhibits NF-κB-p65 DNA binding activity even without altering NF-κB-p65 phosphorylation or expression. These findings thus reveal a novel mechanism by which EGCG inhibits transcriptional activity of NF-κB-p65, that may potentially contribute to anti-inflammatory and health-promoting effects of EGCG and consumption of tea.


Subject(s)
Bronchi/metabolism , Catechin/analogs & derivatives , Epithelial Cells/metabolism , Transcription Factor RelA/metabolism , Transcriptional Activation/drug effects , Catechin/chemistry , Catechin/pharmacology , Cell Line , Humans , Phosphorylation/drug effects , Tea/chemistry
5.
Life Sci ; 259: 118260, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32795541

ABSTRACT

Cigarette smoke (CS), the major risk factor of chronic obstructive pulmonary disease (COPD), contains numerous free radicals that can cause oxidative stress and exaggerated inflammatory responses in the respiratory system. Lipid peroxidation which is oxidative degradation of polyunsaturated fatty acids and results in cell damage has also been associated with COPD pathogenesis. Increased levels of lipid peroxidation as well as its end product 4-hydroxynonenal have indeed been detected in COPD patients. Additionally, reactive oxygen species such as those contained in CS can activate nuclear factor-κB signaling pathway, initiating cascades of proinflammatory mediator expression. As emerging evidence attests to the antioxidative and anti-inflammatory properties of tea catechins, we sought to determine whether epigallocatechin gallate, the most abundant tea catechin, can provide protection against oxidative stress, lipid peroxidation, and inflammatory responses caused by CS. We found that EGCG treatment blocked cigarette smoke extract (CSE)-induced oxidative stress as indicated by decreased production and accumulation of reactive oxygen species in airway epithelial cells (AECs). Likewise, lipid peroxidation in CSE-stimulated AECs was suppressed by EGCG. Our findings further suggest that EGCG sequestered 4-hydroxynonenal and interfered with its protein adduct formation. Lastly, we show that EGCG inhibited nuclear factor-κB activation and the downstream expression of proinflammatory mediators. In summary, our study describing the antioxidative and anti-inflammatory effects of EGCG in CSE-exposed AECs provide valuable information about the therapeutic potential of this tea catechin for COPD.


Subject(s)
Alveolar Epithelial Cells/drug effects , Catechin/analogs & derivatives , Cigarette Smoking/drug therapy , Aldehydes/pharmacology , Alveolar Epithelial Cells/metabolism , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Bronchi/metabolism , Catechin/metabolism , Catechin/pharmacology , Cell Line , Cigarette Smoking/adverse effects , Cigarette Smoking/physiopathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Inflammation/metabolism , Lipid Peroxidation/drug effects , NF-kappa B/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Reactive Oxygen Species , Signal Transduction/drug effects , Smoke/adverse effects , Smoking/adverse effects
6.
Life Sci ; 258: 118136, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32726662

ABSTRACT

The endothelium is a critical regulator of vascular homeostasis, controlling vascular tone and permeability as well as interactions of leukocytes and platelets with blood vessel walls. Consequently, endothelial dysfunction featuring inflammation and reduced vasodilation are considered central to cardiovascular disease (CVD) pathogenesis and have become a therapeutic area of focus. Type II endothelial cell (EC) activation by stress-related stimuli such as tumor necrosis factor-α (TNF-α) initiates the nuclear factor-κB (NF-κB) signaling pathway, a master regulator of inflammatory responses. Because dysregulated NF-κB signaling has been tightly linked to several CVDs, EC-specific inhibition of NF-κB represents an attractive pharmacological strategy. As accumulating evidence highlights the clinical benefits of tea catechin for multiple diseases including CVDs, we sought to determine whether the tea catechin epigallocatechin gallate (EGCG) that displays antioxidative, anti-inflammatory, hypolipidemic, anti-thrombogenic, and anti-hypertensive properties offers protection against CVDs by suppressing the canonical NF-κB pathway. Our findings indicate that EGCG downregulates multiple components of the TNF-α-induced NF-κB signaling pathway and thereby reduces the consequent increase in inflammatory gene transcription and protein expression. Furthermore, EGCG blocked type II EC activation, evidenced by diminished EC leakage and monocyte adhesion in EGCG-treated cells. In summary, our study advances knowledge of EGCG's anti-inflammatory effects on the NF-κB pathway and hence its benefits on endothelial health, supporting its therapeutic potential for CVDs.


