Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Immunol ; 197(11): 4228-4239, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27794000

ABSTRACT

KIF3A, the gene encoding kinesin family member 3A, is a susceptibility gene locus associated with asthma; however, mechanisms by which KIF3A might influence the pathogenesis of the disorder are unknown. In this study, we deleted the mouse Kif3a gene in airway epithelial cells. Both homozygous and heterozygous Kif3a gene-deleted mice were highly susceptible to aeroallergens from Aspergillus fumigatus and the house dust mite, resulting in an asthma-like pathology characterized by increased goblet cell metaplasia, airway hyperresponsiveness, and Th2-mediated inflammation. Deletion of the Kif3a gene increased the severity of pulmonary eosinophilic inflammation and expression of cytokines (Il-4, Il-13, and Il-17a) and chemokine (Ccl11) RNAs following pulmonary exposure to Aspergillus extract. Inhibition of Kif3a disrupted the structure of motile cilia and impaired mucociliary clearance, barrier function, and epithelial repair, demonstrating additional mechanisms by which deficiency of KIF3A in respiratory epithelial cells contributes to pulmonary pathology. Airway epithelial KIF3A suppresses Th2 pulmonary inflammation and airway hyperresponsiveness following aeroallergen exposure, implicating epithelial microtubular functions in the pathogenesis of Th2-mediated lung pathology.


Subject(s)
Allergens/immunology , Aspergillus fumigatus/immunology , Asthma/immunology , Epithelial Cells/immunology , Kinesins/immunology , Respiratory Mucosa/immunology , Th2 Cells/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Cytokines/genetics , Cytokines/immunology , Epithelial Cells/pathology , Kinesins/genetics , Lung/immunology , Lung/pathology , Mice , Mice, Transgenic , Respiratory Mucosa/pathology , Th2 Cells/pathology
2.
J Biol Chem ; 290(30): 18559-74, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26048993

ABSTRACT

Pulmonary surfactant, a mixture of proteins and phospholipids, plays an important role in facilitating gas exchange by maintaining alveolar stability. Saturated phosphatidylcholine (SatPC), the major component of surfactant, is synthesized both de novo and by the remodeling of unsaturated phosphatidylcholine (PC) by lyso-PC acyltransferase 1 (LPCAT1). After synthesis in the endoplasmic reticulum, SatPC is routed to lamellar bodies (LBs) for storage prior to secretion. The mechanism by which SatPC is transported to LB is not understood. The specificity of LPCAT1 for lyso-PC as an acyl acceptor suggests that formation of SatPC via LPCAT1 reacylation is a final step in SatPC synthesis prior to transport. We hypothesized that LPCAT1 forms a transient complex with SatPC and specific phospholipid transport protein(s) to initiate trafficking of SatPC from the endoplasmic reticulum to the LB. Herein we have assessed the ability of different StarD proteins to interact with LPCAT1. We found that LPCAT1 interacts with StarD10, that this interaction is direct, and that amino acids 79-271 of LPCAT1 and the steroidogenic acute regulatory protein-related lipid transfer (START) domain of START domain-containing protein 10 (StarD10) are sufficient for this interaction. The role of StarD10 in trafficking of phospholipid to LB was confirmed by the observation that knockdown of StarD10 significantly reduced transport of phospholipid to LB. LPCAT1 also interacted with one isoform of StarD7 but showed no interaction with StarD2/PC transfer protein.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Lipid Metabolism , Phospholipids/biosynthesis , Phosphoproteins/metabolism , Pulmonary Alveoli/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Humans , Mice , Phosphatidylcholines/biosynthesis , Phosphatidylcholines/metabolism , Phospholipids/metabolism , Phosphoproteins/genetics , Protein Interaction Maps/genetics , Pulmonary Alveoli/cytology , Pulmonary Surfactants/metabolism
3.
J Biol Chem ; 290(18): 11246-57, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25762723

ABSTRACT

Diarrhea is one of the most common adverse side effects observed in Ć¢ĀˆĀ¼7% of individuals consuming Food and Drug Administration (FDA)-approved drugs. The mechanism of how these drugs alter fluid secretion in the gut and induce diarrhea is not clearly understood. Several drugs are either substrates or inhibitors of multidrug resistance protein 4 (MRP4), such as the anti-colon cancer drug irinotecan and an anti-retroviral used to treat HIV infection, 3'-azido-3'-deoxythymidine (AZT). These drugs activate cystic fibrosis transmembrane conductance regulator (CFTR)-mediated fluid secretion by inhibiting MRP4-mediated cAMP efflux. Binding of drugs to MRP4 augments the formation of MRP4-CFTR-containing macromolecular complexes that is mediated via scaffolding protein PDZK1. Importantly, HIV patients on AZT treatment demonstrate augmented MRP4-CFTR complex formation in the colon, which defines a novel paradigm of drug-induced diarrhea.


