STING Is Involved in Antiviral Immune Response against VZV Infection via the Induction of Type I and III IFN Pathways  Ji-Ae Kim, Seul-Ki Park, Seong-Wook.

Slides:



Advertisements
Similar presentations
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Advertisements

Up-Regulation of Activating Transcription Factor-5 Suppresses SAP Expression to Activate T Cells in Hemophagocytic Syndrome Associated with Epstein-Barr.
Ganglioside GM3 Mediates Glucose-Induced Suppression of IGF-1 Receptor–Rac1 Activation to Inhibit Keratinocyte Motility  Duncan Hieu M. Dam, Xiao-Qi Wang,
Juewon Kim, Hyunjung Choi, Eun-Gyung Cho, Tae R. Lee 
Volume 28, Issue 5, Pages (May 2008)
IFN-γ Primes Keratinocytes for HSV-1–Induced Inflammasome Activation
A Signal Transduction Pathway from TGF-β1 to SKP2 via Akt1 and c-Myc and its Correlation with Progression in Human Melanoma  Xuan Qu, Liangliang Shen,
Ganglioside GM3 Mediates Glucose-Induced Suppression of IGF-1 Receptor–Rac1 Activation to Inhibit Keratinocyte Motility  Duncan Hieu M. Dam, Xiao-Qi Wang,
MiR-29 Regulates Type VII Collagen in Recessive Dystrophic Epidermolysis Bullosa  Michael Vanden Oever, Daniel Muldoon, Wendy Mathews, Ron McElmurry, Jakub.
Interferon (IFN)-Induced Protein 35 (IFI35), a Type I Interferon-Dependent Transcript, Upregulates Inflammatory Signaling Pathways by Activating Toll-Like.
IL-1β–Induced Protection of Keratinocytes against Staphylococcus aureus-Secreted Proteases Is Mediated by Human β-Defensin 2  Bingjie Wang, Brian J. McHugh,
TWEAK/Fn14 Activation Contributes to the Pathogenesis of Bullous Pemphigoid  Yale Liu, Lingling Peng, Liang Li, Chengfei Liu, Xiao Hu, Shengxiang Xiao,
MiR-29 Regulates Type VII Collagen in Recessive Dystrophic Epidermolysis Bullosa  Michael Vanden Oever, Daniel Muldoon, Wendy Mathews, Ron McElmurry, Jakub.
Requirement of Gαi1/3–Gab1 Signaling Complex for Keratinocyte Growth Factor– Induced PI3K–AKT–mTORC1 Activation  Yi-ming Zhang, Zhi-qing Zhang, Yuan-yuan.
Absence of γ-Chain in Keratinocytes Alters Chemokine Secretion, Resulting in Reduced Immune Cell Recruitment  Karolin Nowak, Daniela Linzner, Adrian J.
Volume 142, Issue 4, Pages (April 2012)
Development of Cell-Penetrating Asymmetric Interfering RNA Targeting Connective Tissue Growth Factor  Jihye Hwang, Chanil Chang, Ji Hyun Kim, Chang Taek.
TSLP Down-Regulates S100A7 and ß-Defensin 2 Via the JAK2/STAT3-Dependent Mechanism  Hana Lee, Woo-In Ryu, Hee Joo Kim, Hyun Cheol Bae, Hwa Jung Ryu, Jung.
Volume 21, Issue 2, Pages (February 2017)
Characterization of TNF-α– and IL-17A–Mediated Synergistic Induction of DEFB4 Gene Expression in Human Keratinocytes through IκBζ  Claus Johansen, Trine.
Requirement of Zinc Transporter SLC39A7/ZIP7 for Dermal Development to Fine-Tune Endoplasmic Reticulum Function by Regulating Protein Disulfide Isomerase 
Generic and Personalized RNAi-Based Therapeutics for a Dominant-Negative Epidermal Fragility Disorder  Deena M. Leslie Pedrioli, Dun Jack Fu, Emilio Gonzalez-Gonzalez,
TIEG1 Represses Smad7-Mediated Activation of TGF-β1/Smad Signaling in Keloid Pathogenesis  Zhi-Cheng Hu, Fen Shi, Peng Liu, Jian Zhang, Dong Guo, Xiao-Ling.
STING Is Involved in Antiviral Immune Response against VZV Infection via the Induction of Type I and III IFN Pathways  Ji-Ae Kim, Seul-Ki Park, Seong-Wook.
Interferon-γ Protects from Staphylococcal Alpha Toxin-Induced Keratinocyte Death through Apolipoprotein L1  Anne M. Brauweiler, Elena Goleva, Donald Y.M.
IL-1β–Induced Protection of Keratinocytes against Staphylococcus aureus-Secreted Proteases Is Mediated by Human β-Defensin 2  Bingjie Wang, Brian J. McHugh,
Expression and Function of RIG-I in Oral Keratinocytes and Fibroblasts
Volume 11, Issue 6, Pages (June 2012)
