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
. 2019 Aug;29(8):641-654.
doi: 10.1038/s41422-019-0193-0. Epub 2019 Jun 18.

Interferon-inducible cytoplasmic lncLrrc55-AS promotes antiviral innate responses by strengthening IRF3 phosphorylation

Affiliations

Interferon-inducible cytoplasmic lncLrrc55-AS promotes antiviral innate responses by strengthening IRF3 phosphorylation

Yumei Zhou et al. Cell Res. 2019 Aug.

Abstract

Type I interferon (IFN-I) production is efficiently induced to ensure a potent innate immune response to viral infection. How this response can be enhanced, however, remains to be explored. Here, we identify a new cytoplasmic long non-coding RNA (lncRNA), lncLrrc55-AS, that drives a positive feedback loop to promote interferon regulatory factor 3 (IRF3) signaling and IFN-I production. We show that lncLrrc55-AS is virus-induced in multiple cell types via the IFN-JAK-STAT pathway. LncLrrc55-AS-deficient mice display a weakened antiviral immune response and are more susceptible to viral challenge. Mechanistically, lncLrrc55-AS binds phosphatase methylesterase 1 (PME-1), and promotes the interaction between PME-1 and the phosphatase PP2A, an inhibitor of IRF3 signaling. LncLrrc55-AS supports PME-1-mediated demethylation and inactivation of PP2A, thereby enhancing IRF3 phosphorylation and signaling. Loss of PME-1 phenocopies lncLrrc55-AS deficiency, leading to diminished IRF3 phosphorylation and IFN-I production. We have identified an IFN-induced lncRNA as a positive regulator of IFN-I production, adding mechanistic insight into lncRNA-mediated regulation of signaling in innate immunity and inflammation.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Identification of virus-induced IFN-I-dependent lncLrrc55-AS in macrophages. a cDNA of lncLrrc55-AS in VSV-infected mouse peritoneal macrophages. b RT-qPCR analysis of lncLrrc55-AS in macrophages infected with SeV for 12 h. c RT-qPCR analysis (left) of the distribution of different RNAs following nucleus/cytoplasm fractionation of macrophages infected with SeV for 12 h. Western blot analysis (right) of nucleus/cytoplasm fractionation. d FISH analysis of lncLrrc55-AS in macrophages infected with SeV for 12 h. Scale bar, 20 µm. e, f RT-qPCR analysis of lncLrrc55-AS expression in macrophages infected with HSV for 12 h, or stimulated with poly(I:C) for 12 h, LPS for 3 h (e), or IFNβ with the indicated dose for 8 h (f). g, h RT-qPCR analysis of lncLrrc55-AS expression in WT and Irf3−/− macrophages infected with SeV or VSV for 12 h (g), in WT and Ifnar1−/− macrophages treated with IFNα4, IFNβ (500 U/mL) for 8 h or infected with SeV for 12 h (h). i RT-qPCR analysis of lncLrrc55-AS expression in macrophages pretreated for 1 h with the JAK inhibitor Baricitinib (2 μM) or the STAT1 inhibitor Fludarabine (1 μM), then infected with SeV or VSV for 12 h or treated with IFNα4 for 8 h. Data are from three independent experiments (b, c left, ei, means ± SEM) or are representative of three independent experiments with similar results (a, c right, d). ***P < 0.005 (Student’s t-test or ANOVA)
Fig. 2
Fig. 2
LncLrrc55-AS enhances IFN-I production in response to viral infection. a RT-qPCR analysis of lncLrrc55-AS silencing efficiency by two siRNAs (#1, #2). NC, a non-targeting control siRNA. b Fluorescence analysis of VSV replication in NC- or lncLrrc55-AS-silenced peritoneal macrophages infected with VSV-GFP for the indicated hours. c Determination of virus loads by TCID50 assay of the supernatant from macrophages infected with VSV for the indicated hours. d ELISA of IFNα and IFNβ in supernatant of control or lncLrrc55-AS-silenced macrophages infected with VSV for 12 h. e RT-qPCR analysis of lncLrrc55-AS or Ifnb mRNA expression in the selected WT or lncLrrc55-AS KO NIH/3T3 cell clones infected with VSV for 12 h. f RT-qPCR analysis of Ifna4 and Ifnb mRNA expression in empty vector control (EV Ctrl) or lncLrrc55-AS transfected lncLrrc55-AS KO NIH/3T3 cells infected with VSV for 12 h. Data are from three independent experiments (a, b right, cf, means ± SEM) or are representative of three independent experiments with similar results (b left). *P < 0.05, **P < 0.01 and ***P < 0.005; ns, no significance (Student’s t-test or ANOVA)