Subject(s)
Catechin/analogs & derivatives , Coronary Vessels/pathology , Endothelial Cells/pathology , Inflammation/drug therapy , Catechin/pharmacology , Catechin/therapeutic use , Cell Adhesion/drug effects , Cell Membrane Permeability/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation Mediators/metabolism , Monocytes/drug effects , Monocytes/pathology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Clin Sci (Lond) ; 134(9): 1063-1079, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32369100

ABSTRACT

Asthma is a chronic disease of the airways that has long been viewed predominately as an inflammatory condition. Accordingly, current therapeutic interventions focus primarily on resolving inflammation. However, the mainstay of asthma therapy neither fully improves lung function nor prevents disease exacerbations, suggesting involvement of other factors. An emerging concept now holds that airway remodeling, another major pathological feature of asthma, is as important as inflammation in asthma pathogenesis. Structural changes associated with asthma include disrupted epithelial integrity, subepithelial fibrosis, goblet cell hyperplasia/metaplasia, smooth muscle hypertrophy/hyperplasia, and enhanced vascularity. These alterations are hypothesized to contribute to airway hyperresponsiveness, airway obstruction, airflow limitation, and progressive decline of lung function in asthmatic individuals. Consequently, targeting inflammation alone does not suffice to provide optimal clinical benefits. Here we review asthmatic airway remodeling, focusing on airway epithelium, which is critical to maintaining a healthy respiratory system, and is the primary defense against inhaled irritants. In asthma, airway epithelium is both a mediator and target of inflammation, manifesting remodeling and resulting obstruction among its downstream effects. We also highlight the potential benefits of therapeutically targeting airway structural alterations. Since pathological tissue remodeling is likewise observed in other injury- and inflammation-prone tissues and organs, our discussion may have implications beyond asthma and lung disease.


Subject(s)
Airway Remodeling/drug effects , Anti-Asthmatic Agents/pharmacology , Asthma/physiopathology , Inflammation/drug therapy , Animals , Asthma/drug therapy , Epithelium/drug effects , Humans , Inflammation/physiopathology , Lung/drug effects , Lung/physiopathology
8.
PLoS One ; 15(2): e0229256, 2020.
Article in English | MEDLINE | ID: mdl-32084204

ABSTRACT

Cigarette smoke (CS) contains multiple gaseous and particulate materials that can cause lung inflammation, and smoking is the major cause of chronic obstructive pulmonary disease (COPD). We sought to determine the mechanisms of how CS triggers lung inflammation. Nur77, a nuclear hormone receptor belonging to the immediate-early response gene family, controls inflammatory responses, mainly by suppressing the NF-κB signaling pathway. Because it is unknown if Nur77's anti-inflammatory role modulates COPD, we assessed if and how Nur77 expression and activity are altered in CS-induced airway inflammation. In lung tissues and bronchial epithelial cells from COPD patients, we found Nur77 was downregulated. In a murine model of CS-induced airway inflammation, CS promoted lung inflammation and also reduced Nur77 activity in wild type (WT) mice, whereas lungs of Nur77-deficient mice showed exaggerated CS-induced inflammatory responses. Our findings in in vitro studies of human airway epithelial cells complemented those in vivo data in mice, together showing that CS induced threonine-phosphorylation of Nur77, which is known to interfere with its anti-inflammatory functions. In summary, our findings point to Nur77 as an important regulator of CS-induced inflammatory responses and support the potential benefits of Nur77 activation for COPD treatment.


