Irfs As Competing Pioneers in T-Cell Differentiation

Total Page:16

File Type:pdf, Size:1020Kb

Irfs As Competing Pioneers in T-Cell Differentiation Cellular & Molecular Immunology (2017) 14, 649–651 & 2017 CSI and USTC All rights reserved 2042-0226/17 $32.00 www.nature.com/cmi RESEARCH HIGHLIGHT IRFs as competing pioneers in T-cell differentiation Andrea Kröger Cellular & Molecular Immunology (2017) 14, 649–651; doi:10.1038/cmi.2017.37; published online 19 June 2017 CD4-expressing T lymphocytes are combination with IL-6 or IL-21 promote engagement of TCRs on naive CD4+ among of the best-characterized immune TH17 development. These cells produce T cells cultured under Tr1 polarizing system cells.1 They develop in the thy- IL-17, IL-21 and IL-22 and express the conditions. Irf1 and Batf mRNA mus and acquire the potential to differ- lineage-specific transcription factor showed a dual wave expression profile entiate into distinct subpopulations. RORγt. Regulatory T cells (nTreg) can with an early peak between 1 and 6 h Upon leaving the thymus, T helper cells develop directly in the thymus; they after activation. Moreover, stimulation further differentiate in the periphery in express FOXP3 and inhibit T effector of naive CD4+ T cells from knockout response to signals from T-cell receptors, cell functions. Tr1 cells are poorly failed to produce Il10 mRNA or differ- cytokines and other ligands in the micro- defined regulatory T cells;2 they are entiation into Tr1 cells. environment. CD4+ T cells have high FOXP3 negative and express high levels IL-27 receptor signaling is known to plasticity and can differentiate into sev- of anti-inflammatory cytokines like induce Tr1 cell differentiation. Further- eral different subtypes. This subdivision IL-10. more, both STAT1 and STAT3 are is based on the expression of lineage- The diversity of the differentiation required for Tr1 differentiation. Karwacz specific transcription factors, which possibilities of CD4+ T cells raises the and colleagues showed that STAT1 sig- function as ‘mater regulators’ for specific question as to the degree to which a naling was responsible for Irf1 induction, TH subsets. single master regulator contributes to the whereas STAT3 was essential for BATF, TH1 differentiation is driven by IL-12, differentiation, especially because in indicating a distinct regulation of both which promotes the expression of IFNγ some cells similar factors are expressed. transcription factors. The authors also and transcription factor T-BET (encoded Gene expression analysis is typically car- investigated whether both transcription by T-box 21). TH2 cells express the ried out by analyzing induced genes but factors cooperate in Tr1-specificgene master regulator GATA-binding protein does not explain why expression of the expression, whereas IRF1 expression 3 (GATA3) when induced by IL-4 and same transcription factor in the same cell leads to high induction of IL-10. Batf secrete IL-4, IL-5 and IL-13. Transform- types leads to different gene subsets. The overexpression strongly induced Maf and ing growth factor β (TGFβ), in combina- combination of gene expression and Il12,whereasAhr expression was tion with IL-4, induces differentiation to chromatin accessibility analysis allows induced by both. These results suggest TH9 cells, which produce IL-9 and IL-10. analysis of another level of gene regula- that both transcription factors cooperate PU.1 has been suggested as a master tion. Pioneering factors that alter chro- in gene induction and induce a distinct transcription factor. T follicular helper matin also affect the accessibility of set of genes. (TfH) cells are induced by IL-6 and IL-21 transcription factors to specificgenes. To assess the role of IRF1 in auto- and express master regulator BCL6. For the differentiation of TH17 cells, immune disease, Karwacz and coauthors In addition, TGFβ and IL-23 in the cooperative binding of BATF and used an experimental autoimmune ence- interferon regulatory factors (IRF4) phalomyelitis (EAE) model. In