Nicastrin/miR-30a-3p/RAB31 Axis Regulates Keratinocyte Differentiation by Impairing EGFR Signaling in Familial Acne Inversa  Yanyan He, Haoxiang Xu, Chengrang.
Volume 71, Issue 5, Pages e5 (September 2018)
IL-4 Inhibits the Melanogenesis of Normal Human Melanocytes through the JAK2– STAT6 Signaling Pathway  Hyun Choi, Hyunjung Choi, Jiyeon Han, Sun Hee Jin,
Inhibition of DNA Methylation in the COL1A2 Promoter by Anacardic Acid Prevents UV- Induced Decrease of Type I Procollagen Expression  Min-Kyoung Kim,
Histamine Contributes to Tissue Remodeling via Periostin Expression
IGF-II-Mediated COX-2 Gene Expression in Human Keratinocytes Through Extracellular Signal-Regulated Kinase Pathway  Hye Jung Kim, Tae-Yoon Kim  Journal.
Tomoyasu Hattori, Lukasz Stawski, Sashidhar S
Volume 8, Issue 6, Pages (December 2010)
Volume 18, Issue 2, Pages (August 2015)
TGFβ/SMAD/microRNA-486-3p Signaling Axis Mediates Keratin 17 Expression and Keratinocyte Hyperproliferation in Psoriasis  Man Jiang, Zhongbin Sun, Erle.
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
GPR56/ADGRG1 Activation Promotes Melanoma Cell Migration via NTF Dissociation and CTF-Mediated Gα12/13/RhoA Signaling  Nien-Yi Chiang, Yen-Ming Peng,
Volume 24, Issue 11, Pages e3 (September 2018)
Cyclic Dinucleotides Trigger ULK1 (ATG1) Phosphorylation of STING to Prevent Sustained Innate Immune Signaling  Hiroyasu Konno, Keiko Konno, Glen N. Barber 
Volume 11, Issue 8, Pages (May 2015)
Amadeus S. Zhu, Ang Li, Tabetha S. Ratliff, Martha Melsom, Luis A
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
Upregulation of Tenascin-C Expression by IL-13 in Human Dermal Fibroblasts via the Phosphoinositide 3-kinase/Akt and the Protein Kinase C Signaling Pathways 
TET3 Inhibits Type I IFN Production Independent of DNA Demethylation
Cyclooxygenase-2 Inhibitor Enhances Whereas Prostaglandin E2Inhibits the Production of Interferon-Induced Protein of 10 kDa in Epidermoid Carcinoma A431 
All-Trans-Retinoic Acid Induces Interleukin-8 via the Nuclear Factor-κB and p38 Mitogen-Activated Protein Kinase Pathways in Normal Human Keratinocytes 
Nrf2 Promotes Keratinocyte Proliferation in Psoriasis through Up-Regulation of Keratin 6, Keratin 16, and Keratin 17  Luting Yang, Xueli Fan, Tingting.
Reduced Expression of Connective Tissue Growth Factor (CTGF/CCN2) Mediates Collagen Loss in Chronologically Aged Human Skin  TaiHao Quan, Yuan Shao, Tianyuan.
Chi-Hyun Park, Youngji Moon, Chung Min Shin, Jin Ho Chung 
dsRNA Sensing Induces Loss of Cell Identity
SIRT1, a Class III Histone Deacetylase, Regulates LPS-Induced Inflammation in Human Keratinocytes and Mediates the Anti-Inflammatory Effects of Hinokitiol 
Cell Type-Specific Involvement of RIG-I in Antiviral Response
Insulin-Like Growth Factor-Binding Protein 7 Regulates Keratinocyte Proliferation, Differentiation and Apoptosis  Janna Nousbeck, Ofer Sarig, Nili Avidan,
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
TWEAK/Fn14 Activation Contributes to the Pathogenesis of Bullous Pemphigoid  Yale Liu, Lingling Peng, Liang Li, Chengfei Liu, Xiao Hu, Shengxiang Xiao,
Akt Kinase-Mediated Checkpoint of cGAS DNA Sensing Pathway
Collagen Synthesis Is Suppressed in Dermal Fibroblasts by the Human Antimicrobial Peptide LL-37  Hyun Jeong Park, Dae Ho Cho, Hee Jung Kim, Jun Young.
Volume 21, Issue 6, Pages (November 2017)
Volume 20, Issue 12, Pages (September 2017)
Volume 40, Issue 4, Pages (April 2014)
MELK Promotes Melanoma Growth by Stimulating the NF-κB Pathway
Volume 33, Issue 5, Pages (November 2010)
Volume 68, Issue 2, Pages e4 (October 2017)
Volume 28, Issue 5, Pages (May 2008)
Hyun Jeong Park, Hee Jung Kim, Jun Young Lee, Baik Kee Cho, Richard L
Volume 14, Issue 1, Pages (July 2013)
Volume 38, Issue 3, Pages (March 2013)
Presentation transcript:

STING Is Involved in Antiviral Immune Response against VZV Infection via the Induction of Type I and III IFN Pathways  Ji-Ae Kim, Seul-Ki Park, Seong-Wook Seo, Chan-Hee Lee, Ok Sarah Shin  Journal of Investigative Dermatology  Volume 137, Issue 10, Pages 2101-2109 (October 2017) DOI: 10.1016/j.jid.2017.03.041 Copyright © 2017 The Authors Terms and Conditions

Figure 1 The antiviral role of STING in human dermal cells. (a) Control (Ctl), STING (ST), or STAT6 siRNA-transfected HaCaTs were infected with mock or VZV for 48 hours. Western blotting was performed for VZV glycoprotein E (gE), immediate early gene (IE62), STING, and p-IRF3. Control scrambled shRNA- (ctl) or STING-specific shRNA-expressing knockdown fibroblasts (ST1 and ST2) were infected with mock or VZV. (b) Protein expressions of VZV IE62, STING, and p-IRF3 were measured. (c) Viral titers were measured by plaque assay. ∗P < 0.05 compared with control shRNA-knockdown stable cells; n = 3. (d, e) TBK1 or IRF3 siRNA-transfected cells were infected with mock or VZV for 24 hours. Western blotting was performed for VZV gE or IE62. A representative of three independent experiments is shown. p-, phosphorylated; PFU, plaque forming units; shR, short hairpin RNA; siRNA, small interfering RNA; VZV, varicella zoster virus. Journal of Investigative Dermatology 2017 137, 2101-2109DOI: (10.1016/j.jid.2017.03.041) Copyright © 2017 The Authors Terms and Conditions

Figure 2 STING agonist pretreatment results in the reduction of virus replication. (a) HEK293T cells were transfected with an IFN-β–luciferase reporter plasmid and either the empty vector (EV), wild-type STING, or STING-h232 mutant plasmid. At 24 hours after transfection, the cells were stimulated with STING agonists and VZV gDNA. The normalized relative IFN-β-luciferase activity is a representative of two independent experiments. HDF cells were pretreated with 2′3′ cGAMP, 3′3′ cGAMP, or poly I:C HMW at the indicated concentrations and subsequently infected with VZV (multiplicity of infection = 0.01) for 24 hours. (b) Plaque assays were performed. ∗P < 0.05, ∗∗P < 0.01, compared with VZV-infected control-treated cells. (c) gE expression was determined by Western blotting. (d) EV, VZV ORF47, and ORF68 were co-transfected with the STING plasmid into HEK293T cells, which were then stimulated with STING agonists. A representative IFN-β–luciferase activity is shown. cGAMP, cyclic guanosine monophosphate-adenosine monophosphate; Ctl, control; gDNA, genomic DNA; HMW, high molecular weight; ORF, open reading frame; PFU, plaque forming units; poly I:C HMW, polyinosinic:polycytidylic acid high molecular weight; pIC, polyinosinic:polycytidylic acid; poly dA:dT, polydeoxyadenylic-polydeoxythymidylic acid; VZV, varicella zoster virus. Journal of Investigative Dermatology 2017 137, 2101-2109DOI: (10.1016/j.jid.2017.03.041) Copyright © 2017 The Authors Terms and Conditions

Figure 3 The effect of RIG-I on VZV replication. (a) Subcellular localization of STING and VZV ORF68 in transfected HEK293T cells, visualized by confocal microscopy. Anti-calnexin antibody was used to stain endoplasmic reticulum. Scale bar = 20 μm. (b) MRC-5 cells were transfected with control, RIG-I–, or STING-specific siRNAs and then infected with VZV. Viral titers were determined by plaque assay. n = 3, mean ± standard error of the mean. ∗P < 0.05. (c) Western blotting analysis of RIG-I siRNA transfected cells to assess the expression of VZV gE, VZV IE62, RIG-I, MAVS, STING, pTBK, pIRF3, and β-actin. The blot is representative of three independent experiments. (d) HDFs were transiently transfected with control, wild-type RIG-I, RIG-I DN (dominant negative), wild-type STING, and STING-h232; mutant and plaque assays were then performed. n = 3, mean ± standard error of the mean. ∗P < 0.05. Ctl, control; gE, glycoprotein E; p-, phosphorylated; PFU, plaque forming units; siR, small interfering RNA; siRNA, small interfering RNA; WT, wild type; VZV, varicella zoster virus. Journal of Investigative Dermatology 2017 137, 2101-2109DOI: (10.1016/j.jid.2017.03.041) Copyright © 2017 The Authors Terms and Conditions

Figure 4 VZV infection of human dermal cells results in distinctive transcriptomic changes. (a) Dual RNA sequencing was performed with VZV-infected HaCaTs and human dermal fibroblasts at the indicated times. The heatmap depicting fragment per kilobase of exon per million (FPKM) values of 71 VZV ORFs is shown. Shading intensity indicates the degree to which each gene was up-regulated or down-regulated. Range of activation z-score is depicted on the figure. (b) To validate the RNA sequencing data, quantitative real-time reverse transcriptase–PCR assays were performed with mock or VZV-infected (48 hours after infection) HaCaTs. n = 3, mean ± standard error of the mean. ∗P < 0.05 compared with mock control-treated cells. (c) HaCaTs were transiently transfected with AIM2-specific siRNAs and infected with VZV. Quantitative real-time reverse transcriptase–PCR assays were performed. (d) Protein expression of VZV IE62 was analyzed by Western blotting. A representative image of three independent experiments is shown with normalized densitometric units. Ctl, control; h, hour; NHDF, normal human dermal fibroblast; ORF, open reading frame; siRNA, small interfering RNA; VZV, varicella zoster virus. Journal of Investigative Dermatology 2017 137, 2101-2109DOI: (10.1016/j.jid.2017.03.041) Copyright © 2017 The Authors Terms and Conditions

Figure 5 IFN-λ is important for VZV replication control. (a) The concentration of secreted IFN-λ in the culture supernatants of control or STING-specific siRNA-transfected VZV-infected cells was determined by ELISA. ∗P < 0.05 versus control siRNA-transfected VZV-infected cells. (b) IFN-λ–treated HDF and MeWo cells were infected with VZV (multiplicity of infection = 0.01), and plaque assays were performed. ∗P < 0.05, ∗∗P < 0.01, versus control-treated VZV-infected cells. (c) gE-specific staining and protein expression in IFN-λ–treated VZV-infected cells are shown. Quantitative densitometric analysis is presented (shown as numbers). (d) mRNA expression levels of VZV ORF63, MxA, and OAS in IFN-λ–treated VZV-infected cells. (e) HDF cells were first treated with the indicated recombinant IFNs to measure IE62 expression. A representative image of three independent experiments is shown with normalized densitometric units. Ctl, control; gE, glycoprotein E; HDF, human dermal fibroblast; MeWo, malignant melanoma cells; p-, phosphorylated; PFU, plaque forming units; siR, small interfering RNA; siRNA, small interfering RNA; VZV, varicella zoster virus. Journal of Investigative Dermatology 2017 137, 2101-2109DOI: (10.1016/j.jid.2017.03.041) Copyright © 2017 The Authors Terms and Conditions

Figure 6 Proposed schematics for our data show STING-mediated antiviral signaling. VZV DNA can be sensed by cytosolic DNA sensors, such as cGAS or AIM2. cGAS-mediated sensing can lead to production of cGAMP and subsequent STING/TBK1/IRF3 signaling pathway. This event can stimulate gene transcription of type I and III IFNs. Upon binding to their cognate receptors, type I and III IFNs induce the JAK/STAT pathway, leading to phosphorylation of STAT1 and STAT2 to induce ISG expression. Both type I and III IFNs can act as antiviral factors capable of blocking VZV replication in dermal compartments. cGAMP, cyclic guanosine monophosphate-adenosine monophosphate; cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase; dsDNA, double-stranded DNA; ISG, IFN-stimulated genes; JAK, Janus kinase; VZV, varicella zoster virus. Journal of Investigative Dermatology 2017 137, 2101-2109DOI: (10.1016/j.jid.2017.03.041) Copyright © 2017 The Authors Terms and Conditions