Subject(s)
Down-Regulation/drug effects , Nicotiana/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Smoke/adverse effects , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Inflammation/genetics , Lung/pathology , Mice , Phosphorylation/drug effects , Pulmonary Disease, Chronic Obstructive/pathology , Threonine/metabolism
9.
Article in English | MEDLINE | ID: mdl-32021151

ABSTRACT

Background: Glucocorticoids are commonly prescribed to treat inflammation of the respiratory system; however, they are mostly ineffective for controlling chronic obstructive pulmonary disease (COPD)-associated inflammation. This study aimed to elucidate the molecular mechanisms responsible for such glucocorticoid inefficacy in COPD, which may be instrumental to providing better patient outcomes. Roflumilast is a selective phosphodiesterase-4 (PDE4) inhibitor with anti-inflammatory properties in severe COPD patients who have a history of exacerbations. Roflumilast has a suggested ability to mitigate glucocorticoid resistance, but the mechanism is unknown. Methods: To understand the mechanism that mediates roflumilast-induced restoration of glucocorticoid sensitivity in COPD, we tested the role of glucocorticoid receptor α (GRα). Roflumilast's effects on GRα expression and transcriptional activity were assessed in bronchial epithelial cells from COPD patients. Results: We found that both GRα expression and activity are downregulated in bronchial epithelial cells from COPD patients and that roflumilast stimulates both GRα mRNA synthesis and GRα's transcriptional activity in COPD bronchial epithelial cells. We also demonstrate that roflumilast enhances dexamethasone's ability to suppress pro-inflammatory mediator production, in a GRα-dependent manner. Discussion: Our findings highlight the significance of roflumilast-induced GRα upregulation for COPD therapeutic strategies by revealing that roflumilast restores glucocorticoid sensitivity by sustaining GRα expression.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Glucocorticoids/pharmacology , Lung/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Receptors, Glucocorticoid/agonists , Cells, Cultured , Cyclopropanes/pharmacology , Drug Resistance , Epithelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Lung/metabolism , Lung/physiopathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
10.
Biochem J ; 476(19): 2757-2767, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31492735

ABSTRACT

Nur77 is a transcription factor belonging to the NR4A subfamily of nuclear hormone receptors. Upon induction, Nur77 modulates the expression of its target genes and controls a variety of biological and pathophysiological processes. Prior research that revealed a structurally atypical ligand-binding domain (LBD) and failed to locate an endogenous ligand had led to a classification of Nur77 as an orphan receptor. However, several more recent studies indicate that small synthetic molecules and unsaturated fatty acids can bind to Nur77. Discovery of additional endogenous ligands will facilitate our understanding of the receptor's functions and regulatory mechanisms. Our data have identified prostaglandin A2 (PGA2), a cyclopentenone prostaglandin (PG), as such a ligand. Cyclopentenone PGs exert their biological effects primarily by forming protein adducts via the characteristic electrophilic ß-carbon(s) located in their cyclopentenone rings. Our data show that PGA2 induces Nur77 transcriptional activity by forming a covalent adduct between its endocyclic ß-carbon, C9, and Cys566 in the receptor's LBD. The importance of this endocyclic ß-carbon was substantiated by the failure of PGs without such electrophilic properties to react with Nur77. Calculated chemical properties and data from reactive molecular dynamic simulations, intrinsic reaction co-ordinate modeling, and covalent molecular docking also corroborate the selectivity of PGA2's C9 ß-carbon towards Nur77's Cys. In summary, our molecular, chemical, and structural characterization of the PGA2-Nur77 interaction provides the first evidence that PGA2 is an endogenous Nur77 agonist.