Subject(s)
Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Diarrhea/chemically induced , Multidrug Resistance-Associated Proteins/metabolism , Animals , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Drug Approval , HT29 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Irinotecan , Mice , Models, Molecular , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/deficiency , Protein Conformation , United States , United States Food and Drug Administration
4.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L364-74, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27261451

ABSTRACT

Cystic fibrosis (CF) is a life-shortening disease caused by the mutations that generate nonfunctional CF transmembrane conductance regulator (CFTR) protein. A rare serine-to-tyrosine (S1045Y) CFTR mutation was earlier reported to result in CF-associated fatality. We identified an African-American patient with the S1045Y mutation in CFTR, as well as a stop-codon mutation, who has a mild CF phenotype. The underlying mechanism of CF caused by S1045Y-CFTR has not been elucidated. In this study, we determined that S1045Y-CFTR exhibits twofold attenuated function compared with wild-type (WT)-CFTR. We report that serine-to-tyrosine mutation leads to increased tyrosine phosphorylation of S1045Y-CFTR, followed by recruitment and binding of E3-ubiquitin ligase c-cbl, resulting in enhanced ubiquitination and passage of S1045Y-CFTR in the endosome/lysosome degradative compartments. We demonstrate that inhibition of tyrosine phosphorylation partially rescues S1045Y-CFTR surface expression and function. Based on our findings, it could be suggested that consuming genistein (a tyrosine phosphorylation inhibitor) would likely ameliorate CF symptoms in individuals with S1045Y-CFTR, providing a unique personalized therapy for this rare CF mutation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/diagnostic imaging , Genistein/pharmacology , Protein Kinase Inhibitors/pharmacology , Child, Preschool , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , DNA Mutational Analysis , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Male , Mutation, Missense , Phosphorylation , Precision Medicine , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Proto-Oncogene Proteins c-cbl/metabolism
5.
J Biol Chem ; 289(52): 35757-69, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25542932

ABSTRACT

Chemotactic migration of fibroblasts toward growth factors relies on their capacity to sense minute extracellular gradients and respond to spatially confined receptor-mediated signals. Currently, mechanisms underlying the gradient sensing of fibroblasts remain poorly understood. Using single-particle tracking methodology, we determined that a lysophosphatidic acid (LPA) gradient induces a spatiotemporally restricted decrease in the mobility of LPA receptor 2 (LPA2) on chemotactic fibroblasts. The onset of decreased LPA2 mobility correlates to the spatial recruitment and coupling to LPA2-interacting proteins that anchor the complex to the cytoskeleton. These localized PDZ motif-mediated macromolecular complexes of LPA2 trigger a Ca(2+) puff gradient that governs gradient sensing and directional migration in response to LPA. Disruption of the PDZ motif-mediated assembly of the macromolecular complex of LPA2 disorganizes the gradient of Ca(2+) puffs, disrupts gradient sensing, and reduces the directional migration of fibroblasts toward LPA. Our findings illustrate that the asymmetric macromolecular complex formation of chemoattractant receptors mediates gradient sensing and provides a new mechanistic basis for models to describe gradient sensing of fibroblasts.


Subject(s)
Fibroblasts/physiology , Membrane Microdomains/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Calcium Signaling , Chemotaxis , Lysophospholipids/physiology , Mice , NIH 3T3 Cells , Phospholipase C beta/metabolism , Protein Multimerization , Protein Transport
6.
Pharmacol Res ; 102: 107-112, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26429773

