Vitamin D Receptor Inhibits NLRP3 Activation by Impeding Its BRCC3-Mediated Deubiquitination

Zebing Rao, Xin Chen, Junxian Wu, Mengjun Xiao, Jing Zhang, Binghao Wang, Lei Fang, Hongjie Zhang, Xiaoming Wang, Shuo Yang, Yunzi Chen, Zebing Rao, Xin Chen, Junxian Wu, Mengjun Xiao, Jing Zhang, Binghao Wang, Lei Fang, Hongjie Zhang, Xiaoming Wang, Shuo Yang, Yunzi Chen

Abstract

The NLRP3 inflammasome is a multiprotein oligomer responsible for activation of the inflammatory response by promoting the maturation and secretion of the pro-inflammatory cytokines IL-1β and IL-18. Dysregulation of this inflammasome has been linked to several autoimmune diseases, indicating that NLRP3 is tightly regulated to prevent aberrant activation. The regulation of NLRP3 activation remains unclear. Here, we report the identification of vitamin D receptor (VDR) as a negative regulator of NLRP3 oligomerization and activation. VDR can physically bind NLRP3 and block the association of NLRP3 with BRCC3. When BRCC3-mediated deubiquitination of NLRP3 is inhibited by VDR, NLRP3 activation is subsequently inhibited. In the absence of VDR, caspase-1 activation and IL-1β release are increased in response to LPS-induced inflammation or alum-induced peritoneal inflammation, indicating that VDR is a negative regulator of NLRP3 inflammasome activation in vivo. In addition, vitamin D negatively regulates the NLRP3 inflammasome via VDR signaling to effectively inhibit IL-1β secretion. These studies demonstrate that VDR signaling constrains NLRP3 inflammasome activation and might be a potential treatment target for NLRP3 inflammasome-related diseases.

Keywords: BRCC3; NLRP3 inflammasome; VDR; cytokines; deubiquitinating.

Copyright © 2019 Rao, Chen, Wu, Xiao, Zhang, Wang, Fang, Zhang, Wang, Yang and Chen.