Subject(s)
Nuclear Receptor Subfamily 4, Group A, Member 1/agonists , Prostaglandins A/chemistry , Prostaglandins A/physiology , Cell Line , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Prostaglandins A/metabolism , Protein Binding , Protein Domains
12.
Am J Pathol ; 189(3): 482-491, 2019 03.
Article in English | MEDLINE | ID: mdl-30414411

ABSTRACT

The transcription factor Nur77 belongs to the NR4A subfamily of nuclear hormone receptors. It features an atypical ligand-binding site that precludes canonical ligand binding, leading to the designation orphan nuclear receptor. However, recent studies show that small molecules can interact with the receptor and modulate its activity by inducing a conformational change in the Nur77 ligand-binding site. Nur77 expression and activation are rapidly induced by various physiological and pathologic stimuli. Once expressed, Nur77 initiates transcriptional activity and modulates expression of its target genes. Both in vitro and in vivo evidence shows that Nur77 dampens the immune response to proinflammatory stimuli, such as tumor necrosis factor-α, Toll-like receptor ligands, and oxidized lipids, primarily by suppressing NF-κB signaling. Although studies focusing on Nur77's role in lung pathophysiology are currently incomplete, available data support its involvement in the pathogenesis of lung diseases, including asthma, acute lung injury, and pulmonary fibrosis, and thus suggest a therapeutic potential for Nur77 activation in these diseases. This review addresses the mechanisms that control Nur77 as well as its known roles in inflammation-related lung diseases. Evidence regarding the therapeutic potential of Nur77-targeting molecules will also be presented. Although current knowledge is limited, additional research followed by clinical studies may firmly identify Nur77 as a pharmacologic target for inflammation-related lung diseases.


Subject(s)
Lung Diseases/metabolism , Lung/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Signal Transduction , Transcription, Genetic , Animals , Humans , Inflammation/metabolism , Inflammation/pathology , Lung/pathology , Lung Diseases/pathology , Lung Diseases/therapy , NF-kappa B/biosynthesis , Toll-Like Receptors/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
13.
ACS Chem Biol ; 13(12): 3269-3278, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30398845

ABSTRACT

PPARδ belongs to the peroxisome proliferator-activated receptor (PPAR) family of nuclear receptors. Upon activation by an agonist, PPARδ controls a variety of physiological processes via regulation of its target genes. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) is a cyclopentenone prostaglandin that features an electrophilic, α,ß-unsaturated ketone (an enone) in the cyclopentenone ring. Many of 15d-PGJ2's biological effects result from covalent interaction between C9 and the thiol group of a catalytic cysteine (Cys) in target proteins. In this study, we investigated whether 15d-PGJ2 activates PPARδ by forming a covalent adduct. Our data show that 15d-PGJ2 activates PPARδ's transcriptional activity through formation of a covalent adduct between its endocyclic enone at C9 and Cys249 in the receptor's ligand-binding domain. As expected, no adduct formation was seen following a Cys-to-Ser mutation at residue 249 (C249S) of PPARδ or with a PGD2/PGJ2 analogue that lacks the electrophilic C9. Furthermore, the PPARδ C249S mutation weakened induction of the receptor's DNA binding activity by 15d-PGJ2, which highlights the biological significance of our findings. Calculated chemical properties as well as data from molecular orbital calculations, reactive molecular dynamics simulations, and intrinsic reaction coordinate modeling also supported the selectivity of 15d-PGJ2's C9 toward PPARδ's Cys thiol. In summary, our results provide the molecular, chemical, and structural basis of 15d-PGJ2-mediated PPARδ activation, designating 15d-PGJ2 as the first covalent PPARδ ligand to be identified.


Subject(s)
PPAR delta/agonists , PPAR delta/metabolism , Prostaglandin D2/analogs & derivatives , Alkylation , Cell Line , Cysteine/chemistry , Density Functional Theory , Humans , Ligands , Models, Chemical , Molecular Dynamics Simulation , Mutation , PPAR delta/chemistry , PPAR delta/genetics , Prostaglandin D2/chemistry , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Protein Binding , Protein Domains
14.
Free Radic Biol Med ; 126: 350-357, 2018 10.
Article in English | MEDLINE | ID: mdl-30118830

