Induction of GADD34 is necessary for dsRNA-dependent interferon-β production and participates in the control of Chikungunya virus infection

Giovanna Clavarino, Nuno Cláudio, Thérèse Couderc, Alexandre Dalet, Delphine Judith, Voahirana Camosseto, Enrico K Schmidt, Till Wenger, Marc Lecuit, Evelina Gatti, Philippe Pierre, Giovanna Clavarino, Nuno Cláudio, Thérèse Couderc, Alexandre Dalet, Delphine Judith, Voahirana Camosseto, Enrico K Schmidt, Till Wenger, Marc Lecuit, Evelina Gatti, Philippe Pierre

Abstract

Nucleic acid sensing by cells is a key feature of antiviral responses, which generally result in type-I Interferon production and tissue protection. However, detection of double-stranded RNAs in virus-infected cells promotes two concomitant and apparently conflicting events. The dsRNA-dependent protein kinase (PKR) phosphorylates translation initiation factor 2-alpha (eIF2α) and inhibits protein synthesis, whereas cytosolic DExD/H box RNA helicases induce expression of type I-IFN and other cytokines. We demonstrate that the phosphatase-1 cofactor, growth arrest and DNA damage-inducible protein 34 (GADD34/Ppp1r15a), an important component of the unfolded protein response (UPR), is absolutely required for type I-IFN and IL-6 production by mouse embryonic fibroblasts (MEFs) in response to dsRNA. GADD34 expression in MEFs is dependent on PKR activation, linking cytosolic microbial sensing with the ATF4 branch of the UPR. The importance of this link for anti-viral immunity is underlined by the extreme susceptibility of GADD34-deficient fibroblasts and neonate mice to Chikungunya virus infection.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Translation inhibition and IFN-β production…
Figure 1. Translation inhibition and IFN-β production are induced by poly I:C in MEFs.
A) Protein synthesis was monitored in poly I:C(pI:C)-stimulated MEFs using puromycin labelling followed by immunoblot with the anti-puromycin mAb 12D10. Controls are cells not treated with puromycin (No puro) and cells treated with cycloheximide (chx) 5 min prior puromycin incorporation. β-actin immunoblot is shown for equal loading control. Quantification of puromycin signal was quantified with ImageJ software and is represented above the immunoblot. Phosphorylation of eIF2α (P-eIF2α) was assessed in the same MEFs extracts. B) Immunofluorescence staining for puromycin, P-eIF2α and dsRNA of MEFs treated with poly I:C for 4 h and labeled with puromycin for 1 h. Scale bar, 10 µm. C) WT and PKR−/− MEFs were stimulated for 8 h with poly I:C (pI:C), thapsigargin (th) or arsenite (as). PKR and P-eIF2α were detected by immunoblot. D) WT and PKR−/− MEFs were stimulated for 8 h with poly I:C and protein synthesis was monitored like in (A). β-actin immunoblot is shown for equal loading control. E) IFN-β levels were measured, by ELISA, in cell culture supernatants of WT, PKR−/−, eIF2αA/A and control eIF2αS/S MEFs after 4 and 8 h of poly I:C stimulation. Data are mean ± standard deviation of 3 independent experiments. F) Protein synthesis was measured in NIH3T3 cells by puromycin incorporation after 7 h of poly I:C treatment. Where indicated, a chase of 1 h with fresh media was performed prior to puromycin labeling and immunoblotting. Samples with cycloheximide (chx) and arsenite (as) added respectively 5 min and 30 min before the puromycin pulse are shown as controls. G) IFN-β was quantified by ELISA in culture supernatants in the conditions described above after 7 h of poly I:C stimulation or 7 h of poly I:C stimulation followed by 1 h with fresh media (chase). Data are mean ± standard deviation of 4 independent experiments.
Figure 2. PKR is required for ATF4…
Figure 2. PKR is required for ATF4 and GADD34 expression in response to poly I:C.
A) WT and PKR−/− MEFs were stimulated for 8 h with poly I:C (pI:C), or the UPR-inducing drug, thapsigargin (th) for 6 h. ATF4 protein expression was detected by immunoblot on nuclear extracts. Nuclear HDAC1 immunoblot is shown for equal loading control. * indicates unspecific band. B) GADD34 mRNA levels were quantified by qPCR after 6 h of poly I:C (pI:C) treatment in WT and PKR−/− MEFs. For the same cell extracts, immunoblots of GADD34 (middle panel), PKR and P-eIF2α (right panel) were performed. C) The same analysis was performed in eIF2α A/A and control eIF2α S/S MEFs. Treatment with thapsigargin (th), for 6 h was used as control to induce GADD34 and P-eIF2α. eIF2α and β-actin immunoblots are shown for equal loading control. Quantitative PCR data are the mean ± standard deviation of 3 independent experiments.
