Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 280(26): 24462-71, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15878863

ABSTRACT

Transcription factor CCAAT/enhancer-binding protein-beta (C/EBP-beta) regulates a variety of cellular functions in response to exogenous stimuli. We have reported earlier that C/EBP-beta induces gene transcription through a novel interferon (IFN)-response element called gamma-IFN-activated transcriptional element. We show here that IFN-gamma-induced, C/EBP-beta/gamma-IFN-activated transcriptional element-dependent gene expression is regulated by mixed lineage kinases (MLKs), members of the mitogen-activated protein kinase kinase kinase family. MLK3 appears to activate C/EBP-beta in response to IFN-gamma by a mechanism involving decreased phosphorylation of a specific phosphoacceptor residue, Ser(64), within the transactivation domain. Decreased phosphorylation of Ser(64) was independent of IFN-gamma-stimulated ERK1/2 activation and did not require the ERK phosphorylation site Thr(189) located in regulatory domain 2 of C/EBP-beta. Together these studies provide the first evidence that MLK3 is involved in IFN-gamma signaling and identify a novel mechanism of transcriptional activation by IFN-gamma.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation, Enzymologic , Interferon-gamma/metabolism , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/physiology , Animals , Binding Sites , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Enzyme Activation , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Models, Genetic , Mutation , Phosphorylation , Plasmids/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Serine/chemistry , Signal Transduction , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Mitogen-Activated Protein Kinase Kinase Kinase 11
2.
FASEB J ; 18(11): 1258-60, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15208274

ABSTRACT

The molecular mechanisms of pulmonary fibrosis are poorly understood, although reactive oxygen species are thought to have an important role. NRF2 is a transcription factor that protects cells and tissues from oxidative stress by activating protective antioxidant and detoxifying enzymes. We hypothesized that NRF2 protects lungs from injury and fibrosis induced by bleomycin, an anti-neoplastic agent that causes pulmonary fibrosis in susceptible patients. To test this hypothesis, mice with targeted deletion of Nrf2 (Nrf2-/-) and wild-type (Nrf2+/+) mice were treated with bleomycin or vehicle, and pulmonary injury and fibrotic responses were compared. Bleomycin-induced increases in lung weight, epithelial cell death, and inflammation were significantly greater in Nrf2-/- mice than in Nrf2+/+ mice. Indices of lung fibrosis (hydroxyproline content, collagen accumulation, fibrotic score, cell proliferation) were significantly greater in bleomycin-treated Nrf2-/- mice, compared with Nrf2+/+ mice. NRF2 expression and activity were elevated in Nrf2+/+ mice by bleomycin. Bleomycin caused greater up-regulation of several NRF2-inducible antioxidant enzyme genes and protein products in Nrf2+/+ mice compared with Nrf2-/- mice. Further, bleomycin-induced transcripts and protein levels of lung injury and fibrosis markers were significantly attenuated in Nrf2+/+ mice compared with Nrf2-/- mice. Results demonstrated that NRF2 has a critical role in protection against pulmonary fibrosis, presumably through enhancement of cellular antioxidant capacity. This study has important implications for the development of intervention strategies against fibrosis.


Subject(s)
Pulmonary Fibrosis/metabolism , Animals , Biomarkers , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/chemistry , Cell Division/drug effects , Collagen/analysis , Enzyme Induction/drug effects , Hydroxyproline/analysis , Lung/chemistry , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Organ Size , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/prevention & control , RNA/biosynthesis , Tenascin/analysis , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics
3.
J Biol Chem ; 278(24): 21378-87, 2003 Jun 13.
Article in English | MEDLINE | ID: mdl-12682075