ABSTRACT

Cigarette smoke, a source of numerous oxidants, produces oxidative stress and exaggerated inflammatory responses that lead to irreversible lung tissue damage. It is the single, most significant risk factor for chronic obstructive pulmonary disease (COPD). Although an intrinsic defense system that includes both enzymatic and non-enzymatic modulators exists to protect lung tissues against oxidative stress, impairment of these protective mechanisms has been demonstrated in smokers and COPD patients. The antioxidant enzyme GSH peroxidase (GPx) is an important part of this intrinsic defense system. Although cigarette smoke has been shown to downregulate its expression and activity, the underlying mechanism is not known. Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear hormone receptor with antioxidant effects. PPARγ activation has demonstrated protective effects against cigarette smoke-induced oxidative stress and inflammation. Molecular mechanisms for PPARγ's antioxidant function likewise remain to be elucidated. This study explored the link between PPARγ and GPx3 and found a positive association in cigarette smoke extract (CSE)-exposed human bronchial epithelial cells. Moreover, we provide evidence that identifies GPx3 as a PPARγ transcriptional target. Attenuation of antioxidant effects in the absence of GPx3 highlights the antioxidant's prominent role in mediating PPARγ's function. We also demonstrate that ligand-mediated PPARγ activation blocks CSE-induced reactive oxygen species and hydrogen peroxide production via upregulation of GPx3. In summary, our findings describing the molecular mechanisms involving GPx3 and PPARγ in CSE-induced oxidative stress and inflammation may provide valuable information for the development of more effective therapeutics for COPD.


Subject(s)
Cigarette Smoking/adverse effects , Glutathione Peroxidase/genetics , PPAR gamma/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Antioxidants/metabolism , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glutathione Peroxidase/metabolism , Humans , Oxidative Stress/genetics , PPAR gamma/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Smoking/adverse effects
15.
J Immunol ; 201(6): 1775-1783, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30061200

ABSTRACT

Airway epithelial cells (AECs) orchestrate inflammatory responses to airborne irritants that enter the respiratory system. A viscous mucus layer produced by goblet cells in the airway epithelium also contributes to a physiological defense mechanism through the physical and chemical barriers it provides. Dysregulation or impairment in these functions has been implicated as a cause of the chronic inflammation and tissue remodeling that constitute major pathological features of asthma. In particular, mucus hypersecretion leading to airway obstruction and impaired pulmonary function is associated with morbidity and mortality in asthma patients. Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated transcription factor involved in a variety of cellular processes. Accumulating evidence indicates that PPARγ agonists antagonize exaggerated inflammatory responses, yet PPARγ's precise role in airway remodeling/mucus hypersecretion has yet to be defined. In this study, we created an AEC-specific PPARγ (AEC-PPARγ) deletion to investigate PPARγ's functions in a murine model of allergic airway disease. AEC-PPARγ deficiency exaggerated airway hyperresponsiveness, inflammation, cytokine expression, and tissue remodeling. We also found that PPARγ directly bound to a PPAR response element found in MUC5AC and repressed gene expression. Likewise, PPARγ regulated mucin and inflammatory factors in primary human bronchial epithelial cells. In light of the current standard therapies' limited and inadequate direct effect on airway mucus hypersecretion, our study showing AEC-PPARγ's role as a transcriptional repressor of MUC5AC highlights this receptor's potential as a pharmacological target for asthma.


Subject(s)
Asthma/immunology , Epithelial Cells/immunology , Gene Expression Regulation/immunology , Mucin 5AC/immunology , PPAR gamma/immunology , Respiratory Mucosa/immunology , Animals , Asthma/genetics , Asthma/pathology , Cells, Cultured , Epithelial Cells/pathology , Female , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Knockout , Mucin 5AC/genetics , PPAR gamma/genetics , Respiratory Mucosa/pathology , Response Elements/immunology
16.
Int J Biol Macromol ; 119: 380-392, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30026096