ABSTRACT

Many medications induce diarrhea as a side effect, which can be a major obstacle to therapeutic efficacy and also a life-threatening condition. Secretory diarrhea can be caused by excessive fluid secretion in the intestine under pathological conditions. The cAMP/cGMP-regulated cystic fibrosis transmembrane conductance regulator (CFTR) is the primary chloride channel at the apical membrane of intestinal epithelial cells and plays a major role in intestinal fluid secretion and homeostasis. CFTR forms macromolecular complexes at discreet microdomains at the plasma membrane, and its chloride channel function is regulated spatiotemporally through protein-protein interactions and cAMP/cGMP-mediated signaling. Drugs that perturb CFTR-containing macromolecular complexes in the intestinal epithelium and upregulate intracellular cAMP and/or cGMP levels can hyperactivate the CFTR channel, causing excessive fluid secretion and secretory diarrhea. Inhibition of CFTR chloride-channel activity may represent a novel approach to the management of drug-induced secretory diarrhea.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Diarrhea/chemically induced , Drug-Related Side Effects and Adverse Reactions/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Diarrhea/metabolism , Humans
7.
Biochemistry ; 53(25): 4169-79, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24945463

ABSTRACT

Cystic fibrosis (CF) is a recessive genetic disease caused by mutations in CFTR, a plasma-membrane-localized anion channel. The most common mutation in CFTR, deletion of phenylalanine at residue 508 (ΔF508), causes misfolding of CFTR resulting in little or no protein at the plasma membrane. The CFTR corrector VX-809 shows promise for treating CF patients homozygous for ΔF508. Here, we demonstrate the significance of protein-protein interactions in enhancing the stability of the ΔF508 CFTR mutant channel protein at the plasma membrane. We determined that VX-809 prolongs the stability of ΔF508 CFTR at the plasma membrane. Using competition-based assays, we demonstrated that ΔF508 CFTR interacts poorly with Na(+)/H(+) exchanger regulatory factor 1 (NHERF1) compared to wild-type CFTR, and VX-809 significantly increased this binding affinity. We conclude that stabilized CFTR-NHERF1 interaction is a determinant of the functional efficiency of rescued ΔF508 CFTR. Our results demonstrate the importance of macromolecular-complex formation in stabilizing rescued mutant CFTR at the plasma membrane and suggest this to be foundational for the development of a new generation of effective CFTR-corrector-based therapeutics.


Subject(s)
Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Male , Mice, Inbred C57BL , Mutation , Protein Conformation , Protein Stability
8.
J Biol Chem ; 288(17): 12325-34, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23504457

ABSTRACT

The PDZ (postsynaptic density-95/discs large/zona occludens-1) domain-based interactions play important roles in regulating the expression and function of the cystic fibrosis transmembrane conductance regulator (CFTR). Several PDZ domain-containing proteins (PDZ proteins for short) have been identified as directly or indirectly interacting with the C terminus of CFTR. To better understand the regulation of CFTR processing, we conducted a genetic screen and identified MAST205 (a microtubule-associated serine/threonine kinase with a molecular mass of 205 kDa) as a new CFTR regulator. We found that overexpression of MAST205 increased the expression of CFTR and augmented CFTR-mediated fluid transport in a dose-dependent manner. Conversely, knockdown of MAST205 inhibited CFTR function. The PDZ motif of CFTR is required for the regulatory role of MAST205 in CFTR expression and function. We further demonstrated that MAST205 and the CFTR-associated ligand competed for binding to CFTR, which facilitated the processing of CFTR and consequently up-regulated the expression and function of CFTR at the plasma membrane. More importantly, we found that MAST205 could facilitate the processing of F508del-CFTR mutant and augment its quantity and channel function at the plasma membrane. Taken together, our data suggest that MAST205 plays an important role in regulating CFTR expression and function. Our findings have important clinical implications for treating CFTR-associated diseases such as cystic fibrosis and secretory diarrheas.


Subject(s)
Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression Regulation , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Water-Electrolyte Balance , Amino Acid Sequence , Biological Transport, Active/genetics , Cell Membrane/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Diarrhea/genetics , Diarrhea/metabolism , Diarrhea/pathology , Gene Knockdown Techniques , HEK293 Cells , Humans , Microtubule-Associated Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Sequence Deletion
9.
J Biol Chem ; 288(6): 3786-94, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23264633