ABSTRACT

Overexpression of SPRR1B in bronchial epithelial cells is a marker for early metaplastic changes induced by various toxicants/carcinogens. Previously, we have shown that the transcriptional stimulation of SPRR1B expression by phorbol 12-myristate 13-acetate (PMA) is mainly mediated by a -150/-94 bp enhancer harboring two critical 12-O-tetradecanoylphorbol-13-acetate-responsive elements (TREs) and by Jun.Fra-1 dimers. Here, we show that a region between -54 and -39 bp containing an ETS-binding site (EBS) and a GC box is essential for both basal and PMA-inducible SPRR1B transcription. In vivo footprinting demonstrated binding of transcription factors to these elements. However, unlike enhancer TREs, exposure of cells to PMA did not significantly alter the footprinting pattern at these elements. Mutations that crippled both the EBS and GC box suppressed both basal and PMA-inducible SPRR1B transcription. Consistent with this, overexpression of EBS-binding proteins ESE-1 and ESE-3 significantly stimulated SPRR1B promoter activity. Furthermore, preceding SPRR1B transcription, PMA up-regulated mRNA expression of ETS family members such as ESE-1 and ESE-3. Although ESE-1 synergistically activated c-Jun- and PMA-enhanced SPRR1B transcription, coexpression of Sp1 and ESE-1 showed no synergistic or additive effect on promoter activity, indicating an obligatory role for AP-1 proteins in such regulation. In support of this notion, deletion or mutation of two functional TREs inhibited ESE-1- and Sp1-enhanced promoter activation. Thus, the interaction between ESE-1 and Sp1, and AP-1 proteins that bind to the proximal and distal promoter regions, respectively, play a critical role in the induction of squamous differentiation marker expression in bronchial epithelial cells.


Subject(s)
Bronchial Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins , Epithelial Cells/cytology , Promoter Regions, Genetic , Proteins/genetics , Proteins/physiology , Proto-Oncogene Proteins , Sp1 Transcription Factor/physiology , Trans-Activators/physiology , Transcription Factor AP-1/physiology , Transcription Factors , Base Sequence , Binding Sites , Biomarkers, Tumor , Bronchial Neoplasms/metabolism , Carcinogens , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cornified Envelope Proline-Rich Proteins , Epithelial Cells/metabolism , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases , Membrane Proteins , Mitogen-Activated Protein Kinases/metabolism , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oligonucleotides/chemistry , Proto-Oncogene Proteins c-ets , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetradecanoylphorbol Acetate , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Up-Regulation
4.
Am J Physiol Lung Cell Mol Physiol ; 283(6): L1161-78, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12424143

ABSTRACT

Aberrant cell proliferation and differentiation after toxic injury to airway epithelium can lead to the development of various lung diseases including cancer. The activator protein-1 (AP-1) transcription factor, composed of mainly Jun-Jun and Jun-Fos protein dimers, acts as an environmental biosensor to various external toxic stimuli and regulates gene expression involved in various biological processes. Gene disruption studies indicate that the AP-1 family members c-jun, junB, and fra1 are essential for embryonic development, whereas junD, c-fos, and fosB are required for normal postnatal growth. However, broad or target-specific transgenic overexpression of the some of these proteins gives very distinct phenotype(s), including tumor formation. This implies that, although they are required for normal cellular processes, their abnormal activation after toxic injury can lead to the pathogenesis of the lung disease. Consistent with this view, various environmental toxicants and carcinogens differentially regulate Jun and Fos expression in cells of the lung both in vivo and in vitro. Moreover, Jun and Fos proteins distinctly bind to the promoter regions of a wide variety of genes to differentially regulate their expression in epithelial injury, repair, and differentiation. Importantly, lung tumors induced by various carcinogens display a sustained expression of certain AP-1 family members. Therefore a better understanding of the mechanisms of regulation and functional role(s), as well as identification of target genes of members of the AP-1 family in airway epithelial cells, will provide additional insight into toxicant-induced lung diseases. These studies might offer a unique opportunity to use AP-1 family members and transactivation as potential diagnostic markers or drug targets for early detection and/or prevention of various lung diseases.


Subject(s)
Lung/drug effects , Lung/physiopathology , Poisons/pharmacology , Transcription Factor AP-1/physiology , Animals , Gene Expression Regulation , Humans , Lung Neoplasms/etiology , Multigene Family , Transcription Factor AP-1/genetics
5.
Environ Health Perspect ; 110(7): 665-70, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12117643