Fig. 3
Fig. 3
LncLrrc55-AS protects mice against viral infection. a Determination of VSV loads by TCID50 in organs of WT or lncLrrc55-AS−/− mice (n = 6 per group, 7 weeks old) 12 or 18 h after intraperitoneal injection of VSV (5 × 107 pfu/g). b Determination of VSV replication by RT-qPCR in organs of WT or lncLrrc55-AS−/− mice 18 h after infection with VSV (as in a). c Hematoxylin and eosin staining of lung sections from WT or lncLrrc55-AS−/− mice 18 h after infection with VSV (as in a). Scale bar, 50 µm. d ELISA of cytokines in serum from WT or lncLrrc55-AS−/− mice 12 h after infection with VSV (as in a, n = 4). e RT-qPCR analysis of lncLrrc55-AS, Ifna4 and Ifnb mRNA expression in peritoneal macrophages from WT or lncLrrc55-AS−/− mice 12 h after infection with VSV (as in a, n = 3). f Survival of 7-week-old WT or lncLrrc55-AS−/− mice (n = 10) after intraperitoneal injection of VSV (1 × 108 pfu/g). Kaplan–Meier curve was used to evaluate survival rate. Data are shown as means ± SEM (a, b, d, e), or are representative of three independent experiments with similar results (c). *P < 0.05, **P < 0.01 and ***P < 0.005 (Student’s t-test)
Fig. 4
Fig. 4
LncLrrc55-AS enhances IRF3 signaling in antiviral innate responses. a Dual luciferase analysis of the activity of the IFNβ promoter after co-transfecting plasmids expressing lncLrrc55-AS (or empty vector control, EV Ctrl), an IFNβ reporter and TK-renilla with RIG-I, MAVS, TBK1, IRF3 or IRF3-5A in 293T cells. b, c Western blot analysis of phosphorylated (p-) or total protein in lysates of NC- and lncLrrc55-AS-silenced peritoneal macrophages infected with SeV (b) or WT control and lncLrrc55-AS KO NIH/3T3 cells infected with VSV (c) for the indicated hours. d Western blot analysis of phosphorylated (p-) or total IRF3 in lysates of lncLrrc55-AS KO NIH/3T3 cells ectopically expressing EV Ctrl or lncLrrc55-AS after VSV infection for the indicated hours. e Western blot analysis of phosphorylated (p-) or total IRF3 in lysates of WT and Irf3−/− peritoneal macrophages ectopically expressing lncLrrc55-AS or EV Ctrl after SeV infection for 8 h. f RT-qPCR analysis of lncLrrc55-AS, Ifna4 and Ifnb mRNA expression of WT and Irf3−/− peritoneal macrophages ectopically expressing lncLrrc55-AS or EV Ctrl after SeV infection for 8 h. Data are from three independent experiments (a, f, means ± SEM) or are representative of three independent experiments with similar results (be). *P < 0.05, **P < 0.01; ns, no significance (Student’s t-test or ANOVA)
Fig. 5
Fig. 5
Identification of protein phosphatase methylesterase PME-1 as a binding partner of lncLrrc55-AS. a RT-qPCR analysis of the RNA retrieval ratio of lncLrrc55-AS purified by hybridizing with antisense lncLrrc55-AS probes from whole cell extracts of peritoneal macrophages infected with SeV for 12 h (left). SDS-PAGE with a silver nitrate staining analysis of proteins co-purified with lncLrrc55-AS (right). The arrow indicates protein bands that were subjected to mass spectrometry analysis. NC, no-targeting probe control. b, c RNA pull-down experiments using in vitro transcribed biotinylated lncLrrc55-AS or negative strand RNA (NS RNA) as a control to retrieve PME-1 from HEK293T cell extracts (b) or recombinant