Gene Regulation and Suppression of Type I Interferon Signaling by STAT3 in Diffuse Large B Cell Lymphoma

Total Page:16

File Type:pdf, Size:1020Kb

Gene Regulation and Suppression of Type I Interferon Signaling by STAT3 in Diffuse Large B Cell Lymphoma Gene regulation and suppression of type I interferon signaling by STAT3 in diffuse large B cell lymphoma Li Lua,b,1, Fen Zhua,b,1, Meili Zhangc,d,1, Yangguang Lia,b, Amanda C. Drennana,b, Shuichi Kimparaa,b, Ian Rumballa,b, Christopher Selzera,b, Hunter Camerona,b, Ashley Kellicuta,b, Amanda Kelma,b, Fangyu Wanga,b, Thomas A. Waldmannc,2, and Lixin Ruia,b,2 aDepartment of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792; bCarbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792; cLymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and dLaboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick, MD 21702 Contributed by Thomas A. Waldmann, December 7, 2017 (sent for review August 28, 2017; reviewed by Vu N. Ngo and Demin Wang) STAT3 is constitutively activated in many cancers and regulates cell lines and control GCB DLBCL cell lines that lack STAT3 gene expression to promote cancer cell survival, proliferation, activation, a recent study has identified a total of 10,337 STAT3- invasion, and migration. In diffuse large B cell lymphoma (DLBCL), binding regions corresponding to 8,531 genes, of which 1,545 activation of STAT3 and its kinase JAK1 is caused by autocrine genes are differently expressed between the two subtypes and production of IL-6 and IL-10 in the activated B cell–like subtype largely associated with ABC DLBCL biology (14). However, (ABC). However, the gene regulatory mechanisms underlying the biological significance and functional association of these STAT3 pathogenesis of this aggressive lymphoma by STAT3 are not well targets in ABC DLBCL cells remain to be studied. characterized. Here we performed genome-wide analysis and Here, we use genetic and pharmacological inhibition of STAT3 in identified 2,251 STAT3 direct target genes, which involve B cell ABC DLBCL cells and identify genes that are directly regulated by activation, survival, proliferation, differentiation, and migration. STAT3. These STAT3 target genes are involved in multiple sig- Whole-transcriptome profiling revealed that STAT3 acts as both naling pathways, which promote cancer cell survival and pro- a transcriptional activator and a suppressor, with a comparable liferation. In addition, STAT3 negatively regulates lethal type I number of up- and down-regulated genes. STAT3 regulates multi- signaling by inhibiting expression of IRF7, IRF9, STAT1,and ple oncogenic signaling pathways, including NF-κB, a cell-cycle STAT2, key transcription factors for IFNβ production and signaling. checkpoint, PI3K/AKT/mTORC1, and STAT3 itself. In addition, STAT3 negatively regulates the lethal type I IFN signaling pathway Results by inhibiting expression of IRF7, IRF9, STAT1, and STAT2. Inhibition Genome-Wide Analysis Identifies STAT3 Transcriptional Target Genes of STAT3 activity by ruxolitinib synergizes with the type I IFN in- in ABC DLBCL Cells. To identify STAT3 target genes genome-wide, ducer lenalidomide in growth inhibition of ABC DLBCL cells in vitro we performed STAT3 ChIP-seq in the ABC DLBCL cell lines and in a xenograft mouse model. Therefore, this study provides a TMD8 and OCI-Ly10, in which high levels of STAT3 phosphor- mechanistic rationale for clinical trials to evaluate ruxolitinib or a ylation were detected by immunoblot analysis (Fig. 1A). Since specific JAK1 inhibitor combined with lenalidomide in ABC DLBCL. STAT3 activity was sufficiently inhibited by the JAK1/JAK2 in- hibitor AZD1480 (Fig. 1A) and this inhibitor was used for H3Y41- STAT3 | interferon | diffuse large B cell lymphoma P ChIP-seq analysis (3), AZD1480-treated