ABSTRACT

It has long been known that cyclic nucleotides and cyclic nucleotide-dependent signaling molecules control cell migration. However, the concept that it is not just the absence or presence of cyclic nucleotides, but a highly coordinated balance between these molecules that regulates cell migration, is new and revolutionary. In this study, we used multidrug resistance protein 4 (MRP4)-expressing cell lines and MRP4 knock-out mice as model systems and wound healing assays as the experimental system to explore this unique and emerging concept. MRP4, a member of a large family of ATP binding cassette transporter proteins, localizes to the plasma membrane and functions as a nucleotide efflux transporter and thus plays a role in the regulation of intracellular cyclic nucleotide levels. Here, we demonstrate that mouse embryonic fibroblasts (MEFs) isolated from Mrp4(-/-) mice have higher intracellular cyclic nucleotide levels and migrate faster compared with MEFs from Mrp4(+/+) mice. Using FRET-based cAMP and cGMP sensors, we show that inhibition of MRP4 with MK571 increases both cAMP and cGMP levels, which results in increased migration. In contrast to these moderate increases in cAMP and cGMP levels seen in the absence of MRP4, a robust increase in cAMP levels was observed following treatment with forskolin and isobutylmethylxanthine, which decreases fibroblast migration. In response to externally added cell-permeant cyclic nucleotides (cpt-cAMP and cpt-cGMP), MEF migration appears to be biphasic. Altogether, our studies provide the first experimental evidence supporting the novel concept that balance between cyclic nucleotides is critical for cell migration.


Subject(s)
Cell Movement/physiology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Fibroblasts/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Animals , Cell Movement/drug effects , Cyclic AMP/genetics , Cyclic GMP/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Leukotriene Antagonists/pharmacology , Mice , Mice, Knockout , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/genetics , NIH 3T3 Cells , Propionates/pharmacology , Quinolines/pharmacology , Tissue Array Analysis
10.
Eur Respir J ; 40(2): 424-35, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22441736

ABSTRACT

Apoptotic cell clearance by macrophages and neighbouring tissue cells induces hepatocyte growth factor (HGF) secretion. HGF plays a key role in alveolar epithelial regeneration and reconstruction after lung injury. Direct in vivo exposure to apoptotic cells enhances HGF production, resulting in attenuation of pulmonary injury. We investigated the direct effect of in vivo exposure to apoptotic cells in bleomycin-stimulated lungs (2 days old) on HGF induction. Furthermore, sequential changes of bleomycin-induced HGF production following apoptotic cell instillation related to the changes in inflammatory and fibrotic responses were assessed. At 2 h after apoptotic cell instillation into bleomycin-stimulated lungs, the levels of HGF mRNA and protein production, and apoptotic cell clearance by alveolar macrophages were enhanced. Furthermore, HGF induction persistently increased following apoptotic cell instillation up to 21 days after bleomycin treatment. Apoptotic cell instillation attenuated bleomycin-induced pro-inflammatory mediator production, inflammatory cell recruitment and total protein levels. Apoptotic cell instillation also induced antiapoptotic and antifibrotic effects. These anti-inflammatory and antiapoptotic effects could be reversed by co-administration of HGF-neutralising antibody. These findings indicate that in vivo exposure to apoptotic cells enhances transcriptional HGF production in bleomycin-stimulated lungs, resulting in attenuation of lung injury and fibrosis.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis , Bleomycin/pharmacology , Hepatocyte Growth Factor/metabolism , Lung Injury/drug therapy , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Proliferation , Epithelial Cells/cytology , Humans , Inflammation/metabolism , Jurkat Cells , Lung/pathology , Macrophages/metabolism , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , RNA, Messenger/metabolism , Time Factors
11.
Am J Respir Crit Care Med ; 181(4): 374-87, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19965809

