Abstract

Peroxisome proliferator-activated receptor-γ (PPARγ) agonists, a new class of antidiabetic agents, have been shown to possess antiinflammatory activity. In this study, we investigated the molecular mechanism by which PPARγ agonists inhibit proinflammatory cytokine expression in rat glomerular mesangial cells. Both natural and synthetic PPARγ agonists potently inhibited RANTES (regulated upon activation, normal T cell expressed and secreted) and monocyte chemoattractant protein-1 expression induced by TNF-α in mesangial cells, which was dependent on NF-κB signaling. However, PPARγ agonists had little effect on TNF-α-triggered IκBα phosphorylation and its subsequent degradation, p65 phosphorylation, and nuclear translocation. In the absence of PPARγ ligand, TNF-α induced a physical interaction between nuclear p65 and PPARγ, as demonstrated by co-immunoprecipitation. Such an interaction was mediated by the C-terminal region of p65. Activation of PPARγ by its agonist prevented PPARγ·p65 complex formation. Chromatin immunoprecipitation assay revealed that TNF-α induced p65 binding to the cis-acting κB elements in rat RANTES promoter, whereas disruption of PPARγ·p65 by its agonist blocked p65 interaction with its cognate κB sites. Knockdown of PPARγ via siRNA strategy completely abolished TNF-α-mediated p65 binding to κB sites and negated RANTES induction, suggesting that unliganded PPARγ is obligatory for NF-κB signaling. Consistently, overexpression of PPARγ in the absence of its ligand sensitized mesangial cells to TNF-α stimulation. These results uncover a paradoxical action of the unliganded and ligand-activated PPARγ in regulating NF-κB signaling and demonstrate PPARγ ligand as a molecular switch that controls its ability to modulate inflammatory responses in opposite directions.

Highlights

  • Peroxisome proliferator-activated receptor-␥ (PPAR␥),2 a ligand-dependent transcription factor that belongs to a subclass of the nuclear hormone receptor superfamily, plays a pivotal role in regulating a wide variety of biological processes such as insulin sensitivity, immune response, adipogenesis, and glu

  • Mesangial Cells—We first examined the effect of PPAR␥ ago- preincubation with 15d-PGJ2 did not significantly affect the nists on the expression of proinflammatory cytokines in mes- TNF-␣-induced I␬B␣ phosphorylation, its degradation, and Arrowheads denote nuclear staining of p65

  • Synthetic PPAR␥ agonist troglitazone failed to disrupt I␬B␣ degradation and p65 activation (Fig. 3C). These results suggest that PPAR␥ activation by its agonists does not affect the early events of nuclear factor-␬B (NF-␬B) signaling, such as I␬B␣ phosphorylation, its subsequent degradation, and p65 phosphorylation

Read more

Summary

FUNCTIONAL SWITCH BY ITS LIGAND*

We investigated the molecular mechanism by which PPAR␥ agonists inhibit proinflammatory cytokine expression in rat glomerular mesangial cells. Evidence shows that PPAR␥ agonists are able to inhibit the expression and/or biological effects of tumor necrosis factor-␣ (TNF-␣), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), plasminogen activator inhibitor-1, and angiotensinogen [9]. These studies underscore that the antiinflammatory potential of PPAR␥ agonists may play a crucial role in mediating their beneficial actions. Our studies uncover a novel mechanism by which unliganded and ligand-activated PPAR␥ exert an opposite action in regulating NF-␬B activity and inflammatory cytokine expression in mesangial cells

EXPERIMENTAL PROCEDURES
We also assessed the effects of
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