Figures

Figure 1
Figure 1
VDR interacts with NLRP3. (A) Mass spectrometry analysis of NLRP3 and VDR peptides after immunoprecipitation with Flag to pull down NLRP3-associated proteins in Flag-NLRP3-overexpressing HEK293T cells. (B) HA-VDR was co-expressed with Flag-tagged NLRP3 in HEK293T cells; proteins were immunoprecipitated and analyzed by immunoblotting. Whole-cell lysates are shown as the input. (C) LPS-primed BMDMs were unstimulated or stimulated with nigericin for 30 min. Cell lysates were immunoprecipitated (IP) and immunoblotted (IB) with the indicated antibodies. (D) Immunofluorescent staining for VDR and NLRP3 in LPS-primed BMDMs treated with or without nigericin. Scale bar, 10 μm. (E) Purified GST-VDR was incubated with purified His-NLRP3 for 2 h. His-NLRP3-Flag bound to GST-VDR was pulled down by glutathione beads and subjected to immunoblot analysis. (F) Wild-type or mutant NLRP3 (PYD, NACHT, or LRR) and HA-VDR were expressed in HEK293T cells, immunoprecipitated, and analyzed by immunoblotting. (G) Wild-type or mutant VDR (DBD or LBD) and Flag-NLRP3 were expressed in HEK293T cells, immunoprecipitated, and analyzed by immunoblotting.
Figure 2
Figure 2
VDR inhibits NLRP3 inflammasome activation. (A) Immunoblot analysis of IL-1β and cleaved caspase-1 (p20) in culture supernatants (SN) of LPS-primed BMDMs (wild-type and Vdr−/−) treated for 4 h and then stimulated with ATP, nigericin, or alum. Immunoblot analysis of NLRP3, ASC, pro-IL-1β and pro-caspase-1 in cell lysates (Lysate). (B–D) IL-1β (B), IL-18 (C), and TNF-α (D) ELISAs using supernatants from LPS-primed BMDMs (wild-type and Vdr−/−) treated for 4 h and then stimulated with ATP, nigericin, or alum. (E) Immunoblot analysis of IL-1β and cleaved caspase-1 (p20) in culture supernatants (SN) of LPS-primed PMs (wild-type and Vdr−/−) treated for 4 h and then stimulated with nigericin or alum. Immunoblot analysis of pro-IL-1β and pro-caspase-1 in cell lysates (Lysate). (F,G) Immunoblot analysis of IL-1β and cleaved caspase-1 (p20) in culture supernatants (SN) of LPS-primed BMDMs (F). Supernatants were analyzed by IL-1β ELISA (G). (H) Pam3CSK4-primed BMDMs (wild-type and Vdr−/−) were stimulated by LPS transfection. Supernatants (SN) and cell extracts (Lysate) were analyzed by immunoblotting. Data are presented as the mean ± SEM; *p < 0.05, **p < 0.01, and ***p < 0.001. Data in (B–D,G) are representative of three independent experiments.
Figure 3
Figure 3
Vitamin D receptor blocks NLRP3 oligomerization and ASC speck formation. (A,B) Representative immunofluorescence images and quantification of endogenous ASC specks (arrows). The data show representative results from three combined independent experiments. Scale bar, 10 μm. (C) ASC oligomerization induced by the indicated stimuli at 0, 5, 10, and 15 min in WT and Vdr−/− macrophages primed with LPS. Data are presented as the mean ± SEM; *p < 0.05. Data in panel B is representative of three independent experiments.
Figure 4
Figure 4