ABSTRACT

RATIONALE: The resolution of pulmonary inflammation seen in various inflammatory lung conditions depends on the clearance of apoptotic cells to prevent permanent tissue damage or progressive disease. Uptake of apoptotic cells by alveolar macrophages is suppressed by oxidants through the activation of Rho signaling. OBJECTIVES: We hypothesized that antioxidant exposure would increase the ability of alveolar macrophages to clear pulmonary apoptotic cells through the inhibition of RhoA. METHODS: The effects of the antioxidant N-acetylcysteine (NAC) on the pulmonary immune response were seen in mice treated intratracheally with LPS, LPS + NAC, or saline. Apoptotic cell clearance, RhoA activity, and changes in the lung inflammatory responses were analyzed in vivo or ex vivo. MEASUREMENTS AND MAIN RESULTS: Neutrophil accumulation, apoptosis, necrosis, and oxidant production peaked at 3 days post LPS treatment. NAC enhanced the clearance of apoptotic cells and inhibited RhoA activity in alveolar macrophages at 3 days post LPS treatment. NAC suppressed LPS-induced proinflammatory mediators, enhanced the production of transforming growth factor-beta1, reduced the accumulation of inflammatory cells, and reduced levels of protein and lactate dehydrogenase in bronchoalveolar lavage fluid. In the presence of ex vivo apoptotic cells, alveolar macrophages exposed to LPS or LPS + NAC had reduced tumor necrosis factor-alpha levels and increased transforming growth factor-beta1 levels. A Rho kinase inhibitor mimicked the effects of NAC on the clearance of apoptotic cells and the inflammatory responses. CONCLUSIONS: These results indicate that NAC can expedite the resolution of LPS-induced pulmonary inflammation through the inhibition of RhoA activity and the enhancement of apoptotic cell clearance.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Pneumonia/physiopathology , rhoA GTP-Binding Protein/antagonists & inhibitors , Amides/pharmacology , Animals , Apoptosis/physiology , Blotting, Western , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , L-Lactate Dehydrogenase/metabolism , Macrophages, Alveolar/physiology , Male , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Pneumonia/drug therapy , Pneumonia/pathology , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
12.
J Toxicol Environ Health A ; 73(5): 396-409, 2010.
Article in English | MEDLINE | ID: mdl-20155581

ABSTRACT

Nanoparticles are widely used in nanomedicines, including for targeted delivery of pharmacological, therapeutic, and diagnostic agents. Since nanoparticles might translocate across cellular barriers from the circulation into targeted organs, it is important to obtain information concerning the pathophysiologic effects of these particles through systemic migration. In the present study, acute pulmonary responses were examined after intraperitoneal (ip) administration of ultrafine titanium dioxide (TiO(2), 40 mg/kg) in mice at rest or in lungs primed with lipopolysaccharide (LPS, ip, 5 mg/kg). Ultrafine TiO(2) exposure increased neutrophil influx, protein levels in bronchoalveolar lavage (BAL) fluid, and reactive oxygen species (ROS) activity of BAL cells 4 h after exposure. Concomitantly, the levels of proinflammatory mediators, such as tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, and macrophage inflammatory protein (MIP)-2 in BAL fluid and mRNA expression of TNF-alpha and IL-1beta in lung tissue were elevated post ultrafine TiO(2) exposure. Ultrafine TiO(2) exposure resulted in significant activation of inflammatory signaling molecules, such as c-Src and p38 MAP kinase, in lung tissue and alveolar macrophages, and the nuclear factor (NF)-kappaB pathway in pulmonary tissue. Furthermore, ultrafine TiO(2) additively enhanced these inflammatory parameters and this signaling pathway in lungs primed with lipopolysaccharide (LPS). Contrary to this trend, a synergistic effect was found for TNF-alpha at the level of protein and mRNA expression. These results suggest that ultrafine TiO(2) (P25) induces acute lung inflammation after ip administration, and exhibits additive or synergistic effects with LPS, at least partly, via activation of oxidant-dependent inflammatory signaling and the NF-kappaB pathway, leading to increased production of proinflammatory mediators.


Subject(s)
Lipopolysaccharides/pharmacology , Metal Nanoparticles/toxicity , Particulate Matter/toxicity , Pneumonia/chemically induced , Titanium/toxicity , Animals , Bronchi/drug effects , Bronchi/metabolism , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2/metabolism , Drug Synergism , Injections, Intraperitoneal , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , Metal Nanoparticles/administration & dosage , Mice , Mice, Inbred BALB C , Particulate Matter/administration & dosage , Pneumonia/immunology , Pneumonia/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Rest , Titanium/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Respir Res ; 10: 18, 2009 Mar 09.
Article in English | MEDLINE | ID: mdl-19272161

