Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L321-L335, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34105359

ABSTRACT

Bacterial pneumonia is a major cause of morbidity and mortality worldwide despite the use of antibiotics, and novel therapies are urgently needed. Building on previous work, we aimed to 1) develop a baboon model of severe pneumococcal pneumonia and sepsis with organ dysfunction and 2) test the safety and efficacy of a novel extracorporeal blood filter to remove proinflammatory molecules and improve organ function. After a dose-finding pilot study, 12 animals were inoculated with Streptococcus pneumoniae [5 × 109 colony-forming units (CFU)], given ceftriaxone at 24 h after inoculation, and randomized to extracorporeal blood purification using a filter coated with surface-immobilized heparin sulfate (n = 6) or sham treatment (n = 6) for 4 h at 30 h after inoculation. For safety analysis, four uninfected animals also underwent purification. At 48 h, necropsy was performed. Inoculated animals developed severe pneumonia and septic shock. Compared with sham-treated animals, septic animals treated with purification displayed significantly less kidney injury, metabolic acidosis, hypoglycemia, and shock (P < 0.05). Purification blocked the rise in peripheral blood S. pneumoniae DNA, attenuated bronchoalveolar lavage (BAL) CCL4, CCL2, and IL-18 levels, and reduced renal oxidative injury and classical NLRP3 inflammasome activation. Purification was safe in both uninfected and infected animals and produced no adverse effects. We demonstrate that heparin-based blood purification significantly attenuates levels of circulating S. pneumoniae DNA and BAL cytokines and is renal protective in baboons with severe pneumococcal pneumonia and septic shock. Purification was associated with less severe acute kidney injury, metabolic derangements, and shock. These results support future clinical studies in critically ill septic patients.


Subject(s)
Hemofiltration , Heparin/chemistry , Pneumonia, Pneumococcal/therapy , Shock, Septic/therapy , Streptococcus pneumoniae/metabolism , Animals , Cytokines/metabolism , Male , Papio , Pilot Projects , Pneumonia, Pneumococcal/blood , Shock, Septic/blood
2.
Crit Care Med ; 47(5): 651-658, 2019 05.
Article in English | MEDLINE | ID: mdl-30730439

ABSTRACT

OBJECTIVES: Metabolic derangements in sepsis stem from mitochondrial injury and contribute significantly to organ failure and mortality; however, little is known about mitochondrial recovery in human sepsis. We sought to test markers of mitochondrial injury and recovery (mitochondrial biogenesis) noninvasively in peripheral blood mononuclear cells from patients with sepsis and correlate serial measurements with clinical outcomes. DESIGN: Prospective case-control study. SETTING: Academic Medical Center and Veterans Affairs Hospital. PATIENTS: Uninfected control patients (n = 20) and septic ICU patients (n = 37). INTERVENTIONS: Blood samples were collected once from control patients and serially with clinical data on days 1, 3, and 5 from septic patients. Gene products for HMOX1, NRF1, PPARGC1A, and TFAM, and mitochondrial DNA ND1 and D-loop were measured by quantitative reverse transcriptase-polymerase chain reaction. Proinflammatory cytokines were measured in plasma and neutrophil lysates. MEASUREMENTS AND MAIN RESULTS: Median (interquartile range) Acute Physiology and Chronic Health Evaluation II and Sequential Organ Failure Assessment scores were 21 (8) and 10 (4), respectively, and 90-day mortality was 19%. Transcript levels of all four genes in peripheral blood mononuclear cells were significantly reduced in septic patients on day 1 (p < 0.05), whereas mitochondrial DNA copy number fell and plasma D-loop increased (both p < 0.05), indicative of mitochondrial damage. D-loop content was directly proportional to tumor necrosis factor-α and high-mobility group protein B1 cytokine expression. By day 5, we observed transcriptional activation of mitochondrial biogenesis and restoration of mitochondrial DNA copy number (p < 0.05). Patients with early activation of mitochondrial biogenesis were ICU-free by 1 week. CONCLUSIONS: Our findings support data that sepsis-induced mitochondrial damage is reversed by activation of mitochondrial biogenesis and that gene transcripts measured noninvasively in peripheral blood mononuclear cells can serve as novel biomarkers of sepsis recovery.