ABSTRACT

Polyhydroxyalkanoates (PHAs) are aliphatic polyesters produced by bacteria from renewable resources which serve as a substitute of synthetic plastics. In the present study isolation, screening, identification of PHA producing bacteria from marine water samples and optimization of process variables for increased PHA production were accomplished. The potent isolate identified as Bacillus cereus MCCB 281 synthesized PHA co-polymer with 13 mol% 3-hydroxyvalerate in presence of glycerol. Process parameters optimized using central composite design for enhanced PHA production showed 1.5 fold higher PHA yield. Cell dry weight of 3.72 ±â€¯0.04 g L-1, PHA yield 2.54 ±â€¯0.07 g L-1 and PHA content of 68.27 ±â€¯1.2% (w/w) was achieved in fermenter at the optimized conditions. Purified polymer was characterized by Fourier-transform infrared spectroscopy, Nuclear magnetic resonance spectroscopy, Gas chromatography-Mass spectrometry, X-ray powder diffraction techniques and molecular weight of PHA was found to be 2.56 × 105 Da. PHA nanoparticles with average particle size 179 nm were synthesized for medical applications and biocompatibility analysis was performed with L929 mouse fibroblast cell line. This is the first report of a moderately halophilic B. cereus, which utilizes glycerol as the sole carbon source for PHA co-polymer production.


Subject(s)
Bacillus cereus/metabolism , Glycerol/metabolism , Polyhydroxyalkanoates/biosynthesis , Animals , Cell Line , Fermentation , Hydrogen-Ion Concentration , Materials Testing , Mice , Nanoparticles/chemistry , Phylogeny , Polyhydroxyalkanoates/chemistry , Polyhydroxyalkanoates/toxicity , Temperature
17.
Int J Biol Macromol ; 119: 393-401, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29908272

ABSTRACT

A novel esterase, designated as EstSP was identified by function based screening from a soil metagenomic fosmid library of solar saltern of Goa. EstSP gene of 1065 bp encoding a putative esterase of 354 amino acids showing 55% identity to esterase from gamma proteobacterium HIMB55 was identified. The enzyme EstSP belongs to family IV hormone sensitive lipase with novel sequence characteristics and a unique motif GDSGG. EstSP expressed as a His-tag fusion protein of mass 58 kDa was visualized on SDS PAGE and confirmed by Western blot analysis. The enzyme is an alkaline esterase that exhibited highest catalytic activity towards p-nitrophenyl acetate with optimum temperature 40 °C and pH 8.0. The catalytic efficiency and specific activity of EstSP for p-nitrophenyl acetate was 7407.4 min-1 mM-1 and 915.23 U mg-1 respectively. EstSP showed remarkable stability in the presence of polar and non-polar solvents, retaining >80% of its activity after 72 h. Furthermore, the enzyme is halotolerant with optimum activity at 1 M NaCl and maintained 60% residual activity after 24 h exposure to 5 M NaCl. This novel enzyme with remarkable properties could be a promising candidate for industrial bioprocesses in non-aqueous media as well as pharmaceutical, food and biotechnological applications.


Subject(s)
Esterases/chemistry , Esterases/metabolism , Metagenomics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solvents/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Cloning, Molecular , Enzyme Inhibitors/pharmacology , Esterases/antagonists & inhibitors , Esterases/genetics , Gene Expression , Hydrogen-Ion Concentration , Metals/pharmacology , Recombinant Proteins/genetics , Salts/pharmacology , Sequence Analysis , Soil , Substrate Specificity , Temperature
18.
Article in English | MEDLINE | ID: mdl-29450204

ABSTRACT

Asthma affects approximately 300 million people worldwide, significantly impacting quality of life and healthcare costs. While current therapies are effective in controlling many patients' symptoms, a large number continue to experience exacerbations or treatment-related adverse effects. Alternative therapies are thus urgently needed. Accumulating evidence has shown that the peroxisome proliferator-activated receptor (PPAR) family of nuclear hormone receptors, comprising PPARα, PPARß/δ, and PPARγ, is involved in asthma pathogenesis and that ligand-induced activation of these receptors suppresses asthma pathology. PPAR agonists exert their anti-inflammatory effects primarily by suppressing pro-inflammatory mediators and antagonizing the pro-inflammatory functions of various cell types relevant to asthma pathophysiology. Experimental findings strongly support the potential clinical benefits of PPAR agonists in the treatment of asthma. We review current literature, highlighting PPARs' key role in asthma pathogenesis and their agonists' therapeutic potential. With additional research and rigorous clinical studies, PPARs may become attractive therapeutic targets in this disease.

