STAT3 Activation in Response to IL-6 Is Prolonged by the Binding of IL-6 Receptor to EGF Receptor

Total Page:16

File Type:pdf, Size:1020Kb

STAT3 Activation in Response to IL-6 Is Prolonged by the Binding of IL-6 Receptor to EGF Receptor STAT3 activation in response to IL-6 is prolonged by the binding of IL-6 receptor to EGF receptor Yuxin Wanga,b, Anette H. H. van Boxel-Dezaireb, HyeonJoo Cheonb, Jinbo Yanga,b,1, and George R. Starkb,1 aSchool of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, People’s Republic of China; and bDepartment of Molecular Genetics, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195 Contributed by George R. Stark, August 28, 2013 (sent for review July 19, 2013) The activation of STAT3 by tyrosine phosphorylation, essential for (10), indicating the involvement of IL-6 in the growth and dif- normal development and for a normal inflammatory response to ferentiation of the preimplantation mouse embryo (11). One of invading pathogens, is kept in check by negative regulators. Abnor- the most notable activities of the IL-6 family of cytokines, from mal constitutive activation of STAT3, which contributes to the the viewpoint of embryonic development, is the expression of the pathology of cancer and to chronic inflammatory diseases such as IL-6 family member leukemia inhibitory factor (LIF) in the mu- rheumatoid arthritis, occurs when negative regulation is not fully rine uterine endometrial glands of pregnant mice, coincident with effective. SOCS3, the major negative regulator of STAT3, is induced the onset of blastocyst implantation (12, 13). Mice in which the by tyrosine-phosphorylated STAT3 and terminates STAT3 phosphor- IL-6 or LIF genes have been knocked out show much less severe ylation about 2 h after initial exposure of cells to members of the IL-6 defects than might have been anticipated from the widespread family of cytokines by binding cooperatively to the common receptor effects of these cytokines (14, 15), perhaps reflecting the shared subunit gp130 and JAKs 1 and 2. We show here that when the use of common gp130 signaling pathways by other gp130- epidermal growth factor receptor (EGFR) is present and active, STAT3 dependent cytokines. In addition, intriguingly, mice lacking SOCS3 is rephosphorylated about 4 h after exposure of cells to IL-6 or die between days 11 and 13 of gestation (16), showing the impor- oncostatin M and remains active for many hours. Newly synthesized tance of down-regulation of STAT3 activation in development. IL-6 drives association of the IL-6 receptor and gp130 with EGFR, The STAT transcription factors, and especially STAT3, play leading to EGFR-dependent rephosphorylation of STAT3, which is not vital roles in tumor initiation and progression (17, 18). Aberrant inhibited by the continued presence of SOCS3. This second wave of activation of STAT3 leads to the overexpression of oncogenes STAT3 activation supports sustained expression of a subset of IL-6- that drive proliferation and metastasis and inhibit apoptosis (19). induced proteins, several of which play important roles in in- As an activator of the JAK-STAT signaling pathway, IL-6 can act flammation and cancer, in which both IL-6 secretion and EGFR in both a proinflammatory and anti-inflammatory manner. De- levels are often elevated. regulation of IL-6 production can cause severe disease, such as rheumatoid arthritis, psoriasis, arteriosclerosis, and cancer (20, fter ligand-induced dimerization of the IL-6 receptor (IL-6R), 21). Through the JAK/STAT signaling pathway, IL-6 mediates Athe associated kinases JAK1 and JAK2 cross-phosphorylate the production of IL-17 and IL-10 while regulating TH17, which tyrosine residues of adjacent glycoprotein 130 (gp130) subunits of is important in autoimmunity (22). IL-6 elevation and the fl the complex. The SH2 domain of STAT3 binds to newly phos- chronic in ammation that follows are protumorigenic. IL-6 phorylated tyrosines, followed by the phosphorylation of Y705 of exerts powerful antiapoptotic functions through the induction of STAT3 (1). Two phosphorylated STAT3 monomers then dimerize B-cell lymphoma 2 (BCL-2), B-cell lymphoma-extra large (BCL- – and translocate to the nucleus, where they activate the transcription xL), and related inhibitors (23). Therefore, anti