Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiol Immunol ; 59(8): 452-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26146866

ABSTRACT

Induction of mammalian heme oxygenase (HO)-1 and exposure of animals to carbon monoxide (CO) ameliorates experimental colitis. When enteric bacteria, including Escherichia coli, are exposed to low iron conditions, they express an HO-like enzyme, chuS, and metabolize heme into iron, biliverdin and CO. Given the abundance of enteric bacteria residing in the intestinal lumen, our postulate was that commensal intestinal bacteria may be a significant source of CO and those that express chuS and other Ho-like molecules suppress inflammatory immune responses through release of CO. According to real-time PCR, exposure of mice to CO results in changes in enteric bacterial composition and increases E. coli 16S and chuS DNA. Moreover, the severity of experimental colitis correlates positively with E. coli chuS expression in IL-10 deficient mice. To explore functional roles, E. coli were genetically modified to overexpress chuS or the chuS gene was deleted. Co-culture of chuS-overexpressing E. coli with bone marrow-derived macrophages resulted in less IL-12p40 and greater IL-10 secretion than in wild-type or chuS-deficient E. coli. Mice infected with chuS-overexpressing E. coli have more hepatic CO and less serum IL-12 p40 than mice infected with chuS-deficient E. coli. Thus, CO alters the composition of the commensal intestinal microbiota and expands populations of E. coli that harbor the chuS gene. These bacteria are capable of attenuating innate immune responses through expression of chuS. Bacterial HO-like molecules and bacteria-derived CO may represent novel targets for therapeutic intervention in inflammatory conditions.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/immunology , Heme Oxygenase (Decyclizing)/immunology , Heme Oxygenase (Decyclizing)/metabolism , Immune Evasion , Immunity, Innate , Animals , Carbon Monoxide/metabolism , Cells, Cultured , Coculture Techniques , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Escherichia coli/metabolism , Gene Deletion , Gene Expression , Heme Oxygenase (Decyclizing)/genetics , Interleukin-10/metabolism , Interleukin-12 Subunit p40/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics
2.
Immunity ; 24(5): 601-10, 2006 May.
Article in English | MEDLINE | ID: mdl-16713977

ABSTRACT

Carbon monoxide (CO) suppresses proinflammatory responses in macrophages reacting to LPS. We hypothesize that CO acts by inducing a molecule(s) that suppresses the inflammatory response to subsequent stress. Exposure of macrophages to CO alone in vitro produced a brief burst of mitochondrial-derived ROS, which led to expression of PPARgamma. PPARgamma expression proved essential for mediating the anti-inflammatory effects of CO. Blocking the CO-mediated increase in ROS generation prevented PPARgamma induction, and blocking PPARgamma prevented CO's anti-inflammatory effects. In a model of acute lung injury in mice, CO blocked expression of Egr-1, a central mediator of inflammation, and decreased tissue damage; inhibition of PPARgamma abrogated both effects. These data identify the mitochondrial oxidases as an (perhaps the) initial cellular target of CO and demonstrate that CO upregulates expression of PPARgamma via the mitochondria, which assures that a subsequent stress stimulus will lead to a cytoprotective as opposed to a proinflammatory phenotype.


Subject(s)
Carbon Monoxide/pharmacology , Inflammation/prevention & control , Oxidative Stress/drug effects , PPAR alpha/drug effects , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Carbon Monoxide/immunology , Disease Models, Animal , Early Growth Response Protein 1/drug effects , Early Growth Response Protein 1/immunology , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Gene Expression/drug effects , Inflammation/immunology , Lung/drug effects , Lung/pathology , Lung Diseases/immunology , Lung Diseases/metabolism , Lung Injury , Macrophages/drug effects , Macrophages/immunology , Mice , Mitochondria/drug effects , Mixed Function Oxygenases/drug effects , Oxidative Stress/immunology , PPAR alpha/immunology , PPAR alpha/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection
3.
Proc Natl Acad Sci U S A ; 101(41): 14895-900, 2004 Oct 12.
Article in English | MEDLINE | ID: mdl-15469929

