Cystic fibrosis (CF) is the most common, lethal autosomal recessive disorder in the U.S. Novel therapeutic approaches to CF lung disease, the major cause of morbidity and mortality, are clearly needed. Published studies indicate significant heritability of lung disease severity in CF, independent of CFTR genotype. To search for genes modifying CF lung disease, we performed a genome-wide association scan in one cohort of CF patients, with replication of top candidates in an independent cohort. Using this approach, genetic variation in IFRD1 was identified and replicated as a modifier of lung disease severity in CF. IFRD1 is a transcriptional co-regulator that acts in a histone deacetylase (HDAC)-dependent manner. While expressed in numerous cell types, IFRD1 appears to be expressed most highly in neutrophils (PMNs). Our data indicate that PMN differentiation is associated with upregulation of IFRD1 expression;and that PMNs (but not macrophages) from Ifrd1-/- mice have impaired effector function. The inflammatory response in the CF airway is persistently neutrophilic;in turn, the products of activated PMNs appear to be responsible for airway destruction in CF. Our data demonstrate that, after airway infection with P. aeruginosa, Ifrd1-/- mice exhibit significantly slower bacterial clearance;but also significantly less neutrophilic inflammation and disease. This phenotype is strictly dependent upon hematopoietic cell expression of IFRD1. Further, HDAC inhibition leads to specific blunting of airway inflammatory responses in wild type, not Ifrd1-/- mice, indicating that IFRD1 acts in an HDAC-dependent fashion to regulate airway inflammation. Bone marrow transfer techniques and intracellular analysis of cytokine production localized these effects to PMNs. Finally, analysis of PMNs from normal human donors has revealed significant association of IFRD1 polymorphisms with quantitative measures of PMN effector function. Together, these data suggest the inter-related hypotheses that underlie this proposal: (a) IFRD1 is a modifier gene for CF lung disease;(b) IFRD1 modulates the pathogenesis of airway disease in CF through regulation of PMN effector function;and (c) polymorphisms in IFRD1 alter PMN function. The studies in this proposal will define the biological consequences and cellular and molecular mechanisms underlying IFRD1- mediated modulation of PMN function, as well as identify the causal variants in IFRD1 that modify the expression of CF lung disease.

Public Health Relevance

Cystic fibrosis is the most common, lethal autosomal recessive disorder in those with European backgrounds-with an incidence in the United Statess of approximately 30,000. Lung disease is the major cause of morbidity and mortality in cystic fibrosis. Despite considerable progress in the therapy of cystic fibrosis in recent decades, the median survival of patients with this disease remains around 35 years. There is thus a pressing need for new therapeutic approaches to cystic fibrosis lung disease. We have identified (and validated) a gene that modifies the severity of cystic fibrosis lung disease: IFRD1. The studies in this proposal will define the molecular mechanisms by which IFRD1 modifies lung disease severity in cystic fibrosis, with the long-term goal of developing novel therapeutic approaches to this devastating disease.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL094576-04
Application #
8316178
Study Section
Lung Cellular, Molecular, and Immunobiology Study Section (LCMI)
Program Officer
Banks-Schlegel, Susan P
Project Start
2009-08-01
Project End
2014-07-31
Budget Start
2012-08-01
Budget End
2014-07-31
Support Year
4
Fiscal Year
2012
Total Cost
$380,271
Indirect Cost
$123,484
Name
Cincinnati Children's Hospital Medical Center
Department
Type
DUNS #
071284913
City
Cincinnati
State
OH
Country
United States
Zip Code
45229
McAlees, J W; Whitehead, G S; Harley, I T W et al. (2015) Distinct Tlr4-expressing cell compartments control neutrophilic and eosinophilic airway inflammation. Mucosal Immunol 8:863-73
Boespflug, Nicholas D; Kumar, Sachin; McAlees, Jaclyn W et al. (2014) ATF3 is a novel regulator of mouse neutrophil migration. Blood 123:2084-93