ABSTRACT

Of the several factors believed to exacerbate asthmatic symptoms, air pollution and viral infections are considered to be particularly important. Although evidence indicates that each of these respiratory insults individually can increase asthma severity in susceptible individuals, we know little about the extent to which exposure to environmental oxidant pollutants can influence the course of respiratory viral infection and its associated inflammation. To investigate the interaction of these two stimuli within their common epithelial cell targets in the upper and lower respiratory tracks, we infected primary human nasal epithelial cells and cells of the BEAS-2B line grown at the air-liquid interface with human rhinovirus type 16 (RV16) and exposed them to NO2 (2.0 ppm) or O3 (0.2 ppm) for 3 hr. Independently, RV16, NO2, and O3 rapidly increased release of the inflammatory cytokine interleukin-8 through oxidant-dependent mechanisms. The combined effect of RV16 and oxidant ranged from 42% to 250% greater than additive for NO2 and from 41% to 67% for O3. We abrogated these effects by treating the cells with the antioxidant N-acetylcysteine. Surface expression of intercellular adhesion molecule 1 (ICAM-1) underwent additive enhancement in response to combined stimulation. These data indicate that oxidant pollutants can amplify the generation of proinflammatory cytokines by RV16-infected cells and suggest that virus-induced inflammation in upper and lower airways may be exacerbated by concurrent exposure to ambient levels of oxidants commonly encountered the indoor and outdoor environments.


Subject(s)
Cytokines/biosynthesis , Oxidants, Photochemical/adverse effects , Ozone/adverse effects , Picornaviridae Infections/immunology , Cell Culture Techniques , Epithelial Cells/drug effects , Epithelial Cells/immunology , Humans , Inflammation , Nitrogen Dioxide , Nose/cytology , Picornaviridae Infections/physiopathology , Rhinovirus/pathogenicity
6.
Am J Respir Cell Mol Biol ; 27(1): 64-70, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12091247

ABSTRACT

Various toxicants and carcinogens upregulate the expression of small proline-rich protein 1B (SPRR1B), a squamous differentiation marker, in bronchial epithelial cells both in vivo and in vitro. We have recently shown that phorbol 13-myristate 12-acetate (PMA)-stimulated SPRR1B transcription in Clara-like H441 cells is mainly mediated by activator protein-1 (AP-1) and c-Jun N-terminal kinase-1 (JNK1). Though mitogen-activated protein kinase (MAPK) kinase (MEK)-1/2 pathway inhibitors strongly suppressed both basal and PMA-inducible SPRR1B transcription, overexpression of dominant negative (dn) forms of extracellular signal-regulated kinase (ERK)-1 and/or -2 did not have any significant effect indicating the involvement of another ERK-like MAPK in this pathway. Here, we report for the first time the involvement of ERK5 in PMA-inducible SPRR1B transcription in H441 cells. PMA significantly induced ERK5 activation in H441 cells. Overexpression of dn-ERK5 strongly suppressed both basal and PMA-inducible SPRR1B transcription, whereas wild-type ERK5 upregulated it. Consistent with this, a mutant form of MEK-5, an upstream activator of ERK5, strongly suppressed PMA-inducible promoter activity. However, coexpression of c-Jun restored promoter activation suppressed by dn-ERK5. Thus, in addition to JNK1, the activation of MEK5-ERK5 MAPK pathway probably plays a pivotal role in transcriptional regulation of AP-1-mediated SPRR1B expression in the distal bronchiolar region.


Subject(s)
Gene Expression Regulation , Mitogen-Activated Protein Kinases/metabolism , Proteins/genetics , Transcription, Genetic , Bronchi/cytology , Bronchi/metabolism , Cornified Envelope Proline-Rich Proteins , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Membrane Proteins , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/physiology , Point Mutation , Promoter Regions, Genetic , Proteins/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , ras Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 99(12): 7945-50, 2002 Jun 11.
Article in English | MEDLINE | ID: mdl-12048245

ABSTRACT

IFN-gamma induces a number of genes to up-regulate cellular responses by using specific transcription factors and the cognate elements. We recently discovered that CCAAT/enhancer-binding protein-beta (C/EBP-beta) induces gene transcription through an IFN-response element called gamma-IFN-activated transcriptional element (GATE). Using mutant cells, chemical inhibitors, and specific dominant negative inhibitors, we show that induction of GATE-driven gene expression depends on MEK1 (mitogen-activated protein kinase kinase/extracellular signal-regulated protein kinase kinase) and ERKs (extracellular signal-regulated protein kinases) but is independent of Raf-1. Interestingly in cells lacking the MEKK1 gene or expressing the dominant negative MEKK1, ERK activation, and GATE dependent gene expression is inhibited. A dominant negative MEKK1 blocks C/EBP-beta-driven gene expression stimulated by IFN-gamma. These studies describe an IFN-gamma-stimulated pathway that involves MEKK1-MEK1-ERK1/2 kinases to regulate C/EBP-beta-dependent gene expression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , MAP Kinase Kinase Kinase 1 , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Gene Deletion , MAP Kinase Signaling System/physiology , Macrophages , Mice , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Transcription, Genetic/drug effects , Transcriptional Activation
8.
Am J Respir Cell Mol Biol ; 26(5): 627-35, 2002 May.
Article in English | MEDLINE | ID: mdl-11970916