His-tagged PME-1 protein collections (c). RNA retrieval ratio of the RNA pull-down assay was assessed by RT-qPCR (b left). PME-1 retrieval by the RNA pull-down assay was assessed by western blot (b right and c). d RNA immunoprecipitation (RIP) experiments were performed with anti-PME-1 antibody in macrophages infected with SeV for 12 h. Immunoprecipitation of PME-1 was assessed by western blot. Co-purified RNAs were assessed by RT-qPCR analysis. e RIP experiments were performed with anti-PME-1 antibody in NIH/3T3 cells infected with VSV for 10 h. Immunoprecipitation of PME-1 and co-immunoprecipitation of PP2A-C were assessed by western blot. Co-purified RNA was assessed by RT-qPCR analysis. f RNA FISH-immunofluorescence detection of endogenous lncLrrc55-AS molecules and PME-1 in macrophages infected with SeV for 12 h. Scale bar, 10 µm. g RIP experiments were performed with anti-Flag antibody-coupled beads in NIH/3T3 cells transfected with full-length or truncated PME-1-Flag plasmids. LncLrrc55-AS retrieval was assessed by RT-qPCR. Schematic of PME-1 and truncated PME-1 used in the study (left). ΔC-arm, carboxyl-terminal arm (residues 371–385) truncated; ΔN-arm, amino-terminal arm (residues 1–64) truncated; ΔC&N-arm, residues 1–64 and 371–385 truncated; ΔCD, cap domain (residues 191–301) deleted; Δ107–113 aa, residues 107–113 deleted. Data are from three independent experiments (a, b left, d left, e, g means ± SEM) or are representative of three independent experiments with similar results (a, b, d right, c, e left, f). *P < 0.05, ***P < 0.005, ND, not detected (Student’s t-test or ANOVA)
Fig. 6
Fig. 6
LncLrrc55-AS inhibits PP2A-mediated negative regulation of IRF3-induced IFN-I production via PME-1. a RT-qPCR analysis of Ifna4 and Ifnb mRNA expression in non-target siRNA control (NC)- or PME-1-silenced peritoneal macrophages infected with VSV for 12 h. The silencing efficiency of three siRNAs targeting PME-1 was confirmed by western blot (right). b RT-qPCR analysis of Ifna4 and Ifnb mRNA expression in WT or PME-1 KO RAW264.7 cells infected with VSV for 12 h. c Determination of virus loads by TCID50 assay in the supernatant from VSV-infected WT or PME-1 KO RAW264.7 cells for 12 h. d Western blot analysis of phosphorylated (p-) or total proteins in lysates of WT and PME-1 KO RAW264.7 cells infected with VSV for the indicated hours. e RT-qPCR analysis of Ifna4 and Ifnb mRNA expression in NC- and PME-1-, lncLrrc55-AS- or double-silenced peritoneal macrophages infected with VSV for 12 h. f Western blot analysis of p-IRF3 or total IRF3 proteins in lysates of NC- or lncLrrc55-AS-silenced WT and PME-1 KO RAW264.7 cells infected with VSV for the indicated hours. g RT-qPCR analysis of Ifna4 and Ifnb mRNA expression in NC- and PP2A-C-, lncLrrc55-AS- or double-silenced peritoneal macrophages infected with VSV for 12 h. h RT-qPCR analysis of Ifna4 and Ifnb mRNA expression in NC- or lncLrrc55-AS-silenced macrophages pre-treated with Okadaol (50 nM) or DMSO 1 h before infection with VSV for 12 h. i Determination of virus loads by TCID50 in the supernatant from NC- or lncLrrc55-AS-silenced macrophages pre-treated with Okadaol (50 nM) or DMSO 1 h before infection with VSV for 12 h. Data are from three independent experiments (ac, e, gi, means ± SEM) or are representative of three independent experiments with similar results (d, f). *P < 0.05, **P < 0.01, ***P < 0.005; ns, no significance (Student’s t-test or ANOVA)
Fig. 7
Fig. 7