previous enables chromatin accessibility and the studies, C57/BL6J Irf1-deficient mice Institute for Medical Microbiology, Molecular subsequent recruitment of RORγtto were found to be resistant to EAE upon Microbiology, Otto-von-Guericke University of regulate selected sets of TH17-relevant immunization with MOG35-55 when Magdeburg, Magdeburg D-39120, Germany genes3–5 (Figure 1). compared with wild-type mice.7,8 This Correspondence: Professor A Kröger, PhD, Group leader, Principle Investigator, Institute for Medical In a recent issue of Nature Immunol- phenotypic characterization was asso- Microbiology, Molecular Microbiology, Otto-von- ogy, Karwacz et al.6 exploited the role ciated with impaired Th1 cell differentia- Guericke University of Magdeburg, Leipziger of IRF1 and BATF as pioneering factors tion and biased development toward Th2 Straße 44, Magdeburg D-39120, Germany. for CD4+ T-cell differentiation to cell phenotype. Thus, Karwacz showed E-mail: [email protected] Received: 21 April 2017; Accepted: 23 April Tr1 cells. The investigators analyzed that loss of IRF1 led to an increased 2017 global gene expression following clinical core upon EAE induction and Research Highlight 650 BATF IRF4 IRF1 IL12 TH1 IFNγ T-BET TNFα IL4 IL4 TH2 GATA3 IL5 IL13 IL4 TH9 IL9 +TGFβ IL10 Naive IL6 T 17 IL17 CD4+ H IL21 +TGFβ RORγt T cell IL22 IL21 Tfh IL21 IL6 BCL6 Tr1 IL10 Treg FOXP3 IL10 + + Figure 1 IRFs are crucial for the differentiation of CD4 T-cell subsets. CD4 T cells developed in functionally different subsets (TH1, TH2, TH9, TH17, Tfh, Tr1 Treg), some of them are characterized by the expression of lineage-specific transcription factors (T-BET, GATA3, RORγt or FOXP3). BATF, as a pioneer factor, is expressed in different lineages. IRF1 and IRF4 could cooperate with BATF to landscape the chromatin to promote lineage-specific gene expression. IRF, interferon regulatory factors. increased numbers of IL-17- and IFNγ- IRF1 in TH17 cells and vice versa. Over- Karwacz and colleagues used intensive expressing T cells in the central nervous expression of IRF1 in TH17 cells antag- ATAC-seq analyses of Tr1 polarized system of Irf1−/− mice. More impor- onized IRF4-induced IL-14, and the knockout cells to assess the potential tantly, the transfer of Irf1−/− Tcellsinto overexpression of IRF4 in Tr1 cells pioneering roles of BATF and IRF1 Rag2−/− mice and induction of EAE inhibited IRF1-mediated IL-10. Thus, during Tr1 differentiation. They detected resulted in increased disease scores; IRF1 may play the same role as epige- ~ 180 000 accessible regions across the higher numbers of IL-17 producing cells netic pioneer factors in Tr1 cells, such as samples. In contrast to Irf1, c-Maf and −/− were isolated. These data provide evi- IRF4 in TH17 cells. AhR, Batf TH0 and Tr1 cells had dence that IRF1 inhibited Th17 cell To investigate whether BATF or IRF1 distinct chromatin accessibility patterns, differentiation in vivo. Furthermore, could affect epigenetic markers, ChIP- providing high-level support for BATF as Irf1-andBatf-deficient mice were not qPCR assays on the IL-10 locus were a pioneering factor in Tr1 cells. DESeq2 able to generate functional Tr1 cells, performed in Tr1 polarized knockout analysis was used to quantify the number whereas the adaptive transfer of wt cells. Irf1 and Batf deficiency resulted in of transcription-factor-dependent acces- Tr1 cells inhibits disease and the transfer decreased recruitment of activating fac- sible peaks per transcription factor in of Irf1-deficient Tr1 cells exacerbated tors and increased repressive factors. Tr1 conditions. BATF deficiency led to disease. Thus, IRF1 and BATF control Thus, both transcription factors seem altered accessibility at over 20 000 loci, the in vivo generation of Tr1 cells. to induce epigenetic changes during Tr1 IRF1 deficiency of 1200 loci and only Tr1 cells are characterized by high cell differentiation. Reporter assays using minor alterations in Ahr and c-Mafs cells. expression levels of IL-10. To determine promoter elements with