ABSTRACT

BACKGROUND: Synthetic peptides containing the RGD sequence inhibit integrin-related functions in different cell systems. Here, we investigated the effects of synthetic Arg-Gly-Asp-Ser (RGDS) peptide on key inflammatory responses to intratracheal (i.t.) lipopolysaccharide (LPS) treatment and on the integrin signaled mitogen-activated protein (MAP) kinase pathway during the development of acute lung injury. METHODS: Saline or LPS (1.5 mg/kg) was administered i.t. with or without a single dose of RGDS (1, 2.5, or 5 mg/kg, i.p.), anti-alphav or anti-beta3 mAb (5 mg/kg, i.p.). Mice were sacrificed 4 or 24 h post-LPS. RESULTS: A pretreatment with RGDS inhibited LPS-induced increases in neutrophil and macrophage numbers, total protein levels and TNF-alpha and MIP-2 levels, and matrix metalloproteinase-9 activity in bronchoalveolar lavage (BAL) fluid at 4 or 24 h post-LPS treatment. RGDS inhibited LPS-induced phosphorylation of focal adhesion kinase and MAP kinases, including ERK, JNK, and p38 MAP kinase, in lung tissue. Importantly, the inhibition of the inflammatory responses and the kinase pathways were still evident when this peptide was administered 2 h after LPS treatment. Similarly, a blocking antibody against integrin alphav significantly inhibited LPS-induced inflammatory cell migration into the lung, protein accumulation and proinflammatory mediator production in BAL fluid, at 4 or 24 h post-LPS. Anti-beta3 also inhibited all LPS-induced inflammatory responses, except the accumulation of BAL protein at 24 h post-LPS. CONCLUSION: These results suggest that RGDS with high specificity for alphavintegrins attenuates inflammatory cascade during LPS-induced development of acute lung injury.


Subject(s)
Acute Lung Injury/prevention & control , Integrin alphaVbeta3/metabolism , Lung/drug effects , MAP Kinase Signaling System/drug effects , Oligopeptides/pharmacology , Pneumonia/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Chemokine CXCL2/metabolism , Chemotaxis/drug effects , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides , Lung/enzymology , Lung/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Phosphorylation , Pneumonia/chemically induced , Pneumonia/enzymology , Pneumonia/immunology , Time Factors , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Sci Rep ; 9(1): 9808, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31285458

ABSTRACT

The molecular mechanism of Endoplasmic Reticulum-associated degradation (ERAD) of Cystic fibrosis transmembrane-conductance regulator (CFTR) is largely unknown. Particularly, it is unknown what ER luminal factor(s) are involved in ERAD. Herein, we used ProtoArray to identify an ER luminal co-chaperone, DNAJB9, which can directly interact with CFTR. For both WT- and ΔF508 (deletion of phenylalanine at position 508, the most common CF-causing mutant)-CFTR, knockdown of DNAJB9 by siRNA increased their expression levels on the cell surface and, consequently, upregulated their function. Furthermore, genetic ablation of DNAJB9 in WT mice increased CFTR expression and enhanced CFTR-dependent fluid secretion in enteroids. Importantly, DNAJB9 deficiency upregulated enteroids' fluid secretion in CF mice (homozygous for ΔF508), and silencing one allele of DNAJB9 is sufficient to rescue ΔF508-CFTR in vitro and in vivo, suggesting that DNAJB9 may be a rate-limiting factor in CFTR ERAD pathway. Our studies identified the first ER luminal co-chaperone involved in CFTR ERAD, and DNAJB9 could be a novel therapeutic target for CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Sequence Deletion , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endoplasmic Reticulum-Associated Degradation , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Protein Array Analysis
16.
J Toxicol Environ Health A ; 71(8): 478-85, 2008.
Article in English | MEDLINE | ID: mdl-18338282

ABSTRACT

Ultrafine or fine titanium dioxide (TiO(2)) particles are widely used in the production of white pigments, for sunscreens, and in cleanup techniques. However, currently knowledge is deficient concerning cellular responses to these particles. The study evaluated and compared the biological activity of ultrafine and fine TiO(2) particles in RAW 264.7 macrophages according to an oxidative stress paradigm. In vitro exposure of macrophages to ultrafine or fine TiO(2) in the range of 0.5-200 microg/ml did not significantly alter cell viability. However, ultrafine TiO(2) enhanced intracellular generation of reactive oxygen species (ROS) to a greater extent than fine TiO(2) at each exposure concentration. Ultrafine TiO(2) induced ERK1/2 activation in a concentration-dependent manner, while the fine TiO(2)-induced changes were minimal. Phosphorylation of ERK1/2 occurred following 10 min exposure to higher concentrations of ultrafine TiO(2) (> or = 25 microg/ml). Similarly, ultrafine TiO(2) exposure significantly enhanced tumor necrosis factor (TNF)-alpha and macrophage inflammatory protein (MIP)-2 secretion in a concentration-dependent manner, and its potency was higher than fine TiO(2). These findings suggest that when exposure concentration is based upon equivalent mass, ultrafine TiO(2) exerts greater biological activity as measured by ROS generation, ERK 1/2 activation, and proinflammatory mediator secretion in RAW 264.7 macrophages than fine TiO(2).