LncLrrc55-AS promotes the deactivation and demethylation of PP2A. a Phosphatase activity analysis of PP2A in lysates from non-target siRNA control (NC)- or PME-1-silenced macrophages infected with SeV for 12 h. b Phosphatase activity analysis of PP2A in lysates of NC- or lncLrrc55-AS-silenced macrophages infected with SeV for 12 h (left), and in WT or lncLrrc55-AS−/− macrophages infected with VSV for 12 h (right). c, d Western blot analysis of demethylated PP2A-C (dmPP2A-C) or total PP2A-C proteins in lysates of NC- and PME-1- or lncLrrc55-AS-silenced peritoneal macrophages with SeV infection for the indicated hours. e, f Western blot analysis of dmPP2A-C or total PP2A-C proteins in lysates from WT and lncLrrc55-AS KO NIH/3T3 cells (e), and from EV Ctrl- or lncLrrc55-AS-transfected lncLrrc55-AS KO NIH/3T3 cells (f) infected with VSV for the indicated hours. g, h Demethylation level by western blot analysis (g) and phosphatase activity assay (h) of recombinant PP2A-C proteins catalyzed by PME-1 in vitro, with increasing amount of lncLrrc55-AS added to the catalytic reaction system. The nlncLrrc55-AS: nPME-1 indicates the molar ratio of lncLrrc55-AS to PME-1. Data are from three independent experiments (a, b, h means ± SEM) or are representative of three independent experiments with similar results (cg). **P < 0.01, ***P < 0.005 (Student’s t-test)
Fig. 8
Fig. 8
LncLrrc55-AS promotes the interaction between PME-1 and PP2A. a RNA FISH-immunofluorescence detection of endogenous lncLrrc55-AS, PME-1, and PP2A-C in peritoneal macrophages from WT or lncLrrc55-AS−/− non-infected mice and mice infected with SeV for 12 h. Scale bars, 10 µm. b Quantitative analysis of colocalization rate for PME-1 and PP2A in NC- or lncLrrc55-AS-silenced peritoneal macrophages after SeV infection for 12 h. c Interaction between PME-1 and PP2A in RAW264.1 cells. NC- or lncLrrc55-AS-silenced cells were infected with VSV for the indicated hours, followed by immunoprecipitation with anti-PME-1 antibody. The immunoprecipitated PME-1 and PP2A were detected by western blot. d Interaction between PME-1 and PP2A in WT and lncLrrc55-AS−/− peritoneal macrophages infected with SeV for the indicated hours. Samples isolated from cells were equally divided into three parts. All parts were digested with Rnase A or Rnase T1 (10 U/mL), followed by immunoprecipitation with anti-PME-1 antibody (two parts) or with normal IgG antibody (one part; control). The immunoprecipitated PME-1 and PP2A were detected by western blot. e Interactions of PME-1 and PP2A with lncLrrc55-AS in an overexpression system. HEK293T cells were transfected with plasmids encoding PME-1-Flag, PP2A-C-Myc, and lncLrrc55-AS, followed by immunoprecipitation with anti-Flag antibody-coupled beads. The immunoprecipitated PME-1-Flag and PP2A-C-Myc were detected by western blot analysis. HEK293T cells transfected with empty vector plasmids were used as controls. Data are from three independent experiments (b, means ± SEM) or are representative of three independent experiments with similar results (a, ce). *P < 0.05 (Student’s t-test)

Comment in

Similar articles

Cited by

References

    1. McNab F, et al. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015;15:87–103. doi: 10.1038/nri3787. - DOI - PMC - PubMed
    1. Hoffmann HH, Schneider WM, Rice CM. Interferons and viruses: an evolutionary arms race of molecular interactions. Trends Immunol. 2015;36:124–138. doi: 10.1016/j.it.2015.01.004. - DOI - PMC - PubMed
    1. Wu J, Chen ZJ. Innate immune sensing and signaling of cytosolic nucleic acids. Annu. Rev. Immunol. 2014;32:461–488. doi: 10.1146/annurev-immunol-032713-120156. - DOI - PubMed
    1. Liu S, et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science. 2015;347:aaa2630. doi: 10.1126/science.aaa2630. - DOI - PubMed
    1. Liu J, Qian C, Cao X. Post-translational modification control of innate immunity. Immunity. 2016;45:15–30. doi: 10.1016/j.immuni.2016.06.020. - DOI - PubMed

Publication types

MeSH terms