Figure 3. GADD34 mediates eIF2α dephosphorylation but…
Figure 3. GADD34 mediates eIF2α dephosphorylation but not global translation recovery in response to poly I:C.
A) After treatment with poly I:C, protein extracts of WT and GADD34ΔC/ΔC MEFs were immunobloted for GADD34 and P-eIF2α. B) Protein synthesis was analyzed in WT cells treated for 1 to 6 hours with poly I:C (pI:C) alone or together with thapsigargin (th). Controls are cells not treated with puromycin (co) and cells treated with cycloheximide (chx) 5 min before puromycin incorporation. C) Protein synthesis was analyzed in GADD34ΔC/ΔC cells treated for 1 to 6 hours with poly I:C (pI:C) alone or together with thapsigargin (th). Tubulin or β-actin immunoblot are shown for equal loading control. In GADD34ΔC/ΔC cells translation is strongly impacted by thapsigargin, but not poly I:C.
Figure 4. GADD34 is required for cytokine…
Figure 4. GADD34 is required for cytokine production in poly I:C-stimulated MEFs.
A) Left Panel, immunoblot for cystatin C after treatment or not with brefeldin A (BFA) in poly I:C-stimulated WT MEFs. Arrow indicates N-glycosylated-Cystatin C. Right panel, WT and GADD34ΔC/ΔC MEFs were treated with poly I:C (pI:C), thapsigargin (th) or tunicamycin (tun) for the indicated times. Levels of GADD34 and Cystatin C (CysC) were examined by immunoblot. β-actin immunoblot is shown as equal loading control. B) Immunoblots for GADD34 and PKR in WT and GADD34-inactivated cells treated with poly I:C for the indicated periods of time. The UPR-inducing drugs, Thapsigargin (th) and tunicamycin (tun), were used as controls to induce GADD34. Immunoblot of tubulin is shown as equal loading control. C) Amount of IFN-β (left panel) and IL-6 (right panel) in cell culture supernatants of WT and GADD34ΔC/ΔC MEFs after 6 h of poly I:C stimulation. Mock are samples treated with lipofectamine alone. Data are mean ± standard deviation of five (IFN-β) and three (IL-6) independent experiments. D) Transcription of IFN-β, IL-6, PKR and Cystatin C was analyzed by qPCR in samples of WT and GADD34ΔC/ΔC MEFs treated with poly I:C (pI:C). Mock represent samples treated with lipofectamine alone. E) WT and GADD34ΔC/ΔC MEFs were transfected overnight with an expression plasmid carrying the murine GADD34 (G34) cDNA and then treated with poly I:C for 6 h. IFN-β production was quantified by ELISA, left panel, in cell culture supernatants and plotted as a ratio of IFN-β to total cell proteins to compensate for different cell mortality levels induced by the transfection. In the right panel immunoblots for GADD34 and P-eIF2α in the same experimental conditions are shown. One representative analysis of 3 independent experiments is shown.
Figure 5. PKR is required to control…
Figure 5. PKR is required to control CHIKV infection and IFN-β production in MEFs.
A) WT and PKR−/− MEFs were infected with CHIKV-GFP at an MOI of 10 or 50, for 24 h and 48 h. The amount of infected cells was determined by GFP expression, left panel. Interferon β present in the cell culture supernatants was measured by ELISA, right panel. Data represented are mean ± standard deviation from 3 experiments. B) WT (top panel) and PKR−/− MEFs (bottom panel) were infected for 24 h with CHIKV-GFP, then labeled with puromycin for 1 h prior fixation. GFP-CHIKV positive (green) were visualized by confocal microscopy after staining with specific antibodies for puromycin (cyan) and phospho-eIF2α (red). Cell Nuclei are stained with Hoechst 33258 (blue). Infection by CHIKV inhibits protein synthesis (visualized by puromycin incorporation) in WT, but not in PKR-deficient cells (arrows). In WT MEFs, eIF2α phosphorylation levels correlate with translation inhibition, although variability is observed among different infected cells, presumably due to GADD34 activity and time of infection. Non-infected WT and PKR−/− cells serve as a reference for normal translation activity and are indicated by an arrowhead. Scale bar 10 µm.