ABSTRACT

Human thioredoxin (Trx) is a 12-kD protein known to be involved in various reduction/oxidation reactions essential for cell growth and cellular injury repair. We previously demonstrated, based on nuclear run-on assay, that retinoic acid (RA) stimulated Trx gene expression in airway epithelial cells at the transcriptional level. Nucleotide sequencing of the 5'-flanking region of the human Trx gene revealed the presence of a TATA box at -28 and four RA response element (RARE)-like half sites at -426, -453, -507, and -626 nt. Transient transfection assays with a Trx promoter-reporter gene, chloramphenicol acetyltransferase (CAT), demonstrated a dose-dependent involvement of these four RARE-like half sites in RA-enhanced promoter activity. When the DNA fragment that flanks these four RARE-like half sites from -357 to -671 nt was introduced into a heterologous promoter of the tk-CAT2 vector, both basal and RA-stimulated CAT activities were observed. A site-directed mutagenesis approach demonstrated an essential role for RARE-I and RARE-II at -426 and -453 nt, respectively, and an auxiliary role for RARE-III at -507 nt in both basal and RA-stimulated CAT activities. Both in vivo and in vitro genomic footprinting experiments further demonstrated specific protein-DNA interactions in these "putative" RARE-I/II/III half sites. Gel electrophoretic mobility shift assays demonstrated specific interactions of these RARE-like half sites with the nuclear extracts obtained from RA-treated cultures. The anti-RAR-alpha antibody super-shift experiment further confirmed the interactions of RARE-I/II sites with RAR-alpha nuclear receptor. These results suggest a classic RARE/RAR interaction involved in RA-stimulated Trx gene expression in human airway epithelium.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Thioredoxins/metabolism , Vitamin A/pharmacology , 5' Flanking Region/physiology , Cells, Cultured , DNA Footprinting , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Epithelial Cells/cytology , Genes, Reporter , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Retinoic Acid/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Response Elements/physiology , Retinoic Acid Receptor alpha , Sequence Analysis, DNA , Thioredoxins/genetics , Transfection
9.
Am J Physiol Lung Cell Mol Physiol ; 282(2): L215-25, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11792626

ABSTRACT

Exposure of distal bronchiolar region to various toxicants and pollutants suppresses Clara cell differentiation marker expression and greatly enhances the induction of squamous cell differentiation (SCD). Here, we demonstrate for the first time phorbol 13-myristate 12-acetate (PMA)-inducible expression of SCD markers, SPRRs, in Clara-like H441 cells. The transcriptional stimulation of human SPRR1B expression is mainly mediated by a -150- to -84-bp region that harbors two critical activator protein (AP)-1 sites. In unstimulated cells, the -150- to -84-bp region is weakly bound by AP-1 proteins, mainly JunD and Fra1. However, PMA prominently induced the binding of JunB and Fra1. Consistent with this, overexpression of wild-type Jun proteins upregulated the SPRR1B promoter activity. Conversely, a c-jun mutant suppressed both basal and PMA-inducible reporter gene expression. Intriguingly, overexpression of fra2 suppressed PMA-inducible reporter activity, whereas fra1 significantly enhanced basal level activity, indicating an opposing role for these proteins in SPRR1B expression in a manner similar to that observed in proximal tracheobronchial epithelial cells (BEAS-2B clone S6). Interestingly, unlike in S6 cells, a catalytically inactive c-Jun NH(2)-terminal kinase (JNK) 1 mutant significantly reduced the PMA-inducible SPRR1B promoter activity in H441 cells. Thus either temporal expression and/or spatial activation of AP-1 proteins by JNK1 might contribute to the induction of SCD in Clara cells.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar , Lung Neoplasms , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/metabolism , Biomarkers , Carcinogens/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cornified Envelope Proline-Rich Proteins , Gene Expression/drug effects , Gene Expression/physiology , Humans , MAP Kinase Kinase 1 , Membrane Proteins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase Kinases/metabolism , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-raf/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/enzymology , p38 Mitogen-Activated Protein Kinases , ras Proteins/metabolism
10.
Am J Respir Cell Mol Biol ; 26(2): 175-82, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11804867