Subject(s)
DNA, Mitochondrial/blood , Leukocytes, Mononuclear/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Sepsis/metabolism , Adult , Biomarkers/blood , Case-Control Studies , Female , Humans , Male , Middle Aged , Mitochondria/genetics , Mitochondrial Diseases/blood , Mitochondrial Diseases/genetics , Prospective Studies , Real-Time Polymerase Chain Reaction , Sepsis/blood , Sepsis/genetics
3.
Lab Anim (NY) ; 48(2): 57-65, 2019 02.
Article in English | MEDLINE | ID: mdl-30643274

ABSTRACT

Sepsis involves a disordered host response to systemic infection leading to high morbidity and mortality. Despite intense research, targeted sepsis therapies beyond antibiotics have remained elusive. The cornerstone of sepsis research is the development of animal models to mimic human bacterial infections and test novel pharmacologic targets. Nonhuman primates (NHPs) have served as an attractive, but expensive, animal to model human bacterial infections due to their nearly identical cardiopulmonary anatomy and physiology, as well as host response to infection. Several NHP species have provided substantial insight into sepsis-mediated inflammation, endothelial dysfunction, acute lung injury, and multi-organ failure. The use of NHPs has usually focused on translating therapies from early preclinical models to human clinical trials. However, despite successful sepsis interventions in NHP models, there are still no FDA-approved sepsis therapies. This review highlights major NHP models of bacterial sepsis and their relevance to clinical medicine.


Subject(s)
Bacteremia , Disease Models, Animal , Primates , Animals , Anti-Bacterial Agents/therapeutic use , Bacteremia/drug therapy , Bacteremia/microbiology , Bacteremia/physiopathology , Humans
4.
JCI Insight ; 3(23)2018 12 06.
Article in English | MEDLINE | ID: mdl-30518685

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a prevalent disease with significant mortality for which no effective pharmacologic therapy exists. Low-dose inhaled carbon monoxide (iCO) confers cytoprotection in preclinical models of sepsis and ARDS. METHODS: We conducted a phase I dose escalation trial to assess feasibility and safety of low-dose iCO administration in patients with sepsis-induced ARDS. Twelve participants were randomized to iCO or placebo air 2:1 in two cohorts. Four subjects each were administered iCO (100 ppm in cohort 1 or 200 ppm in cohort 2) or placebo for 90 minutes for up to 5 consecutive days. Primary outcomes included the incidence of carboxyhemoglobin (COHb) level ≥10%, prespecified administration-associated adverse events (AEs), and severe adverse events (SAEs). Secondary endpoints included the accuracy of the Coburn-Forster-Kane (CFK) equation to predict COHb levels, biomarker levels, and clinical outcomes. RESULTS: No participants exceeded a COHb level of 10%, and there were no administration-associated AEs or study-related SAEs. CO-treated participants had a significant increase in COHb (3.48% ± 0.7% [cohort 1]; 4.9% ± 0.28% [cohort 2]) compared with placebo-treated subjects (1.97% ± 0.39%). The CFK equation was highly accurate at predicting COHb levels, particularly in cohort 2 (R2 = 0.9205; P < 0.0001). Circulating mitochondrial DNA levels were reduced in iCO-treated participants compared with placebo-treated subjects. CONCLUSION: Precise administration of low-dose iCO is feasible, well-tolerated, and appears to be safe in patients with sepsis-induced ARDS. Excellent agreement between predicted and observed COHb should ensure that COHb levels remain in the target range during future efficacy trials. TRIAL REGISTRATION: ClinicalTrials.gov NCT02425579. FUNDING: NIH grants P01HL108801, KL2TR002385, K08HL130557, and K08GM102695.


