Limited differential expression of miRNAs and other small RNAs in LPS-stimulated human monocytes

PLoS One. 2019 Mar 25;14(3):e0214296. doi: 10.1371/journal.pone.0214296. eCollection 2019.

Abstract

Monocytes are a distinct subset of myeloid cells with diverse functions in early inflammatory immune modulation. While previous studies have surveyed the role of miRNA regulation on different myeloid cell lines and primary cultures, the time-dependent kinetics of inflammatory stimulation on miRNA expression and the relationship between miRNA-to-target RNA expression have not been comprehensively profiled in monocytes. In this study, we use next-generation sequencing and RT-PCR assays to analyze the non-coding small RNA transcriptome of unstimulated and lipopolysaccharide (LPS)-stimulated monocytes at 6 and 24 hours. We identified a miRNA signature consisting of five mature miRNAs (hsa-mir-146a, hsa-mir-155, hsa-mir-9, hsa-mir-147b, and hsa-mir-193a) upregulated by LPS-stimulated monocytes after 6 hours and found that most miRNAs were also upregulated after 24 hours of stimulation. Only one miRNA gene was down-regulated and no other small RNAs were found dysregulated in monocytes after LPS treatment. In addition, novel tRNA-derived fragments were also discovered in monocytes although none showed significant changes upon LPS stimulation. Interrogation of validated miRNA targets by transcriptomic analysis revealed that absolute expression of most miRNA targets implicating in innate immune response decreased over time in LPS-stimulated monocytes although their expression patterns along the treatment were heterogeneous. Our findings reveal a potential role by which selective miRNA upregulation and stable expression of other small RNAs enable monocytes to develop finely tuned cellular responses during acute inflammation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Gene Expression Profiling / methods*
  • Gene Expression Regulation / drug effects
  • High-Throughput Nucleotide Sequencing
  • Humans
  • Lipopolysaccharides / pharmacology*
  • MicroRNAs / genetics*
  • Monocytes / drug effects
  • Monocytes / metabolism*
  • Sequence Analysis, RNA

Substances

  • Lipopolysaccharides
  • MicroRNAs

Grants and funding

This study was funded and supported by Amgen Inc., which played a role in study design, data collection and analysis, decision to publish, and preparation of the manuscript.