high BATF and Although the scale of IRF1- and BATF- whether IRF1 and BATF have to interact IRF1 co-occupancy showed that only dependent changes differs, both tran- on the Il10 promoter, Karwacz et al. IRF1 was able to directly induce the scription factors altered chromatin acces- performed sequential ChIP assays and IL-10 promoter. Moreover, both factors sibility, whereas c-MAF and AhR did showed a strong co-occupancy of IRF-1 provide binding sites for other transcrip- not. Thus, both IRF1 and BATF are and BATF. Because IRF4 and IRF1 share tion factors, suggesting that both factors necessary for additional Tr1 transcription the same DBD, and IRF4 and BATF could induce chromatin changes, which factorstobindchromatinandsupport show a similar dependency in TH17 cells, allow the exclusive or cooperative bind- the role for IRF1 and BATF as pioneer- they also investigated the function of ing of other transcription factors. ing factors. Cellular & Molecular Immunology Research Highlight 651 Taking the findings together this CONFLICT OF INTEREST 5 Ciofani M, Madar A, Galan C, Sellars M, study uncovered the critical role of Theauthordeclaresnoconflict of interest. Mace K, Pauli F et al. A validated regulatory network for Th17 cell specification. Cell IRF1 and BATF in preparing the chro- 2012; 151:289–303. matin landscape for the induction of 6 Karwacz K, Miraldi ER, Pokrovskii M, the Tr1 gene network in response to 1 Zhu J, Yamane H, Paul WE. Differentiation of Madi A, Yosef N, Wortman I et al. Critical IL-27 signaling. Where BATF acts as a effector CD4 T cell populations. Annu Rev role of IRF1 and BATF in forming chromatin Immunol 2010; 28:445–489. landscape during type 1 regulatory cell dif- pioneer factor and prepares the geno- 2 Zeng H, Zhang R, Jin B, Chen L. Type 1 ferentiation. Nat Immunol 2017; 18: mic landscape for the binding of addi- regulatory T cells: a new mechanism of 412–421. tional transcription factors that define peripheral immune tolerance. Cell Mol 7 Tada Y, Ho A, Matsuyama T, Mak TW.
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]
  • 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]
  • 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]
  • 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]
  • Transcription Factor IRF4 Drives Dendritic Cells to Promote Th2 Differentiation
    ARTICLE Received 30 May 2013 | Accepted 21 Nov 2013 | Published 20 Dec 2013 DOI: 10.1038/ncomms3990 Transcription factor IRF4 drives dendritic cells to promote Th2 differentiation Jesse W. Williams1, Melissa Y. Tjota2,3, Bryan S. Clay2, Bryan Vander Lugt4, Hozefa S. Bandukwala2, Cara L. Hrusch5, Donna C. Decker5, Kelly M. Blaine5, Bethany R. Fixsen5, Harinder Singh4, Roger Sciammas6 & Anne I. Sperling1,2,5 Atopic asthma is an inflammatory pulmonary disease associated with Th2 adaptive immune responses triggered by innocuous antigens. While dendritic cells (DCs) are known to shape the adaptive immune response, the mechanisms by which DCs promote Th2 differentiation remain elusive. Herein we demonstrate that Th2-promoting stimuli induce DC expression of IRF4. Mice with conditional deletion of Irf4 in DCs show a dramatic defect in Th2-type lung inflammation, yet retain the ability to elicit pulmonary Th1 antiviral responses. Using loss- and gain-of-function analysis, we demonstrate that Th2 differentiation is dependent on IRF4 expression in DCs. Finally, IRF4 directly targets and activates the Il-10 and Il-33 genes in DCs. Reconstitution with exogenous IL-10 and IL-33 recovers the ability of Irf4-deficient DCs to promote Th2 differentiation. These findings reveal a regulatory module in DCs by which IRF4 modulates IL-10 and IL-33 cytokine production to specifically promote Th2 differentiation and inflammation. 1 Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, 924 E. 57th Street, Chicago, Illinois 60637 USA. 2 Committee on Immunology, University of Chicago, 924 E. 57th Street, Chicago, Illinois 60637 USA. 3 Medical Scientist Training Program, University of Chicago, 924 E.
