Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Oct 6;117(40):25008-25017.
doi: 10.1073/pnas.2008214117. Epub 2020 Sep 23.

14-3-3ζ-TRAF5 axis governs interleukin-17A signaling

Affiliations

14-3-3ζ-TRAF5 axis governs interleukin-17A signaling

Jenna McGowan et al. Proc Natl Acad Sci U S A. .

Abstract

IL-17A is a therapeutic target in many autoimmune diseases. Most nonhematopoietic cells express IL-17A receptors and respond to extracellular IL-17A by inducing proinflammatory cytokines. The IL-17A signal transduction triggers two broad, TRAF6- and TRAF5-dependent, intracellular signaling pathways to produce representative cytokines (IL-6) and chemokines (CXCL-1), respectively. Our limited understanding of the cross-talk between these two branches has generated a crucial gap of knowledge, leading to therapeutics indiscriminately blocking IL-17A and global inhibition of its target genes. In previous work, we discovered an elevated expression of 14-3-3 proteins in inflammatory aortic disease, a rare human autoimmune disorder with increased levels of IL-17A. Here we report that 14-3-3ζ is essential for IL-17 signaling by differentially regulating the signal-induced IL-6 and CXCL-1. Using genetically manipulated human and mouse cells, and ex vivo and in vivo rat models, we uncovered a function of 14-3-3ζ. As a part of the molecular mechanism, we show that 14-3-3ζ interacts with several TRAF proteins; in particular, its interaction with TRAF5 and TRAF6 is increased in the presence of IL-17A. In contrast to TRAF6, we found TRAF5 to be an endogenous suppressor of IL-17A-induced IL-6 production, an effect countered by 14-3-3ζ. Furthermore, we observed that 14-3-3ζ interaction with TRAF proteins is required for the IL-17A-induced IL-6 levels. Together, our results show that 14-3-3ζ is an essential component of IL-17A signaling and IL-6 production, an effect that is suppressed by TRAF5. To the best of our knowledge, this report of the 14-3-3ζ-TRAF5 axis, which differentially regulates IL-17A-induced IL-6 and CXCL-1 production, is unique.

Keywords: 14-3-3ζ; IL-17A; TRAF.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interest.