Subject(s)
Administration, Inhalation , Carbon Monoxide/administration & dosage , Respiratory Distress Syndrome/drug therapy , Respiratory Therapy/methods , Sepsis/drug therapy , Adult , Aged , Biomarkers/blood , Blood Gas Analysis , Carboxyhemoglobin , DNA, Mitochondrial , Female , Humans , Male , Middle Aged
5.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L699-L709, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28663335

ABSTRACT

Mitochondrial damage is often overlooked in acute lung injury (ALI), yet most of the lung's physiological processes, such as airway tone, mucociliary clearance, ventilation-perfusion (Va/Q) matching, and immune surveillance require aerobic energy provision. Because the cell's mitochondrial quality control (QC) process regulates the elimination and replacement of damaged mitochondria to maintain cell survival, we serially evaluated mitochondrial biogenesis and mitophagy in the alveolar regions of mice in a validated Staphylococcus aureus pneumonia model. We report that apart from cell lysis by direct contact with microbes, modest epithelial cell death was detected despite significant mitochondrial damage. Cell death by TdT-mediated dUTP nick-end labeling staining occurred on days 1 and 2 postinoculation: apoptosis shown by caspase-3 cleavage was present on days 1 and 2, while necroptosis shown by increased levels of phospho- mixed lineage kinase domain-like protein (MLKL) and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) was present on day 1 Cell death in alveolar type I (AT1) cells assessed by bronchoalveolar lavage fluid receptor for advanced glycation end points (RAGE) levels was high, yet AT2 cell death was limited while both mitochondrial biogenesis and mitophagy were induced. These mitochondrial QC mechanisms were evaluated mainly in AT2 cells by localizing increases in citrate synthase content, increases in nuclear mitochondrial biogenesis regulators nuclear respiratory factor-1 (NRF-1) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and increases in light chain 3B protein (LC3-I)/LC3II ratios. Concomitant changes in p62, Pink 1, and Parkin protein levels indicated activation of mitophagy. By confocal microscopy, mitochondrial biogenesis and mitophagy were often observed on day 1 within the same AT2 cells. These findings imply that mitochondrial QC activation in pneumonia-damaged AT2 cells promotes cell survival in support of alveolar function.


Subject(s)
Alveolar Epithelial Cells/pathology , Mitochondria/pathology , Pneumonia, Staphylococcal/etiology , Pneumonia, Staphylococcal/pathology , Staphylococcal Infections/complications , Staphylococcus aureus/pathogenicity , Alveolar Epithelial Cells/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Pneumonia, Staphylococcal/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
6.
Ann Am Thorac Soc ; 13(3): 401-13, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700878

ABSTRACT

RATIONALE: Limitations in methods for the rapid diagnosis of hospital-acquired infections often delay initiation of effective antimicrobial therapy. New diagnostic approaches offer potential clinical and cost-related improvements in the management of these infections. OBJECTIVES: We developed a decision modeling framework to assess the potential cost-effectiveness of a rapid biomarker assay to identify hospital-acquired infection in high-risk patients earlier than standard diagnostic testing. METHODS: The framework includes parameters representing rates of infection, rates of delayed appropriate therapy, and impact of delayed therapy on mortality, along with assumptions about diagnostic test characteristics and their impact on delayed therapy and length of stay. Parameter estimates were based on contemporary, published studies and supplemented with data from a four-site, observational, clinical study. Extensive sensitivity analyses were performed. The base-case analysis assumed 17.6% of ventilated patients and 11.2% of nonventilated patients develop hospital-acquired infection and that 28.7% of patients with hospital-acquired infection experience delays in appropriate antibiotic therapy with standard care. We assumed this percentage decreased by 50% (to 14.4%) among patients with true-positive results and increased by 50% (to 43.1%) among patients with false-negative results using a hypothetical biomarker assay. Cost of testing was set at $110/d. MEASUREMENTS AND MAIN RESULTS: In the base-case analysis, among ventilated patients, daily diagnostic testing starting on admission reduced inpatient mortality from 12.3 to 11.9% and increased mean costs by $1,640 per patient, resulting in an incremental cost-effectiveness ratio of $21,389 per life-year saved. Among nonventilated patients, inpatient mortality decreased from 7.3 to 7.1% and costs increased by $1,381 with diagnostic testing. The resulting incremental cost-effectiveness ratio was $42,325 per life-year saved. Threshold analyses revealed the probabilities of developing hospital-acquired infection in ventilated and nonventilated patients could be as low as 8.4 and 9.8%, respectively, to maintain incremental cost-effectiveness ratios less than $50,000 per life-year saved. CONCLUSIONS: Development and use of serial diagnostic testing that reduces the proportion of patients with delays in appropriate antibiotic therapy for hospital-acquired infections could reduce inpatient mortality. The model presented here offers a cost-effectiveness framework for future test development.


