Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Am J Respir Crit Care Med ; 185(8): 851-61, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22312014

ABSTRACT

RATIONALE: Mitochondrial damage is an important component of multiple organ failure syndrome, a highly lethal complication of severe sepsis that lacks specific therapy. Mitochondrial quality control is regulated in part by the heme oxygenase-1 (HO-1; Hmox1) system through the redox-regulated NF-E2-related factor-2 (Nrf2) transcription factor, but its role in mitochondrial biogenesis in Staphylococcus aureus sepsis is unknown. OBJECTIVES: To test the hypothesis that Nrf2-dependent up-regulation of the HO-1/carbon monoxide (CO) system would preserve mitochondrial biogenesis and rescue mice from lethal S. aureus sepsis. METHODS: A controlled murine S. aureus peritonitis model with and without inhaled CO was examined for HO-1 and Nrf2 regulation of mitochondrial biogenesis and the resolution of hepatic mitochondrial damage. MEASUREMENTS AND MAIN RESULTS: Sepsis survival was significantly enhanced using inhaled CO (250 ppm once-daily for 1 h), and linked mechanistically to Hmox1 induction and mitochondrial HO activity through Nrf2 transcriptional and Akt kinase activity. HO-1/CO stimulated Nrf2-dependent gene expression and nuclear accumulation of nuclear respiratory factor-1, -2α (Gabpa), and peroxisome proliferator-activated receptor gamma coactivator-1α; increased mitochondrial transcription factor-A and citrate synthase protein levels; and augmented mtDNA copy number. CO enhanced antiinflammatory IL-10 and reduced proinflammatory tumor necrosis factor-α production. By contrast, Nrf2(-/-) and Akt1(-/-) mice lacked CO induction of Hmox1 and mitochondrial biogenesis, and CO rescued neither strain from S. aureus sepsis. CONCLUSIONS: We identify an inducible Nrf2/HO-1 regulatory cycle for mitochondrial biogenesis that is prosurvival and counter-inflammatory in sepsis, and describe targeted induction of mitochondrial biogenesis as a potential multiple organ failure therapy.


Subject(s)
Carbon Monoxide/pharmacology , Heme Oxygenase-1/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Sepsis/enzymology , Staphylococcal Infections/therapy , Administration, Inhalation , Animals , Blotting, Western , Carbon Monoxide/metabolism , Disease Models, Animal , Female , Heme Oxygenase-1/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria/genetics , NF-E2-Related Factor 2/genetics , Organelle Biogenesis , Peritonitis/drug therapy , Peritonitis/microbiology , Peritonitis/mortality , Random Allocation , Real-Time Polymerase Chain Reaction , Risk Assessment , Sepsis/genetics , Sepsis/mortality , Staphylococcal Infections/genetics , Staphylococcal Infections/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Survival Rate , Up-Regulation
2.
J Biol Chem ; 286(18): 16374-85, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21454555

ABSTRACT

The induction of heme oxygenase-1 (HO-1; Hmox1) by inflammation, for instance in sepsis, is associated both with an anti-inflammatory response and with mitochondrial biogenesis. Here, we tested the idea that HO-1, acting through the Nfe2l2 (Nrf2) transcription factor, links anti-inflammatory cytokine expression to activation of mitochondrial biogenesis. HO-1 induction after LPS stimulated anti-inflammatory IL-10 and IL-1 receptor antagonist (IL-1Ra) expression in mouse liver, human HepG2 cells, and mouse J774.1 macrophages but blunted tumor necrosis factor-α expression. This was accompanied by nuclear Nfe2l2 accumulation and led us to identify abundant Nfe2l2 and other mitochondrial biogenesis transcription factor binding sites in the promoter regions of IL10 and IL1Ra compared with pro-inflammatory genes regulated by NF-κΒ. Mechanistically, HO-1, through its CO product, enabled these transcription factors to bind the core IL10 and IL1Ra promoters, which for IL10 included Nfe2l2, nuclear respiratory factor (NRF)-2 (Gabpa), and MEF2, and for IL1Ra, included NRF-1 and MEF2. In cells, Hmox1 or Nfe2l2 RNA silencing prevented IL-10 and IL-1Ra up-regulation, and HO-1 induction failed post-LPS in Nfe2l2-silenced cells and post-sepsis in Nfe2l2(-/-) mice. Nfe2l2(-/-) mice compared with WT mice, showed more liver damage, higher mortality, and ineffective CO rescue in sepsis. Nfe2l2(-/-) mice in sepsis also generated higher hepatic TNF-α mRNA levels, lower NRF-1 and PGC-1α mRNA levels, and no enhancement of anti-inflammatory Il10, Socs3, or bcl-x(L) gene expression. These findings disclose a highly structured transcriptional network that couples mitochondrial biogenesis to counter-inflammation with major implications for immune suppression in sepsis.