Figures

Fig. 1.
Fig. 1.
14-3-3ζ is required for IL-17A–mediated gene induction. (A) ARPE-19 cells, treated with hIL-17A in the presence of 14-3-3 pan inhibitor (BV02), were analyzed for IL6 mRNA levels by qRT-PCR. (B) CRISPR/Cas9-mediated 14-3-3ζKO ARPE-19 cells were analyzed for the endogenous levels of additional 14-3-3 isoforms, as indicated, using qRT-PCR (Upper) or immunoblot (Lower). (C and D) The WT and 14-3-3ζKO ARPE-19 cells, treated with hIL-17A, were analyzed for IL6 (C) or IL8 (D) mRNA levels using qRT-PCR. (E) The WT and 14-3-3ζKO ARPE-19 cells, treated with hIL-17A, were analyzed for IL-6 protein levels using ELISA. (F and G) The WT and 14-3-3ζKO MEFs, treated with mIL-17A, were analyzed for Il6 (F) or Il8 (G) mRNA levels using qRT-PCR. The loss of 14-3-3ζ protein in the KO MEFs is also shown as an immunoblot. (H and I) Rescuing Flag-tagged 14-3-3ζ (shown by immunoblot) by lentiviral transduction of 14-3-3ζKO ARPE-19 cells support IL-17A–induced IL6 (H) and IL8 (I) mRNA levels, analyzed by qRT-PCR. All results are representative of at least three experiments, *P < 0.05, ***P < 0.0005, and ****P < 0.0001; ns, not significant.
Fig. 2.
Fig. 2.
14-3-3ζ is required for IL-17A–mediated NF-κB activation. (A) Dual-luciferase activity was measured in NSC or 14-3-3ζKD HeLa cells, transiently coexpressing NF-κB, thymidine kinase (TK), and HA-tagged 14-3-3ζ, were treated with hIL-17A for 20 h. (B) WT or 14-3-3ζKO ARPE-19 cells, treated with hIL-17A for 1 h, were immunostained for endogenous p65 and analyzed by confocal microscopy. (Scale bar: 10 μm.) (C) WT or 14-3-3ζKO MEFs, treated with mIL-17A for 1 h, were immunostained for endogenous p65 and analyzed by confocal microscopy and the nuclear p65-expressing cells were quantified from at least 100 cells. (D and E) Nuclear fractions from WT and 14-3-3ζKO MEFs (D) or ARPE-19 cells (E), treated with IL-17A for 1 h, were analyzed for p65 and HDAC1 by immunoblots (shown in SI Appendix, Fig. S3), which were quantified using imageJ software. The results are representative of at least three experiments, *P < 0.05, **P < 0.005, and ***P < 0.0005.
Fig. 3.
Fig. 3.
14-3-3ζ is required for IL-17A–mediated pERK. (AC) The WT and 14-3-3ζKO ARPE-19 (A and C) treated with hIL-17A, or MEFs treated with mIL-17A (B), for the indicated times and doses, were analyzed for pERK by immunoblot. (D) The 14-3-3ζ (Flag-tagged) restored 14-3-3ζKD HeLa cells, treated with IL-17A, were analyzed for pERK by immunoblot. EV, empty vector. All results are representative of at least three experiments, *P < 0.05, **P < 0.005, ***P < 0.0005, and ****P < 0.0001.
Fig. 4.
Fig. 4.
14-3-3ζ forms a complex with TRAF proteins in cells and tissues. (A) HEK293T cells were cotransfected with HA–14-3-3ζ and Flag-TRAFs, as indicated. The cell lysates were immunoprecipitated with anti-HA, and the IPs were analyzed by immunoblot. The expression of the proteins is shown in the input. (B) Rat aortic lysates were immunoprecipitated with anti–14-3-3ζ or a control IgG, and the IPs were analyzed by immunoblot, as indicated. (C and D) ARPE-19 cells, treated with IL-17A, were immunoprecipitated with anti–14-3-3ζ or TRAF6, and the IPs were analyzed by immunoblot, as indicated. (E) Confocal analyses of TRAF6 and TRAF5 colocalization with 14-3-3ζ in hIL-17–treated ARPE-19 cells. (Scale bar, 10 µm.) (F and G) HA–14-3-3ζ or Flag-TRAF proteins were ectopically expressed in HEK293T cells, and isolated to near purity, and were subjected to cell-free interaction assay (SI Appendix, Fig. S5), and the complex was analyzed by co-IP.
Fig. 5.
Fig. 5.
TRAF5 suppresses IL-17A–stimulated 14-3-3ζ–dependent IL6 induction. (A and B) ARPE-19 cells, expressing shRNA against TRAF5 or a nonsilencing control (NSC) sequence, were analyzed for hIL-17A–induced IL6 by qRT-PCR (A) or ELISA (B). The KD of TRAF5 is shown by immunoblot (A). (C) The qRT-PCR analyses of CXCL1 mRNA in TRAF5KD ARPE-19 cells when compared to control cells, upon IL-17A treatment. (D) The qRT-PCR analyses of Cxcl1 mRNA induction in WT and 14-3-3ζKO MEFs, stimulated with mIL-17A. (E) IL-17A–stimulated pERK was analyzed in control (NSC) or TRAF5KD ARPE-19 cells by immunoblot. (F) The qRT-PCR analyses of IL6 induction in control (NSC) or TRAF5KD ARPE-19 cells in the presence or the absence of 30 µM of U0126 (MEK inhibitor), upon IL-17A. (G) The ARPE-19–derived cells, as indicated, were analyzed for IL6 by qRT-PCR and the protein expression was validated in the immunoblot. All results are representative of at least three experiments, *P < 0.05, **P < 0.005, ***P < 0.0005, and ****P < 0.0001.