Subject(s)
Cross Infection/diagnosis , Cross Infection/economics , Early Diagnosis , Pneumonia, Ventilator-Associated/diagnosis , Pneumonia, Ventilator-Associated/economics , Adult , Aged , Aged, 80 and over , Cost-Benefit Analysis , Critical Illness , Decision Support Techniques , Female , Humans , Male , Middle Aged , North Carolina , Prospective Studies , Quality-Adjusted Life Years , Young Adult
7.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L834-46, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26320156

ABSTRACT

Inhaled carbon monoxide (CO) gas has therapeutic potential for patients with acute respiratory distress syndrome if a safe, evidence-based dosing strategy and a ventilator-compatible CO delivery system can be developed. In this study, we used a clinically relevant baboon model of Streptococcus pneumoniae pneumonia to 1) test a novel, ventilator-compatible CO delivery system; 2) establish a safe and effective CO dosing regimen; and 3) investigate the local and systemic effects of CO therapy on inflammation and acute lung injury (ALI). Animals were inoculated with S. pneumoniae (10(8)-10(9) CFU) (n = 14) or saline vehicle (n = 5); in a subset with pneumonia (n = 5), we administered low-dose, inhaled CO gas (100-300 ppm × 60-90 min) at 0, 6, 24, and/or 48 h postinoculation and serially measured blood carboxyhemoglobin (COHb) levels. We found that CO inhalation at 200 ppm for 60 min is well tolerated and achieves a COHb of 6-8% with ambient CO levels ≤ 1 ppm. The COHb level measured at 20 min predicted the 60-min COHb level by the Coburn-Forster-Kane equation with high accuracy. Animals given inhaled CO + antibiotics displayed significantly less ALI at 8 days postinoculation compared with antibiotics alone. Inhaled CO was associated with activation of mitochondrial biogenesis in the lung and with augmentation of renal antioxidative programs. These data support the feasibility of safely delivering inhaled CO gas during mechanical ventilation and provide preliminary evidence that CO may accelerate the resolution of ALI in a clinically relevant nonhuman primate pneumonia model.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/therapy , Carbon Monoxide/administration & dosage , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/therapy , Acute Lung Injury/blood , Administration, Inhalation , Animals , Anti-Bacterial Agents/administration & dosage , Antioxidants/metabolism , Carboxyhemoglobin/metabolism , Disease Models, Animal , Equipment Design , Humans , Kidney/metabolism , Lung/pathology , Male , Papio , Pneumonia, Pneumococcal/blood , Respiration, Artificial , Respiratory Therapy/instrumentation , Sepsis/etiology , Sepsis/metabolism , Sepsis/therapy
8.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L857-71, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26186946

ABSTRACT

The heme oxygenase-1 (HO-1)/carbon monoxide (CO) system induces mitochondrial biogenesis, but its biological impact in human skeletal muscle is uncertain. The enzyme system generates CO, which stimulates mitochondrial proliferation in normal muscle. Here we examined whether CO breathing can be used to produce a coordinated metabolic and vascular response in human skeletal muscle. In 19 healthy subjects, we performed vastus lateralis muscle biopsies and tested one-legged maximal O2 uptake (V̇o2max) before and after breathing air or CO (200 ppm) for 1 h daily for 5 days. In response to CO, there was robust HO-1 induction along with increased mRNA levels for nuclear-encoded mitochondrial transcription factor A (Tfam), cytochrome c, cytochrome oxidase subunit IV (COX IV), and mitochondrial-encoded COX I and NADH dehydrogenase subunit 1 (NDI). CO breathing did not increase V̇o2max (1.96 ± 0.51 pre-CO, 1.87 ± 0.50 post-CO l/min; P = not significant) but did increase muscle citrate synthase, mitochondrial density (139.0 ± 34.9 pre-CO, 219.0 ± 36.2 post-CO; no. of mitochondrial profiles/field), myoglobin content and glucose transporter (GLUT4) protein level and led to GLUT4 localization to the myocyte membrane, all consistent with expansion of the tissue O2 transport system. These responses were attended by increased cluster of differentiation 31 (CD31)-positive muscle capillaries (1.78 ± 0.16 pre-CO, 2.37 ± 0.59 post-CO; capillaries/muscle fiber), implying the enrichment of microvascular O2 reserve. The findings support that induction of the HO-1/CO system by CO not only improves muscle mitochondrial density, but regulates myoglobin content, GLUT4 localization, and capillarity in accordance with current concepts of skeletal muscle plasticity.


