Oncotarget

Research Papers:

Peptide inhibition of the SETD6 methyltransferase catalytic activity

Michal Feldman and Dan Levy _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2018; 9:4875-4885. https://doi.org/10.18632/oncotarget.23591

Metrics: PDF 1840 views  |   HTML 2510 views  |   ?  


Abstract

Michal Feldman1,2 and Dan Levy1,2

1The Shraga Segal Department of Microbiology, Immunology and Genetics, National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er-Sheva 84105, Israel

2National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er-Sheva 84105, Israel

Correspondence to:

Dan Levy, email: [email protected]

Michal Feldman, email: [email protected]

Keywords: Lysine methylation; SETD6

Received: September 19, 2017     Accepted: December 04, 2017     Published: December 21, 2017

ABSTRACT

A large body of evidence accumulating in the past few years indicates the physiological significance of non-histone proteins lysine methylation, catalyzed by protein lysine methyl transferases (PKMTs). Dysregulation of these enzymes was shown to contribute to the development and progression of numerous diseases. SETD6 lysine methylatransferase was recently shown to participate in essential cellular processes, such as the NFkB pathway, oxidative stress and also the Wnt signaling cascade. In order to test the effect of blocking SETD6 catalytic activity, we used the peptide inhibition method, which is considered highly specific and can potentially target almost any protein. We designed a 15 amino acids peptide based on the sequence of the RelA protein (residues 302-316), containing the lysine that is methylated by SETD6. To enable cellular intake, the designed peptide was fused to a cell penetrating peptide (CPP) vp22. The vp22-RelA302-316 peptide showed direct and specific interaction with SETD6 in vitro. This interaction was shown to inhibit SETD6 methyltransferase activity. SETD6 catalytic blockage by the peptide was also observed in cells upon treatment with the vp22-RelA302-316, resulting in induced cellular migration and proliferation. This new insight into the activity of a methylation inhibitory peptide could represent a milestone in the development of therapeutic tools, which can be of use in physiological cases where administration of cell proliferation is required.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 23591