cells served as a control for STAT3 ChIP-seq experiments. Using the model-based analysis iffuse large B cell lymphoma (DLBCL), the most common Dnon-Hodgkin lymphoma, includes two main molecular sub- Significance types: an activated B cell–like (ABC) and a germinal center B cell– like (GCB) (1, 2). ABC DLBCL is more aggressive, with autocrine We demonstrate that STAT3 is a critical transcriptional regulator signaling from the cytokines IL-6 and IL-10 that constitutively ac- of the activated B cell–like subtype of diffuse large B cell lym- – tivates JAK1 (3) and STAT3 (4 7) to promote cell survival. ABC phoma (ABC DLBCL), the most common, aggressive, non-Hodgkin DLBCL cells have high NF-κB activity (8) due to genetic alterations lymphoma. By genome-wide assessment, we have identified tar- in the Toll-like receptor (TLR) and B cell receptor signaling get genes of STAT3. Gene regulation by STAT3 in ABC DLBCL ac- pathways (9–11). Somatic mutations of MYD88 (mainly L265P), a centuates survival signaling pathways while dampening the lethal key signaling adaptor in TLR signaling, engage the NF-κB pathway type I interferon pathway. Knowledge of these STAT3-regulated and induce production of IL-6, IL-10, and IFNβ (11). In contrast to genes has led to our demonstration that a small-molecule in- IL-6 and IL-10, IFNβ is proapoptotic, and its basal level is low to hibitor in the JAK1-STAT3 signaling pathway synergizes with the undetectable in ABC DLBCL cells (12). IFNβ production can be type I interferon inducer lenalidomide, suggesting a new thera- prevented by the transcription factors IRF4 and SPIB through re- peutic strategy for ABC DLBCL, a subtype that is particularly dif- pression of IRF7, a transcription factor for IFNβ expression (12). ficult to treat and has poor prognosis. Recently, we revealed that, in addition to STAT3 activation, JAK1 is present in the nucleus and directly phosphorylates his- Author contributions: L.R. designed research; L.L., F.Z., M.Z., Y.L., A.C.D., S.K., I.R., C.S., tone H3 on tyrosine 41 (H3Y41) to induce expression of nearly H.C., A. Kellicut, A. Kelm, and F.W. performed research; T.A.W. and L.R. supervised re- κ search; L.L., F.Z., M.Z., Y.L., S.K., and L.R. analyzed data; and L.L., F.Z., M.Z., T.A.W., and 3,000 genes, including the NF- B pathway genes MYD88 and L.R. wrote the paper. IRF4 (3, 13). These findings suggest a positive feedback loop Reviewers: V.N.N., City of Hope National Medical Center; and D.W., BloodCenter between the cytokine and NF-κB signaling pathways that pro- of Wisconsin. motes the malignant phenotype of ABC DLBCL cells. This The authors declare no conflict of interest. epigenetic gene regulatory mechanism by JAK1 is distinct from Published under the PNAS license. the canonical JAK-STAT signaling pathway, given that more 1L.L., F.Z., and M.Z. contributed equally to this work. than 90% of these genes do not bear a STAT motif in their 2To whom correspondence may be addressed. Email: [email protected] or lrui@ promoter region (3). Using STAT3 chromatin immunoprecipi- medicine.wisc.edu. tation followed by DNA sequencing (ChIP-seq) and whole- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. genome transcriptome (RNA-seq) analysis in ABC DLBCL 1073/pnas.1715118115/-/DCSupplemental. E498–E505 | PNAS | Published online January 2, 2018 www.pnas.org/cgi/doi/10.1073/pnas.1715118115 Downloaded by guest on September 28, 2021 OCI-Ly10 PNAS PLUS A B TMD8 DMSO AZD1480 High DMSO AZD1480 High AZD1480 TMD8 (2 M): 0 15' 30' 1h 2h 4h 6h 0 pSTAT3 STAT3 Low Low -actin AZD1480 OCI-Ly10 (2 M): 0 15' 30' 1h 2h 4h 6h pSTAT3 STAT3 -actin Density heatmap of 22,856 peaks Density heatmap of 11,487 peaks -5K Apex +5K -5K Apex +5K C STAT3 motif enrichment ((TMD8)TMD8) STAT3 motif enrichment (OCI-Ly10) 2 2 Probability Probability Probability 