Subject(s)
Carbon Monoxide/metabolism , Heme Oxygenase-1/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Adolescent , Adult , Capillaries/anatomy & histology , DNA, Mitochondrial/genetics , Exercise Test , Female , Glucose Transporter Type 4/metabolism , Humans , Male , Microscopy, Electron, Transmission , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle Proteins/metabolism , Muscle, Skeletal/ultrastructure , Oxygen Consumption , Quadriceps Muscle/blood supply , Quadriceps Muscle/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
9.
Ann Am Thorac Soc ; 12(3): 349-56, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25568929

ABSTRACT

RATIONALE: The 2011 combined Global Initiative for Chronic Obstructive Lung Disease (GOLD) assessment incorporates symptoms, exacerbation history, and spirometry in discriminating risk of exacerbations in patients with chronic obstructive pulmonary disease (COPD). Six-minute-walk distance (6MWD) and accelerometry also have been used to assess disease severity in COPD. The association between these measures and the risks of hospitalization and mortality in the context of GOLD 2011 is unknown. OBJECTIVES: To describe changes in exercise tolerance and physical activity over time in patients with COPD and to test the hypothesis that lower baseline 6MWD or accelerometry step count is associated with increased risk of COPD-related hospitalization or all-cause mortality, independent of GOLD 2011 group. METHODS: Physical function and medical outcomes were prospectively assessed in 326 patients with moderate to severe COPD in INSPIRE-II, a randomized controlled trial of a coping skills training intervention. Cox models were used to determine if GOLD 2011 group, 6MWD, or accelerometry steps were associated with risk of COPD-related hospitalization or all-cause mortality. MEASUREMENTS AND MAIN RESULTS: Physical function declined over time in GOLD group D but remained stable in groups A, B, and C. GOLD classification was associated with time to death or first COPD-related hospitalization. Baseline 6MWD was more strongly associated with time to death or first COPD-related hospitalization (hazard ratio, 0.50 [95% confidence interval, 0.34, 0.73] per 150 m, P=0.0003) than GOLD 2011 classification. A similar relationship was observed for accelerometry steps (hazard ratio, 0.80 [95% confidence interval, 0.70, 0.92] per 1,000 steps, P=0.002). CONCLUSIONS: Exercise tolerance and daily physical activity are important predictors of hospitalization and mortality in COPD, independent of GOLD 2011 classification. Physical function may represent a modifiable risk factor that warrants increased attention as a target for interventions to improve clinically meaningful outcomes in COPD.


Subject(s)
Accelerometry/methods , Exercise Tolerance/physiology , Motor Activity/physiology , Pulmonary Disease, Chronic Obstructive/diagnosis , Walking/physiology , Aged , Disease Progression , Female , Follow-Up Studies , Humans , Male , Pulmonary Disease, Chronic Obstructive/physiopathology , Retrospective Studies , Severity of Illness Index , Spirometry , Time Factors
10.
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
12.
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
13.
PLoS One ; 6(9): e25249, 2011.
Article in English | MEDLINE | ID: mdl-21966468