Figure 6. CHIKV infection and IFN-β production…
Figure 6. CHIKV infection and IFN-β production are controlled by GADD34 in MEFs.
A) WT and GADD34ΔC/ΔC cells were infected with CHIKV-GFP for a period of 24 and 48 h. The percentage of infected, GFP positive, cells and viral titers were analyzed. B) Levels of IFN-β in cell culture supernatants of WT and GADD34ΔC/ΔC MEFs infected with CHIKV-GFP for 24 h and 48 h. C) Murine IFN-β was added 3 h before infection of WT and GADD34ΔC/ΔC MEFs with CHIKV-GFP (10 MOI). Productive infection was estimated by GFP expression 24 h after CHIKV exposure. D) Cells were treated with guanabenz 3 h before and during infection (24 h) with CHIKV-GFP, percentage of infected cells (left) and corresponding IFN-β production (middle), are shown. Immunoblot of GADD34 and P-eIF2α are shown on the right. NI stands for non-infected. Percentage of infected cells, viral titers and IFN-β measurements data represent mean ± standard deviation of 3 experiments. p values shown in (D) were obtained applying a Student's t test.
Figure 7. CHIKV infection in mouse neonates.
Figure 7. CHIKV infection in mouse neonates.
A) Kaplan–Meier plots representing the survival of FVB (WT) and GADD34ΔC/ΔC mouse neonates 9-day-old (n = 11 per group) (upper panel) or 12-day-old (n = 14 per group) (lower panel) after intradermal inoculation with 106 PFU of CHIKV and observed for 21 days. B) Left panel, viral titers in different tissues and serum of 12-day-old mice inoculated with 106 PFU of CHIKV via the intradermal route. Mice were sacrificed 5 days after infection and the amount of infectious virus in serum and tissues quantified by TCID50 (see methods) (n = 5). In addition of considerably increased levels of viral replication in CHIKV target tissues, GADD34ΔC/ΔC neonates also display signs of heart infection. Right panel, Quantification of IFN-β for the same different tissues, CHIKV-infected target tissues of GADD34ΔC/ΔC mice produced less IFN-β than WT. C) 17-day-old mice were infected with 106 PFU of CHIKV via the intradermal route, and sacrificed 72 h later. Quantification of viral titers and IFN-β/viral titers ratio is presented for different tissues. A broken line indicates the detection threshold. In B and C represented data are arithmetic mean ± standard deviation, n = 5. In B and Cp values were calculated using a Student's t test, *p≤0.1, **p≤0.05.
Figure 8. CHIKV infection causes severe myocarditis…
Figure 8. CHIKV infection causes severe myocarditis in mouse neonates.
Histological appearance of horizontal sections of the heart through left and right ventricles of 12-day FVB (A, C and E) and GADD34ΔC/ΔC mice at D5 pi (B, D and F). Normal appearance of heart of FVB infected mice, at low magnification (A, ×10) with normal cardiomyocytes (C, ×100) and exceptional small foci of lymphocytes (E, ×400). Numerous foci of necrosis in the heart of GADD34ΔC/ΔC infected mice, at low magnification (B, ×10) and extensive through the ventricular wall (D, ×100). Higher magnification shows few residual cardiomyocytes (arrow head) and inflammation mainly composed of monocytes as well as extensive deposition of calcium (F, ×400). The mice were inoculated with 106 PFU of CHIKV via the intradermal route.

References

    1. Kawai T, Akira S. Innate immune recognition of viral infection. Nat Immunol. 2006;7:131–137.
    1. Kawai T, Akira S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int Immunol. 2009;21:317–337.
    1. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801.
    1. Garcia MA, Meurs EF, Esteban M. The dsRNA protein kinase PKR: virus and cell control. Biochimie. 2007;89:799–811.
    1. Malathi K, Dong B, Gale M, Jr, Silverman RH. Small self-RNA generated by RNase L amplifies antiviral innate immunity. Nature. 2007;448:816–819.
    1. Gitlin L, Barchet W, Gilfillan S, Cella M, Beutler B, et al. Essential role of mda-5 in type I IFN responses to polyriboinosinic:polyribocytidylic acid and encephalomyocarditis picornavirus. Proc Natl Acad Sci U S A. 2006;103:8459–8464.
    1. Zhang Z, Kim T, Bao M, Facchinetti V, Jung SY, et al. DDX1, DDX21, and DHX36 helicases form a complex with the adaptor molecule TRIF to sense dsRNA in dendritic cells. Immunity. 2011;34:866–878.
