Abstract

Human alveolar macrophages are critical components of the innate immune system. Cigarette smoking-induced changes in alveolar macrophage gene expression are linked to reduced resistance to pulmonary infections and to the development of emphysema/COPD. We hypothesized that microRNAs (miRNAs) could control, in part, the unique messenger RNA (mRNA) expression profiles found in alveolar macrophages of cigarette smokers. Activation of macrophages with different stimuli in vitro leads to a diverse range of M1 (inflammatory) and M2 (anti-inflammatory) polarized phenotypes that are thought to mimic activated macrophages in distinct tissue environments. Microarray mRNA data indicated that smoking promoted an “inverse” M1 mRNA expression program, defined by decreased expression of M1-induced transcripts and increased expression of M1-repressed transcripts with few changes in M2-regulated transcripts. RT-PCR arrays identified altered expression of many miRNAs in alveolar macrophages of smokers and a decrease in global miRNA abundance. Stratification of human subjects suggested that the magnitude of the global decrease in miRNA abundance was associated with smoking history. We found that many of the miRNAs with reduced expression in alveolar macrophages of smokers were predicted to target mRNAs upregulated in alveolar macrophages of smokers. For example, miR-452 is predicted to target the transcript encoding MMP12, an important effector of smoking-related diseases. Experimental antagonism of miR-452 in differentiated monocytic cells resulted in increased expression of MMP12. The comprehensive mRNA and miRNA expression profiles described here provide insight into gene expression regulation that may underlie the adverse effects cigarette smoking has on alveolar macrophages.

Highlights

  • Cigarette smoking is a prominent risk factor for many respiratory diseases including emphysema/COPD, respiratory bronchiolitis, interstitial lung disease, and desquamative interstitial pneumonitis [1,2,3]

  • Cigarette smoking has been shown to alter the transcriptional profile of human alveolar macrophages in a consistent manner as reported by two independent groups [1,2,25]

  • In vitro polarization of monocyte-derived macrophages (MDMs) leads to distinct phenotypes that have been categorized as M1, M2a, M2b, and M2c [24,55]

Read more

Summary

Introduction

Cigarette smoking is a prominent risk factor for many respiratory diseases including emphysema/COPD, respiratory bronchiolitis, interstitial lung disease, and desquamative interstitial pneumonitis [1,2,3]. Aberrant gene expression in alveolar macrophages has been shown to alter the protease/anti-protease balance in the lung contributing to the development of emphysema [7,8,9,10]. Alveolar macrophages are essential immune effector cells in the lung with functions that include pathogen clearance and responses to inhaled environmental exposures [8,12,13,14,15,16]. Smoking causes alveolar macrophage defects in phagocytosis, responses to pathogen-associated molecular patterns, and microbicidal activity [17,18,19,20,21]. These defects compromise alveolar macrophage-mediated protection from infectious agents [22]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call