4.7 Article

Unspliced XBP1 contributes to cholesterol biosynthesis and tumorigenesis by stabilizing SREBP2 in hepatocellular carcinoma

Journal

CELLULAR AND MOLECULAR LIFE SCIENCES
Volume 79, Issue 9, Pages -

Publisher

SPRINGER BASEL AG
DOI: 10.1007/s00018-022-04504-x

Keywords

XBP1; Unspliced XBP1 (XBP1-u); Cholesterol biosynthesis; Tumorigenesis; SREBP2

Funding

  1. National Natural Science Foundation of China [31871367, 32070715, 81872273, 82173029]

Ask authors/readers for more resources

XBP1-u promotes tumor growth in hepatocellular carcinoma cells by enhancing cholesterol biosynthesis through the XBP1-u/SREBP2/HMGCR axis. The stabilization of SREBP2 by XBP1-u leads to increased transcription of HMGCR, the rate-limiting enzyme in cholesterol biosynthesis.
Cholesterol biosynthesis plays a critical role in rapidly proliferating tumor cells. X-box binding protein 1 (XBP1), which was first characterized as a basic leucine zipper-type transcription factor, exists in an unspliced (XBP1-u) and spliced (XBP1-s) form. Recent studies showed that unspliced XBP1 (XBP1-u) has unique biological functions independent from XBP1-s and could promote tumorigenesis; however, whether it is involved in tumor metabolic reprogramming remains unknown. Herein, we found that XBP1-u promotes tumor growth by enhancing cholesterol biosynthesis in hepatocellular carcinoma (HCC) cells. Specifically, XBP1-u colocalizes with sterol regulatory element-binding protein 2 (SREBP2) and inhibits its ubiquitination/proteasomal degradation. The ensuing stabilization of SREBP2 activates the transcription of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), a rate-limiting enzyme in cholesterol biosynthesis. We subsequently show that the XBP1-u/SREBP2/HMGCR axis is crucial for enhancing cholesterol biosynthesis and lipid accumulation as well as tumorigenesis in HCC cells. Taken together, these findings reveal a novel function of XBP1-u in promoting tumorigenesis through increased cholesterol biosynthesis in hepatocarcinoma cells. Hence, XBP1-u might be a potential target for anti-tumor therapeutic strategies that focus on cholesterol metabolism in HCC.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available