ABSTRACT

To better understand the molecular basis of chronic obstructive pulmonary disease (COPD), we used serial analysis of gene expression (SAGE) and microarray analysis to compare the gene expression patterns of lung tissues from COPD and control smokers. A total of 59,343 tags corresponding to 26,502 transcripts were sequenced in SAGE analyses. A total of 327 genes were differentially expressed (1.5-fold up- or down-regulated). Microarray analysis using the same RNA source detected 261 transcripts that were differentially expressed to a significant degree between GOLD-2 and GOLD-0 smokers. We confirmed the altered expression of a select number of genes by using real-time quantitative RT-PCR. These genes encode for transcription factors (EGR1 and FOS), growth factors or related proteins (CTGF, CYR61, CX3CL1, TGFB1, and PDGFRA), and extracellular matrix protein (COL1A1). Immunofluorescence studies on the same lung specimens localized the expression of Egr-1, CTGF, and Cyr61 to alveolar epithelial cells, airway epithelial cells, and stromal and inflammatory cells of GOLD-2 smokers. Cigarette smoke extract induced Egr-1 protein expression and increased Egr-1 DNA-binding activity in human lung fibroblast cells. Cytomix (tumor necrosis factor alpha, IL-1beta, and IFN-gamma) treatment showed that the activity of matrix metalloproteinase-2 (MMP-2) was increased in lung fibroblasts from EGR1 control (+/+) mice but not detected in that of EGR1 null (-/-) mice, whereas MMP-9 was regulated by EGR1 in a reverse manner. Our study represents the first comprehensive analysis of gene expression on GOLD-2 versus GOLD-0 smokers and reveals previously unreported candidate genes that may serve as potential molecular targets in COPD.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation/physiology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , Aged , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Humans , Immediate-Early Proteins/genetics , Lung/pathology , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Smoking/pathology , Transcription Factors/genetics
4.
J Exp Med ; 198(11): 1707-16, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14657222

ABSTRACT

Carbon monoxide (CO) and nitric oxide (NO) each have mechanistically unique roles in various inflammatory disorders. Although it is known that CO can induce production of NO and that NO can induce expression of the cytoprotective enzyme heme oxygenase 1 (HO-1), there is no information whether the protective effect of CO ever requires NO production or whether either gas must induce expression of HO-1 to exert its functional effects. Using in vitro and in vivo models of tumor necrosis factor alpha-induced hepatocyte cell death in mice, we find that activation of nuclear factor kappaB and increased expression of inducible NO are required for the protective effects of CO, whereas the protective effects of NO require up-regulation of HO-1 expression. When protection from cell death is initiated by CO, NO production and HO-1 activity are each required for the protective effect showing for the first time an essential synergy between these two molecules in tandem providing potent cytoprotection.


Subject(s)
Carbon Monoxide/pharmacology , Heme Oxygenase (Decyclizing)/biosynthesis , Liver Failure/prevention & control , Nitric Oxide/physiology , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Enzyme Induction , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism
5.
Am J Pathol ; 163(6): 2555-63, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633627

ABSTRACT

The stress-inducible gene heme oxygenase (HO-1) has previously been shown to provide cytoprotection against oxidative stress. The mechanism(s) by which HO-1 provides this cytoprotection is poorly understood. We demonstrate here that carbon monoxide (CO), a byproduct released during the degradation of heme by HO, plays a major role in mediating the cytoprotection against oxidant-induced lung injury. We show in vitro that CO protects cultured epithelial cells from hyperoxic damage. By using dominant negative mutants and mice deficient in the genes for the various MAP kinases, we demonstrate that the cytoprotective effects of CO are mediated by selective activation of the MKK3/p38 beta protein MAP kinase pathway. In vivo, our experiments demonstrate that CO at a low concentration protects the lungs, extends the survival of the animals, and exerts potent anti-inflammatory effects with reduced inflammatory cell influx into the lungs and marked attenuation in the expression of pro-inflammatory cytokines.