    1. Friedman RM. Role of Interferon in Viral Interference. Nature. 1964;201:848–849.
    1. Wagner RR. Inhibition of Interferon Biosynthesis by Actinomycin D. Nature. 1964;204:49–51.
    1. Schilte C, Couderc T, Chretien F, Sourisseau M, Gangneux N, et al. Type I IFN controls chikungunya virus via its action on nonhematopoietic cells. J Exp Med. 2010;207:429–442.
    1. Deuber SA, Pavlovic J. Virulence of a mouse-adapted Semliki Forest virus strain is associated with reduced susceptibility to interferon. J Gen Virol. 2007;88:1952–1959.
    1. Despres P, Griffin JW, Griffin DE. Antiviral activity of alpha interferon in Sindbis virus-infected cells is restored by anti-E2 monoclonal antibody treatment. J Virol. 1995;69:7345–7348.
    1. Burke CW, Gardner CL, Steffan JJ, Ryman KD, Klimstra WB. Characteristics of alpha/beta interferon induction after infection of murine fibroblasts with wild-type and mutant alphaviruses. Virology. 2009;395:121–132.
    1. Frolova EI, Fayzulin RZ, Cook SH, Griffin DE, Rice CM, et al. Roles of nonstructural protein nsP2 and Alpha/Beta interferons in determining the outcome of Sindbis virus infection. J Virol. 2002;76:11254–11264.
    1. White LK, Sali T, Alvarado D, Gatti E, Pierre P, et al. Chikungunya virus induces IPS-1-dependent innate immune activation and protein kinase R-independent translational shutoff. J Virol. 2011;85:606–620.
    1. Aguilar PV, Weaver SC, Basler CF. Capsid protein of eastern equine encephalitis virus inhibits host cell gene expression. J Virol. 2007;81:3866–3876.
    1. Her Z, Malleret B, Chan M, Ong EK, Wong SC, et al. Active infection of human blood monocytes by Chikungunya virus triggers an innate immune response. J Immunol. 2010;184:5903–5913.
    1. Proud CG. PKR: a new name and new roles. Trends Biochem Sci. 1995;20:241–246.
    1. Meurs E, Chong K, Galabru J, Thomas NS, Kerr IM, et al. Molecular cloning and characterization of the human double-stranded RNA-activated protein kinase induced by interferon. Cell. 1990;62:379–390.
    1. Williams BR. PKR; a sentinel kinase for cellular stress. Oncogene. 1999;18:6112–6120.
    1. Williams BR. Signal integration via PKR. Sci STKE. 2001;2001:re2.
    1. Balachandran S, Roberts PC, Brown LE, Truong H, Pattnaik AK, et al. Essential role for the dsRNA-dependent protein kinase PKR in innate immunity to viral infection. Immunity. 2000;13:129–141.
    1. Diebold SS, Montoya M, Unger H, Alexopoulou L, Roy P, et al. Viral infection switches non-plasmacytoid dendritic cells into high interferon producers. Nature. 2003;424:324–328.
    1. Langland JO, Cameron JM, Heck MC, Jancovich JK, Jacobs BL. Inhibition of PKR by RNA and DNA viruses. Virus Res. 2006;119:100–110.
    1. Domingo-Gil E, Toribio R, Najera JL, Esteban M, Ventoso I. Diversity in viral anti-PKR mechanisms: a remarkable case of evolutionary convergence. PLoS One. 2011;6:e16711.
    1. Ventoso I, Sanz MA, Molina S, Berlanga JJ, Carrasco L, et al. Translational resistance of late alphavirus mRNA to eIF2alpha phosphorylation: a strategy to overcome the antiviral effect of protein kinase PKR. Genes Dev. 2006;20:87–100.
    1. Pichlmair A, Reis e Sousa C. Innate recognition of viruses. Immunity. 2007;27:370–383.
    1. Takeuchi O, Akira S. MDA5/RIG-I and virus recognition. Curr Opin Immunol. 2008;20:17–22.
    1. McAllister CS, Samuel CE. The RNA-activated protein kinase enhances the induction of interferon-beta and apoptosis mediated by cytoplasmic RNA sensors. J Biol Chem. 2009;284:1644–1651.
    1. Yang YL, Reis LF, Pavlovic J, Aguzzi A, Schafer R, et al. Deficient signaling in mice devoid of double-stranded RNA-dependent protein kinase. Embo J. 1995;14:6095–6106.
