Dimethyl itaconate inhibits TNF-α induced NF-κB signaling pathway in epithelial cells
DMI has been successfully used to moderate IL-17-IκBζ-driven skin inflammation in mouse model of psoriasis. As NF-κB plays crucial role in psoriasis, we investigated if DMI also acts as an anti-inflammatory agent in psoriasis by inhibiting NF-κB signaling pathway. We first used NF-κB luciferase assay to evaluate the effects of DMI on NF-κB-dependent transcriptional activity upon TNF-α stimulation. NF-κB-luciferase reporter plasmid (pGL4.32) and Renilla luciferase plasmid (pGL4.74) were transiently transfected in several epithelial cell types. As shown in Figure 1, DMI potently inhibited TNF-α-induced NF-κB-dependent transcriptional activity in several epithelial cells, including human kidney epithelial HEK293T cell, breast epithelial MCF-7 cells, neuroblast epithelial SK-N-AS cells and cervix epithelial Hela cells in a dose-dependent manner. Dose-response analysis revealed that DMI inhibited TNF-α-stimulated NF-κB signaling pathway with an approximate IC50 value of 0.25 mM. Cell viability analysis showed that DMI did not have significant effect on cell viability at this concentration, eliminating the interference from cytotoxicity.
Figure 1. DMI inhibited TNF-α-induced NF-κB-dependent transcriptional activity in several epithelial cells. Human HEK293T (A) or MCF-7 (B), SK-N-AS (C) and Hela cells (D) transfected with plasmids were pretreated with DMI for 12 h before treatment with TNF-α (10 ng mL-1). Data represent the mean ± SD of at least three independent experiments, and each experiment was performed in triplicate.
Dimethyl itaconate inhibits TNF-α-induced nuclear translocation of NF-κB
We then tested whether DMI can abolish the TNF-α-induced nuclear translocation of p65. In this experiment, we used a SK-N-AS monoclonal stable cell line transfected with NF-κB/p65 labeled by red fluorescent protein and nuclear H2B labeled by green fluorescent were pre-incubated with 0.25 mM DMI for 1 h before stimulation with 10 ng mL-1 TNF-α at 37℃ (5% CO2). The movement of NF-κB/p65 was captured every 5 min for 4 h as described before14. We measured the nuclear amounts of fluorescent protein-RelA by time-lapse imaging adding DMI or not. About 120 live cells were quantified for each condition. We found that addition of DMI significantly reduced the percentage of activated cells (Supplementary Figure 1) and nuclear NF-κB intensity (Figure 2A, B). Collectively, these data suggested that DMI inhibited TNF-α-induced nuclear translocation of p65.
Figure 2. DMI inhibited TNF-α-induced NF-κB/p65 nuclear translocation in SK-N-AS cells. (A) Time-lapses images of stable transfected SK-N-AS cells with p65-mcherry and H2B-EGFP treated with DMSO or DMI (0.25 mM, 1 h) and then stimulated with 10 ng mL-1 TNF-α. Scale bar, 10 μm. (B) Average nuclear NF-κB intensity of cells pretreated with DMSO or DMI and then stimulated with TNF-α (10 ng mL-1).
DMI inhibits TNF-α-induced NF-κB Activation by Targeting IKK
From NF-κB luciferase assay and time-lapse confocal imaging experiment, we showed that DMI inhibited the NF-κB signaling pathway. We next identified the molecular target of DMI. Because IKKβ plays central role in NF-κB signaling pathway, we first evaluated the effect of DMI on IKKβ-dependent phosphorylation and degradation of IκBα in TNF-α-induced activation of NF-κB in Hela cells. As shown in Figure 3, phosphorylated IκBα and IκBα were detected in Hela cells pretreated with or without DMI for 12 h and then stimulated with TNF-α for indicated time by Western blot analysis using antiphospho-Ser32/Ser36 IκBα antibody and no phosphorylated IκBα antibody. We found TNF-α induced phosphorylation and degradation of IκBα were in time-dependent manner as described before15. Strikingly, pretreatment with DMI for 12 h could markedly inhibit TNF-α-induced IκBα phosphorylation and degradation. These results indicated that DMI inhibits TNF-α-induced NF-κB activation by preventing IκBα phosphorylation and degradation by targeting IKKβ.