IL-6/IL-6R mono- of many downstream genes whose products mediate the diverse clonal antibodies are used to inhibit aberrant and prolonged IL-6 effects of STAT3 in development and disease (2). Suppressor of fi cytokine signaling 3 (SOCS3), the major negative regulator of IL-6- Signi cance IMMUNOLOGY dependent signaling, is highly induced by activated STAT3, strongly inhibiting further STAT3 phosphorylation and thus collaborating An important part of the response to invading pathogens is with additional negative regulatory mechanisms to prevent excessive activation of the transcription factor STAT3 in response to IL-6. activation of potentially deleterious gene expression (3). SOCS3 This normal response is terminated rapidly by the major neg- down-regulates the phosphorylation of STAT3 about 90 min after ative regulator suppressor of cytokine signaling 3 (SOCS3). an initial exposure to IL-6 (4), using two opposing surfaces to bind Abnormal prolonged activation of STAT3 contributes to the to JAK2 and gp130 simultaneously (5). However, even in the pathology of cancer and to chronic inflammatory diseases. We continued presence of SOCS3, STAT3 is rephosphorylated in re- show that prolonged activation of STAT3 results from associ- sponse to IL-6 after about 4 h through a mechanism that has been ation of the IL-6 receptor with the epidermal growth factor obscure until now. Rephosphorylation of STAT3 might occur be- receptor, which in turn activates STAT3 in a way that is not cause the interaction between SOCS3 and the IL-6 receptor is inhibited by SOCS3. Prolonged STAT3 activation supports somehow prevented or because continued STAT3 phosphor- sustained expression of a subset of proteins that play impor- ylation occurs through a mechanism that is immune to SOCS3. tant roles in inflammation and in cancer, where both IL-6 The epidermal growth factor receptor (EGFR) is well known to secretion and epidermal growth factor receptor levels are catalyze the tyrosine phosphorylation of STAT3 in response to EGF often elevated. (6), and the intrinsic kinase activity of the receptor, but not any of Author contributions: Y.W., A.H.H.v.B.-D., H.C., J.Y., and G.R.S. designed research; Y.W. the JAKs, is required for this reaction (7). On activation by EGF, performed research; Y.W., A.H.H.v.B.-D., H.C., J.Y., and G.R.S. analyzed data; and Y.W. the EGFR dimerizes to facilitate cross-phosphorylation of several and G.R.S. wrote the paper. tyrosine residues, including Y1068, the binding site for STAT3 (8). The authors declare no conflict of interest. Multiple tyrosine phosphorylations of EGFR initiate downstream Freely available online through the PNAS open access option. signaling cascades that facilitate proliferation, regeneration, and 1To whom correspondence may be addressed. E-mail: [email protected] or [email protected]. tumorigenesis (9). cn. During the early phase of mouse development, IL-6 mRNA is This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. differentially expressed and translated into the mature protein 1073/pnas.1315862110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1315862110 PNAS | October 15, 2013 | vol. 110 | no. 42 | 16975–16980 Downloaded by guest on October 5, 2021 signaling in the treatment of autoimmunity and cancer (21). Be- that factors that are newly synthesized and secreted mediate the cause EGFR activation can mediate both IL-6 production (24, 25) second wave. We used a cytokine array to identify such factors by and STAT3 activation, we investigated the role of EGFR in reg- analyzing media conditioned for 4 h after washout from cells ulating STAT3 activation in response to IL-6. treated with IL-6 for 30 min, finding that IL-6 is the dominant We confirm the observation of others (3, 26) that, after the induced factor (Fig. 2C). The supernatant media were also an- initial phosphorylation of STAT3 in response to IL-6 and sub- alyzed by ELISA to quantify IL-6 production. The lack of IL-6 in sequent inhibition by SOCS3, a second wave of activation leads to media collected 10 min after washout shows that the removal of the rephosphorylation of STAT3. We now find that rephosphor- the IL-6 added initially is efficient (Fig. 2D, Left). IL-6 was se- ylation requires new IL-6 synthesis and secretion, leading to