ABSTRACT

NRF2 is a transcription factor important in the protection against carcinogenesis and oxidative stress through antioxidant response element (ARE)-mediated transcriptional activation of several phase 2 detoxifying and antioxidant enzymes. This study was designed to determine the role of NRF2 in the pathogenesis of hyperoxic lung injury by comparing pulmonary responses to 95-98% oxygen between mice with site-directed mutation of the gene for NRF2 (Nrf2-/-) and wild-type mice (Nrf2+/+). Pulmonary hyperpermeability, macrophage inflammation, and epithelial injury in Nrf2-/- mice were 7.6-fold, 47%, and 43% greater, respectively, compared with Nrf2+/+ mice after 72 h hyperoxia exposure. Hyperoxia markedly elevated the expression of NRF2 mRNA and DNA-binding activity of NRF2 in the lungs of Nrf2+/+ mice. mRNA expression for ARE- responsive lung antioxidant and phase 2 enzymes was evaluated in both genotypes of mice to identify potential downstream molecular mechanisms of NRF2 in hyperoxic lung responses. Hyperoxia-induced mRNA levels of NAD(P)H:quinone oxidoreductase 1 (NQO1), glutathione-S-transferase (GST)-Ya and -Yc subunits, UDP glycosyl transferase (UGT), glutathione peroxidase-2 (GPx2), and heme oxygenase-1 (HO-1) were significantly lower in Nrf2-/- mice compared with Nrf2+/+ mice. Consistent with differential mRNA expression, NQO1 and total GST activities were significantly lower in Nrf2-/- mice compared with Nrf2+/+ mice after hyperoxia. Results demonstrated that NRF2 has a significant protective role against pulmonary hyperoxic injury in mice, possibly through transcriptional activation of lung antioxidant defense enzymes.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Lung/enzymology , Oxygen/toxicity , Trans-Activators/metabolism , Animals , Antioxidants/metabolism , Bronchoalveolar Lavage Fluid/chemistry , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Leucine Zippers , Lung/cytology , Lung/physiopathology , Lung Injury , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2 , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidation-Reduction , RNA, Messenger/metabolism , Tissue Extracts/chemistry , Trans-Activators/genetics
11.
Am J Respir Cell Mol Biol ; 26(1): 42-51, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751202

ABSTRACT

A strong role for reactive oxygen species (ROS) has been proposed in the pathogenesis of a number of lung diseases. Hyperoxia (> 95% oxygen) generates ROS and extensive lung damage, and has been used as a model of oxidant injury. However, the precise mechanisms of hyperoxia-induced toxicity have not been completely clarified. This study was designed to identify hyperoxia susceptibility genes in C57BL/6J (susceptible) and C3H/HeJ (resistant) mice. The quantitative phenotypes used for this analysis were pulmonary inflammatory cell influx, epithelial cell sloughing, and hyperpermeability. Genome-wide linkage analyses of intercross (F2) and recombinant inbred cohorts identified significant and suggestive quantitative trait loci on chromosomes 2 (hyperoxia susceptibility locus 1 [Hsl1]) and 3 (Hsl2), respectively. Comparative mapping of Hsl1 identified a strong candidate gene, Nfe2l2 (nuclear factor, erythroid derived 2, like 2 or Nrf2) that encodes a transcription factor NRF2 which regulates antioxidant and phase 2 gene expression. Strain-specific variation in lung Nrf2 messenger RNA expression and a T --> C substitution in the B6 Nrf2 promoter that cosegregated with susceptibility phenotypes in F2 animals supported Nrf2 as a candidate gene. Results from this study have important implications for understanding the mechanisms through which oxidants mediate the pathogenesis of lung disease.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Linkage , Genetic Predisposition to Disease , Hypoxia , Reactive Oxygen Species , Trans-Activators/genetics , Animals , Antioxidants/pharmacology , Bronchoalveolar Lavage , Chromosome Mapping , Crosses, Genetic , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Female , Genes, Dominant , Genetic Variation , Genome , Genotype , Lung/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation , NF-E2-Related Factor 2 , Oxygen/metabolism , Phenotype , Polymorphism, Genetic , Polymorphism, Restriction Fragment Length , Promoter Regions, Genetic , RNA, Messenger/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...