Basic Study
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Biol Chem. Feb 26, 2016; 7(1): 178-187
Published online Feb 26, 2016. doi: 10.4331/wjbc.v7.i1.178
High levels of homocysteine downregulate apolipoprotein E expression via nuclear factor kappa B
Violeta G Trusca, Adina D Mihai, Elena V Fuior, Ioana M Fenyo, Anca V Gafencu
Violeta G Trusca, Elena V Fuior, Ioana M Fenyo, Anca V Gafencu, Department of Genomics, Transcriptomics and Molecular Therapies, Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
Adina D Mihai, School of Biological and Biomedical Sciences, Durham University, Durham DH1 3LE, United Kingdom
Author contributions: Trusca VG and Mihai AD performed the research; Trusca VG, Fuior EV, Fenyo IM and Gafencu AV wrote the paper; Gafencu AV designed the experiments, provided the reagents and analyzed the data.
Supported by The grant of the Romanian National Authority for Scientific Research, National Research Council - Executive Agency for Higher Education, Research, Development and Innovation Funding, No. PN-II-ID-PCE-2011-3-0591 (grant awarded to Gafencu AV); the Romanian Academy, and the strategic grant financed by the European Social Found within the Sectorial Operational Program Human Resources Development 2007-2013, No. POSDRU/159/1.5/S/133391 (Fenyo IM and Trusca VG).
Conflict-of-interest statement: Violeta G Trusca, Adina D Mihai, Elena V Fuior, Ioana M Fenyo, Anca V Gafencu, authors of the manuscript “High levels of homocysteine downregulate apolipoprotein E expression via nuclear factor kappa B” declare to have no conflict of interest.
Data sharing statement: Not applicable.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Anca V Gafencu, PhD, Department of Genomics, Transcriptomics and Molecular Therapies, Institute of Cellular Biology and Pathology, “Nicolae Simionescu” of the Romanian Academy, 8, BP Hasdeu Street, 050568 Bucharest, Romania. anca.gafencu@icbp.ro
Telephone: +40-21-3194518 Fax: +40-21-3194519
Received: May 29, 2015
Peer-review started: June 2, 2015
First decision: July 27, 2015
Revised: October 12, 2015
Accepted: November 3, 2015
Article in press: November 4, 2015
Published online: February 26, 2016
Abstract

AIM: To investigate the effect of high homocysteine (Hcy) levels on apolipoprotein E (apoE) expression and the signaling pathways involved in this gene regulation.

METHODS: Reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot were used to assess apoE expression in cells treated with various concentrations (50-500 μmol/L) of Hcy. Calcium phosphate-transient transfections were performed in HEK-293 and RAW 264.7 cells to evaluate the effect of Hcy on apoE regulatory elements [promoter and distal multienhancer 2 (ME2)]. To this aim, plasmids containing the proximal apoE promoter [(-500/+73)apoE construct] alone or in the presence of ME2 [ME2/(-500/+73)apoE construct] to drive the expression of the reporter luciferase gene were used. Co-transfection experiments were carried out to investigate the downstream effectors of Hcy-mediated regulation of apoE promoter by using specific inhibitors or a dominant negative form of IKβ. In other co-transfections, the luciferase reporter was under the control of synthetic promoters containing multiple specific binding sites for nuclear factor kappa B (NF-κB), activator protein-1 (AP-1) or nuclear factor of activated T cells (NFAT). Chromatin immunoprecipitation (ChIP) assay was accomplished to detect the binding of NF-κB p65 subunit to the apoE promoter in HEK-293 treated with 500 μmol/L Hcy. As control, cells were incubated with similar concentration of cysteine. NF-κB p65 proteins bound to DNA were immunoprecipitated with anti-p65 antibodies and DNA was identified by PCR using primers amplifying the region -100/+4 of the apoE gene.

RESULTS: RT-PCR revealed that high levels of Hcy (250-750 μmol/L) induced a 2-3 fold decrease in apoE mRNA levels in HEK-293 cells, while apoE gene expression was not significantly affected by treatment with lower concentrations of Hcy (100 μmol/L). Immunoblotting data provided additional evidence for the negative role of Hcy in apoE expression. Hcy decreased apoE promoter activity, in the presence or absence of ME2, in a dose dependent manner, in both RAW 264.7 and HEK-293 cells, as revealed by transient transfection experiments. The downstream effectors of the signaling pathways of Hcy were also investigated. The inhibitory effect of Hcy on the apoE promoter activity was counteracted by MAPK/ERK kinase 1/2 (MEK1/2) inhibitor U0126, suggesting that MEK1/2 is involved in the downregulation of apoE promoter activity by Hcy. Our data demonstrated that Hcy-induced inhibition of apoE took place through activation of NF-κB. Moreover, we demonstrated that Hcy activated a synthetic promoter containing three NF-κB binding sites, but did not affect promoters containing AP-1 or NFAT binding sites. ChIP experiments revealed that NF-κB p65 subunit is recruited to the apoE promoter following Hcy treatment of cells.

CONCLUSION: Hcy-induced stress negatively modulates apoE expression via MEK1/2 and NF-κB activation. The decreased apoE expression in peripheral tissues may aggravate atherosclerosis, neurodegenerative diseases and renal dysfunctions.

Keywords: Apolipoprotein E, Homocysteine, Nuclear factor kappa B, Gene regulation, MAPK/ERK kinase

Core tip: This original manuscript investigates the effect of high homocysteine (Hcy) levels on apoE expression, and the signaling pathways involved in this gene regulation. Our novel findings show that high doses of Hcy decrease apolipoprotein E (apoE) expression. We revealed that this regulation involves nuclear factor kappa B activation, via MAPK/ERK kinase. The Hcy-mediated decrease of apoE expression in peripheral tissues may aggravate atherosclerosis, neurodegenerative diseases and renal dysfunctions. Thus, the current manuscript may be of interest for scientists working in the field of cardiovascular disease and related inflammatory disorders.