Fig. 6.
Fig. 6.
The interaction with TRAF is required for 14-3-3ζ–mediated IL6 induction by IL-17A. (A) Two putative TRAF5 binding motifs (in red) of 14-3-3ζ and the mutants (M1 and M2) are shown. (B) The modeled structures of the TRAF-domain of TRAF5 (4GJH.pdb, pink color) complex with the dimeric 14-3-3ζ (5D2D.pdb, gray color) with restricted interaction to either of the two TBMs, SNEE or AYQE, are shown. The TBMs of 14-3-3ζ are shown in yellow color. (C) HEK293T cells, coexpressing V5-TRAF5 or Flag–14-3-3ζ, as indicated, were immunorecipitated with anti-Flag and elutes were analyzed by immunoblot. (D) HEK293T cells, coexpressing Flag-TRAF5 and HA–14-3-3ζ (WT or M2), were analyzed by PLA followed by confocal microscopy. (Scale bar, 5 µm.) (E) ARPE-19 cells, ectopically expressing 14-3-3ζ (WT, M1 or M2), were analyzed for IL-17A–induced IL6 by qRT-PCR. The expression of Flag-tagged 14-3-3ζ is shown by immunoblot. All results are representative of at least three experiments, ****P < 0.005.
Fig. 7.
Fig. 7.
Functional significance of 14-3-3ζ participation in IL-17A signaling. (A) Culture supernatants from either untreated or IL-17A–treated WT or 14-3-3ζKO MEFs were analyzed for the killing of C. albicans. (B) WT or 14-3-3ζKO MEFs, treated with mIL-17A, were analyzed for Defb3 by qRT-PCR. (C) The schematic design of 14-3-3ζ KO Lewis rat generation is shown; the exon 3 of rat 14-3-3ζ was targeted by the gRNAs, as indicated. A representative genotyping result is shown from the tail DNA isolated from the indicated rat strains (+/+, +/−, and −/−). An immunoblot of 14-3-3ζ from the tail biopsy of the indicated strains is shown. (D and E) The duodenum of WT or heterozygous 14-3-3ζ Lewis rats were either untreated or treated with rat (r) IL-17A for 20 h, and the Il6 (D) and Cxcl1 (E) mRNA levels were analyzed by qRT-PCR. (F and G) The WT and 14-3-3ζKO rats (n = 8) were injected with rIL-17A intraperitoneally for the indicated times, and cytokine (Il-6 and Cxcl-1) levels in the sera were analyzed by ELISA. The results are representative of at least three experiments, *P < 0.05, **P < 0.005, ***P < 0.0005, and ****P < 0.0001.
Fig. 8.
Fig. 8.
A proposed model for the identified 14-3-3ζ–TRAF5 axis in regulating IL-17A signaling pathways. IL-17A binding triggers a receptor complex (IL-17RA–ACT1–TRAF) activation that transduces the signal to generate transcriptional output (IL-6/IL-8/CXCL-1). Our results show that 14-3-3ζ is a TRAF-binding component that is needed for IL-17A-stimulated pERK, NF-κB activation, and IL-6/IL-8 production, but not for CXCL-1. Importantly, we uncovered a role of TRAF5 in suppressing the IL-17A–stimulated IL-6 production. The IL-17A stimulation increases the 14-3-3ζ–TRAF5 interaction that potentially inhibits the TRAF5 function and supports the IL-6 production. Overall, our model suggests that IL-17A stimulation increases 14-3-3ζ interaction to sequester TRAF5, which results in increased IL-6 levels. These results have broad implications covering IL-17A’s role in antifungal immunity to regulation of inflammation.

References

    1. Amatya N., Garg A. V., Gaffen S. L., IL-17 signaling: The yin and the yang. Trends Immunol. 38, 310–322 (2017). - PMC - PubMed
    1. Ishigame H. et al. ., Differential roles of interleukin-17A and -17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity 30, 108–119 (2009). - PubMed
    1. Leonardi C. et al. ., Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N. Engl. J. Med. 366, 1190–1199 (2012). - PubMed
    1. van Baarsen L. G. et al. ., Heterogeneous expression pattern of interleukin 17A (IL-17A), IL-17F and their receptors in synovium of rheumatoid arthritis, psoriatic arthritis and osteoarthritis: Possible explanation for nonresponse to anti-IL-17 therapy? Arthritis Res. Ther. 16, 426 (2014). - PMC - PubMed
    1. Pacheco-Lugo L. et al. ., Plasma cytokines as potential biomarkers of kidney damage in patients with systemic lupus erythematosus. Lupus 28, 34–43 (2019). - PubMed

Publication types