ABSTRACT

Activation of the host antibacterial defenses by the toll-like receptors (TLR) also selectively activates energy-sensing and metabolic pathways, but the mechanisms are poorly understood. This includes the metabolic and mitochondrial biogenesis master co-activators, Ppargc1a (PGC-1α) and Ppargc1b (PGC-1ß) in Staphylococcus aureus (S. aureus) sepsis. The expression of these genes in the liver is markedly attenuated inTLR2(-/-) mice and markedly accentuated in TLR4(-/-) mice compared with wild type (WT) mice. We sought to explain this difference by using specific TLR-pathway knockout mice to test the hypothesis that these co-activator genes are directly regulated through TLR2 signaling. By comparing their responses to S. aureus with WT mice, we found that MyD88-deficient and MAL-deficient mice expressed hepatic Ppargc1a and Ppargc1b normally, but that neither gene was activated in TRAM-deficient mice. Ppargc1a/b activation did not require NF-kß, but did require an interferon response factor (IRF), because neither gene was activated in IRF-3/7 double-knockout mice in sepsis, but both were activated normally in Unc93b1-deficient (3d) mice. Nuclear IRF-7 levels in TLR2(-/-) and TLR4(-/-) mice decreased and increased respectively post-inoculation and IRF-7 DNA-binding at the Ppargc1a promoter was demonstrated by chromatin immunoprecipitation. Also, a TLR2-TLR4-TRAM native hepatic protein complex was detected by immunoprecipitation within 6 h of S. aureus inoculation that could support MyD88-independent signaling to Ppargc1a/b. Overall, these findings disclose a novel MyD88-independent pathway in S. aureus sepsis that links TLR2 and TLR4 signaling in innate immunity to Ppargc1a/b gene regulation in a critical metabolic organ, the liver, by means of TRAM, TRIF, and IRF-7.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Sepsis/metabolism , Sepsis/microbiology , Staphylococcus aureus/pathogenicity , Toll-Like Receptor 2/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Blotting, Western , Chromatin Immunoprecipitation , Female , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic , Receptors, Interleukin , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Trans-Activators/genetics , Transcription Factors
14.
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
15.
Free Radic Biol Med ; 48(5): 736-46, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20043987

ABSTRACT

Nitric oxide synthase-2 (NOS2) plays a critical role in reactive nitrogen species generation and cysteine modifications that influence mitochondrial function and signaling during inflammation. Here, we investigated the role of NOS2 in hepatic mitochondrial biogenesis during Escherichia coli peritonitis in mice. NOS2(-/-) mice displayed smaller mitochondrial biogenesis responses than Wt mice during E. coli infection according to differences in mRNA levels for the PGC-1 alpha coactivator, nuclear respiratory factor-1, mitochondrial transcription factor-A (Tfam), and mtDNA polymerase (Pol gamma). NOS2(-/-) mice did not significantly increase mitochondrial Tfam and Pol gamma protein levels during infection in conjunction with impaired mitochondrial DNA (mtDNA) transcription, loss of mtDNA copy number, and lower State 3 respiration rates. NOS2 blockade in mitochondrial-GFP reporter mice disrupted Hsp60 localization to mitochondria after E. coli exposure. Mechanistically, biotin-switch and immunoprecipitation studies demonstrated NOS2 binding to and S-nitros(yl)ation of Hsp60 and Hsp70. Specifically, NOS2 promoted Tfam accumulation in mitochondria by regulation of Hsp60-Tfam binding via S-nitros(yl)ation. In hepatocytes, site-directed mutagenesis identified (237)Cys as a critical residue for Hsp60 S-nitros(yl)ation. Thus, the role of NOS2 in inflammation-induced mitochondrial biogenesis involves both optimal gene expression for nuclear-encoded mtDNA-binding proteins and functional regulation of the Hsp60 chaperone that enables their importation for mtDNA transcription and replication.


Subject(s)
Chaperonin 60/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Mitochondria, Liver/metabolism , Nitric Oxide Synthase Type II/metabolism , Peritonitis/immunology , Animals , Cells, Cultured , Chaperonin 60/genetics , DNA, Mitochondrial/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Escherichia coli/pathogenicity , Escherichia coli Infections/complications , Escherichia coli Infections/genetics , Escherichia coli Infections/pathology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/metabolism , Mutagenesis, Site-Directed , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Nuclear Respiratory Factor 1/metabolism , Peritonitis/etiology , Peritonitis/genetics , Peritonitis/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Binding/genetics , Protein Transport/genetics , Respiratory Rate/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism
16.
Free Radic Biol Med ; 46(5): 564-72, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19073249