Vitamin D receptor interferes with the BRCC3–NLRP3 interaction. (A) Both WT and Vdr−/− BMDMs were treated with LPS for 4 h. NLRP3 ubiquitination was analyzed. (B) Immunoblot analysis of BRCC3 protein in mock or LPS-primed WT and Vdr−/− BMDMs lysates immunoprecipitated with the anti-NLRP3 antibody. (C,D) HEK293T cells were transfected with the indicated vectors. Samples were immunoprecipitated with the anti-Flag antibody and analyzed by immunoblotting. (E) LPS-primed BMDMs (wild-type and Vdr−/−) transfected with the indicated non-targeting or BRCC3-specific siRNA were unstimulated or stimulated with nigericin for 30 min. Supernatants (SN) and cell extracts (Lysate) were analyzed by immunoblotting. IL-1β ELISA (F). Data are presented as the mean ± SEM; **p < 0.01. Data in (F) is representative of three independent experiments.
Figure 5
Figure 5
Vitamin D receptor inhibits NLRP3 deubiquitination. (A) HEK293T cells were transfected with the indicated vectors. NLRP3 ubiquitination was analyzed. (B) Immunoblot analysis of NLRP3 after treatment of LPS-primed BMDMs (wild-type and Vdr−/−) with CHX (1 μM) for the indicated time. (C) HEK293T cells expressing HA-BRCC3 and Myc-NLRP3 PYD, NACHT, or LRR were co-transfected with VDR as indicated. Myc-NLRP3 immunoprecipitants were analyzed for ubiquitination. (D) HEK293T cells expressing Flag-NLRP3, HA-BRCC3, and HA-Ubiquitin K63 or K48 were collected, and the cell lysates were immunoprecipitated with the anti-Flag antibody to detect ubiquitination.
Figure 6
Figure 6
Vitamin D receptor deficiency promotes LPS-induced systemic inflammation and Alum-induced peritoneal inflammation via suppression of the NLRP3 inflammasome. (A) Survival of VDR+/+NLRP3+/+, VDR−/−NLRP3+/+, VDR+/+NLRP3−/−, and VDR−/−NLRP3−/− mice (n = 10 mice/group) intraperitoneally injected with LPS (8 mg/kg body weight) over a period of 72 h. (B–D) Serum IL-1β, IL-18, and TNFα ELISAs at 6 h after intraperitoneal injection of LPS (8 mg/kg body weight) into VDR+/+NLRP3+/+, VDR−/−NLRP3+/+, VDR+/+NLRP3−/−, and VDR−/−NLRP3−/− mice. (E,F) Representative FACS plots of peritoneal CD11b+ Ly6G+ cells (neutrophils) from VDR+/+NLRP3+/+, VDR−/−NLRP3+/+, VDR+/+NLRP3−/−, and VDR−/−NLRP3−/− mice at 12 h after an intraperitoneal injection with alum (n = 5 mice/group). *p < 0.05, **p < 0.01, ***p < 0.001, NS p > 0.05. Values are the mean ± SEM of five mice per group. Both male and female mice were randomly assigned. Data in (B–D,F) are representative of three independent experiments.
Figure 7
Figure 7
Vitamin D enhances VDR-mediated inhibition of NLRP3 inflammasome activation. (A) Immunoblot analysis of IL-1β and cleaved caspase-1 (p20) in culture supernatants (SN) of LPS-primed BMDMs treated for 3 h with various doses (upper lanes) of 1,25 VD and then stimulated with nigericin. Immunoblot analysis of NLRP3, ASC, pro-IL-1β and pro-caspase-1 in cell lysates (Lysate). (B–D) IL-1β (B), IL-18 (C), and TNF-α (D) in supernatants. (E) LPS-primed BMDMs (wild type and Vdr−/−) were treated with 1,25 vitamin D (1,25 VD, 20 μM) and then stimulated with nigericin. Supernatants (SN) and cell extracts (Lysate) were analyzed by immunoblotting. (F) LPS-primed THP-1 cells were treated with different doses of 1,25 vitamin D (1,25VD) as indicated and then stimulated with nigericin. Supernatants (SN) and cell extracts (Lysate) were analyzed by immunoblotting. (G) The induction of ASC oligomerization by the indicated stimuli in WT and Vdr−/− LPS-primed macrophages. Nigericin stimulation lasted 15 min. (H) LPS-primed BMDMs were treated with or without 20 μM 1,25VD for 3 h. NLRP3 ubiquitination was analyzed. Data are presented as the mean ± SEM; *p < 0.05, **p < 0.01, and ***p < 0.001. Data in (B–D) are representative of three independent experiments.