Figure 3. DMI inhibited TNF-α-induced NF-κB signaling pathway by preventing IκBα phosphorylation and degradation. Hela cells were incubated in the presence of TNF-α (10 ng mL-1) for indicated time (0 min, 5 min, 10 min, 15 min, 30 min) in the culture medium with or without 1 mM DMI. (A) Western blot imaging of dynamic of phosphorylation and degradation of IκBα in total lysates. IκBα and p-IκBα band integrated intensity were shown in (B) and (C).
We further tested whether DMI can directly interact with IKKβ. We generated N-10xHis-tagged IKKβ recombinant protein from HEK293T cells transfected with a pCMV-N-His-IKKβ expression plasmid for further analysis. The N-10xHis-tagged IKKβ was captured by NTA agarose beads. The purified recombinant IKKβ protein was incubated with or without DMI for 12 h at 4 ℃, and then the mixtures were resolved by SDS-PAGE. As shown in Supplementary Figure 3, one clear band was observed at around 88 kDa. After that, in-gel digestion of IKKβ was performed with trypsin and then analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Notably, we identified several tryptic peptides having mass shift, as shown in Supplementary Figure 2. Tandem mass spectrometry revealed that treatment of recombinant human IKK-β with 0.25 mM DMI led to a combination of dimethyl and monomethyl itaconate at cysteines 412, 370 and 716 (Supplementary Table 1). Neither of these modifications was observed at any cysteine in vehicle-treated IKK-β.
To test whether the DMI can modify IKK-β in cell, we treated HEK293T cells overexpressing recombinant IKK-β with 0.25 mM DMI for 3h. After incubation, we extracted whole-cell lysate using NTA agarose beads and digested the protein with trypsin. As shown in Figure 4, LC-MS/MS analysis identified a peptide with calculated mass of 2876.46 Da, which is 132.34 Da larger than the C412-containing peptide ITYETQISPRPQPESVSCILQEPK that has a calculated mass of 2744.12 Da. The mass difference of 132.34 Da matches the molecular weight of itaconate. LC-MS/MS spectra demonstrated that a 132.34 Da mass shift occurred starting from y5 to the y12 fragment, indicating that the C412 residue was covalently modified by DMI. However, the modified molecule was itaconate rather than DMI. It was shown that DMI was rapidly degraded in cells without releasing itaconate12, hence it was possible that DMI covalently binds to IKKβ first and the complex undergoes further esterase digestion. As the MMI modified IKKβ was also observed in vitro, the other possibility is the fragmentation may happen in the mass spectrometry detection. In addition to this, we also hypothesized that the inhibitory effect of NF-κB signaling pathway from DMI treatment was comprehensive results of both DMI and itaconate. We supposed that only a few DMI may convert to itaconate, and this part of itaconate alkylate cysteine residues on proteins rapidly. It has been discovered that IRG1, an enzyme that produces itaconate in cells, can suppress NF-κB signaling pathway by decreasing IKKα/β activation upon LPS stimulation16. We then tested whether itaconate can bind to IKKβ covalently in vitro. As shown in Supplementary Table 2, itaconate can modify several cysteines in IKKβ. Next, to determine whether IRG1-mediated itaconate production can affect NF-κB signaling pathway upon TNF-α stimulation in cells, we overexpressed IRG1 in HEK293T cells using a pCMV3-N-Flag-IRG1. IRG1 and itaconate was only detected in pCMV3-N-Flag-IRG1-overexpressing cells (for detail, see Supplementary Figure 5A, B). Not surprisingly, itaconate produced by IRG1 inhibited TNF-α-induced NF-κB signaling pathway (Supplementary Figure 5B), which was consistent with the results before16. To eliminate the effect of IRG1 itself, we also used 4-octyl itaconate (OI), a recently described cell-permeable derivative of itaconate which can release itaconate in cells17, to treat cells and then detected the activation of NF-κB pathway. Cell luciferase assay and Western blot analysis showed that OI can inhibit TNF-α-stimulated NF-κB signaling pathway (for detail, see the Supplementary Figure 4 and Supplementary Figure 6). However, the inhibitory effect of OI on NF-κB signaling pathway was weaker than that of DMI on NF-κB signaling pathway. These results indicated that the inhibitory effect of DMI on NF-κB signaling pathway may from both DMI and itaconate. Despite of these possibilities, our results suggested that DMI treatment indeed modified IKKβ in cells.
