This is a revised application to study the regulation and the role of mitochondrial biogenesis and mitophagy in experimental sepsis and acute lung injury (ALI) caused by S. aureus. It is relevant to ICU patients who survive an initial episode of severe sepsis and ARDS/multiple organ dysfunction syndrome (MODS), but die with so- called immune paralysis characterized by effector cell apoptosis, anti-inflammatory cytokine over-expression, suppression of pro-inflammatory cytokine synthesis and recurrent infections. One important pro-resolution mechanism discovered by our group is the powerful control over innate immunity by the redox-regulated bi- genomic transcriptional network of mitochondrial biogenesis, which is strongly activated by the induction of the heme oxygenase-1/carbon monoxide system (HO-1/CO) to protect energy metabolism and mitochondrial mass, but which we think may also promote the clearance of damaged organelles (mitophagy) and limit further inflammatory damage in MODS. Published and preliminary data raise the novel possibility that the transcriptional program for mitochondrial biogenesis integrates mitophagy, counter-inflammation, and anti- oxidant defenses into a coherent injury resolution network in alveolar epithelium, the major site of lung damage in ALI. We propose that the program of mitochondrial biogenesis mediates lung protection through HO-1/CO activation of Nfe2l2 and NRF-1 leading to 1) anti-inflammatory Socs3 and IL10 gene expression, 2) suppression of inflammasome-mediated IL-1? production, and 3) activation of mitophagy through Bnip3 and Atg5, promoting alveolar epithelial cell survival and resolution of barrier dysfunction. Using live S. aureus sepsi and pneumonia in mice and complementary lung cell studies, we will investigate how this integrated genetic network of mitochondrial biogenesis impacts on lung inflammation and ALI resolution. Proof-of-concept would mean the lung in sepsis/pneumonia has counter-regulatory safeguards involving the induction of mitochondrial biogenesis to prevent further mitochondrial damage from the systemic inflammatory response and clear damaged mitochondria to restore mitochondrial health and capacity for alveolar epithelial repair, e.g. though the type 2 (AT2) cell We propose translational studies to test the concept in diffuse alveolar damage (DAD) in human lung, which if successful, would open up therapeutic avenues for the improvement of mitochondrial function and the resolution of ALI/ARDS.
Our Specific Aims are:
Aim 1 : Determine whether Nfe2l2 and NRF1 induction of lung mitochondrial biogenesis in murine S. aureus sepsis and pneumonia up-regulates Socs3 and Il10 anti-inflammatory gene expression, suppresses caspase1 cleavage and IL-1? production and mitigates lung inflammation and ALI.
Aim 2 : Use gain and loss of function studies to determine whether Nfe2l2 and NRF1 induction of lung of mitochondrial biogenesis a) regulates the autophagy genes Bnip3 and Atg5 and b) activates pro- survival mitophagy through HO-1/CO-related mitochondrial ROS generation in murine S. aureus pneumonia.
Aim 3 : Assess the extent, location, and relationship of mitochondrial biogenesis to mitophagy in the alveolar epithelium of human ALI/ARDS patients compared with healthy human lung. Completion of these Aims would link transcriptional regulation of mitochondrial biogenesis to mitophagy and to immune counter-regulation, anti-oxidant defenses, and cell survival. Positive predictive studies in human ALI/ARDS would have a high impact on our understanding of the resolution of sepsis and MODS.

Public Health Relevance

This is a proposal to study the regulation and the role of mitochondrial turnover in sepsis and acute lung injury (ALI) caused by S. aureus. It is relevant to ICU patients who survive an initial episode of severe sepsis and acute respiratory distress syndrome (ARDS) with multiple organ dysfunction syndrome (MODS), but often die with so-called 'immune paralysis' and recurrent infections. One important pro-resolution mechanism discovered by our group is the powerful control over inflammation by the network of mitochondrial biogenesis, which is strongly activated by the induction of the heme oxygenase-1/carbon monoxide system (HO-1/CO) to protect energy metabolism and mitochondrial mass, but which we think also promotes the clearance of damaged organelles (mitophagy) and limits further inflammatory damage in MODS. We have preliminary evidence that the process of mitochondrial biogenesis may integrate the clearance of damaged mitochondria, anti-inflammation, and anti-oxidant defenses into a coherent injury resolution network at the major sites of lung damage in ARDS. We would like to understand how the genetic program of mitochondrial biogenesis mediates lung protection through anti-inflammatory gene expression, suppression of inflammatory interleukin-1b (IL-1?) production, and activates mitophagy leading to resolution of lung capillary leak in ARDS patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
4R01AI095424-05
Application #
9081492
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Davidson, Wendy F
Project Start
2012-07-01
Project End
2017-06-30
Budget Start
2016-07-01
Budget End
2017-06-30
Support Year
5
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Duke University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
044387793
City
Durham
State
NC
Country
United States
Zip Code
27705
Suliman, Hagir B; Zobi, Fabio; Piantadosi, Claude A (2016) Heme Oxygenase-1/Carbon Monoxide System and Embryonic Stem Cell Differentiation and Maturation into Cardiomyocytes. Antioxid Redox Signal 24:345-60
Suliman, Hagir B; Piantadosi, Claude A (2016) Mitochondrial Quality Control as a Therapeutic Target. Pharmacol Rev 68:20-48
Hull, Travis D; Boddu, Ravindra; Guo, Lingling et al. (2016) Heme oxygenase-1 regulates mitochondrial quality control in the heart. JCI Insight 1:e85817
Piantadosi, Claude A (2016) Cardioprotective role of S-nitrosylated hemoglobin from rbc. J Clin Invest 126:4402-4403
Chang, Alan L; Ulrich, Allison; Suliman, Hagir B et al. (2015) Redox regulation of mitophagy in the lung during murine Staphylococcus aureus sepsis. Free Radic Biol Med 78:179-89
Schumacker, Paul T; Gillespie, Mark N; Nakahira, Kiichi et al. (2014) Mitochondria in lung biology and pathology: more than just a powerhouse. Am J Physiol Lung Cell Mol Physiol 306:L962-74
Bartz, Raquel R; Fu, Ping; Suliman, Hagir B et al. (2014) Staphylococcus aureus sepsis induces early renal mitochondrial DNA repair and mitochondrial biogenesis in mice. PLoS One 9:e100912
Cherry, Anne D; Suliman, Hagir B; Bartz, Raquel R et al. (2014) Peroxisome proliferator-activated receptor ? co-activator 1-? as a critical co-activator of the murine hepatic oxidative stress response and mitochondrial biogenesis in Staphylococcus aureus sepsis. J Biol Chem 289:41-52
Suliman, Hagir B; Piantadosi, Claude A (2014) Mitochondrial biogenesis: regulation by endogenous gases during inflammation and organ stress. Curr Pharm Des 20:5653-62