References

    1. Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. (2016) 16:407–20. 10.1038/nri.2016.58
    1. He Y, Hara H, Nunez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. (2016) 41:1012–21. 10.1016/j.tibs.2016.09.002
    1. Baroja-Mazo A, Martin-Sanchez F, Gomez AI, Martinez CM, Amores-Iniesta J, Compan V, et al. . The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. (2014) 15:738–48. 10.1038/ni.2919
    1. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. (2014) 157:1013–22. 10.1016/j.cell.2014.04.007
    1. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. . NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. (2013) 493:674–8. 10.1038/nature11729
    1. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, et al. . NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. (2010) 464:1357–61. 10.1038/nature08938
    1. Mortimer L, Moreau F, MacDonald JA, Chadee K. NLRP3 inflammasome inhibition is disrupted in a group of auto-inflammatory disease CAPS mutations. Nat Immunol. (2016) 17:1176–86. 10.1038/ni.3538
    1. Kotas ME, Medzhitov R. Homeostasis, inflammation, and disease susceptibility. Cell. (2015) 160:816–27. 10.1016/j.cell.2015.02.010
    1. Rathinam VA, Fitzgerald KA. Inflammasome complexes: emerging mechanisms and effector functions. Cell. (2016) 165:792–800. 10.1016/j.cell.2016.03.046
    1. Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F, Alnemri ES. Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation. J Biol Chem. (2012) 287:36617–22. 10.1074/jbc.M112.407130
    1. Py BF, Kim MS, Vakifahmetoglu-Norberg H, Yuan J. Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol Cell. (2013) 49:331–8. 10.1016/j.molcel.2012.11.009
    1. Carlberg C, Campbell MJ. Vitamin D receptor signaling mechanisms: integrated actions of a well-defined transcription factor. Steroids. (2013) 78:127–36. 10.1016/j.steroids.2012.10.019
    1. Sassi F, Tamone C, D'Amelio P. Vitamin D: nutrient, hormone, and immunomodulator. Nutrients. (2018) 10:E1656. 10.3390/nu10111656
    1. Dimitrov V, White JH. Vitamin D signaling in intestinal innate immunity and homeostasis. Mol Cell Endocrinol. (2017) 453:68–78. 10.1016/j.mce.2017.04.010
    1. Landel V, Millet P, Baranger K, Loriod B, Feron F. Vitamin D interacts with Esr1 and Igf1 to regulate molecular pathways relevant to Alzheimer's disease. Mol Neurodegener. (2016) 11:22. 10.1186/s13024-016-0087-2
    1. Sly LM, Lopez M, Nauseef WM, Reiner NE. 1alpha,25-Dihydroxyvitamin D3-induced monocyte antimycobacterial activity is regulated by phosphatidylinositol 3-kinase and mediated by the NADPH-dependent phagocyte oxidase. J Biol Chem. (2001) 276:35482–93. 10.1074/jbc.M102876200
    1. Shin DM, Yuk JM, Lee HM, Lee SH, Son JW, Harding CV, et al. . Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cell Microbiol. (2010) 12:1648–65. 10.1111/j.1462-5822.2010.01497.x
    1. Bellastella G, Maiorino MI, Petrizzo M, De Bellis A, Capuano A, Esposito K, et al. . Vitamin D and autoimmunity: what happens in autoimmune polyendocrine syndromes? J Endocrinol Invest. (2015) 38:629–33. 10.1007/s40618-014-0233-z
    1. Vanherwegen AS, Eelen G, Ferreira GB, Ghesquiere B, Cook DP, Nikolic T, et al. . Vitamin D controls the capacity of human dendritic cells to induce functional regulatory T cells by regulation of glucose metabolism. J Steroid Biochem Mol Biol. (2018) 187:134–45. 10.1016/j.jsbmb.2018.11.011
    1. Bryan NB, Dorfleutner A, Kramer SJ, Yun C, Rojanasakul Y, Stehlik C. Differential splicing of the apoptosis-associated speck like protein containing a caspase recruitment domain (ASC) regulates inflammasomes. J Inflamm. (2010) 7:23. 10.1186/1476-9255-7-23
    1. Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, et al. . The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation. Nat Immunol. (2014) 15:727–37. 10.1038/ni.2913
    1. Shim DW, Lee KH. Posttranslational regulation of the NLR family pyrin domain-containing 3 inflammasome. Front Immunol. (2018) 9:1054. 10.3389/fimmu.2018.01054
    1. Stutz A, Kolbe CC, Stahl R, Horvath GL, Franklin BS, van Ray O, et al. . NLRP3 inflammasome assembly is regulated by phosphorylation of the pyrin domain. J Exp Med. (2017) 214:1725–36. 10.1084/jem.20160933
    1. He Y, Zeng MY, Yang D, Motro B, Nunez G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature. (2016) 530:354–7. 10.1038/nature16959