ligand- creted by 1 h after washout and remained in the media at dependent association of IL-6R and EGFR. STAT3 phosphory- a similar level for at least 4 h (Fig. 2D, Left). An analysis by real- lation continues to be driven for many hours by this two-receptor time PCR showed that IL-6 mRNA was induced within the first complex, which is immune to inhibition by SOCS3, thus increasing 30 min of initial treatment with IL-6 and then declined (Fig. 2D, and prolonging the expression of many STAT3-dependent proteins Right). An antibody against human IL-6 totally ablated the sec- that are involved in tumorigenesis and immune regulation. ond wave of STAT3 activation in DLD1 cells, showing that no other secreted protein is responsible (Fig. 2E). The same phe- Results nomenon was observed in HepG2 cells (Fig. 2F). In summary, we IL-6 Signaling Is Prolonged in a Biphasic Pattern. To examine the show that treatment with IL-6 induces the synthesis and secre- second wave of STAT3 activation in detail, we treated DLD1 tion of more IL-6 and that this induction is necessary for the (colon cancer cells) with IL-6 for 30 min, followed by washout, second wave of STAT3 activation. and measured STAT3 phosphorylation as a function of time (Fig. 1 A and B). Phosphorylation lasts for many hours, with EGFR Is Required for the Second Wave of STAT3 Phosphorylation.
Recommended publications
  • 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 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]
  • SOCS3 Antibody A
    Revision 1 C 0 2 - t SOCS3 Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 3 2 Web: [email protected] 9 www.cellsignal.com 2 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB H M R Endogenous 26 Rabbit O14543 9021 Product Usage Information 5. Bjørbaek, C. et al. (1998) Mol Cell 1, 619-25. 6. Adams, T.E. et al. (1998) J Biol Chem 273, 1285-7. Application Dilution 7. Soriano, S.F. et al. (2002) J Exp Med 196, 311-21. 8. Emanuelli, B. et al. (2000) J Biol Chem 275, 15985-91. Western Blotting 1:1000 9. Stoiber, D. et al. (1999) J Immunol 163, 2640-7. 10. Stoiber, D. et al. (2001) J Immunol 166, 466-72. Storage 11. Roberts, A.W. et al. (2001) Proc Natl Acad Sci USA 98, 9324-9. 12. Seki, Y. et al. (2003) Nat Med 9, 1047-54. Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% 13. Shouda, T. et al. (2001) J Clin Invest 108, 1781-8. glycerol. Store at –20°C. Do not aliquot the antibody. 14. Fang, M. et al. (2005) Cell Mol Immunol 2, 373-7. 15. Goren, I. et al. (2006) J Invest Dermatol 126, 477-85. Specificity / Sensitivity 16. Mori, H. et al. (2004) Nat Med 10, 739-43. 17.
    [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]
  • Japanese Encephalitis Virus Upregulates the Expression of SOCS3 in Mouse Brain and Raw264.7 Cells
    Viruses2014, 6, 4280-4293; doi:10.3390/v6114280 OPEN ACCESS viruses ISSN 1999-4915 www.mdpi.com/journal/viruses Article Japanese Encephalitis Virus Upregulates the Expression of SOCS3 in Mouse Brain and Raw264.7 Cells Xiangmin Li 1,2, Qiaoyan Zhu 2, Qishu Cao 2, Huanchun Chen 1,2 and Ping Qian 1,2,* 1 State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei, China; E-Mails: [email protected] (X.L.); [email protected] (H.C.) 2 Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China; E-Mails: [email protected] (Q.Z.); [email protected] (Q.C.) * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel./Fax: +86-27-87282608. External Editor: Eric O. Freed Received: 29 August 2014; in revised form: 21 October 2014 / Accepted: 23 October 2014 / Published: 10 November 2014 Abstract: Japanese encephalitis virus (JEV) is one of the pathogens that can invade the central nervous system, causing acute infection and inflammation of brain. SOCS3 protein plays a vital role in immune processes and inflammation of the central nervous system. In this study, Raw264.7 cells and suckling mice were infected with JEV, and SOCS3 expression was analyzed by the gene expression profile, semiquantitative RT-PCR, qRT-PCR, immunohistochemistry (IHC) and Western blot. Results indicated that 520 genes were found to be differentially expressed (fold change ≥ 2.0, p < 0.05) in total. The differentially regulated genes were involved in biological processes, such as stimulus response, biological regulation and immune system processes.
