MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1

W Pan, S Zhu, M Yuan, H Cui, L Wang… - The journal of …, 2010 - journals.aai.org
W Pan, S Zhu, M Yuan, H Cui, L Wang, X Luo, J Li, H Zhou, Y Tang, N Shen
The journal of immunology, 2010journals.aai.org
Systemic lupus erythematosus is a complex autoimmune disease caused by genetic and
epigenetic alterations. DNA methylation abnormalities play an important role in systemic
lupus erythematosus disease processes. MicroRNAs (miRNAs) have been implicated as
fine-tuning regulators controlling diverse biological processes at the level of
posttranscriptional repression. Dysregulation of miRNAs has been described in various
disease states, including human lupus. Whereas previous studies have shown miRNAs can …
Abstract
Systemic lupus erythematosus is a complex autoimmune disease caused by genetic and epigenetic alterations. DNA methylation abnormalities play an important role in systemic lupus erythematosus disease processes. MicroRNAs (miRNAs) have been implicated as fine-tuning regulators controlling diverse biological processes at the level of posttranscriptional repression. Dysregulation of miRNAs has been described in various disease states, including human lupus. Whereas previous studies have shown miRNAs can regulate DNA methylation by targeting the DNA methylation machinery, the role of miRNAs in aberrant CD4+ T cell DNA hypomethylation of lupus is unclear. In this study, by using high-throughput microRNA profiling, we identified that two miRNAs (miR-21 and miR-148a) overexpressed in CD4+ T cells from both patients with lupus and lupus-prone MRL/lpr mice, which promote cell hypomethylation by repressing DNA methyltransferase 1 (DNMT1) expression. This in turn leads to the overexpression of autoimmune-associated methylation-sensitive genes, such as CD70 and LFA-1, via promoter demethylation. Further experiments revealed that miR-21 indirectly downregulated DNMT1 expression by targeting an important autoimmune gene, RASGRP1, which mediated the Ras–MAPK pathway upstream of DNMT1; miR-148a directly downregulated DNMT1 expression by targeting the protein coding region of its transcript. Additionally, inhibition of miR-21 and miR-148a expression in CD4+ T cells from patients with lupus could increase DNMT1 expression and attenuate DNA hypomethylation. Together, our data demonstrated a critical functional link between miRNAs and the aberrant DNA hypomethylation in lupus CD4+ T cells and could help to develop new therapeutic approaches.
journals.aai.org