19.
Arch Pharm Res ; 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29043602

ABSTRACT

In aim of obtaining novel bio-active compounds, a new series of fluorinated 1-(4-(aryl)thiazol-2-yl)-2-((1-(aryl)-2,5-dimethyl-1H-pyrrol-3-yl)methylene)hydrazines (5a-t) and 1-(4-(4-aryl)thiazol-2-yl)-2-((3-(2,4-dichlorophenyl)-1-phenyl-1H-pyrazol-4-yl)methylene)hydrazines (8a-d) were synthesized and screened for their antibacterial and antifungal activities. The potent compounds were further screened in vitro for anti-tuberculosis activity against Mycobacterium tuberculosis H37Rv strain. Compounds 5a, 5c-5h and 5m were found to be good inhibitors of B. subtilis with MIC ranging from 0.2 to 0.8 µg mL-1, which are nearly three to ten times more potent than the standard drug Ciprofloxacin. Compounds 5a, 5h-5k and 5o exhibited potent antifungal activity against C. albicans strain with MIC ranging from 0.4 to 1.6 µg mL-1. Compounds 8a-8c were found to be excellent inhibitors of A. niger. Compounds 5a and 5k showed significant anti tubercular activity with MIC 3.12 and 6.25 µg mL-1 respectively. Furthermore, highly active compounds were tested for their preliminary toxicity profile by hemolytic assay and were found to be non hemolytic at higher concentration with good selectivity index. Cytotoxicity of the potent compounds 5a, 5d, 5g, 5i and 5k was checked by MTT assay using normal embryonic kidney cell line HEK 293 and found to be non-toxic up to 50-200 times the MIC for antibacterial activity.

20.
Int J Chron Obstruct Pulmon Dis ; 12: 2141-2156, 2017.
Article in English | MEDLINE | ID: mdl-28790817

ABSTRACT

COPD, for which cigarette smoking is the major risk factor, remains a worldwide burden. Current therapies provide only limited short-term benefit and fail to halt progression. A variety of potential therapeutic targets are currently being investigated, including COPD-related proinflammatory mediators and signaling pathways. Other investigational compounds target specific aspects or complications of COPD such as mucus hypersecretion and pulmonary hypertension. Although many candidate therapies have shown no significant effects, other emerging therapies have improved lung function, pulmonary hypertension, glucocorticoid sensitivity, and/or the frequency of exacerbations. Among these are compounds that inhibit the CXCR2 receptor, mitogen-activated protein kinase/Src kinase, myristoylated alanine-rich C kinase substrate, selectins, and the endothelin receptor. Activation of certain transcription factors may also be relevant, as a large retrospective cohort study of COPD patients with diabetes found that the peroxisome proliferator-activated receptor γ (PPARγ) agonists rosiglitazone and pioglitazone were associated with reduced COPD exacerbation rate. Notably, several therapies have shown efficacy only in identifiable subgroups of COPD patients, suggesting that subgroup identification may become more important in future treatment strategies. This review summarizes the status of emerging therapeutic pharmaceuticals for COPD and highlights those that appear most promising.


Subject(s)
Drug Design , Lung/drug effects , Molecular Targeted Therapy/methods , Pulmonary Disease, Chronic Obstructive/drug therapy , Respiratory System Agents/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Bronchodilator Agents/therapeutic use , Drug Resistance , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lung/enzymology , Lung/physiopathology , Molecular Targeted Therapy/adverse effects , PPAR gamma/agonists , PPAR gamma/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory System Agents/adverse effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...