ABSTRACT

Mitochondrial biogenesis protects metabolism from mitochondrial dysfunction produced by activation of innate immunity by lipopolysaccharide (LPS) or other bacterial products. Here we tested the hypothesis in mouse heart that activation of toll-like receptor-4 (TLR4), which induces early-phase genes that damage mitochondria, also activates mitochondrial biogenesis through induction of nitric oxide synthase (NOS2). We compared three strains of mice: wild type (Wt) C57BL/6J, TLR4(-/-), and NOS2(-/-)for cardiac mitochondrial damage and mitochondrial biogenesis by real-time RT-PCR, Western analysis, immunochemistry, and isoform analysis of myosin heavy chain (MHC) after sublethal heat-killed Escherichia coli (HkEC). After HkEC, Wt mice displayed significant myocardial mtDNA depletion along with enhanced TLR4 and NOS2 gene and protein expression that normalized in 72 h. HkEC generated less cytokine stress in TLR4(-/-)and NOS2(-/-)than Wt mice, NOS2(-/-)mice had mtDNA damage comparable to Wt, and both knockout strains failed to restore mtDNA copy number because of mitochondrial transcriptosome dysfunction. Wt mice also showed the largest beta-MHC isoform switch, but MHC recovery lagged in the NOS2(-/-)and TLR4(-/-)strains. The NOS2(-/-)mice also unexpectedly revealed the codependency of TLR4 expression on NOS2. These findings demonstrate the decisive participation of NOS2 induction by TLR4 in optimization of mitochondrial biogenesis and MHC expression after gram-negative challenge.


Subject(s)
Endotoxemia/enzymology , Escherichia coli Infections/enzymology , Mitochondria, Heart/physiology , Nitric Oxide Synthase Type II/physiology , Toll-Like Receptor 4/physiology , Ventricular Myosins/metabolism , Animals , Cell Respiration , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytokines/genetics , Cytokines/metabolism , DNA Damage/physiology , DNA, Mitochondrial/analysis , Endotoxemia/genetics , Endotoxemia/immunology , Enzyme Induction , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Heart/microbiology , Heart/physiology , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/microbiology , Myocardial Contraction , Proteins/genetics , Proteins/metabolism , Recovery of Function , Ventricular Myosins/genetics
18.
Antioxid Redox Signal ; 10(2): 269-75, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17999632

ABSTRACT

Cell survival and injury repair is facilitated by mitochondrial biogenesis; however, the role of this process in lung repair is unknown. We evaluated mitochondrial biogenesis in the mouse lung in two injuries that cause acute inflammation and in two that cause chronic inflammation and pulmonary fibrosis. By using reporter mice that express green fluorescent protein (GFP) exclusively in mitochondria, we tracked mitochondrial biogenesis and correlated it with histologic lung injury, proliferation, and fibrosis. At 72 hours after acute LPS or continuous exposure to hyperoxia (Fio2, 1.0), the lungs showed diffuse infiltration by inflammatory cells in the alveolar region. In reporter mice, patchy new mitochondrial fluorescence was found in the alveolar region but was most prominent and unexpected in perivascular regions. At 14 days after instillation of asbestos or bleomycin, diffuse chronic inflammation had developed, and green fluorescence appeared in inflammatory cells in the expanded interstitium and was most intense in smooth muscle cells of pulmonary vessels. In all four lung injuries, mitochondrial fluorescence colocalized with mitochondrial superoxide dismutase, but not with proliferating cell nuclear antigen. These data indicate that vascular mitochondrial biogenesis is activated in diverse inhalational lung injuries along with oxidative stress. This finding indicates a unique and unexpected mechanism of metabolic adaptation to pulmonary fibrotic injuries.


Subject(s)
Lung Injury , Mitochondria/physiology , Pulmonary Circulation/physiology , Pulmonary Fibrosis/physiopathology , Animals , Asbestos/toxicity , Bleomycin/toxicity , Disease Models, Animal , Lung/pathology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitochondria/enzymology , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Superoxide Dismutase/metabolism
19.
J Clin Invest ; 117(12): 3730-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18037988