Subject(s)
Cytokines/biosynthesis , Gene Expression Regulation , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Mitochondria, Liver/enzymology , Sepsis/enzymology , Transcription Factors/metabolism , Animals , Carbon Monoxide/metabolism , Cytokines/genetics , Heme Oxygenase-1/genetics , Hep G2 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Sepsis/genetics , Transcription Factors/genetics
3.
Am J Respir Crit Care Med ; 183(2): 226-33, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20732986

ABSTRACT

RATIONALE: Damage to mitochondrial DNA (mtDNA) by the production of reactive oxygen species during inflammatory states, such as sepsis, is repaired by poorly understood mechanisms. OBJECTIVES: To test the hypothesis that the DNA repair enzyme, 8-oxoguanine DNA glycosylase (OGG1), contributes to mtDNA repair in sepsis. METHODS: Using a well-characterized mouse model of Staphylococcus aureus sepsis, we analyzed molecular markers for mitochondrial biogenesis and OGG1 translocation into liver mitochondria as well as OGG1 mRNA expression at 0, 24, 48, and 72 hours after infection. The effects of OGG1 RNA silencing on mtDNA content were determined in control, tumor necrosis factor-α, and peptidoglycan-exposed rat hepatoma cells. Based on in situ analysis of the OGG1 promoter region, chromatin immunoprecipitation assays were performed for nuclear respiratory factor (NRF)-1 and NRF-2α GA-binding protein (GABP) binding to the promoter of OGG1. MEASUREMENTS AND MAIN RESULTS: Mice infected with 10(7) cfu S. aureus intraperitoneally demonstrated hepatic oxidative mtDNA damage and significantly lower hepatic mtDNA content as well as increased mitochondrial OGG1 protein and enzyme activity compared with control mice. The infection also caused increases in hepatic OGG1 transcript levels and NRF-1 and NRF-2α transcript and protein levels. A bioinformatics analysis of the Ogg1 gene locus identified several promoter sites containing NRF-1 and NRF-2α DNA binding motifs, and chromatin immunoprecipitation assays confirmed in situ binding of both transcription factors to the Ogg1 promoter within 24 hours of infection. CONCLUSIONS: These studies identify OGG1 as an early mitochondrial response protein during sepsis under regulation by the NRF-1 and NRF-2α transcription factors that regulate mitochondrial biogenesis.


Subject(s)
DNA Glycosylases/metabolism , DNA Repair , DNA, Mitochondrial/metabolism , Mitochondria, Liver/enzymology , Sepsis/enzymology , Staphylococcus aureus , Animals , Blotting, Western , Disease Models, Animal , GA-Binding Protein Transcription Factor/metabolism , Immunoprecipitation/methods , Male , Mice , Mice, Inbred C57BL , Nuclear Respiratory Factor 1/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/microbiology
4.
PLoS One ; 9(7): e100912, 2014.
Article in English | MEDLINE | ID: mdl-24988481