    [Show full text]
  • Suppression of Cytokine Signaling by SOCS3: Characterization of the Mode of Inhibition and the Basis of Its Specificity
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Immunity Article Suppression of Cytokine Signaling by SOCS3: Characterization of the Mode of Inhibition and the Basis of Its Specificity Jeffrey J. Babon,1,2,5,* Nadia J. Kershaw,1,3,5 James M. Murphy,1,2,5 Leila N. Varghese,1,2 Artem Laktyushin,1 Samuel N. Young,1 Isabelle S. Lucet,4 Raymond S. Norton,1,2,6 and Nicos A. Nicola1,2,* 1Walter and Eliza Hall Institute of Medical Research, 1G Royal Pde, Parkville, 3052, VIC, Australia 2The University of Melbourne, Royal Parade, Parkville, 3050, VIC, Australia 3Ludwig Institute for Cancer Research, Royal Pde, Parkville, 3050, VIC, Australia 4Monash University, Wellington Rd, Clayton, 3800, VIC, Australia 5These authors contributed equally to this work 6Present address: Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia *Correspondence: [email protected] (J.J.B.), [email protected] (N.A.N.) DOI 10.1016/j.immuni.2011.12.015 SUMMARY Genetic deletion of each individual JAK leads to various immunological and hematopoietic defects; however, aberrant Janus kinases (JAKs) are key effectors in controlling activation of JAKs can be likewise pathological. Three myelopro- immune responses and maintaining hematopoiesis. liferative disorders (polycythemia vera, essential thrombocythe- SOCS3 (suppressor of cytokine signaling-3) is a mia, and primary myelofibrosis) are caused by a single point major regulator of JAK signaling and here we investi- mutation in JAK2 (JAK2V617F)(James et al., 2005; Levine et al., gate the molecular basis of its mechanism of action.
    [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]
  • Gadd45b Deficiency Promotes Premature Senescence and Skin Aging
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 19 Gadd45b deficiency promotes premature senescence and skin aging Andrew Magimaidas1, Priyanka Madireddi1, Silvia Maifrede1, Kaushiki Mukherjee1, Barbara Hoffman1,2 and Dan A. Liebermann1,2 1 Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA, USA 2 Department of Medical Genetics and Molecular Biochemistry, Temple University School of Medicine, Philadelphia, PA, USA Correspondence to: Dan A. Liebermann, email: [email protected] Keywords: Gadd45b, senescence, oxidative stress, DNA damage, cell cycle arrest, Gerotarget Received: March 17, 2016 Accepted: April 12, 2016 Published: April 20, 2016 ABSTRACT The GADD45 family of proteins functions as stress sensors in response to various physiological and environmental stressors. Here we show that primary mouse embryo fibroblasts (MEFs) from Gadd45b null mice proliferate slowly, accumulate increased levels of DNA damage, and senesce prematurely. The impaired proliferation and increased senescence in Gadd45b null MEFs is partially reversed by culturing at physiological oxygen levels, indicating that Gadd45b deficiency leads to decreased ability to cope with oxidative stress. Interestingly, Gadd45b null MEFs arrest at the G2/M phase of cell cycle, in contrast to other senescent MEFs, which arrest at G1. FACS analysis of phospho-histone H3 staining showed that Gadd45b null MEFs are arrested in G2 phase rather than M phase. H2O2 and UV irradiation, known to increase oxidative stress, also triggered increased senescence in Gadd45b null MEFs compared to wild type MEFs. In vivo evidence for increased senescence in Gadd45b null mice includes the observation that embryos from Gadd45b null mice exhibit increased senescence staining compared to wild type embryos.
    [Show full text]