Figure 4. DMI directly modified IKKβ in cells. HEK293T cells overexpressing recombinant IKKβ were incubated with 0.25 mM DMI for 3 h. LC-MS/MS analysis of DMI-mediated modification of recombinant IKKβ in cells.
Having identified that DMI can directly modify recombinant IKKβ in cells, we next explored how this modification affected IKKβ structure and activity. IKKβ protein exists as a dimer in solution. Each IKKβ monomer has a trimodular linear architecture: the N-terminal kinase domain (KD, 1-309), the central ubiquitin-like domain (ULD, 310-404), and the C-terminal dimerization domain (SDD, 408-664)18. IKKβ activity can be described as a kinase cycle of three states: poised, active, and inactivated. In the absence of stimulation, IKKβ was in the poised state19. After stimulation by TNF-α, the poised IKKβ is activated by phosphorylation on Ser177 and Ser181 in its activation loop. Then the activated IKKβ phosphorylates its substrate IκBα. In addition to this, the activated IKKβ also phosphorylates its own C terminus, thereby inducing conformation change that results in the decrease of kinase activity. Then the inactive hyper-phosphorylated IKKβ becomes available by dephosphorylation. Among all these steps in the IKK kinase cycle, activation of IKKβ is the most important one. Furthermore, IKKβ autophosphorylation seems to be the key step in activation of IKKβ. Thus, we tested whether DMI can directly inhibit the activation of IKKβ. We monitored the level of active IKKβ in Hela cells stimulated with TNF-α pre-treated with or without DMI using Western blot analysis. As shown in Figure 5, DMI dose-dependently and time-dependently inhibited phosphorylation of Ser177/181 on IKKβ, the activated state of IKKβ. This result indicated that DMI indeed inhibits the activation of IKKβ.
Recent crystal structure studies revealed that IKKβ formed higher order homo-oligomers to phosphorylate its dimer partners. The oligomerization surface was a “V shaped” interface including the N-terminal KD-ULD portions and the proximal SDD from the two promoters. The double mutant I413A/L414A within the V-shaped interface was found to disrupt IKKβ activation in cells20. We found the modified residue, C412, in IKKβ was right in this “V-shaped” interface. Moreover, mutation of Cys-412 (C412A) in IKKβ could inhibit its activation. And then DMI had no obvious effect on C412A mutant comparing with wile type IKKβ (Supplementary Figure 7). These evidences all support that DMI inhibits the autophosphorylation of IKKβ by modifying C412 to disrupt the oligomerization surface of IKKβ. Our results also suggest that C412 in IKKβ provides a new and potent druggable binding site for IKKβ based drug design.
Figure 5. DMI suppressed the activation of IKKβ. Hela cells were incubated in the presence of TNF-α for indicated time (0 min, 5 min, 10 min, 15 min, 30 min) with or without 0.25 mM DMI for 12 h. Cell extracts were separated on a 10% SDS gel, and the level of activated IKKβ were determined by Western blotting using phospho-IKKα/β (Ser176/180) antibody. One representative experiment of three was shown (A). Quantitation of the level of phospho-IKKα/β was shown (B). (C) Western blot analysis of phospho-IKKα/β(Ser176/180) in lysates of Hela cells untreated or different concentrations of DMI-pretreated and then stimulated with TNF-α (10 ng mL-1, 10 min). β-actin was used as loading control. Blot shown is representative of three independent experiments. (D) Histogram of intracellular phospho-IKKα/β(Ser176/180) from (C).