    [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]
  • 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]
  • PU.1 Cooperates with IRF4 and IRF8 to Suppress Pre-B-Cell Leukemia
    Leukemia (2016) 30, 1375–1387 © 2016 Macmillan Publishers Limited All rights reserved 0887-6924/16 www.nature.com/leu ORIGINAL ARTICLE PU.1 cooperates with IRF4 and IRF8 to suppress pre-B-cell leukemia SHM Pang1,2, M Minnich3, P Gangatirkar1,2, Z Zheng1, A Ebert3, G Song4, RA Dickins1,2,5, LM Corcoran1,2, CG Mullighan4, M Busslinger3, ND Huntington1,2, SL Nutt1,2 and S Carotta1,2,6 The Ets family transcription factor PU.1 and the interferon regulatory factor (IRF)4 and IRF8 regulate gene expression by binding to composite DNA sequences known as Ets/interferon consensus elements. Although all three factors are expressed from the onset of B-cell development, single deficiency of these factors in B-cell progenitors only mildly impacts on bone marrow B lymphopoiesis. Here we tested whether PU.1 cooperates with IRF factors in regulating early B-cell development. Lack of PU.1 and IRF4 resulted in a partial block in development the pre-B-cell stage. The combined deletion of PU.1 and IRF8 reduced recirculating B-cell numbers. Strikingly, all PU.1/IRF4 and ~ 50% of PU.1/IRF8 double deficient mice developed pre-B-cell acute lymphoblastic leukemia (B-ALL) associated with reduced expression of the established B-lineage tumor suppressor genes, Ikaros and Spi-B. These genes are directly regulated by PU.1/IRF4/IRF8, and restoration of Ikaros or Spi-B expression inhibited leukemic cell growth. In summary, we demonstrate that PU.1, IRF4 and IRF8 cooperate to regulate early B-cell development and to prevent pre-B-ALL formation.
    [Show full text]
  • Phosphorylation of Microglial IRF5 and IRF4 by IRAK4 Regulates Inflammatory Responses to Ischemia
    cells Article Phosphorylation of Microglial IRF5 and IRF4 by IRAK4 Regulates Inflammatory Responses to Ischemia Conelius Ngwa, Abdullah Al Mamun, Yan Xu, Romana Sharmeen and Fudong Liu * Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; [email protected] (C.N.); [email protected] (A.A.M.); [email protected] (Y.X.); [email protected] (R.S.) * Correspondence: [email protected]; Tel.: +1-713-500-7038; Fax: +1-713-500-0660 Abstract: Background: Interferon Regulatory Factor (IRF) 5 and 4 play a determinant role in regu- lating microglial pro- and anti-inflammatory responses to cerebral ischemia. How microglial IRF5 and IRF4 signaling are activated has been elusive. We hypothesized that interleukin-1 receptor associated kinase 4 (IRAK4) phosphorylates and activates IRF5 and IRF4 in ischemic microglia. We aimed to explore the upstream signals of the two IRFs, and to determine how the IRAK4-IRF signaling regulates the expression of inflammatory mediators, and impacts neuropathology. Meth- ods: Spontaneously Immortalized Murine (SIM)-A9 microglial cell line, primary microglia and neurons from C57BL/6 WT mice were cultured and exposed to oxygen-glucose deprivation (OGD), followed by stimulation with LPS or IL-4. An IRAK4 inhibitor (ND2158) was used to examine IRAK40s effects on the phosphorylation of IRF5/IRF4 and the impacts on neuronal morphology by co-immunoprecipitation (Co-IP)/Western blot, ELISA, and immunofluorescence assays. Results: We confirmed that IRAK4 formed a Myddosome with MyD88/IRF5/IRF4, and phosphorylated both IRFs, which subsequently translocated into the nucleus.
    [Show full text]
  • Differential Expression of IFN Regulatory Factor 4 Gene in Human Monocyte-Derived Dendritic Cells and Macrophages
    Differential Expression of IFN Regulatory Factor 4 Gene in Human Monocyte-Derived Dendritic Cells and Macrophages This information is current as Anne Lehtonen, Ville Veckman, Tuomas Nikula, Riitta of September 27, 2021. Lahesmaa, Leena Kinnunen, Sampsa Matikainen and Ilkka Julkunen J Immunol 2005; 175:6570-6579; ; doi: 10.4049/jimmunol.175.10.6570 http://www.jimmunol.org/content/175/10/6570 Downloaded from References This article cites 79 articles, 45 of which you can access for free at: http://www.jimmunol.org/content/175/10/6570.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 27, 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 Differential Expression of IFN Regulatory Factor 4 Gene in Human Monocyte-Derived Dendritic Cells and Macrophages1 Anne Lehtonen,2* Ville Veckman,* Tuomas Nikula,‡ Riitta Lahesmaa,‡ Leena Kinnunen,† Sampsa Matikainen,* and Ilkka Julkunen* In vitro human monocyte differentiation to macrophages or dendritic cells (DCs) is driven by GM-CSF or GM-CSF and IL-4, respectively.
