Elsevier

Redox Biology

Volume 30, February 2020, 101414
Redox Biology

Long isoforms of NRF1 negatively regulate adipogenesis via suppression of PPARγ expression

https://doi.org/10.1016/j.redox.2019.101414Get rights and content
Under a Creative Commons license
open access

Highlights

  • SVF cells isolated from WAT of Nrf1(f)-KO mice displayed augmented adipogenesis.

  • Stable silencing of L-Nrf1 in 3T3-L1 cells resulted in enhanced and accelerated adipogenesis.

  • Overexpression of L-NRF1-741, but not S-NRF1s, attenuated adipogenesis in 3T3-L1 cells.

  • L-NRF1 suppressed adipogenesis via downregulating PPARγ2 expression.

Abstract

Nuclear factor erythroid 2-related factor 1 (NRF1), a ubiquitously expressed CNC-bZIP transcription factor, plays a critical role in white adipocyte (WAC) biology, whereas the underlying mechanisms remain unknown. The mouse Nrf1 gene is transcribed in a number of alternatively spliced forms, resulting in two long protein isoforms (L-NRF1) containing 741 and 742 amino acids (aa) and multiple short isoforms (S-NRF1). Our previous study found that adipocyte-specific knockout of Nrf1 [Nrf1(f)-KO] in mice disturbs the expression of lipolytic genes in adipocytes, leading to adipocyte hypertrophy followed by inflammation, pyroptosis and insulin resistance. In the present study, we found that the stromal vascular fraction (SVF) cells isolated from white adipose tissues (WAT) of Nrf1(f)-KO mice display augmented adipogenesis showing elevated mRNA and protein expression of adipogenic markers and lipid accumulation. In 3T3-L1 cells, stable knockdown (KD) of all or long isoforms of Nrf1 (termed as A-Nrf1-KD and L-Nrf1-KD, respectively) using lentiviral shRNAs resulted in enhanced and accelerated adipogenic differentiation. Conversely, overexpression of L-NRF1-741, but not any of the S-NRF1, substantially attenuated adipogenesis in 3T3-L1 cells. These findings indicate that L-NRF1 might serve as a critical negative regulator of adipogenesis. Mechanistic investigation revealed that L-NRF1 may negatively regulates the transcription of peroxisome proliferator-activated receptor γ (PPARγ), in particular the master regulator of adipogenesis PPARγ2. Taken all together, the findings in the present study provide further evidence for a novel role of NRF1 beyond its participation in cellular antioxidant response and suggest that L-NRF1 is a negative regulator of PPARγ2 expression and thereby can suppress adipogenesis.

Keywords

NRF1
Adipogenesis
PPARγ
White adipocytes

Cited by (0)

1

These authors contributed equally to this work.