Abstract

The small GTPase protein RAC1 participates in innate immunity by activating a complex program that includes cytoskeleton remodeling, chemotaxis, activation of NADPH oxidase, and modulation of gene expression. However, its role in regulating the transcriptional signatures that in term control the cellular inflammatory profiles are not well defined. Here we investigated the functional and mechanistic connection between RAC1 and the transcription factor NRF2 (nuclear factor erythroid 2-related factor 2), master regulator of the anti-oxidant response. Lipopolysaccharide and constitutively active RAC1(Q61L) mutant induced the anti-oxidant enzyme heme-oxygenase-1 (HO-1) through activation of NRF2. The use of KEAP1-insensitive NRF2 mutants indicated that RAC1 regulation of NRF2 is KEAP1-independent. Interestingly, NRF2 overexpression inhibited, whereas a dominant-negative mutant of NRF2 exacerbated RAC1-dependent activation of nuclear factor-κB (NF-κB), suggesting that NRF2 has an antagonistic effect on the NF-κB pathway. Moreover, we found that RAC1 acts through NF-κB to induce NRF2 because either expression of a dominant negative mutant of IκBα that leads to NF-κB degradation or the use of p65-NF-κB-deficient cells demonstrated lower NRF2 protein levels and basally impaired NRF2 signature compared with control cells. In contrast, NRF2-deficient cells showed increased p65-NF-κB protein levels, although the mRNA levels remain unchanged, indicating post-translational alterations. Our results demonstrate a new mechanism of modulation of RAC1 inflammatory pathway through a cross-talk between NF-κB and NRF2.

Highlights

  • RAC1 is a small G-protein of the Rho family that activates the transcription factor nuclear factor-␬B (NF-␬B) to elicit an inflammatory response

  • We found that RAC1 acts through NF-␬B to induce nuclear factor (erythroid-derived 2)-like 2 (NRF2) because either expression of a dominant negative mutant of I␬B␣ that leads to NF-␬B degradation or the use of p65-NF-␬B-deficient cells demonstrated lower NRF2 protein levels and basally impaired NRF2 signature compared with control cells

  • In the case of mouse embryo fibroblasts (MEFs) from p65-knock-out mice, SFN was able to induce HO-1 expression, this effect was greatly attenuated in comparison with wild type MEFs. These results indicate that the regulation of NRF2 by NF-␬B is not related elimination of the KEAP1 constrain and suggest that the changes in NRF2 levels are due to regulation of gene expression by NF-␬B

Read more

Summary

Background

RAC1 is a small G-protein of the Rho family that activates the transcription factor NF-␬B to elicit an inflammatory response. Conclusion: RAC1 modulates inflammation by coordinating the activity of pro-inflammatory NF-␬B and anti-oxidant NRF2 transcription factors. The small GTPase protein RAC1 participates in innate immunity by activating a complex program that includes cytoskeleton remodeling, chemotaxis, activation of NADPH oxidase, and modulation of gene expression. NRF2 regulates the expression of a battery of cytoprotective genes that share in common a cis-acting enhancer sequence termed antioxidant response element (ARE) [8, 9] These genes include those coding the antioxidant enzymes heme oxygenase-1 (HO-1) and NADP(H) quinone oxidoreductase (NQO1), enzymes of glutathione metabolism and protein degradation, through the proteasome and autophagy routes [10, 11]. Cross-talk between RAC1 and NRF2 in Inflammation to the hippocampus, reinforcing the idea of NRF2 anti-inflammatory properties This evidence has been corroborated in other recent reports by analyzing other cells of the monocytemacrophage linage [8, 13,14,15]. These results uncover a new mechanism of regulation of inflammatory events trough a RAC1/NRF2/ HO-1 axis

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call