Subject(s)
Macrophages, Peritoneal/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Titanium/toxicity , Air Pollutants , Animals , Cell Line , Inflammation/metabolism , MAP Kinase Signaling System/drug effects , Mice , Particle Size , Titanium/chemistry , Toxicity Tests
17.
Cell Signal ; 27(9): 1751-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26027517

ABSTRACT

In this study we characterized the effects of radiation injury on the expression and function of the autotaxin (ATX)-LPA2 GPCR axis. In IEC-6 crypt cells and jejunum enteroids quantitative RT-PCR showed a time- and dose-dependent upregulation of lpa2 in response to ƎĀ³-irradiation that was abolished by mutation of the NF-κB site in the lpa2 promoter or by inhibition of ATM/ATR kinases with CGK-733, suggesting that lpa2 is a DNA damage response gene upregulated by ATM via NF-κB. The resolution kinetics of the DNA damage marker ƎĀ³-H2AX in LPA-treated IEC-6 cells exposed to ƎĀ³-irradiation was accelerated compared to vehicle, whereas pharmacological inhibition of LPA2 delayed the resolution of ƎĀ³-H2AX. In LPA2-reconstituted MEF cells lacking LPA1&3 the levels of ƎĀ³-H2AX decreased rapidly, whereas in Vector MEF were high and remained sustained. Inhibition of ERK1&2 or PI3K/AKT signaling axis by pertussis toxin or the C311A/C314A/L351A mutation in the C-terminus of LPA2 abrogated the effect of LPA on DNA repair. LPA2 transcripts in Lin(-)Sca-1(+)c-Kit(+) enriched for bone marrow stem cells were 27- and 5-fold higher than in common myeloid or lymphoid progenitors, respectively. Furthermore, after irradiation higher residual ƎĀ³-H2AX levels were detected in the bone marrow or jejunum of irradiated LPA2-KO mice compared to WT mice. We found that ƎĀ³-irradiation increases plasma ATX activity and LPA level that is in part due to the previously established radiation-induced upregulation of TNFα. These findings identify ATX and LPA2 as radiation-regulated genes that appear to play a physiological role in DNA repair.


Subject(s)
DNA Damage , Gamma Rays , MAP Kinase Signaling System/radiation effects , Phosphoric Diester Hydrolases/metabolism , Radiation Injuries, Experimental/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Cell Line , Jejunum/metabolism , Jejunum/pathology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/pathology , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Diester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/pathology , Rats , Receptors, Lysophosphatidic Acid/genetics , Response Elements
18.
PLoS One ; 7(4): e35156, 2012.
Article in English | MEDLINE | ID: mdl-22529984

ABSTRACT

The entirety of all protein coding sequences is reported to represent a small fraction (~2%) of the mouse and human genomes; the vast majority of the rest of the genome is presumed to be repetitive elements (REs). In this study, the C57BL/6J mouse reference genome was subjected to an unbiased RE mining to establish a whole-genome profile of RE occurrence and arrangement. The C57BL/6J mouse genome was fragmented into an initial set of 5,321 units of 0.5 Mb, and surveyed for REs using unbiased self-alignment and dot-matrix protocols. The survey revealed that individual chromosomes had unique profiles of RE arrangement structures, named RE arrays. The RE populations in certain genomic regions were arranged into various forms of complexly organized structures using combinations of direct and/or inverse repeats. Some of these RE arrays spanned stretches of over 2 Mb, which may contribute to the structural configuration of the respective genomic regions. There were substantial differences in RE density among the 21 chromosomes, with chromosome Y being the most densely populated. In addition, the RE array population in the mouse chromosomes X and Y was substantially different from those of the reference human chromosomes. Conversion of the dot-matrix data pertaining to a tandem 13-repeat structure within the Ch7.032 genome unit into a line map of known REs revealed a repeat unit of ~11.3 Kb as a mosaic of six different RE types. The data obtained from this study allowed for a comprehensive RE profiling, including the establishment of a library of RE arrays, of the reference mouse genome. Some of these RE arrays may participate in a spectrum of normal and disease biology that are specific for mice.