    1. Bullon P, Navarro JM. Inflammasome as a key pathogenic mechanism in endometriosis. Curr Drug Targets. (2017) 18:997–1002. 10.2174/1389450117666160709013850
    1. Abdul-Sater AA, Said-Sadier N, Ojcius DM, Yilmaz O, Kelly KA. Inflammasomes bridge signaling between pathogen identification and the immune response. Drugs Today. (2009) 45(Suppl. B):105–12.
    1. Goncalves AC, Ferreira LS, Manente FA, de Faria C, Polesi MC, de Andrade CR, et al. . The NLRP3 inflammasome contributes to host protection during Sporothrix schenckii infection. Immunology. (2017) 151:154–66. 10.1111/imm.12719
    1. Savage CD, Lopez-Castejon G, Denes A, Brough D. NLRP3-inflammasome activating DAMPs stimulate an inflammatory response in glia in the absence of priming which contributes to brain inflammation after injury. Front Immunol. (2012) 3:288. 10.3389/fimmu.2012.00288
    1. Voet S, Mc Guire C, Hagemeyer N, Martens A, Schroeder A, Wieghofer P, et al. . A20 critically controls microglia activation and inhibits inflammasome-dependent neuroinflammation. Nat Commun. (2018) 9:2036. 10.1038/s41467-018-04376-5
    1. Guo C, Xie S, Chi Z, Zhang J, Liu Y, Zhang L, et al. Bile Acids Control Inflammation and Metabolic Disorder through Inhibition of NLRP3 Inflammasome. Immunity. (2016) 45:802–816. 10.1016/j.immuni.2016.09.008
    1. Yan Y, Jiang W, Liu L, Wang X, Ding C, Tian Z, et al. . Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell. (2015) 160:62–73. 10.1016/j.cell.2014.11.047
    1. Yang CS, Kim JJ, Kim TS, Lee PY, Kim SY, Lee HM, et al. . Small heterodimer partner interacts with NLRP3 and negatively regulates activation of the NLRP3 inflammasome. Nat Commun. (2015) 6:6115. 10.1038/ncomms7115
    1. Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. (2009) 27:519–50. 10.1146/annurev.immunol.021908.132612
    1. Guarda G, Braun M, Staehli F, Tardivel A, Mattmann C, Forster I, et al. . Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. (2011) 34:213–23. 10.1016/j.immuni.2011.02.006
    1. Bauernfeind F, Hornung V. Of inflammasomes and pathogens–sensing of microbes by the inflammasome. EMBO Mol Med. (2013) 5:814–26. 10.1002/emmm.201201771
    1. Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. . Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. (2009) 183:787–91. 10.4049/jimmunol.0901363
    1. Song H, Liu B, Huai W, Yu Z, Wang W, Zhao J, et al. . The E3 ubiquitin ligase TRIM31 attenuates NLRP3 inflammasome activation by promoting proteasomal degradation of NLRP3. Nat Commun. (2016) 7:13727. 10.1038/ncomms13727
    1. Humphries F, Bergin R, Jackson R, Delagic N, Wang B, Yang S, et al. . The E3 ubiquitin ligase Pellino2 mediates priming of the NLRP3 inflammasome. Nat Commun. (2018) 9:1560. 10.1038/s41467-018-03669-z
    1. Han CY, Rho HS, Kim A, Kim TH, Jang K, Jun DW, et al. . FXR inhibits endoplasmic reticulum stress-induced NLRP3 inflammasome in hepatocytes and ameliorates liver injury. Cell Rep. (2018) 24:2985–99. 10.1016/j.celrep.2018.07.068
    1. Pike JW, Meyer MB. Fundamentals of vitamin D hormone-regulated gene expression. J Steroid Biochem Mol Biol. (2013) 144(Pt A):5–11. 10.1016/j.jsbmb.2013.11.004
    1. Hendy GN, Canaff L. Calicium-sensing receptor,proinflammatory cytokines and calcium homestasis. Semin Cell Dev Biol. (2016) 49:37–43 10.1016/j.semcdb.2015.11.006
    1. Deng M, Guo H, Tam JW, Johnson BM, Brickey WJ, New JS, et al. . Platelet-activating factor (PAF) mediates NLRP3-NEK7 inflammasome induction independently of PAFR. J Exp Med. (2019). 10.1084/jem.20190111. [Epub ahead of print].
    1. Rao Z, Zhang N, Xu N, Pan Y, Xiao M, Wu J, et al. . 1,25-Dihydroxyvitamin D inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) secretion via Targeting the NF-E2-related factor 2-hemeoxygenase-1-HMGB1 pathway in macrophages. Front Immunol. (2017) 8:1308. 10.3389/fimmu.2017.01308
    1. Liu W, Chen Y, Golan MA, Annunziata ML, Du J, Dougherty U, et al. . Intestinal epithelial vitamin D receptor signaling inhibits experimental colitis. J Clin Invest. (2013) 123:3983–96. 10.1172/JCI65842
    1. Guo C, Chi Z, Jiang D, Xu T, Yu W, Wang Z, et al. . Cholesterol homeostatic regulator SCAP-SREBP2 integrates NLRP3 inflammasome activation and cholesterol biosynthetic signaling in macrophages. Immunity. (2018) 49:842–56 e847. 10.1016/j.immuni.2018.08.021

Source: PubMed

3
Sottoscrivi