Elsevier

Steroids

Volume 99, Part B, July 2015, Pages 219-229
Steroids

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

https://doi.org/10.1016/j.steroids.2015.02.022Get rights and content
Under a Creative Commons license
open access

Highlights

  • IFN-γ leads to the proteasomal degradation of HMGCR.

  • IFN-γ and 25-HC can transcriptionally and post-translationally alter levels of HMGCR.

  • The reduction of HMGCR through the action of IFN-γ requires the de novo synthesis of 25-HC by CH25H.

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.

Abbreviations

HMGCR
3-hydroxy-3-methylglutaryl-CoA reductase
25-HC
25-hydroxycholesterol
IFN
interferon
SREBP
sterol regulatory element-binding protein
SCAP
SREBP cleavage activating protein
ER
endoplasmic reticulum
ERAD
ER-associated protein degradation
FBS
fetal bovine serum
LPDS
lipoprotein depleted serum
MEV
mevalonate
LS
lanosterol
CHO
cholesterol
CH25H
cholesterol 25-hydroxylase
TLR
Toll-like receptor
BMDMs
bone marrow derived macrophages
DCs
dendritic cells
SBGN
systems biology graphical notation
PRRs
Pattern recognition receptors
IFNAR1
IFN-α/β receptor

Keywords

CH25H
Cholesterol biosynthesis
25-Hydroxycholesterol
Macrophages
Immunity
Infection

Cited by (0)