Cardiff University | Prifysgol Caerdydd ORCA
Online Research @ Cardiff 
WelshClear Cookie - decide language by browser settings

Rapid proteasomal elimination of 3-hydroxy-3-methylglutaryl-CoA reductase by interferon-Rapid proteasomal elimination of 3-hydroxy-3-methylglutaryl-CoA reductase by interferon-y in primary macrophages requires endogenous 25-hydroxycholesterol synthesis in primary macrophages requires endogenous 25-hydroxycholesterol synthesis

Lu, Hongjin, Talbot, Simon, Robertson, Kevin A., Watterson, Steven, Forster, Thorsten, Roy, Douglas and Ghazal, Peter ORCID: https://orcid.org/0000-0003-0035-2228 2015. Rapid proteasomal elimination of 3-hydroxy-3-methylglutaryl-CoA reductase by interferon-Rapid proteasomal elimination of 3-hydroxy-3-methylglutaryl-CoA reductase by interferon-y in primary macrophages requires endogenous 25-hydroxycholesterol synthesis in primary macrophages requires endogenous 25-hydroxycholesterol synthesis. Steroids 99 , pp. 219-229. 10.1016/j.steroids.2015.02.022

[thumbnail of 1-s2.0-S0039128X15000884-main.pdf]
Preview
PDF - Published Version
Available under License Creative Commons Attribution Non-commercial No Derivatives.

Download (887kB) | Preview

Abstract

Interferons (IFNs) play a central role in immunity and emerging evidence suggests that IFN-signalling coordinately regulates sterol biosynthesis in macrophages, via Sterol Regulatory Element-Binding Protein (SREBP) dependent and independent pathways. However, the precise mechanisms and kinetic steps by which IFN controls sterol biosynthesis are as yet not fully understood. Here, we elucidate the molecular circuitry governing how IFN controls the first regulated step in the mevalonate-sterol pathway, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), through the synthesis of 25-Hydroxycholesterol (25-HC) from cholesterol by the IFN-inducible Cholesterol-25-Hydroxylase (CH25H). We show for the first 30-min of IFN stimulation of macrophages the rate of de novo synthesis of the Ch25h transcript is markedly increased but by 120-min becomes transcriptionally curtailed, coincident with induction of the Activating Transcription Factor 3 (ATF3) repressor. We demonstrate ATF3 induction by Toll-like receptors is strictly dependent on IFN-signalling. While the SREBP-pathway dependent rates of de novo transcription of Hmgcr are relatively unchanged in the first 90-min of IFN treatment, we find HMGCR enzyme levels undergo a rapid proteasomal-mediated degradation, defining a previously unappreciated SREBP-independent mechanism for IFN-action. These events precede a sustained marked reduction in Hmgcr RNA levels involving SREBP-dependent mechanisms. We demonstrate that HMGCR proteasomal-degradation by IFN strictly requires the synthesis of endogenous 25-HC and functionally couples HMGCR to CH25H to coordinately suppress sterol biosynthesis. In conclusion, we quantitatively delineate proteomic and transcriptional levels of IFN-mediated control of HMGCR, the primary enzymatic step of the mevalonate-sterol biosynthesis pathway, providing a foundational framework for mathematically modelling the therapeutic outcome of immune-metabolic pathways.

Item Type: Article
Date Type: Published Online
Status: Published
Schools: Medicine
Uncontrolled Keywords: CH25H, Cholesterol biosynthesis, 25-Hydroxycholesterol, Macrophages, Immunity, Infection,
Publisher: Elsevier
ISSN: 0039-128X
Date of First Compliant Deposit: 21 March 2018
Date of Acceptance: 25 February 2015
Last Modified: 06 May 2023 06:35
URI: https://orca.cardiff.ac.uk/id/eprint/110065

Citation Data

Cited 22 times in Scopus. View in Scopus. Powered By Scopus® Data

Actions (repository staff only)

Edit Item Edit Item

Downloads

Downloads per month over past year

View more statistics