1 1 bits E-valueE 3.6e-21 E-value 9.5e-8 bits 0 0 3 2 7 8 1 9 6 4 5 11 3 2 7 8 9 1 4 6 10 5 11 10 Position of Best Site in Sequence Position of Best Site in Sequence D E B cell activation 7.03E-05 OCI-Ly10 Lymphocyte proliferation 3.87E-03 TMD8 MEDICAL SCIENCES (6,058) (4,746) Lymphocyte differentiation 1.27E-02 Intracellular signal transduction 5.81E-12 Apoptotic process 8.49E-04 Regulation of cell migration 4.81E-02 Cell cycle 5.00E-03 2,495 2,251 3,807 Cellular response to stress 5.29E-04 Response to external stimulus 3.97E-03 Regulation of metabolic process 5.38E-06 01234 Enrichment Score Fig. 1. Genome-wide analysis reveals STAT3 transcriptional targets in ABC DLBCL. (A) Immunoblot analysis of phospho-STAT3 and total STAT3 protein in TMD8 and OCI-Ly10 cells treated with 2 μM AZD1480. (B) Heat maps of STAT3 ChIP-seq in TMD8 and OCI-Ly10 cells after 4 h of treatment with either DMSO or 2 μM AZD1480. STAT3 peak summits were centered within 5 kb of the flanking sequence on either side. Darker color indicates a higher density of reads. STAT3 peaks were ranked by signal intensity at the peak center, and the same order is used to display the AZD1480-treated sample. (C) The CentriMo plots show the distribution of known the STAT3 motif in the ChIP-seq peak summit regions (P < 0.001). De novo motif discovery from STAT3 ChIP-seq peaks shows identical sequence logs to the known STAT consensus motif (15). (D) Venn diagram shows 2,251 STAT3-binding genes that are shared in TMD8 and OCI- Ly10 cells. (E) Gene ontology analysis of the 2,251 STAT3-binding genes (P < 0.05). of ChIP-seq (MACS) for peak calling (16), we identified a total of OCI-Ly10 cells compared with the AZD1480-treated control sam- 11,487 STAT3-binding sites (peaks) in TMD8 cells and 22,856 in ple (Fig.
Recommended publications
  • Activated Peripheral-Blood-Derived Mononuclear Cells
    Transcription factor expression in lipopolysaccharide- activated peripheral-blood-derived mononuclear cells Jared C. Roach*†, Kelly D. Smith*‡, Katie L. Strobe*, Stephanie M. Nissen*, Christian D. Haudenschild§, Daixing Zhou§, Thomas J. Vasicek¶, G. A. Heldʈ, Gustavo A. Stolovitzkyʈ, Leroy E. Hood*†, and Alan Aderem* *Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103; ‡Department of Pathology, University of Washington, Seattle, WA 98195; §Illumina, 25861 Industrial Boulevard, Hayward, CA 94545; ¶Medtronic, 710 Medtronic Parkway, Minneapolis, MN 55432; and ʈIBM Computational Biology Center, P.O. Box 218, Yorktown Heights, NY 10598 Contributed by Leroy E. Hood, August 21, 2007 (sent for review January 7, 2007) Transcription factors play a key role in integrating and modulating system. In this model system, we activated peripheral-blood-derived biological information. In this study, we comprehensively measured mononuclear cells, which can be loosely termed ‘‘macrophages,’’ the changing abundances of mRNAs over a time course of activation with lipopolysaccharide (LPS). We focused on the precise mea- of human peripheral-blood-derived mononuclear cells (‘‘macro- surement of mRNA concentrations. There is currently no high- phages’’) with lipopolysaccharide. Global and dynamic analysis of throughput technology that can precisely and sensitively measure all transcription factors in response to a physiological stimulus has yet to mRNAs in a system, although such technologies are likely to be be achieved in a human system, and our efforts significantly available in the near future. To demonstrate the potential utility of advanced this goal. We used multiple global high-throughput tech- such technologies, and to motivate their development and encour- nologies for measuring mRNA levels, including massively parallel age their use, we produced data from a combination of two distinct signature sequencing and GeneChip microarrays.
    [Show full text]
  • The STAT3 Inhibitor Galiellalactone Reduces IL6-Mediated AR Activity in Benign and Malignant Prostate Models
    Author Manuscript Published OnlineFirst on September 25, 2018; DOI: 10.1158/1535-7163.MCT-18-0508 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. The STAT3 inhibitor galiellalactone reduces IL6-mediated AR activity in benign and malignant prostate models Florian Handle1,2, Martin Puhr1, Georg Schaefer3, Nicla Lorito1, Julia Hoefer1, Martina Gruber1, Fabian Guggenberger1, Frédéric R. Santer1, Rute B. Marques4, Wytske M. van Weerden4, Frank Claessens2, Holger H.H. Erb5*, Zoran Culig1* 1 Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria 2 Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium 3 Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria 4 Department of Urology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands 5 Department of Urology and Pediatric Urology, University Medical Center Mainz, Mainz, Germany *Joint senior authors Running title: Galiellalactone reduces IL6-mediated AR activity Conflict of interest statement: Zoran Culig received research funding and honoraria from Astellas. Other authors declare no potential conflict of interest. Keywords (5): Prostate cancer, androgen receptor, Interleukin-6, STAT3, galiellalactone Financial support: This work was supported by grants from the Austrian Cancer Society/Tirol (to F. Handle) and the Austrian Science Fund (FWF): W1101-B12 (to Z. Culig). Corresponding author: Prof. Zoran Culig, Medical University of Innsbruck, Division of Experimental Urology, Anichstr. 35 6020 Innsbruck, Austria, [email protected] Word counts: Abstract: 242 (limit: 250); Text: 4993 (limit: 5000) Handle et al., 2018 1 Downloaded from mct.aacrjournals.org on October 1, 2021.
    [Show full text]
  • Quantitative Modelling Explains Distinct STAT1 and STAT3
    bioRxiv preprint doi: https://doi.org/10.1101/425868; this version posted September 24, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Title Quantitative modelling explains distinct STAT1 and STAT3 activation dynamics in response to both IFNγ and IL-10 stimuli and predicts emergence of reciprocal signalling at the level of single cells. 1,2, 3 1 1 1 1 1 2 Sarma U , Maitreye M , Bhadange S , Nair A , Srivastava A , Saha B , Mukherjee D . 1: National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India. 2 : Corresponding author. [email protected] , [email protected] 3: Present address. Labs, Persistent Systems Limited, Pingala – Aryabhata, Erandwane, Pune, 411004 India. bioRxiv preprint doi: https://doi.org/10.1101/425868; this version posted September 24, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract Cells use IFNγ-STAT1 and IL-10-STAT3 pathways primarily to elicit pro and anti-inflammatory responses, respectively. However, activation of STAT1 by IL-10 and STAT3 by IFNγ is also observed. The regulatory mechanisms controlling the amplitude and dynamics of both the STATs in response to these functionally opposing stimuli remains less understood. Here, our experiments at cell population level show distinct early signalling dynamics of both STAT1 and STAT3(S/1/3) in responses to IFNγ and IL-10 stimulation.
    [Show full text]
  • A Co-Opted ISG15-USP18 Binding Mechanism Normally Reserved for Deisgylation Controls Type I IFN Signalling
    bioRxiv preprint doi: https://doi.org/10.1101/2021.06.01.446527; this version posted June 3, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 A co-opted ISG15-USP18 binding mechanism normally reserved for deISGylation controls 2 type I IFN signalling 3 4 5 6 Andri Vasou1, Katie Nightingale2, Vladimíra Cetkovská 1, Connor G.G. Bamford3‡, Jelena 7 Andrejeva1, Richard E. Randall1, John McLauchlan3, Michael P. Weekes2, David J Hughes1* 8 9 10 11 1Biomedical Sciences Research Complex, School of Biology, University of St Andrews, St 12 Andrews, United Kingdom. 2Cambridge Institute for Medical Research, Hills Road, Cambridge, 13 United Kingdom 3MRC-University of Glasgow Centre for Virus Research, Glasgow, United 14 Kingdom. 15 16 17 ‡ Present addresses: Wellcome-Wolfson Institute for Experimental Medicine, Queen’s 18 University, Belfast UK. 19 20 21 22 * Corresponding author: [email protected]; Tel: +44 1334 467197 23 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.01.446527; this version posted June 3, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 24 Abstract 25 Type I interferon (IFN) signalling induces the expression of several hundred IFN-stimulated 26 genes that provide an unfavourable environment for viral replication.
    [Show full text]
  • A Molecular Switch from STAT2-IRF9 to ISGF3 Underlies Interferon-Induced Gene Transcription
    ARTICLE https://doi.org/10.1038/s41467-019-10970-y OPEN A molecular switch from STAT2-IRF9 to ISGF3 underlies interferon-induced gene transcription Ekaterini Platanitis 1, Duygu Demiroz1,5, Anja Schneller1,5, Katrin Fischer1, Christophe Capelle1, Markus Hartl 1, Thomas Gossenreiter 1, Mathias Müller2, Maria Novatchkova3,4 & Thomas Decker 1 Cells maintain the balance between homeostasis and inflammation by adapting and inte- grating the activity of intracellular signaling cascades, including the JAK-STAT pathway. Our 1234567890():,; understanding of how a tailored switch from homeostasis to a strong receptor-dependent response is coordinated remains limited. Here, we use an integrated transcriptomic and proteomic approach to analyze transcription-factor binding, gene expression and in vivo proximity-dependent labelling of proteins in living cells under homeostatic and interferon (IFN)-induced conditions. We show that interferons (IFN) switch murine macrophages from resting-state to induced gene expression by alternating subunits of transcription factor ISGF3. Whereas preformed STAT2-IRF9 complexes control basal expression of IFN-induced genes (ISG), both type I IFN and IFN-γ cause promoter binding of a complete ISGF3 complex containing STAT1, STAT2 and IRF9. In contrast to the dogmatic view of ISGF3 formation in the cytoplasm, our results suggest a model wherein the assembly of the ISGF3 complex occurs on DNA. 1 Max Perutz Labs (MPL), University of Vienna, Vienna 1030, Austria. 2 Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna 1210, Austria. 3 Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria. 4 Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna 1030, Austria.
    [Show full text]
  • Mechanism of Action Through an IFN Type I-Independent Responses To
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 is online at: average * The Journal of Immunology , 12 of which you can access for free at: 2012; 188:3088-3098; Prepublished online 20 from submission to initial decision 4 weeks from acceptance to publication February 2012; doi: 10.4049/jimmunol.1101764 http://www.jimmunol.org/content/188/7/3088 MF59 and Pam3CSK4 Boost Adaptive Responses to Influenza Subunit Vaccine through an IFN Type I-Independent Mechanism of Action Elena Caproni, Elaine Tritto, Mario Cortese, Alessandro Muzzi, Flaviana Mosca, Elisabetta Monaci, Barbara Baudner, Anja Seubert and Ennio De Gregorio J Immunol cites 33 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription http://www.jimmunol.org/content/suppl/2012/02/21/jimmunol.110176 4.DC1 This article http://www.jimmunol.org/content/188/7/3088.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767
    [Show full text]
  • IFN-Α Wakes up Sleeping Hematopoietic Stem Cells
    NEWS AND VIEWS calcium consumption does not seem to have without increasing the risk of coronary heart 7. Aoki, K. et al. Ann. Epidemiol. 15, 598–606 (2005). such an effect. How to optimize calcium sup- disease, which has been associated with cal- 8. Langhans, N. et al. Gastroenterology 112, 280–286 13 plements for human health deserves further cium supplementation . (1997). investigation. 9. Recker, R.R. N. Engl. J. Med. 313, 70–73 (1985). 10. Yang, Y.-X. et al. J. Am. Med. Assoc. , 2947– Maybe it’s time to determine whether low- 1. Zaidi, M. Nat. Med. 13, 791–801 (2007). 296 2. Del Fattore, A. et al. Bone 42, 19–29 (2008). 2953 (2006). fat milk–based drinks taken as an alternative 3. Sobacchi, C. et al. Hum. Mol. Genet. 10, 1767–1773 11. Collazo-Clavell, M.L., Jimenez, A., Hodgson, S.F. & to the soda so frequently consumed, especially (2001). Sarr, M.G. Endocr. Pract. 10, 195–198 (2004). 4. Amling, M. et al. Nat. Med. 15, 674–681 (2009). 12. Bo-Linn, G.W. et al. J. Clin. Invest. 73, 640–647 during the early adolescent years, can attain 5. Straub, D.A. Nutr. Clin. Pract. 22, 286–296 (2007). (1984). the noble goal of keeping bones healthy— 6. Teitelbaum, S.L. Science 289, 1504–1508 (2000). 13. Bolland, M.J. BMJ 336, 262–266 (2008). IFN-α wakes up sleeping hematopoietic stem cells Emmanuelle Passegué & Patricia Ernst The cytokine interferon-α stimulates the turnover and proliferation of hematopoietic cells in vivo (pages 696–700).
    [Show full text]
  • Inactivation of Type I IFN Jak-STAT Pathway in EBV Latency
    Ning S and Wang L, J Cancer Biol Treat 2016, 3: 009 DOI: 10.24966/CBT-7546/100009 HSOA Journal of Cancer Biology and Treatment Research Article defense system, among which IRF7 is the “master” regulator of type I Inactivation of Type I IFN Interferon (IFN-I) response to pathogenic infection [4]. Robust IFN-I response during infection requires a positive feedback loop between Jak-STAT Pathway in EBV IRF7 and IFN-I [5-6]. Aberrant production of IFN-I, however, is Latency associated with autoimmune disorders and malignancies [7-9]. Thus, tight regulation of IRF7 is important in balancing the appropriate Shunbin Ning and Ling Wang* immune response to clear invading pathogens while preventing Department of Internal Medicine, Center of Excellence for Inflammation, immune-mediated pathogenesis [10]. Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, USA IFNs exert their functions through induction of IFN-Stimulated Genes (ISGs) via Jak-STAT pathways [11-13]. The IFN-I Jak-STAT pathway comprises of IFNAR1/2, Jak1, Tyk2, STAT1/2, and IRF9 [14]. Following IFN-I binding to IFNARs, signaling via protein kinases leads to tyrosine phosphorylation and activation of IFNAR1 Abstract at Y466 [15], Tyk2(Y1054/Y1055) and Jak1(Y1034/Y1035), and then to that Epstein-Barr Virus (EBV) latent infection is associated with a of STAT1(Y701) and STAT2(Y690 and S287) [16]. The phosphorylated variety of lymphomas and carcinomas. Interferon (IFN) Regulatory STAT1/2 then dimerize and associate with IRF9 to form a complex Factors (IRFs) are a family of transcription factors, among which termed interferon-stimulated gene factor 3 (ISGF3).
    [Show full text]
  • Expression of the Tumor Necrosis Factor Receptor-Associated Factors
    Expression of the Tumor Necrosis Factor Receptor- Associated Factors (TRAFs) 1 and 2 is a Characteristic Feature of Hodgkin and Reed-Sternberg Cells Keith F. Izban, M.D., Melek Ergin, M.D, Robert L. Martinez, B.A., HT(ASCP), Serhan Alkan, M.D. Department of Pathology, Loyola University Medical Center, Maywood, Illinois the HD cell lines. Although KMH2 showed weak Tumor necrosis factor receptor–associated factors expression, the remaining HD cell lines also lacked (TRAFs) are a recently established group of proteins TRAF5 protein. These data demonstrate that consti- involved in the intracellular signal transduction of tutive expression of TRAF1 and TRAF2 is a charac- several members of the tumor necrosis factor recep- teristic feature of HRS cells from both patient and tor (TNFR) superfamily. Recently, specific members cell line specimens. Furthermore, with the excep- of the TRAF family have been implicated in promot- tion of TRAF1 expression, HRS cells from the three ing cell survival as well as activation of the tran- HD cell lines showed similar TRAF protein expres- ␬ scription factor NF- B. We investigated the consti- sion patterns. Overall, these findings demonstrate tutive expression of TRAF1 and TRAF2 in Hodgkin the expression of several TRAF proteins in HD. Sig- and Reed–Sternberg (HRS) cells from archived nificantly, the altered regulation of selective TRAF paraffin-embedded tissues obtained from 21 pa- proteins may reflect HRS cell response to stimula- tients diagnosed with classical Hodgkin’s disease tion from the microenvironment and potentially (HD). In a selective portion of cases, examination of contribute both to apoptosis resistance and cell HRS cells for Epstein-Barr virus (EBV)–encoded maintenance of HRS cells.
    [Show full text]
  • Type I IFN Signaling Infection by Up-Regulating Typhimurium
    Enhanced Antibacterial Potential in UBP43-Deficient Mice against Salmonella typhimurium Infection by Up-Regulating Type I IFN Signaling This information is current as of September 24, 2021. Keun Il Kim, Oxana A. Malakhova, Kasper Hoebe, Ming Yan, Bruce Beutler and Dong-Er Zhang J Immunol 2005; 175:847-854; ; doi: 10.4049/jimmunol.175.2.847 http://www.jimmunol.org/content/175/2/847 Downloaded from References This article cites 45 articles, 21 of which you can access for free at: http://www.jimmunol.org/content/175/2/847.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on September 24, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2005 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Enhanced Antibacterial Potential in UBP43-Deficient Mice against Salmonella typhimurium Infection by Up-Regulating Type I IFN Signaling1 Keun Il Kim,2* Oxana A. Malakhova,* Kasper Hoebe,† Ming Yan,* Bruce Beutler,† and Dong-Er Zhang3* ISG15 is an IFN-inducible ubiquitin-like protein and its expression and conjugation to target proteins are dramatically induced upon viral or bacterial infection.
    [Show full text]
  • Beyond Viral Interferon Regulatory Factors: Immune Evasion Strategies Jinjong Myoung1, Shin-Ae Lee2, and Hye-Ra Lee3*
    J. Microbiol. Biotechnol. (2019), 29(12), 1873–1881 https://doi.org/10.4014/jmb.1910.10004 Research Article Review jmb Beyond Viral Interferon Regulatory Factors: Immune Evasion Strategies Jinjong Myoung1, Shin-Ae Lee2, and Hye-Ra Lee3* 1Korea Zoonosis Research Institute, Genetic Engineering Research Institute and Department of Bioactive Material Science, College of Natural Science, Jeonbuk National University, Jeonju 54531, Republic of Korea 2Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA 3Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong 30019, Republic of Korea Received: October 8, 2019 Revised: October 8, 2019 The innate immune response serves as a first-line-of-defense mechanism for a host against Accepted: October 24, 2019 viral infection. Viruses must therefore subvert this anti-viral response in order to establish an First published online: efficient life cycle. In line with this fact, Kaposi’s sarcoma-associated herpesvirus (KSHV) October 25, 2019 encodes numerous genes that function as immunomodulatory proteins to antagonize the host *Corresponding author immune system. One such mechanism through which KSHV evades the host immunity is by Phone: +82-44-860-1831 encoding a viral homolog of cellular interferon (IFN) regulatory factors (IRFs), known as Fax: +82-44-860-1598 E-mail: [email protected] vIRFs. Herein, we summarize recent advances in the study of the immunomodulatory strategies of KSHV vIRFs and their effects on KSHV-associated pathogenesis. pISSN 1017-7825, eISSN 1738-8872 Copyright© 2019 by Keywords: KSHV, viral interferon regulatory factor, immune evasion strategy, PRR-mediated The Korean Society for Microbiology signaling pathway, apoptosis pathway and Biotechnology Introduction various transcription factors.
    [Show full text]
  • Transcription Factor IRF8 Directs a Silencing Programme for TH17 Cell Differentiation
    ARTICLE Received 17 Aug 2010 | Accepted 13 Apr 2011 | Published 17 May 2011 DOI: 10.1038/ncomms1311 Transcription factor IRF8 directs a silencing programme for TH17 cell differentiation Xinshou Ouyang1, Ruihua Zhang1, Jianjun Yang1, Qingshan Li1, Lihui Qin2, Chen Zhu3, Jianguo Liu4, Huan Ning4, Min Sun Shin5, Monica Gupta6, Chen-Feng Qi5, John Cijiang He1, Sergio A. Lira1, Herbert C. Morse III5, Keiko Ozato6, Lloyd Mayer1 & Huabao Xiong1 TH17 cells are recognized as a unique subset of T helper cells that have critical roles in the pathogenesis of autoimmunity and tissue inflammation. Although OR Rγt is necessary for the generation of TH17 cells, the molecular mechanisms underlying the functional diversity of TH17 cells are not fully understood. Here we show that a member of interferon regulatory factor (IRF) family of transcription factors, IRF8, has a critical role in silencing TH17-cell differentiation. Mice with a conventional knockout, as well as a T cell-specific deletion, of the Irf8 gene exhibited more efficient TH17 cells. Indeed, studies of an experimental model of colitis showed that IRF8 deficiency resulted in more severe inflammation with an enhanced TH17 phenotype. IRF8 was induced steadily and inhibited TH17-cell differentiation during TH17 lineage commitment at least in part through its physical interaction with RORγt. These findings define IRF8 as a novel intrinsic transcriptional inhibitor of TH17-cell differentiation. 1 Immunology Institute, Department of Medicine, Mount Sinai School of Medicine, 1 Gustave L. Levy Place, New York, New York 10029, USA. 2 Department of Pathology, Mount Sinai School of Medicine, New York, New York 10029, USA.
    [Show full text]