Apolipoprotein E negatively regulates house dust mite–induced asthma via a low-density lipoprotein receptor–mediated pathway

X Yao, K Fredriksson, ZX Yu, X Xu… - American journal of …, 2010 - atsjournals.org
X Yao, K Fredriksson, ZX Yu, X Xu, N Raghavachari, KJ Keeran, GJ Zywicke, M Kwak…
American journal of respiratory and critical care medicine, 2010atsjournals.org
Rationale: Distinct sets of corticosteroid-unresponsive genes modulate disease severity in
asthma. Objectives: To identify corticosteroid-unresponsive genes that provide new insights
into disease pathogenesis and asthma therapeutics. Methods: Experimental murine asthma
was induced by nasal administration of house dust mite for 5 days per week.
Dexamethasone and apolipoprotein E (apo E) mimetic peptides were administered via
osmotic minipumps. Measurements and Main Results: Genome-wide expression profiling of …
Rationale: Distinct sets of corticosteroid-unresponsive genes modulate disease severity in asthma.
Objectives: To identify corticosteroid-unresponsive genes that provide new insights into disease pathogenesis and asthma therapeutics.
Methods: Experimental murine asthma was induced by nasal administration of house dust mite for 5 days per week. Dexamethasone and apolipoprotein E (apo E) mimetic peptides were administered via osmotic minipumps.
Measurements and Main Results: Genome-wide expression profiling of the lung transcriptome in a house dust mite–induced model of murine asthma identified increases in apo E mRNA levels that persisted despite corticosteroid treatment. House dust mite–challenged apo E−/− mice displayed enhanced airway hyperreactivity and goblet cell hyperplasia, which could be rescued by administration of an apo E(130–149) mimetic peptide. Administration of the apo E(130–149) mimetic peptide to house dust mite–challenged apo E−/− mice also inhibited eosinophilic airway inflammation, IgE production, and the expression of Th2 and Th17 cytokines. House dust mite–challenged low-density lipoprotein receptor (LDLR) knockout mice displayed a similar phenotype as apo E−/− mice with enhanced airway hyperreactivity, goblet cell hyperplasia, and mucin gene expression, but could not be rescued by the apo E(130–149) mimetic peptide, consistent with a LDLR-dependent mechanism.
Conclusions: These findings for the first time identify an apo E–LDLR pathway as an endogenous negative regulator of airway hyperreactivity and goblet cell hyperplasia in asthma. Furthermore, our results demonstrate that strategies that activate the apo E–LDLR pathway, such as apo E mimetic peptides, might be developed into a novel treatment approach for patients with asthma.
ATS Journals