4.8 Article

Nuclear Receptor NR1D1 Regulates Abdominal Aortic Aneurysm Development by Targeting the Mitochondrial Tricarboxylic Acid Cycle Enzyme Aconitase-2

Journal

CIRCULATION
Volume 146, Issue 21, Pages 1591-1609

Publisher

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCULATIONAHA.121.057623

Keywords

alpha-Ketoglutarate; abdominal aortic aneurysm; mitochondria; nuclear receptor; vascular smooth muscle

Funding

  1. National Science Fund for Distinguished Young Scholars [81625002]
  2. National Natural Science Foundation of China [81930007, 82230014, 81800048, 81800378, 82070477, 91839301]
  3. Shanghai Science and Technology Committee [19ZR1430400, 22JC1402100]
  4. Innovative Research Team of High-Level Local Universities in Shanghai
  5. Shanghai Municipal Key Clinical Specialty [shslczdzk06204]

Ask authors/readers for more resources

NR1D1 is upregulated in AAA tissues and its knockout in VSMCs inhibits AAA formation. NR1D1 regulates mitochondrial metabolism by repressing ACO2 expression, and α-KG supplementation has beneficial effects in preventing and treating AAA.
Background:Metabolic disorder increases the risk of abdominal aortic aneurysm (AAA). NRs (nuclear receptors) have been increasingly recognized as important regulators of cell metabolism. However, the role of NRs in AAA development remains largely unknown. Methods:We analyzed the expression profile of the NR superfamily in AAA tissues and identified NR1D1 (NR subfamily 1 group D member 1) as the most highly upregulated NR in AAA tissues. To examine the role of NR1D1 in AAA formation, we used vascular smooth muscle cell (VSMC)-specific, endothelial cell-specific, and myeloid cell-specific conditional Nr1d1 knockout mice in both AngII (angiotensin II)- and CaPO4-induced AAA models. Results:Nr1d1 gene expression exhibited the highest fold change among all 49 NRs in AAA tissues, and NR1D1 protein was upregulated in both human and murine VSMCs from AAA tissues. The knockout of Nr1d1 in VSMCs but not endothelial cells and myeloid cells inhibited AAA formation in both AngII- and CaPO4-induced AAA models. Mechanistic studies identified ACO2 (aconitase-2), a key enzyme of the mitochondrial tricarboxylic acid cycle, as a direct target trans-repressed by NR1D1 that mediated the regulatory effects of NR1D1 on mitochondrial metabolism. NR1D1 deficiency restored the ACO2 dysregulation and mitochondrial dysfunction at the early stage of AngII infusion before AAA formation. Supplementation with alpha KG (alpha-ketoglutarate, a downstream metabolite of ACO2) was beneficial in preventing and treating AAA in mice in a manner that required NR1D1 in VSMCs. Conclusions:Our data define a previously unrecognized role of nuclear receptor NR1D1 in AAA pathogenesis and an undescribed NR1D1-ACO2 axis involved in regulating mitochondrial metabolism in VSMCs. It is important that our findings suggest alpha KG supplementation as an effective therapeutic approach for AAA treatment.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available