    1. Schulz O, Pichlmair A, Rehwinkel J, Rogers NC, Scheuner D, et al. Protein kinase R contributes to immunity against specific viruses by regulating interferon mRNA integrity. Cell Host Microbe. 2010;7:354–361.
    1. Barry G, Breakwell L, Fragkoudis R, Attarzadeh-Yazdi G, Rodriguez-Andres J, et al. PKR acts early in infection to suppress Semliki Forest virus production and strongly enhances the type I interferon response. J Gen Virol. 2009;90:1382–1391.
    1. Gilfoy FD, Mason PW. West Nile virus-induced interferon production is mediated by the double-stranded RNA-dependent protein kinase PKR. J Virol. 2007;81:11148–11158.
    1. Smith EJ, Marie I, Prakash A, Garcia-Sastre A, Levy DE. IRF3 and IRF7 phosphorylation in virus-infected cells does not require double-stranded RNA-dependent protein kinase R or Ikappa B kinase but is blocked by Vaccinia virus E3L protein. J Biol Chem. 2001;276:8951–8957.
    1. Samuel MA, Whitby K, Keller BC, Marri A, Barchet W, et al. PKR and RNase L contribute to protection against lethal West Nile Virus infection by controlling early viral spread in the periphery and replication in neurons. J Virol. 2006;80:7009–7019.
    1. Ryman KD, White LJ, Johnston RE, Klimstra WB. Effects of PKR/RNase L-dependent and alternative antiviral pathways on alphavirus replication and pathogenesis. Viral Immunol. 2002;15:53–76.
    1. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 2003;11:619–633.
    1. Harding HP, Zhang Y, Bertolotti A, Zeng H, Ron D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol Cell. 2000;5:897–904.
    1. Zhang P, McGrath BC, Reinert J, Olsen DS, Lei L, et al. The GCN2 eIF2alpha kinase is required for adaptation to amino acid deprivation in mice. Mol Cell Biol. 2002;22:6681–6688.
    1. Berlanga JJ, Ventoso I, Harding HP, Deng J, Ron D, et al. Antiviral effect of the mammalian translation initiation factor 2alpha kinase GCN2 against RNA viruses. EMBO J. 2006;25:1730–1740.
    1. Chen JJ, London IM. Regulation of protein synthesis by heme-regulated eIF-2 alpha kinase. Trends Biochem Sci. 1995;20:105–108.
    1. Lu L, Han AP, Chen JJ. Translation initiation control by heme-regulated eukaryotic initiation factor 2alpha kinase in erythroid cells under cytoplasmic stresses. Mol Cell Biol. 2001;21:7971–7980.
    1. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–529.
    1. Zhang K, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature. 2008;454:455–462.
    1. Randazzo BP, Tal-Singer R, Zabolotny JM, Kesari S, Fraser NW. Herpes simplex virus 1716, an ICP 34.5 null mutant, is unable to replicate in CV-1 cells due to a translational block that can be overcome by coinfection with SV40. J Gen Virol. 1997;78(Pt 12):3333–3339.
    1. Cheng G, Feng Z, He B. Herpes simplex virus 1 infection activates the endoplasmic reticulum resident kinase PERK and mediates eIF-2alpha dephosphorylation by the gamma(1)34.5 protein. J Virol. 2005;79:1379–1388.
    1. Schmidt EK, Clavarino G, Ceppi M, Pierre P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat Methods. 2009;6:275–277.
    1. Connor JH, Weiser DC, Li S, Hallenbeck JM, Shenolikar S. Growth arrest and DNA damage-inducible protein GADD34 assembles a novel signaling complex containing protein phosphatase 1 and inhibitor 1. Mol Cell Biol. 2001;21:6841–6850.
    1. Lu PD, Harding HP, Ron D. Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response. J Cell Biol. 2004;167:27–33.
    1. Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004;18:3066–3077.
    1. Novoa I, Zeng H, Harding HP, Ron D. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2alpha. J Cell Biol. 2001;153:1011–1022.
    1. Novoa I, Zhang Y, Zeng H, Jungreis R, Harding HP, et al. Stress-induced gene expression requires programmed recovery from translational repression. EMBO J. 2003;22:1180–1187.
    1. Ma Y, Hendershot LM. Delineation of a negative feedback regulatory loop that controls protein translation during endoplasmic reticulum stress. J Biol Chem. 2003;278:34864–34873.
    1. Kojima E, Takeuchi A, Haneda M, Yagi A, Hasegawa T, et al. The function of GADD34 is a recovery from a shutoff of protein synthesis induced by ER stress: elucidation by GADD34-deficient mice. FASEB J. 2003;17:1573–1575.
    1. Cappello F, Gatti E, Camossetto V, David A, Lelouard H, et al. Cystatin F is secreted, but artificial modification of its C-terminus can induce its endocytic targeting. Exp Cell Res. 2004;297:607–618.
    1. Bernasconi R, Molinari M. ERAD and ERAD tuning: disposal of cargo and of ERAD regulators from the mammalian ER. Curr Opin Cell Biol. 2011;23:176–183.
    1. Harding HP, Zhang Y, Scheuner D, Chen JJ, Kaufman RJ, et al. Ppp1r15 gene knockout reveals an essential role for translation initiation factor 2 alpha (eIF2alpha) dephosphorylation in mammalian development. Proc Natl Acad Sci U S A. 2009;106:1832–1837.
    1. Tsaytler P, Harding HP, Ron D, Bertolotti A. Selective inhibition of a regulatory subunit of protein phosphatase 1 restores proteostasis. Science. 2011;332:91–94.
    1. Couderc T, Chretien F, Schilte C, Disson O, Brigitte M, et al. A mouse model for Chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease. PLoS Pathog. 2008;4:e29.
    1. Clavarino G, Claudio N, Dalet A, Terawaki S, Couderc T, et al. Protein phosphatase 1 subunit Ppp1r15a/GADD34 regulates cytokine production in polyinosinic:polycytidylic acid-stimulated dendritic cells. Proc Natl Acad Sci U S A. 2012;91:3006–3011.
    1. Patterson AD, Hollander MC, Miller GF, Fornace AJ., Jr Gadd34 requirement for normal hemoglobin synthesis. Mol Cell Biol. 2006;26:1644–1653.
    1. Wang X, Liao Y, Yap PL, Png KJ, Tam JP, et al. Inhibition of protein kinase R activation and upregulation of GADD34 expression play a synergistic role in facilitating coronavirus replication by maintaining de novo protein synthesis in virus-infected cells. J Virol. 2009;83:12462–12472.
    1. Leber JH, Crimmins GT, Raghavan S, Meyer-Morse NP, Cox JS, et al. Distinct TLR- and NLR-mediated transcriptional responses to an intracellular pathogen. PLoS Pathog. 2008;4:e6.
    1. Lelouard H, Schmidt EK, Camosseto V, Clavarino G, Ceppi M, et al. Regulation of translation is required for dendritic cell function and survival during activation. J Cell Biol. 2007;179:1427–1439.
    1. Boyce M, Py BF, Ryazanov AG, Minden JS, Long K, et al. A pharmacoproteomic approach implicates eukaryotic elongation factor 2 kinase in ER stress-induced cell death. Cell Death Differ. 2008;15:589–599.
    1. Abraham N, Stojdl DF, Duncan PI, Methot N, Ishii T, et al. Characterization of transgenic mice with targeted disruption of the catalytic domain of the double-stranded RNA-dependent protein kinase, PKR. J Biol Chem. 1999;274:5953–5962.
    1. Baltzis D, Li S, Koromilas AE. Functional characterization of pkr gene products expressed in cells from mice with a targeted deletion of the N terminus or C terminus domain of PKR. J Biol Chem. 2002;277:38364–38372.
    1. Vanlandingham DL, Tsetsarkin K, Hong C, Klingler K, McElroy KL, et al. Development and characterization of a double subgenomic chikungunya virus infectious clone to express heterologous genes in Aedes aegypti mosquitoes. Insect Biochem Mol Biol. 2005;35:1162–1170.
    1. Schuffenecker I, Iteman I, Michault A, Murri S, Frangeul L, et al. Genome microevolution of chikungunya viruses causing the Indian Ocean outbreak. PLoS Med. 2006;3:e263.
    1. Todd DJ, Lee AH, Glimcher LH. The endoplasmic reticulum stress response in immunity and autoimmunity. Nat Rev Immunol. 2008;8:663–674.
    1. Martinon F, Chen X, Lee AH, Glimcher LH. TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nat Immunol. 2010;11:411–418.
    1. Dalod M, Pierre P. Integration of ER stress and viral nucleotide sensing in DCs: Mounting a response commensurate to the threat? Eur J Immunol. 2011;41:898–901.

Source: PubMed

3
Abonneren