Subject(s)
Carbon Monoxide/pharmacology , Cytoprotection , Lung Diseases/chemically induced , Lung Diseases/prevention & control , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxidants , Protein-Tyrosine Kinases/metabolism , Animals , Carboxyhemoglobin/metabolism , Cells, Cultured , Cytokines/antagonists & inhibitors , Enzyme Activation , Hyperoxia/mortality , Hyperoxia/pathology , Hyperoxia/physiopathology , Hyperoxia/prevention & control , Inflammation Mediators/antagonists & inhibitors , Lung/pathology , MAP Kinase Kinase 3 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Neutrophil Infiltration/drug effects , Pneumonia/prevention & control , Survival Analysis , p38 Mitogen-Activated Protein Kinases
6.
J Biol Chem ; 278(39): 36993-8, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-12857751

ABSTRACT

The stress-inducible protein heme oxygenase-1 provides protection against oxidative stress and modulates pro-inflammatory cytokines. As the sepsis syndrome results from the release of pro-inflammatory mediators, we postulated that heme oxygenase-1 and its enzymatic product CO would protect against lethality in a murine model of sepsis. Mice treated with a lethal dose of lipopolysaccharide (LPS) and subsequently exposed to inhaled CO had significantly better survival and lower serum interleukin (IL)-6 and IL-1beta levels than their untreated counterparts. In vitro, mouse macrophages exposed to LPS and CO had significantly attenuated IL-6 production; this effect was concentration-dependent and occurred at a transcriptional level. The same effect was seen with increased endogenous CO production through overexpression of heme oxygenase-1. Mutation within the AP-1-binding site in the IL-6 promoter diminished the effect of CO on promoter activity, and treatment of macrophages with CO decreased AP-1 binding in an electrophoretic mobility shift assay. Electrophoretic mobility supershift assay indicated that the JunB, JunD, and c-Fos components of AP-1 were particularly affected. Upstream of AP-1, CO decreased JNK phosphorylation in murine macrophages and lung endothelial cells. Mice deficient in the JNK pathway had decreased serum levels of IL-6 and IL-1beta in response to LPS compared with control mice, and no effect of CO on these cytokine levels was seen in Jnk1 or Jnk2 genedeleted mice. In summary, these results suggest that CO provides protection in a murine model of sepsis through modulation of inflammatory cytokine production. For the first time, the effect of CO is shown to be mediated via the JNK signaling pathway and the transcription factor AP-1.


Subject(s)
Carbon Monoxide/pharmacology , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Mitogen-Activated Protein Kinases/physiology , Signal Transduction/physiology , Transcription Factor AP-1/physiology , Animals , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1 , Lipopolysaccharides/toxicity , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/genetics , Phosphorylation
7.
Am J Respir Cell Mol Biol ; 27(6): 739-45, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12444034

ABSTRACT

The stress-inducible gene heme oxygenase-1 (HO-1) provides protection against oxidative stress. Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Granulocyte macrophage colony-stimulating factor (GM-CSF) is a potent cytokine generated in response to bacterial endotoxin (lipopolysaccharide [LPS]) to stimulate proliferation, maturation, and effector functions of leukocytes, contributing to the proinflammatory responses to LPS. We hypothesized that HO-1 and/or CO could regulate the expression and production of GM-CSF. HO-1 overexpression, as well as exposure to a low concentration of CO, inhibited LPS-induced GM-CSF production in macrophages. Furthermore, CO inhibited LPS-induced GM-CSF induction via inhibition in the activation of the transcription factor NF-kappaB. CO inhibited LPS-induced activation of NF-kappaB, which has been shown to regulate GM-CSF transcription, by preventing the phosphorylation and degradation of the regulatory subunit IkappaB-alpha. These data raise the intriguing possibility that CO at low concentrations may play an important role in inflammatory disease states and thus has potential therapeutic implications.


Subject(s)
Carbon Monoxide/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Animals , Cells, Cultured , Gene Expression/drug effects , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , I-kappa B Proteins/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Membrane Proteins , Mice , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...