ABSTRACT

Acute kidney injury (AKI) contributes to the high morbidity and mortality of multi-system organ failure in sepsis. However, recovery of renal function after sepsis-induced AKI suggests active repair of energy-producing pathways. Here, we tested the hypothesis in mice that Staphyloccocus aureus sepsis damages mitochondrial DNA (mtDNA) in the kidney and activates mtDNA repair and mitochondrial biogenesis. Sepsis was induced in wild-type C57Bl/6J and Cox-8 Gfp-tagged mitochondrial-reporter mice via intraperitoneal fibrin clots embedded with S. aureus. Kidneys from surviving mice were harvested at time zero (control), 24, or 48 hours after infection and evaluated for renal inflammation, oxidative stress markers, mtDNA content, and mitochondrial biogenesis markers, and OGG1 and UDG mitochondrial DNA repair enzymes. We examined the kidneys of the mitochondrial reporter mice for changes in staining density and distribution. S. aureus sepsis induced sharp amplification of renal Tnf, Il-10, and Ngal mRNAs with decreased renal mtDNA content and increased tubular and glomerular cell death and accumulation of protein carbonyls and 8-OHdG. Subsequently, mtDNA repair and mitochondrial biogenesis was evidenced by elevated OGG1 levels and significant increases in NRF-1, NRF-2, and mtTFA expression. Overall, renal mitochondrial mass, tracked by citrate synthase mRNA and protein, increased in parallel with changes in mitochondrial GFP-fluorescence especially in proximal tubules in the renal cortex and medulla. Sub-lethal S. aureus sepsis thus induces widespread renal mitochondrial damage that triggers the induction of the renal mtDNA repair protein, OGG1, and mitochondrial biogenesis as a conspicuous resolution mechanism after systemic bacterial infection.


Subject(s)
DNA Repair , DNA, Mitochondrial/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Mitochondria/metabolism , Sepsis/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus , Animals , DNA, Mitochondrial/genetics , Gene Expression Regulation/genetics , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Multiple Organ Failure/genetics , Multiple Organ Failure/metabolism , Multiple Organ Failure/pathology , Sepsis/genetics , Sepsis/pathology , Staphylococcal Infections/genetics , Staphylococcal Infections/pathology
5.
Free Radic Biol Med ; 53(8): 1584-94, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22940620

ABSTRACT

Acute lung injury (ALI) initiates protective responses involving genes downstream of the Nrf2 (Nfe2l2) transcription factor, including heme oxygenase-1 (HO-1), which stimulates mitochondrial biogenesis and related anti-inflammatory processes. We examined mitochondrial biogenesis during Staphylococcus aureus pneumonia in mice and the effect of Nrf2 deficiency on lung mitochondrial biogenesis and resolution of lung inflammation. S. aureus pneumonia established by nasal insufflation of live bacteria was studied in mitochondrial reporter (mt-COX8-GFP) mice, wild-type (WT) mice, and Nrf2⁻/⁻ mice. Bronchoalveolar lavage, wet/dry ratios, real-time RT-PCR and Western analysis, immunohistochemistry, and fluorescence microscopy were performed on the lung at 0, 6, 24, and 48 h. The mice survived S. aureus inoculations at 5×108 CFU despite diffuse lung inflammation and edema, but the Nrf2⁻/⁻ lung showed increased ALI. In mt-COX8-GFP mice, mitochondrial fluorescence was enhanced in bronchial and alveolar type II (AT2) epithelial cells. WT mice displayed rapid HO-1 upregulation and lower proinflammatory TNF-α, IL-1ß, and CCL2 and, especially in AT2 cells, higher anti-inflammatory IL-10 and suppressor of cytokine signaling-3 than Nrf2⁻/⁻ mice. In the alveolar region, WT but not Nrf2⁻/⁻ mice showed strongly induced nuclear respiratory factor-1, PGC-1α, mitochondrial transcription factor-A, SOD2, Bnip3, mtDNA copy number, and citrate synthase. These findings indicate that S. aureus pneumonia induces Nrf2-dependent mitochondrial biogenesis in the alveolar region, mainly in AT2 cells. Absence of Nrf2 suppresses the alveolar transcriptional network for mitochondrial biogenesis and anti-inflammation, which worsens ALI. The findings link redox activation of mitochondrial biogenesis to ALI resolution.


Subject(s)
Acute Lung Injury/etiology , Mitochondrial Turnover , NF-E2-Related Factor 2/physiology , Pneumonia, Staphylococcal/complications , Pneumonia/etiology , Pulmonary Alveoli/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Blotting, Western , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Humans , Immunoenzyme Techniques , Inflammation Mediators/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Pneumonia/pathology , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/microbiology , Pulmonary Edema/metabolism , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/pathogenicity , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL