Genetically programmed alternative splicing of NEMO mediates an autoinflammatory disease phenotype

Younglang Lee, Alex W Wessel, Jiazhi Xu, Julia G Reinke, Eries Lee, Somin M Kim, Amy P Hsu, Jevgenia Zilberman-Rudenko, Sha Cao, Clinton Enos, Stephen R Brooks, Zuoming Deng, Bin Lin, Adriana A de Jesus, Daniel N Hupalo, Daniela Gp Piotto, Maria T Terreri, Victoria R Dimitriades, Clifton L Dalgard, Steven M Holland, Raphaela Goldbach-Mansky, Richard M Siegel, Eric P Hanson, Younglang Lee, Alex W Wessel, Jiazhi Xu, Julia G Reinke, Eries Lee, Somin M Kim, Amy P Hsu, Jevgenia Zilberman-Rudenko, Sha Cao, Clinton Enos, Stephen R Brooks, Zuoming Deng, Bin Lin, Adriana A de Jesus, Daniel N Hupalo, Daniela Gp Piotto, Maria T Terreri, Victoria R Dimitriades, Clifton L Dalgard, Steven M Holland, Raphaela Goldbach-Mansky, Richard M Siegel, Eric P Hanson

Abstract

Host defense and inflammation are regulated by the NF-κB essential modulator (NEMO), a scaffolding protein with a broad immune cell and tissue expression profile. Hypomorphic mutations in inhibitor of NF-κB kinase regulatory subunit gamma (IKBKG) encoding NEMO typically present with immunodeficiency. Here, we characterized a pediatric autoinflammatory syndrome in 3 unrelated male patients with distinct X-linked IKBKG germline mutations that led to overexpression of a NEMO protein isoform lacking the domain encoded by exon 5 (NEMO-Δex5). This isoform failed to associate with TANK binding kinase 1 (TBK1), and dermal fibroblasts from affected patients activated NF-κB in response to TNF but not TLR3 or RIG-I-like receptor (RLR) stimulation when isoform levels were high. By contrast, T cells, monocytes, and macrophages that expressed NEMO-Δex5 exhibited increased NF-κB activation and IFN production, and blood cells from these patients expressed a strong IFN and NF-κB transcriptional signature. Immune cells and TNF-stimulated dermal fibroblasts upregulated the inducible IKK protein (IKKi) that was stabilized by NEMO-Δex5, promoting type I IFN induction and antiviral responses. These data revealed how IKBKG mutations that lead to alternative splicing of skipping exon 5 cause a clinical phenotype we have named NEMO deleted exon 5 autoinflammatory syndrome (NDAS), distinct from the immune deficiency syndrome resulting from loss-of-function IKBKG mutations.

Trial registration: ClinicalTrials.gov NCT00001788 NCT00001372 NCT02974595.

Keywords: Genetic diseases; Genetics; Immunology; Inflammation; Innate immunity; Signal transduction.

Conflict of interest statement

Conflict of interest: CE has served on an advisory board for Union Chimique Belge. RGM has received study support under government cooperative research and development agreements (CRADAs) from Swedish Orphan Biovitrum AB, Regeneron, Eli Lilly, and Novartis. RMS has been an employee of Novartis since 2018 and currently owns Novartis stock, though he worked for the NIH at the time he worked on this manuscript. EPH’s spouse receives income from Eli Lilly and holds unvested restricted stock options.

Figures

Figure 1. A synonymous exonic mutation and…
Figure 1. A synonymous exonic mutation and 2 intronic splice donor mutations mediate alternative splicing of IKBKG and lead to severe autoinflammatory disease.
(A) Pedigree and clinical features of developmental and inflammatory diseases in patients P1–P3. (B) Schema of mutations identified in patients P1–P3 and other NEMO mutants. Note exon coloring used for C and D. Patient P4 has the well-described NEMO-C417R zinc finger hypomorphic mutation. I DNA gel electrophoresis of IKBKG cDNA from P1, P2, P3, and healthy control (HC) dermal fibroblasts. Chromatograms of Sanger-sequenced FL isoforms from P1 and HCs and the variant isoform from P1–P3. (D) Schema of minigene spanning IKBKG exons 4–6 containing the reference sequence or patient mutations. DNA electrophoresis of cDNA and sequence traces are shown as In C. The 574 bp fragment contains noncoding intronic sequence in addition to a fragment of exon 5. WT = IKBKG minigene containing reference sequence. (E) Western blot of NEMO from patients P1–P3 and NEMO control patient skin fibroblasts, optical densitometry of FL-NEMO relative to loading control is shown in red, of NEMO-Δex5 in blue. (F) Western blot of PBMCs and whole cell lysate from patients P1, P2, and P3 and 2 healthy controls (HCs).
Figure 2. NEMO-Δex5 blocks NF-κB activation in…
Figure 2. NEMO-Δex5 blocks NF-κB activation in response to poly(I:C) but TNF-induced NF-κB remains intact.
(A) NEMO/TANK/TBK1 interaction and function. (B) HEK293T cells transfected with TANK, TBK1, and NEMO followed by TANK IP and Western blot as indicated. (C and D) Patient and healthy control (HC) dermal fibroblasts (DFs) were stimulated with poly(I:C) in a time course followed by Western blot as indicated. Quantitation of phospho-p65/IκBα by optical densitometry, below. Dashed line at 20%. (E) P1 and HC DFs were stimulated with TNF or poly(I:C) for 60 minutes and stained for NF-κB p65 visualized at 200× magnification by immunofluorescence (Keyence BZ9000), quantitation of p65 nuclear intensity, right. Statistical significance by 2-way ANOVA with Tukey’s multiple-comparison test. (F) DFs from HC and P1 were stably transduced with NEMO-Δex5 or FL-NEMO, respectively, and stimulated with poly(I:C) in a time course; lysates probed by Western blot as indicated. Different exposure intensity of NEMO panels (HC/EV, HC/Δex5, P1/EV, and P1/WT) was required to reveal all relevant bands, relative quantitation by optical densitometry, below. (G) P1 and HC DFs stimulated with poly(I:C), TNF, or cultured in media for 3 hours. Average gene expression by NanoString, ratio in stimulated/untreated from n = 3 independent experiments. (H) DFs treated as in G with relevant differentially expressed genes by RNA-Seq and totals shown, n = 3 technical replicates per media and TNF condition, n = 2 for poly(I:C) treatment. (I) P1 and HC DFs stimulated with TNF or poly(I:C) for 12 hours, cytokine production by capture bead assay, representative experiment of n = 3. (J) DFs from patients with NDAS, NEMO controls, and HCs incubated with hPIV3-GFP. Quantitation of total GFP intensity per cell, error bar indicates SEM. Two-way ANOVA applying Dunnett’s multiple-comparison test for adjusted P values, pertaining to 48-hour point. ***P < 0.001; ****P < 0.0001. Poly(I:C) dose 10 μg/mL and TNF 20 ng/mL throughout. All results representative of at least 2 independent experiments.
Figure 3. Increased NF-κB activation and type…
Figure 3. Increased NF-κB activation and type I IFN production in T cells and monocytes expressing NEMO-Δex5.
(A) Relative gene expression by whole-blood RNA-Seq from P1, P2, and P3 obtained at serial clinic visits; patients with systemic lupus erythematosus (SLE) with SLEDAI greater than 6 (SLE hi, n = 22); SLEDAI less than 6 (SLE lo, n = 76), and from healthy controls (n = 13). (B) IFN signature gene score (see Methods) from whole-blood RNA-Seq data, ordinary 1-way ANOVA, Dunnett’s correction for multiple comparisons, group mean indicated by red bar with SD. (C) IFN and NF-κB scores plotted versus NEMO-Δex5 splicing index, least squares linear regression fit, R2 shown. (D) P1 and HC PBMCs cultured in media or stimulated with poly(I:C) 10 μg/mL for 60 minutes and treated with brefeldin for 4 hours, stained with antibodies against IFN-α, IFN-β, p-STAT1, CD14, and CD4. (E) THP1 were stably transduced to express FL or mutant forms of NEMO. NF-κB p65 phosphorylation was measured by intracellular flow cytometry. (F) PMA-differentiated reconstituted THP1 cells were stimulated with LPS 10 ng/mL for 60 minutes, fixed, and stained with anti–NF-κB p65 (red) and nuclear dye (green). Scale bars: 70 μm. Areas of nuclear p65 overlap (pink) were quantitated (below). Means compared by 2-way ANOVA with Dunnett’s correction. (G) Secreted cytokines in cell culture supernatant from NEMO-reconstituted THP1 cells (black symbols) after 72-hour PMA differentiation (red). (H) P1, P4, and HC blasting T cells were infected with GFP-expressing human parainfluenza virus 3 (hPIV3-GFP) for 48 hours; n = 6, 2, and 10 replicates, respectively. Frequency of hPIV3-GFP+ cells, right. P value calculated with Mann-Whitney unpaired, 2-tailed U test. All data except from Figures 3A–C are representative of at least 2 independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001.
Figure 4. NEMO-Δex5 and IKKi form a…
Figure 4. NEMO-Δex5 and IKKi form a stable complex in response to poly(I:C) stimulation.
(A) THP1 cells reconstituted with FL and mutant NEMO forms and stimulated with poly(I:C) for 90 minutes or differentiated with PMA for 72 hours followed by Western blot of whole cell lysates. (B) HEK293T cells were transfected with FL or NEMO-Δex5 followed by stimulation with poly(I:C) 10 μg/mL for 8 hours and IP of IKKi using FLAG epitope. The membrane was probed for IKKi and NEMO with quantitation of pulled down IKKi normalized to NEMO on the right; means compared by paired, 2-tailed t test. (C) HC and P1 blasting T cells stimulated with poly(I:C) 10 μg/mL for 3 hours and IKKi isolated by IP. Western blot to detect co-immunoprecipitated NEMO, and IKKi blot as IP control, (right) quantitation of NEMO pulldown with IKKi by densitometry, black bars correspond to FL-NEMO bands and gray bars to NEMO-Δex5 forms in top panel (ImageJ). (D) Quantitation of total NEMO pulldown (NEMO-FL + NEMO-Δex5) as in C, shown as poly(I:C) response [ratio of poly(I:C) treated/media] (n = 4 independent IPs from NDAS-P1 and 4 IPs from HC T cells; means compared by paired, 2-tailed t test. Lines link paired data obtained from the same experiment. (E) NEMO-reconstituted THP1 cells in media or stimulated with LMW poly(I:C) 10 μg/mL for 60 minutes with NEMO and IKKi proximity ligation assay (PLA) spec intensity (x axis) and total NEMO/IKKi complex intensity (rainbow heatmap) using specific antibodies against IKKi and NEMO. (F) Four representative images taken at 40× original magnification of cells treated for 60 minutes as in E, with average signal area quantified; means compared by 2-way ANOVA with Tukey’s correction (G). EV, empty vector; FL, full length; no 1°, omission of primary antibody control. All results representative of at least 2 independent experiments. *P < 0.05; **P < 0.01.
Figure 5. IKKi can be induced by…
Figure 5. IKKi can be induced by TNF in hPIV3-infected P1 dermal fibroblasts, Ih rescues innate antiviral immunity.
(A) Venn diagram indicates the intersection of TNF-inducible, TLR3/RLR/IRF3 pathway genes that interact with NEMO, both directly (red) and indirectly (black). (B) IKKi expression in primary dermal fibroblasts from HCs in response to TNF 20 ng/mL for 24 hours or TNF 20 ng/mL, LPS 10 ng/mL, or viral infection for 72 hours detected by Western blot. TBK1 and TUBULIN served as loading controls. (C) P1 and HC skin fibroblasts were stimulated with poly(I:C) 10 μg/mL for 3 hours with or without TNF 20 ng/mL costimulation, and IFNB1 gene expression was determined by qPCR, 2-tailed Mann-Whitney U test. (D) P1, P4, and HC dermal fibroblasts were infected with hPIV3-GFP and treated with TNF 20 ng/mL as indicated by red shaded histograms. At 72 hours, cells were fixed and stained intracellularly with antibody to detect IKKi expression (quantitated on far right). (E) NDAS, NEMO control, and HC dermal fibroblasts (or iPSC-derived fibroblast-like cells from P5) were infected with hPIV3-GFP with or without TNF 20 ng/mL costimulation and imaged in time course experiments of 48 to 72 hours duration to measure virus protein expression. AUC of total GFP intensity per cell was measured and within-experiment normalization to HC samples was performed. For HC and NDAS-P1 samples, n = 7 independent experiments, for all other samples n = 3 independent experiments, with total numbers of conditions shown; “+/- TNF” in figure table underneath indicates mean hPIV3-GFP AUC ratio of TNF-stimulated versus unstimulated infected cells. Means compared by 2-way ANOVA with Tukey’s multiple-comparison test. (F) iPSC-derived fibroblast-like cells were transduced with lentiviral overexpression vector encoding IKKi-IRES-mCherry and infected with hPIV3-GFP. Scale bars: 400 μm. Two-way ANOVA with Bonferroni’s correction for multiple comparisons. Data are representative of at least 2 independent experiments. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

References

    1. Zhao T, et al. The NEMO adaptor bridges the nuclear factor-kappaB and interferon regulatory factor signaling pathways. Nat Immunol. 2007;8(6):592–600. doi: 10.1038/ni1465.
    1. Pomerantz JL, Baltimore D. NF-kappaB activation by a signaling complex containing TRAF2, TANK and TBK1, a novel IKK-related kinase. EMBO J. 1999;18(23):6694–6704. doi: 10.1093/emboj/18.23.6694.
    1. Clark K, et al. Novel cross-talk within the IKK family controls innate immunity. Biochem J. 2011;434(1):93–104. doi: 10.1042/BJ20101701.
    1. Bruni D, et al. A novel IRAK1-IKKε signaling axis limits the activation of TAK1-IKKβ downstream of TLR3. J Immunol. 2013;190(6):2844–2856. doi: 10.4049/jimmunol.1202042.
    1. Audry M, et al. NEMO is a key component of NF-κB- and IRF-3-dependent TLR3-mediated immunity to herpes simplex virus. J Allergy Clin Immunol. 2011;128(3):610–617. doi: 10.1016/j.jaci.2011.04.059.
    1. Rodero MP, Crow YJ. Type I interferon-mediated monogenic autoinflammation: the type I interferonopathies, a conceptual overview. J Exp Med. 2016;213(12):2527–2538. doi: 10.1084/jem.20161596.
    1. Thesleff I. The genetic basis of tooth development and dental defects. Am J Med Genet A. 2006;140(23):2530–2535.
    1. Smahi A, et al. Genomic rearrangement in NEMO impairs NF-kappaB activation and is a cause of incontinentia pigmenti. The International Incontinentia Pigmenti (IP) Consortium. Nature. 2000;405(6785):466–472. doi: 10.1038/35013114.
    1. Zonana J, et al. A novel X-linked disorder of immune deficiency and hypohidrotic ectodermal dysplasia is allelic to incontinentia pigmenti and due to mutations in IKK-gamma (NEMO) Am J Hum Genet. 2000;67(6):1555–1562. doi: 10.1086/316914.
    1. Klemann C, et al. Transplantation from a symptomatic carrier sister restores host defenses but does not prevent colitis in NEMO deficiency. Clin Immunol. 2016;164:52–56. doi: 10.1016/j.clim.2016.01.010.
    1. Pachlopnik Schmid J, et al. Transient hemophagocytosis with deficient cellular cytotoxicity, monoclonal immunoglobulin M gammopathy, increased T-cell numbers, and hypomorphic NEMO mutation. Pediatrics. 2006;117(5):e1049–1056. doi: 10.1542/peds.2005-2062.
    1. Zilberman-Rudenko J, et al. Recruitment of A20 by the C-terminal domain of NEMO suppresses NF-κB activation and autoinflammatory disease. Proc Natl Acad Sci U S A. 2016;113(6):1612–1617. doi: 10.1073/pnas.1518163113.
    1. Fusco F, et al. EDA-ID and IP, two faces of the same coin: how the same IKBKG/NEMO mutation affecting the NF-kappaB pathway can cause immunodeficiency and/or inflammation. Int Rev Immunol. 2015;34(6):445–459. doi: 10.3109/08830185.2015.1055331.
    1. Orstavik KH, et al. Novel splicing mutation in the NEMO (IKK-gamma) gene with severe immunodeficiency and heterogeneity of X-chromosome inactivation. Am J Med Genet A. 2006;140(1):31–39.
    1. Hai T, et al. An alternative splice product of IkappaB kinase (IKKgamma), IKKgamma-delta, differentially mediates cytokine and human T-cell leukemia virus type 1 tax-induced NF-kappaB activation. J Virol. 2006;80(9):4227–4241. doi: 10.1128/JVI.80.9.4227-4241.2006.
    1. Jorgensen SE, et al. Ectodermal dysplasia with immunodeficiency caused by a branch-point mutation in IKBKG/NEMO. J Allergy Clin Immunol. 2016;138(6):1706–1709. doi: 10.1016/j.jaci.2016.05.030.
    1. Zhang C, et al. RNA landscape of evolution for optimal exon and intron discrimination. Proc Natl Acad Sci U S A. 2008;105(15):5797–5802. doi: 10.1073/pnas.0801692105.
    1. Zhang XHF, et al. Exon inclusion is dependent on predictable exonic splicing enhancers. Mol Cell Biol. 2005;25(16):7323–7332. doi: 10.1128/MCB.25.16.7323-7332.2005.
    1. Schomacker H, et al. Pathogenesis of acute respiratory illness caused by human parainfluenza viruses. Curr Opin Virol. 2012;2(3):294–299. doi: 10.1016/j.coviro.2012.02.001.
    1. Zhang L, et al. Infection of ciliated cells by human parainfluenza virus type 3 in an in vitro model of human airway epithelium. J Virol. 2005;79(2):1113–1124. doi: 10.1128/JVI.79.2.1113-1124.2005.
    1. Sabbah A, Bose S. Retinoic acid inducible gene I activates innate antiviral response against human parainfluenza virus type 3. Virol J. 2009;6:200. doi: 10.1186/1743-422X-6-200.
    1. Gao D, et al. TLR3 controls constitutive IFN-β antiviral immunity in human fibroblasts and cortical neurons. J Clin Invest. 2021;131(1):134529.
    1. Bennett L, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003;197(6):711–723. doi: 10.1084/jem.20021553.
    1. de Jesus AA, et al. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J Clin Invest. 2020;130(4):1669–1682. doi: 10.1172/JCI129301.
    1. Fitzgerald KA, et al. IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway. Nat Immunol. 2003;4(5):491–496. doi: 10.1038/ni921.
    1. Tenoever BR, et al. Multiple functions of the IKK-related kinase IKKepsilon in interferon-mediated antiviral immunity. Science. 2007;315(5816):1274–1278. doi: 10.1126/science.1136567.
    1. Sharma S, et al. Triggering the interferon antiviral response through an IKK-related pathway. Science. 2003;300(5622):1148–1151. doi: 10.1126/science.1081315.
    1. Shimada T, et al. IKK-I, a novel lipopolysaccharide-inducible kinase that is related to IkappaB kinases. Int Immunol. 1999;11(8):1357–1362. doi: 10.1093/intimm/11.8.1357.
    1. Soderberg O, et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Methods. 2006;3(12):995–1000. doi: 10.1038/nmeth947.
    1. Avin A, et al. Quantitative analysis of protein-protein interactions and post-translational modifications in rare immune populations. Nat Commun. 2017;8(1):1524. doi: 10.1038/s41467-017-01808-6.
    1. Lu LL, et al. Select paramyxoviral V proteins inhibit IRF3 activation by acting as alternative substrates for inhibitor of kappaB kinase epsilon (IKKe)/TBK1. J Biol Chem. 2008;283(21):14269–14276. doi: 10.1074/jbc.M710089200.
    1. Balka KR, et al. TBK1 and IKKε act redundantly to mediate STING-induced NF-κB responses in myeloid cells. Cell Rep. 2020;31(1):107492. doi: 10.1016/j.celrep.2020.03.056.
    1. Canna SW, Goldbach-Mansky R. New monogenic autoinflammatory diseases--a clinical overview. Semin Immunopathol. 2015;37(4):387–394. doi: 10.1007/s00281-015-0493-5.
    1. Liu P, et al. Expression of an IKKg splice variant determines IRF3 and canonical NF-κB pathway utilization in ssRNA virus infection. PLoS One. 2009;4(11):e8079. doi: 10.1371/journal.pone.0008079.
    1. Sankar S, et al. IKK-I signals through IRF3 and NF kappa B to mediate the production of inflammatory cytokines. Cell Signal. 2006;18(7):982–993. doi: 10.1016/j.cellsig.2005.08.006.
    1. Zhang X, et al. IKKε negatively regulates RIG-I via direct phosphorylation. J Med Virol. 2016;88(4):712–718. doi: 10.1002/jmv.24376.
    1. Belgnaoui SM, et al. Linear ubiquitination of NEMO negatively regulates the interferon antiviral response through disruption of the MAVS-TRAF3 complex. Cell Host Microbe. 2012;12(2):211–222. doi: 10.1016/j.chom.2012.06.009.
    1. Vink PM, et al. In vivo knockdown of TAK1 accelerates bone marrow proliferation/differentiation and induces systemic inflammation. PLoS One. 2013;8(3):e57348. doi: 10.1371/journal.pone.0057348.
    1. Liu P, et al. Expression of an IKKgamma splice variant determines IRF3 and canonical NF-kappaB pathway utilization in ssRNA virus infection. PLoS One. 2009;4(11):e8079. doi: 10.1371/journal.pone.0008079.
    1. Emmerich CH, Cohen P. Optimising methods for the preservation, capture and identification of ubiquitin chains and ubiquitylated proteins by immunoblotting. Biochem Biophys Res Commun. 2015;466(1):1–14. doi: 10.1016/j.bbrc.2015.08.109.
    1. Xing J, et al. Identification of a role for TRIM29 in the control of innate immunity in the respiratory tract. Nat Immunol. 2016;17(12):1373–1380. doi: 10.1038/ni.3580.
    1. Zaph C, et al. Epithelial-cell-intrinsic IKK-beta expression regulates intestinal immune homeostasis. Nature. 2007;446(7135):552–556. doi: 10.1038/nature05590.
    1. Nenci A, et al. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature. 2007;446(7135):557–561. doi: 10.1038/nature05698.
    1. Haas TL, et al. Recruitment of the linear ubiquitin chain assembly complex stabilizes the TNF-R1 signaling complex and is required for TNF-mediated gene induction. Mol Cell. 2009;36(5):831–844. doi: 10.1016/j.molcel.2009.10.013.
    1. Laplantine E, et al. NEMO specifically recognizes K63-linked poly-ubiquitin chains through a new bipartite ubiquitin-binding domain. EMBO J. 2009;28(19):2885–2895. doi: 10.1038/emboj.2009.241.
    1. Mikkola ML. TNF superfamily in skin appendage development. Cytokine Growth Factor Rev. 2008;19(3-4):219–230. doi: 10.1016/j.cytogfr.2008.04.008.
    1. Zhang SY, et al. TLR3 deficiency in patients with herpes simplex encephalitis. Science. 2007;317(5844):1522–1527. doi: 10.1126/science.1139522.
    1. Reinert LS, et al. TLR3 deficiency renders astrocytes permissive to herpes simplex virus infection and facilitates establishment of CNS infection in mice. J Clin Invest. 2012;122(4):1368–1376. doi: 10.1172/JCI60893.
    1. Guo Y, et al. Herpes simplex virus encephalitis in a patient with complete TLR3 deficiency: TLR3 is otherwise redundant in protective immunity. J Exp Med. 2011;208(10):2083–2098. doi: 10.1084/jem.20101568.
    1. Rutsch F, et al. A specific IFIH1 gain-of-function mutation causes Singleton-Merten syndrome. Am J Hum Genet. 2015;96(2):275–282. doi: 10.1016/j.ajhg.2014.12.014.
    1. Jang MA, et al. Mutations in DDX58, Ih encodes RIG-I, cause atypical Singleton-Merten syndrome. Am J Hum Genet. 2015;96(2):266–274. doi: 10.1016/j.ajhg.2014.11.019.
    1. Tojima Y, et al. NAK is an IkappaB kinase-activating kinase. Nature. 2000;404(6779):778–782. doi: 10.1038/35008109.
    1. McWhirter SM, et al. IFN-regulatory factor 3-dependent gene expression is defective in Tbk1-deficient mouse embryonic fibroblasts. Proc Natl Acad Sci U S A. 2004;101(1):233–238. doi: 10.1073/pnas.2237236100.
    1. Wang J, et al. NF-kappa B RelA subunit is crucial for early IFN-beta expression and resistance to RNA virus replication. J Immunol. 2010;185(3):1720–1729. doi: 10.4049/jimmunol.1000114.
    1. Panne D, et al. Interferon regulatory factor 3 is regulated by a dual phosphorylation-dependent switch. J Biol Chem. 2007;282(31):22816–22822. doi: 10.1074/jbc.M703019200.
    1. Wessel AW, Hanson EP. A method for the quantitative analysis of stimulation-induced nuclear translocation of the p65 subunit of NF-κB from patient-derived dermal fibroblasts. Methods Mol Biol. 2015;1280:413–426.
    1. Desmet FO, et al. Human Splicing Finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res. 2009;37(9):e67. doi: 10.1093/nar/gkp215.

Source: PubMed

3
구독하다