ABSTRACT

The clinical utility of anthracycline anticancer agents, especially doxorubicin, is limited by a progressive toxic cardiomyopathy linked to mitochondrial damage and cardiomyocyte apoptosis. Here we demonstrate that the post-doxorubicin mouse heart fails to upregulate the nuclear program for mitochondrial biogenesis and its associated intrinsic antiapoptosis proteins, leading to severe mitochondrial DNA (mtDNA) depletion, sarcomere destruction, apoptosis, necrosis, and excessive wall stress and fibrosis. Furthermore, we exploited recent evidence that mitochondrial biogenesis is regulated by the CO/heme oxygenase (CO/HO) system to ameliorate doxorubicin cardiomyopathy in mice. We found that the myocardial pathology was averted by periodic CO inhalation, which restored mitochondrial biogenesis and circumvented intrinsic apoptosis through caspase-3 and apoptosis-inducing factor. Moreover, CO simultaneously reversed doxorubicin-induced loss of DNA binding by GATA-4 and restored critical sarcomeric proteins. In isolated rat cardiac cells, HO-1 enzyme overexpression prevented doxorubicin-induced mtDNA depletion and apoptosis via activation of Akt1/PKB and guanylate cyclase, while HO-1 gene silencing exacerbated doxorubicin-induced mtDNA depletion and apoptosis. Thus doxorubicin disrupts cardiac mitochondrial biogenesis, which promotes intrinsic apoptosis, while CO/HO promotes mitochondrial biogenesis and opposes apoptosis, forestalling fibrosis and cardiomyopathy. These findings imply that the therapeutic index of anthracycline cancer chemotherapeutics can be improved by the protection of cardiac mitochondrial biogenesis.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antimetabolites/pharmacology , Carbon Monoxide/pharmacology , Cardiomyopathies/enzymology , Doxorubicin/adverse effects , Heme Oxygenase (Decyclizing)/metabolism , Mitochondria, Heart/enzymology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Cells, Cultured , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Doxorubicin/pharmacology , Fibrosis , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Silencing , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Heme Oxygenase (Decyclizing)/genetics , Male , Mice , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Myocardium/enzymology , Myocardium/pathology , Necrosis/chemically induced , Necrosis/enzymology , Necrosis/genetics , Necrosis/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sarcomeres/enzymology , Sarcomeres/genetics , Sarcomeres/pathology
20.
Am J Respir Crit Care Med ; 176(8): 768-77, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17600279

ABSTRACT

RATIONALE: The extent, timing, and significance of mitochondrial injury and recovery in bacterial sepsis are poorly characterized, although oxidative and nitrosative mitochondrial damage have been implicated in the development of organ failure. OBJECTIVES: To define the relationships between mitochondrial biogenesis, oxidative metabolism, and recovery from Staphylococcus aureus sepsis. METHODS: We developed a murine model of fibrin clot peritonitis, using S. aureus. The model yielded dose-dependent decreases in survival and resting energy expenditure, allowing us to study recovery from sublethal sepsis. MEASUREMENTS AND MAIN RESULTS: Peritonitis caused by 10(6) colony-forming units of S. aureus induced a low tumor necrosis factor-alpha state and minimal hepatic cell death, but activated prosurvival protein kinase A, B, and C sequentially over 3 days. Basal metabolism by indirect calorimetry was depressed because of selective mitochondrial oxidative stress and subsequent loss of mitochondrial DNA copy number. During recovery, mitochondrial biogenesis was strongly activated by regulated expression of the requisite nuclear respiratory factors 1 and 2 and the coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha, as well as by repression of the biogenesis suppressor nuclear receptor interacting protein-140. Biogenesis reconstituted mitochondrial DNA copy number and transcription, and restored basal metabolism without significant hepatocellular proliferation. These events dramatically increased hepatic mitochondrial density in transgenic mice expressing mitochondrially targeted green fluorescent protein. CONCLUSIONS: This is the first demonstration that mitochondrial biogenesis restores oxidative metabolism in bacterial sepsis and is therefore an early and important prosurvival factor.


Subject(s)
Mitochondria/metabolism , Sepsis/microbiology , Staphylococcus aureus/pathogenicity , Adaptor Proteins, Signal Transducing/metabolism , Animals , Basal Metabolism , Disease Models, Animal , GA-Binding Protein Transcription Factor/metabolism , Hepatocytes/pathology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Nuclear Respiratory Factor 1/metabolism , Oxidative Stress , Peritonitis/microbiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases/biosynthesis , RNA, Messenger/metabolism , Trans-Activators/metabolism , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...