    [Show full text]
  • The Molecular Choreography of IRF4 and IRF8 with Immune System Partners
    The Molecular Choreography of IRF4 and IRF8 with Immune System Partners 1,2,4 1,3 1 HARINDER SINGH, ELKE GLASMACHER, ABRAHAM B. CHANG, 1 AND BRYAN VANDER LUGT 1Department of Discovery Immunology, Genentech Inc., South San Francisco, California 94080 4Correspondence: [email protected] The transcription factors IRF4 and IRF8 represent immune-specific members of the interferon regulatory family. They play major roles in controlling the development and functioning of innate and adaptive cells. Genes encoding these factors appear to have been coopted by the immune system via gene duplication and divergence of regulatory and protein coding sequences to enable the acquisition of unique molecular properties and functions. Unlike other members of the IRF family, IRF4 and IRF8 do not activate transcription of Type 1 interferon genes or positively regulate interferon-induced gene expression. Instead, they bind to unusual composite Ets-IRF or AP-1-IRF elements with specific Ets or AP-1 family transcription factors, respectively, and regulate the expression of diverse sets of immune response genes in innate as well as adaptive cells. The molecular cloning of interferon regulatory factor et al. 2012). Strikingly, this involves cooperative assembly 8 (IRF8) as interferon consensus sequence-binding pro- of IRF4 with AP-1 heterodimers containing a basic leucine tein (ICSBP) led to the realization that it bound inter- zipper transcription factor, AFT-like (BATF) subunit. feron response sequence elements (ISRE), albeit with This molecular property is shared by IRF8 but not by other low affinity, and antagonized ISRE-mediated gene acti- IRF family members, reminiscent of the molecular specif- vation (Driggers et al.
    [Show full text]
  • Deletion of Irf4 in T Cells Suppressed Autoimmune Uveitis and Dysregulated Transcriptional Programs Linked to CD4+ T Cell Differentiation and Metabolism
    International Journal of Molecular Sciences Article Deletion of Irf4 in T Cells Suppressed Autoimmune Uveitis and Dysregulated Transcriptional Programs Linked to CD4+ T Cell Differentiation and Metabolism Minkyung Kang 1,†, Hyun-Su Lee 1,†, Jin Kyeong Choi 1,2, Cheng-Rong Yu 1 and Charles E. Egwuagu 1,* 1 Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institute of Health, Bethesda, MD 20892, USA; [email protected] (M.K.); [email protected] (H.-S.L.); [email protected] (J.K.C.); [email protected] (C.-R.Y.) 2 Department of Immunology, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54907, Korea * Correspondence: [email protected]; Tel.: +301-496-0049; Fax: +301-480-3914 † These authors contributed equally. Abstract: Interferon regulatory factor-4 (IRF4) and IRF8 regulate differentiation, growth and functions of lymphoid and myeloid cells. Targeted deletion of irf8 in T cells (CD4-IRF8KO) has been shown to exacerbate colitis and experimental autoimmune uveitis (EAU), a mouse model of human uveitis. We therefore generated mice lacking irf4 in T cells (CD4-IRF4KO) and investigated whether expression of IRF4 by T cells is also required for regulating T cells that suppress autoimmune diseases. Surprisingly, we found that CD4-IRF4KO mice are resistant to EAU. Suppression of EAU derived in part from inhibiting pathogenic responses of Th17 cells while inducing expansion of regulatory lymphocytes that secrete IL-10 and/or IL-35 in the eye and peripheral lymphoid tissues. Furthermore, CD4- IRF4KO T cells exhibit alterations in cell metabolism and are defective in the expression of two Citation: Kang, M.; Lee, H.-S.; Choi, Ikaros zinc-finger (IKZF) transcription factors (Ikaros, Aiolos) that are required for lymphocyte J.K.; Yu, C.-R.; Egwuagu, C.E.
    [Show full text]