Subject(s)
Genome , Mice, Inbred C57BL/genetics , Repetitive Sequences, Nucleic Acid , Animals , Chromosome Mapping , Chromosomes, Mammalian , Genes, myc , Humans , Immunoglobulin M/genetics , Immunoglobulin Switch Region , Mice , Translocation, Genetic , X Chromosome , Y Chromosome
19.
J Immunol ; 179(10): 7001-11, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982091

ABSTRACT

Src tyrosine kinases (TKs) are signaling proteins involved in cell signaling pathways toward cytoskeletal, membrane and nuclear targets. In the present study, using a selective Src TK inhibitor, PP1, we investigated the roles of Src TKs in the key pulmonary responses, NF-kappaB activation, and integrin signaling during acute lung injury in BALB/C mice intratracheally treated with LPS. LPS resulted in c-Src phosphorylation in lung tissue and the phospho-c-Src was predominantly localized in recruited neutrophils and alveolar macrophages. PP1 inhibited LPS-induced increases in total protein content in bronchoalveolar lavage fluid, neutrophil recruitment, and increases in the production or activity of TNF-alpha and matrix metalloproteinase-9. PP1 also blocked LPS-induced NF-kappaB activation, and phosphorylation and degradation of IkappaB-alpha. The inhibition of NF-kappaB activation by PP1 correlated with a depression of LPS-induced integrin signaling, which included increases in the phosphorylations of integrin beta(3), and of the focal adhesion kinase (FAK) family members, FAK and Pyk2, in lung tissue, and reductions in the fibrinogen-binding activity of alveolar macrophages. Moreover, treatment with anti-alpha(v), anti-beta(3), or Arg-Gly-Asp-Ser (RGDS), inhibited LPS-induced NF-kappaB activation. Taken together, our findings suggest that Src TKs play a critical role in LPS-induced activations of NF-kappaB and integrin (alpha(v)beta(3)) signaling during acute lung injury. Therefore, Src TK inhibition may provide a potential means of ameliorating inflammatory cascade-associated lung injury.


Subject(s)
Integrin alphaVbeta3/immunology , NF-kappa B/immunology , Respiratory Distress Syndrome/immunology , Signal Transduction/immunology , src-Family Kinases/immunology , Animals , Focal Adhesion Kinase 1/immunology , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/immunology , Focal Adhesion Kinase 2/metabolism , I-kappa B Proteins/immunology , I-kappa B Proteins/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Lipopolysaccharides/toxicity , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Oligopeptides/pharmacology , Phosphorylation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptors, Neuropeptide Y/immunology , Receptors, Neuropeptide Y/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/pathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , src-Family Kinases/metabolism
20.
Biochem Biophys Res Commun ; 337(2): 481-9, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16223607

ABSTRACT

Neuronatin (Nnat) is selectively expressed in the neonatal brain and is involved in neuronal differentiation during brain development. However, Nnat also appears to be abundantly expressed in adipose tissue, and is conspicuously elevated in the adipose tissue of obese Zucker diabetic fatty rats compared with control lean Zucker lean control rats shown in our previous report. Here, we examined the expression of Nnat in adipose tissue and demonstrated that the ectopic expression of Nnat mediated by retroviral infection or stable transfection of 3T3-L1 pre-adipocytes stimulated differentiation into mature adipocytes with early induction of adipogenic transcription factors. Moreover, in 3T3-L1 cells overexpressing Nnat, increased intracellular free calcium levels and enhanced phosphorylation of cAMP-response element-binding protein (CREB) were observed, which appears to potentiate CCAAT/enhancer-binding protein (C/EBP)beta, C/EBPdelta, and C/EBPalpha transcriptional activities. Collectively, the data indicate that Nnat enhances CREB phosphorylation through increasing intracellular free calcium levels, which potentiates expression of adipogenic transcription factors resulting in heightened adipocyte differentiation. These findings contribute to a greater fundamental understanding of obesity, a clinically important risk factor in numerous diseases.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Extracellular Fluid/metabolism , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL