IRF8 Dendritic Cells by Inhibition of IRF1 and Lipids Inhibit IL-12P40 In

Total Page:16

File Type:pdf, Size:1020Kb

IRF8 Dendritic Cells by Inhibition of IRF1 and Lipids Inhibit IL-12P40 In Francisella tularensis SchuS4 and SchuS4 Lipids Inhibit IL-12p40 in Primary Human Dendritic Cells by Inhibition of IRF1 and IRF8 This information is current as of September 29, 2021. Robin Ireland, Rong Wang, Joshua B. Alinger, Pamela Small and Catharine M. Bosio J Immunol 2013; 191:1276-1286; Prepublished online 1 July 2013; doi: 10.4049/jimmunol.1300867 Downloaded from http://www.jimmunol.org/content/191/3/1276 Supplementary http://www.jimmunol.org/content/suppl/2013/07/01/jimmunol.130086 http://www.jimmunol.org/ Material 7.DC1 References This article cites 55 articles, 19 of which you can access for free at: http://www.jimmunol.org/content/191/3/1276.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 29, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *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 All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Francisella tularensis SchuS4 and SchuS4 Lipids Inhibit IL-12p40 in Primary Human Dendritic Cells by Inhibition of IRF1 and IRF8 Robin Ireland,* Rong Wang,*,1 Joshua B. Alinger,* Pamela Small,† and Catharine M. Bosio* Induction of innate immunity is essential for host survival of infection. Evasion and inhibition of innate immunity constitute a strat- egy used by pathogens, such as the highly virulent bacterium Francisella tularensis, to ensure their replication and transmission. The mechanism and bacterial components responsible for this suppression of innate immunity by F. tularensis are not defined. In this article, we demonstrate that lipids enriched from virulent F. tularensis strain SchuS4, but not attenuated live vaccine strain, inhibit inflammatory responses in vitro and in vivo. Suppression of inflammatory responses is associated with IkBa-independent inhibition of NF-kBp65 activation and selective inhibition of activation of IFN regulatory factors. Interference with NF-kBp65 Downloaded from and IFN regulatory factors is also observed following infection with viable SchuS4. Together these data provide novel insight into how highly virulent bacteria selectively modulate the host to interfere with innate immune responses required for survival of infection. The Journal of Immunology, 2013, 191: 1276–1286. irulent Francisella tularensis ssp. tularensis is a Gram- allowing the adaptive response time to develop. Unlike attenuated negative bacterium and the causative agent of the fatal subspecies and strains, virulent F. tularensis is not sensed by host http://www.jimmunol.org/ V disease coined tularemia. Francisella has four major receptors or other detection machinery (7–10). In addition to evading subspecies (1). F. tularensis ssp. novicida and ssp. mediasiatica are detection by the host, virulent F. tularensis also suppresses the ability generally considered attenuated for humans. F. tularensis ssp. of host cells to mount inflammatory responses (7, 8, 11). The holarctica (Type B; F. holarctica) causes serious disease in humans ability of the bacterium to both evade and suppress innate immune but is not typically fatal. F. tularensis ssp. tularensis (Type A; F. responses is a primary mechanism of virulence. tularensis) is highly infectious and can cause a lethal infection Generation of novel vaccines and therapeutics for treatment of following inhalation of as few as 10 organisms in both humans and tularemia has been hampered by our lack of understanding of the rodent models (2, 3). F. tularensis ssp. tularensis was previously host pathways associated with innate immunity that are modulated used as a biological weapon and thus presents a viable concern as a by virulent F. tularensis. Similarly, we have not identified those by guest on September 29, 2021 potential bioweapon today (4). Although a vaccine generated from components of the bacterium that facilitate suppression of innate serial passage of F. ho larc ti ca—that is, live vaccine strain (LVS)— inflammatory responses in the host. Identification of both the host was produced in the 1960s, it is no longer licensed as a vaccine for signaling pathways and components uniquely associated with viru- use against tularemia (5). Further, treatment of individuals infected lent F. tularensis that mediate inhibition of inflammation will greatly with F. tularensis with antibiotic (when delivered in a timely man- facilitate development of new therapeutics and vaccines. ner) does not always result in complete clearance of the bacterium In this article, we demonstrate that lipids isolated from fully virulent (6). Thus, a need has arisen for development of both novel vaccines F. tularensis strain SchuS4, but not attenuated LVS, inhibit innate and therapeutics to combat this highly virulent pathogen. immune responses in primary human cells in vitro and in the mouse The effectiveness of vaccines and therapeutics is tied to their lung in vivo. Inhibition of inflammatory responses by SchuS4 lipids ability to trigger innate immune responses. Induction of innate was a result of targeting specific host proteins required for tran- immunity is an important component of host defense because this scription of genes necessary for innate immunity. Importantly, these response slows replication and dissemination of microorganisms, findings were recapitulated following infection with viable SchuS4. Materials and Methods *Immunity to Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Reagents † National Institutes of Health, Hamilton, MT 59840; and Department of Microbiol- Ultrapure E. coli strain K12 LPS, Pam3CSK4, Pam2CSK4, lipoteichoic ogy, University of Tennessee, Knoxville, TN 37996 acid (LTA), ssRNA40/LyoVec, and R848 (imidazoquinolone compound) 1 Current address: U.S. Meat Animal Research Center, Clay Center, NE. were purchased from Invivogen (San Diego, CA). E. coli strain O127:B7 Received for publication April 1, 2013. Accepted for publication May 31, 2013. LPS was purchased from Sigma-Aldrich (St. Louis, MO). Recombinant GM-CSF and IL-4 were purchased from PeproTech (Rocky Hill, NJ). This work was supported by the Intramural Research Program of the National In- stitutes of Health, National Institute of Allergy and Infectious Diseases. Pronase was obtained from Roche Diagnostics (Indianapolis, IN). Address correspondence and reprint requests to Dr. Catharine M. Bosio, Immunity to Bacteria Pulmonary Pathogens Section, Laboratory of Intracellular Parasites, Rocky Mountain Laboratories/NIAID/NIH, Hamilton, MT 59840. E-mail address: bosioc@niaid. Virulent F. tularensis ssp. tularensis strain SchuS4 was kindly provided by nih.gov Jeannine Peterson, Ph.D. (Centers for Disease Control, Fort Collins, CO). The online version of this article contains supplemental material. Attenuated F. tularensis ssp. holarctica LVS was originally obtained from Abbreviations used in this article: BAL, bronchoalveolar lavage; cRPMI, complete Dr. Jean Celli (Rocky Mountain Laboratories, Hamilton, MT). Stock vials RPMI 1640; EtOH, ethanol; hDC, human dendritic cell; IRF, IFN regulatory factor; of SchuS4 and LVS in broth were generated as previously described LTA, lipoteichoic acid; LVS, live vaccine strain; MF, membrane fraction. (10, 12). www.jimmunol.org/cgi/doi/10.4049/jimmunol.1300867 The Journal of Immunology 1277 Isolation of total membrane fraction (10 mg/ml), LTA (100 mg/ml), R848 (5 mg/ml), or LPS (10 ng/ml) + 100 U/ml IFN-g (PeproTech) was added 18 h after MF or lipid treatment, and Total membrane fraction (MF) from LVS and SchuS4 were isolated as cells were cultured for an additional 20 h before supernatants were col- previously described (13–15). Briefly, SchuS4 was grown in modified lected for assessment of cytokines. Mueller–Hinton broth as described earlier (10, 12, 13). Following over- night culture, bacteria were pelleted by centrifugation for 15 min at 8000 3 g. Mice and in vivo assessment of inflammation The resulting pellet was resuspended in the following buffer: 50 mM Tris/ HCl, 0.6 mg/ml DNase, 0.6 mg/ml RNase, 1 mM EDTA (all from Sigma- Specific pathogen–free, 6- to 8-wk-old male C57BL/6J mice (wild type) Aldrich), and 1 complete EDTA-Free Protease Inhibitor Cocktail Tablet (n = 5 per group) were purchased from The Jackson Laboratory (Bar Harbor, (Roche), followed by centrifugation and resuspension in the buffer de- ME). Mice were housed in sterile microisolator cages in the ABSL-2 fa- scribed above. Bacteria were lysed via processing in FastPrep Lysing cility at the National Institute of Allergy and Infectious Diseases/Rocky Matrix B tubes using a FastPrep-24 (MPBio) for 10 cycles of 45 s with 2- Mountain Laboratories. All mice were provided sterile water and food ad min rest periods on ice between each cycle. The resulting slurry was then libitum, and all research involving animals was conducted in accordance centrifuged at 10,000 rpm for 10 min. The supernatant was collected and with Animal Care and Use
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]
  • 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]
  • Review Article the Role of Interferon Regulatory Factor-1 (IRF1) in Overcoming Antiestrogen Resistance in the Treatment of Breast Cancer
    SAGE-Hindawi Access to Research International Journal of Breast Cancer Volume 2011, Article ID 912102, 9 pages doi:10.4061/2011/912102 Review Article The Role of Interferon Regulatory Factor-1 (IRF1) in Overcoming Antiestrogen Resistance in the Treatment of Breast Cancer J.L.Schwartz,A.N.Shajahan,andR.Clarke Georgetown University Medical Center, W401 Research Building, 3970 Reservoir Road, NW, Washington, DC 20057, USA Correspondence should be addressed to R. Clarke, [email protected] Received 18 February 2011; Revised 29 April 2011; Accepted 9 May 2011 Academic Editor: Chengfeng Yang Copyright © 2011 J. L. Schwartz et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Resistance to endocrine therapy is common among breast cancer patients with estrogen receptor alpha-positive (ER+) tumors and limits the success of this therapeutic strategy. While the mechanisms that regulate endocrine responsiveness and cell fate are not fully understood, interferon regulatory factor-1 (IRF1) is strongly implicated as a key regulatory node in the underlying signaling network. IRF1 is a tumor suppressor that mediates cell fate by facilitating apoptosis and can do so with or without functional p53. Expression of IRF1 is downregulated in endocrine-resistant breast cancer cells, protecting these cells from IRF1- induced inhibition of proliferation and/or induction of cell death. Nonetheless, when IRF1 expression is induced following IFNγ treatment, antiestrogen sensitivity is restored by a process that includes the inhibition of prosurvival BCL2 family members and caspase activation.
    [Show full text]
  • Partner-Regulated Interaction of IFN Regulatory Factor 8 with Chromatin Visualized in Live Macrophages
    Partner-regulated interaction of IFN regulatory factor 8 with chromatin visualized in live macrophages Leopoldo Laricchia-Robbio*, Tomohiko Tamura*, Tatiana Karpova†, Brian L. Sprague†, James G. McNally†, and Keiko Ozato*‡ *Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-2753; and †Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5055 Edited by Laurie H. Glimcher, Harvard School of Public Health, Boston, MA, and approved August 18, 2005 (received for review May 17, 2005) IFN regulatory factor (IRF) 8 is a transcription factor that directs protein–protein interactions on fluorescence recovery have not macrophage differentiation. By fluorescence recovery after pho- been extensively studied. Moreover, many FRAP studies so far tobleaching, we visualized the movement of IRF8-GFP in differen- reported are qualitative, lacking quantitative insight into the tiating macrophages. Recovery data fitted to mathematical models chromatin-binding events. More recent efforts to fit FRAP data revealed two binding states for IRF8. The majority of IRF8 was to mathematical models begin to provide a clearer knowledge on highly mobile and transiently interacted with chromatin, whereas the property of FRAP mobility (16, 17). a small fraction of IRF8 bound to chromatin more stably. IRF8 IRF8, a member of the IFN regulatory factor (IRF) family, is mutants that did not stimulate macrophage differentiation a key factor that guides the development of macrophages (18). showed a faster recovery, revealing little interaction with chro- We previously established an in vitro model system where matin. A macrophage activation signal by IFN-␥͞LPS led to a global IRF8Ϫ/Ϫ myeloid progenitor cells called Tot2 differentiate into slowdown of IRF8 movement, leading to increased chromatin macrophages upon IRF8 introduction (19).
    [Show full text]
  • The Proapoptotic Gene Interferon Regulatory Factor-1 Mediates the Antiproliferative Outcome of Paired Box 2 Gene and Tamoxifen
    Oncogene (2020) 39:6300–6312 https://doi.org/10.1038/s41388-020-01435-4 ARTICLE The proapoptotic gene interferon regulatory factor-1 mediates the antiproliferative outcome of paired box 2 gene and tamoxifen 1 1 1 2 3 3 Shixiong Wang ● Venkata S. Somisetty ● Baoyan Bai ● Igor Chernukhin ● Henri Niskanen ● Minna U. Kaikkonen ● 4,5 2 6,7 Meritxell Bellet ● Jason S. Carroll ● Antoni Hurtado Received: 13 November 2019 / Revised: 5 August 2020 / Accepted: 17 August 2020 / Published online: 25 August 2020 © The Author(s) 2020. This article is published with open access Abstract Tamoxifen is the most prescribed selective estrogen receptor (ER) modulator in patients with ER-positive breast cancers. Tamoxifen requires the transcription factor paired box 2 protein (PAX2) to repress the transcription of ERBB2/HER2. Now, we identified that PAX2 inhibits cell growth of ER+/HER2− tumor cells in a dose-dependent manner. Moreover, we have identified that cell growth inhibition can be achieved by expressing moderate levels of PAX2 in combination with tamoxifen treatment. Global run-on sequencing of cells overexpressing PAX2, when coupled with PAX2 ChIP-seq, identified common targets regulated by both PAX2 and tamoxifen. The data revealed that PAX2 can inhibit estrogen-induced gene transcription 1234567890();,: 1234567890();,: and this effect is enhanced by tamoxifen, suggesting that they converge on repression of the same targets. Moreover, PAX2 and tamoxifen have an additive effect and both induce coding genes and enhancer RNAs (eRNAs). PAX2–tamoxifen upregulated genes are also enriched with PAX2 eRNAs. The enrichment of eRNAs is associated with the highest expression of genes that positivity regulate apoptotic processes.
    [Show full text]
  • An Immunoevasive Strategy Through Clinically-Relevant Pan-Cancer Genomic and Transcriptomic Alterations of JAK-STAT Signaling Components
    bioRxiv preprint doi: https://doi.org/10.1101/576645; this version posted March 14, 2019. 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-NC-ND 4.0 International license. An immunoevasive strategy through clinically-relevant pan-cancer genomic and transcriptomic alterations of JAK-STAT signaling components Wai Hoong Chang1 and Alvina G. Lai1, 1Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7FZ, United Kingdom Since its discovery almost three decades ago, the Janus ki- Although cytokines are responsible for inflammation in nase (JAK)-signal transducer and activator of transcription cancer, spontaneous eradication of tumors by endoge- (STAT) pathway has paved the road for understanding inflam- nous immune processes rarely occurs. Moreover, the matory and immunity processes related to a wide range of hu- dynamic interaction between tumor cells and host immu- man pathologies including cancer. Several studies have demon- nity shields tumors from immunological ablation, which strated the importance of JAK-STAT pathway components in overall limits the efficacy of immunotherapy in the clinic. regulating tumor initiation and metastatic progression, yet, the extent of how genetic alterations influence patient outcome is far from being understood. Focusing on 133 genes involved in Cytokines can be pro- or anti-inflammatory and are inter- JAK-STAT signaling, we found that copy number alterations dependent on each other’s function to maintain immune underpin transcriptional dysregulation that differs within and homeostasis(3).
    [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]
  • Vs. BCR-ABL-Positive Cells to Interferon Alpha
    Schubert et al. Journal of Hematology & Oncology (2019) 12:36 https://doi.org/10.1186/s13045-019-0722-9 RESEARCH Open Access Differential roles of STAT1 and STAT2 in the sensitivity of JAK2V617F- vs. BCR-ABL- positive cells to interferon alpha Claudia Schubert1, Manuel Allhoff2, Stefan Tillmann1, Tiago Maié2, Ivan G. Costa2, Daniel B. Lipka3, Mirle Schemionek1, Kristina Feldberg1, Julian Baumeister1, Tim H. Brümmendorf1, Nicolas Chatain1† and Steffen Koschmieder1*† Abstract Background: Interferon alpha (IFNa) monotherapy is recommended as the standard therapy in polycythemia vera (PV) but not in chronic myeloid leukemia (CML). Here, we investigated the mechanisms of IFNa efficacy in JAK2V617F- vs. BCR-ABL-positive cells. Methods: Gene expression microarrays and RT-qPCR of PV vs. CML patient PBMCs and CD34+ cells and of the murine cell line 32D expressing JAK2V617F or BCR-ABL were used to analyze and compare interferon-stimulated gene (ISG) expression. Furthermore, using CRISPR/Cas9n technology, targeted disruption of STAT1 or STAT2, respectively, was performed in 32D-BCR-ABL and 32D-JAK2V617F cells to evaluate the role of these transcription factors for IFNa efficacy. The knockout cell lines were reconstituted with STAT1, STAT2, STAT1Y701F, or STAT2Y689F to analyze the importance of wild-type and phosphomutant STATs for the IFNa response. ChIP-seq and ChIP were performed to correlate histone marks with ISG expression. Results: Microarray analysis and RT-qPCR revealed significant upregulation of ISGs in 32D-JAK2V617F but downregulation in 32D-BCR-ABL cells, and these effects were reversed by tyrosine kinase inhibitor (TKI) treatment. Similar expression patterns were confirmed in human cell lines, primary PV and CML patient PBMCs and CD34+ cells, demonstrating that these effects are operational in patients.
    [Show full text]
  • A Dual Cis-Regulatory Code Links IRF8 to Constitutive and Inducible Gene Expression in Macrophages
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press A dual cis-regulatory code links IRF8 to constitutive and inducible gene expression in macrophages Alessandra Mancino,1,3 Alberto Termanini,1,3 Iros Barozzi,1 Serena Ghisletti,1 Renato Ostuni,1 Elena Prosperini,1 Keiko Ozato,2 and Gioacchino Natoli1 1Department of Experimental Oncology, European Institute of Oncology (IEO), 20139 Milan, Italy; 2Laboratory of Molecular Growth Regulation, Genomics of Differentiation Program, National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Maryland 20892, USA The transcription factor (TF) interferon regulatory factor 8 (IRF8) controls both developmental and inflammatory stimulus-inducible genes in macrophages, but the mechanisms underlying these two different functions are largely unknown. One possibility is that these different roles are linked to the ability of IRF8 to bind alternative DNA sequences. We found that IRF8 is recruited to distinct sets of DNA consensus sequences before and after lipopolysaccharide (LPS) stimulation. In resting cells, IRF8 was mainly bound to composite sites together with the master regulator of myeloid development PU.1. Basal IRF8–PU.1 binding maintained the expression of a broad panel of genes essential for macrophage functions (such as microbial recognition and response to purines) and contributed to basal expression of many LPS-inducible genes. After LPS stimulation, increased expression of IRF8, other IRFs, and AP-1 family TFs enabled IRF8 binding to thousands of additional regions containing low-affinity multimerized IRF sites and composite IRF–AP-1 sites, which were not premarked by PU.1 and did not contribute to the basal IRF8 cistrome.
    [Show full text]
  • Induced IFN Regulatory Factor 1 Transcription Factor by Myd88 in Toll-Like Receptor-Dependent Gene Induction Program
    Evidence for licensing of IFN-␥-induced IFN regulatory factor 1 transcription factor by MyD88 in Toll-like receptor-dependent gene induction program Hideo Negishi*, Yasuyuki Fujita*, Hideyuki Yanai*, Shinya Sakaguchi*, Xinshou Ouyang*, Masahiro Shinohara†, Hiroshi Takayanagi†, Yusuke Ohba*, Tadatsugu Taniguchi*‡, and Kenya Honda* *Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan; and †Department of Cell Signaling, Graduate School, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan Contributed by Tadatsugu Taniguchi, August 18, 2006 The recognition of microbial components by Toll-like receptors (TLRs) In the present study we investigated how IFN-␥-induced IRF1 initiates signal transduction pathways, which trigger the expression contributes to TLR-mediated signaling. We demonstrate that of a series of target genes. It has been reported that TLR signaling is IRF1 forms a complex with MyD88, similar to the case of IRF4, enhanced by cytokines such as IFN-␥, but the mechanisms underlying IRF5, and IRF7. We also provide evidence that IRF1 induced this enhancement remain unclear. The MyD88 adaptor, which is by IFN-␥ is activated by MyD88, which we refer to as ‘‘licensing,’’ essential for signaling by many TLRs, recruits members of the IFN and migrates rapidly into the nucleus to mediate an efficient regulatory factor (IRF) family of transcription factors, such as IRF5 and induction of IFN-␤, iNOS, and IL-12p35. Our study therefore IRF7, to evoke the activation of TLR target genes. In this study we revealed that IRF1 is a previously unidentified member of the demonstrate that IRF1, which is induced by IFN-␥, also interacts with multimolecular complex organized via MyD88 and that the IRF1 and is activated by MyD88 upon TLR activation.
    [Show full text]
  • 2958.Full.Pdf
    The Orphan Nuclear Receptor NR4A3 Is Involved in the Function of Dendritic Cells Masanori Nagaoka, Takuya Yashiro, Yuna Uchida, Tomoaki Ando, Mutsuko Hara, Hajime Arai, Hideoki Ogawa, Ko This information is current as Okumura, Kazumi Kasakura and Chiharu Nishiyama of September 27, 2021. J Immunol 2017; 199:2958-2967; Prepublished online 11 September 2017; doi: 10.4049/jimmunol.1601911 http://www.jimmunol.org/content/199/8/2958 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2017/09/09/jimmunol.160191 Material 1.DCSupplemental http://www.jimmunol.org/ References This article cites 23 articles, 10 of which you can access for free at: http://www.jimmunol.org/content/199/8/2958.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 27, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *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 © 2017 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology The Orphan Nuclear Receptor NR4A3 Is Involved in the Function of Dendritic Cells Masanori Nagaoka,*,1 Takuya Yashiro,*,1 Yuna Uchida,* Tomoaki Ando,† Mutsuko Hara,† Hajime Arai,† Hideoki Ogawa,† Ko Okumura,† Kazumi Kasakura,* and Chiharu Nishiyama*,† NR4A3/NOR1 belongs to the NR4A subfamily of the nuclear hormone receptor superfamily, which is activated in a ligand- independent manner.
    [Show full text]
  • Irf1) Signaling Regulates Apoptosis and Autophagy to Determine Endocrine Responsiveness and Cell Fate in Human Breast Cancer
    INTERFERON REGULATORY FACTOR-1 (IRF1) SIGNALING REGULATES APOPTOSIS AND AUTOPHAGY TO DETERMINE ENDOCRINE RESPONSIVENESS AND CELL FATE IN HUMAN BREAST CANCER A Dissertation Submitted to the Faculty of the Graduate School of Arts and Sciences of Georgetown University in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Physiology & Biophysics By Jessica L. Roberts, B.S. Washington, DC September 27, 2013 Copyright 2013 by Jessica L. Roberts All Rights Reserved ii INTERFERON REGULATORY FACTOR-1 (IRF1) SIGNALING REGULATES APOPTOSIS AND AUTOPHAGY TO DETERMINE ENDOCRINE RESPONSIVENESS AND CELL FATE IN HUMAN BREAST CANCER Jessica L. Roberts, B.S. Thesis Advisor: Robert Clarke, Ph.D. ABSTRACT Interferon regulatory factor-1 (IRF1) is a nuclear transcription factor and pivotal regulator of cell fate in cancer cells. While IRF1 is known to possess tumor suppressive activities, the role of IRF1 in mediating apoptosis and autophagy in breast cancer is largely unknown. Here, we show that IRF1 inhibits antiapoptotic B-cell lymphoma 2 (BCL2) protein expression, whose overexpression often contributes to antiestrogen resistance. We proposed that directly targeting the antiapoptotic BCL2 members with GX15-070 (GX; obatoclax), a BH3-mimetic currently in clinical development, would be an attractive strategy to overcome antiestrogen resistance in some breast cancers. Inhibition of BCL2 activity, through treatment with GX, was more effective in reducing the cell density of antiestrogen resistant breast cancer cells versus sensitive cells, and this increased sensitivity correlated with an accumulation of autophagic vacuoles. While GX treatment promoted autophagic vacuole and autolysosome formation, p62/SQSTM1, a marker for autophagic degradation, levels accumulated.
    [Show full text]