Epigenetic Disruption of Interferon-Γ Response Through Silencing The

Total Page:16

File Type:pdf, Size:1020Kb

Epigenetic Disruption of Interferon-Γ Response Through Silencing The Oncogene (2008) 27, 5267–5276 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $30.00 www.nature.com/onc ONCOGENOMICS Epigenetic disruption of interferon-c response through silencing the tumor suppressor interferon regulatory factor 8 in nasopharyngeal, esophageal and multiple other carcinomas KY Lee1,11, H Geng1,11,KMNg1,JYu2, A van Hasselt3, Y Cao4, Y-X Zeng5, AHY Wong1, X Wang1, J Ying1, G Srivastava6, ML Lung7, L-D Wang8, TT Kwok9, B-Z Levi10, ATC Chan1, JJY Sung2 and Q Tao1 1Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir YK Pao Center for Cancer, Department of Clinical Oncology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong, China; 2Department of Medicine and Therapeutics, Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China; 3Department of Surgery, Chinese University of Hong Kong, Hong Kong, China; 4Hunan Yale (Xiang Ya) School of Medicine, Central South University, Changsha, China; 5State Key Laboratory in Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China; 6Department of Pathology, University of Hong Kong, Hong Kong, China; 7Department of Biology, Hong Kong University of Science and Technology, Hong Kong, China; 8Henan Key Laboratory for Esophageal Cancer, Zhengzhou University College of Medicine and Cancer Research Center of Xinxiang Medical College, Henan, China; 9Department of Biochemistry, Chinese University of Hong Kong, Hong Kong, China and 10Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel 16q24 is frequently deleted in multiple tumors including frequently silenced by epigenetic mechanism in multiple cancers of nasopharynx, esophagus, breast, prostate and carcinomas. liver. By array comparative genomic hybridization Oncogene (2008) 27, 5267–5276; doi:10.1038/onc.2008.147; (aCGH), we refined a 16q24 hemizygous deletion in published online 12 May 2008 nasopharyngeal carcinoma (NPC) cell lines. Semi-quanti- tative RT–PCR analysis revealed interferon regulatory Keywords: IRF8; methylation; CpG island; tumor factor 8 (IRF8) as the only downregulated gene within this suppressor gene; carcinoma deletion. IRF8 belongs to a family of interferon (IFN) regulatory factors that modulate various important physiologic processes including host defense, cell growth and differentiation and immune regulation. In contrast to the broad expression of IRF8 in normal adult and fetal Introduction tissues, transcriptional silencing and promoter methyla- tion of IRF8 were frequently detected in multiple Carcinogenesis is a multistep process with the accumu- carcinoma (except for hepatocellular) cell lines (100% lation of multiple genetic and epigenetic changes (Jones in NPC, 88% in esophageal and 18–78% in other and Baylin, 2002). In addition to the contribution of carcinoma cell lines) and in a large collection of oncogenes, inactivation of tumor suppressor genes primary carcinomas (78% in NPC, 36–71% in other (TSGs) is also frequently involved (Jones and Baylin, carcinomas). Methylation of the IRF8 promoter led to the 2002; Baylin and Ohm, 2006). Genetic mutations disruption of its response to IFN-c stimulation. Pharma- including deletions disrupt TSG functions (Knudson, cological and genetic demethylation could restore IRF8 2001), whereas epigenetic silencing also inactivates expression, indicating a direct epigenetic mechanism. TSGs through methylation of promoter CpG islands Ectopic expression of IRF8 in tumor cells lacking its (CGIs), which is a characteristic epigenetic feature of ONCOGENOMICS expression strongly inhibited their clonogenicity, confirm- tumor DNA (Jones and Baylin, 2002). ing its tumor suppressor function. Thus, IRF8 was Nasopharyngeal carcinoma (NPC) is prevalent in identified as a functional tumor suppressor, which is Southern China and Southeast Asia (Tao and Chan, 2007). Although its pathogenesis has been shown to be strongly associated with Epstein–Barr virus, its mole- Correspondence: Professor Q Tao, Cancer Epigenetics Laboratory, cular mechanism is still poorly elucidated (Lo et al., Department of Clinical Oncology, Sir YK Pao Center for Cancer, 2004; Tao and Chan, 2007). Numerous genetic altera- Prince of Wales Hospital, Chinese University of Hong Kong, tions have been detected in NPC cell lines and tumors, Hong Kong, China. including 3p14–22, 11q13.3–24, 13q14.3–22, 14q24.3–32.1 E-mail: [email protected] and 16q21–24 (Chen et al., 1999; Lo et al., 2000; 11These authors contributed equally to this work. Received 7 November 2007; revised 21 March 2008; accepted 1 April Tao and Chan, 2007). These regions thus represent 2008; published online 12 May 2008 the critical TSG loci for NPC. Previous searches for Methylation of IRF8 in multiple carcinomas KY Lee et al 5268 putative TSGs in these regions have identified only few clone to locus 16q24.1, spanning B160-kb, with three candidates, with tumor-specific promoter methylation, genes, COX4NB, COX4I1 and IRF8 residing at this such as BLU and RASSF1A at 3p21 (Lo et al., 2001; locus (Figure 1a). Semi-quantitative RT–PCR analysis Qiu et al., 2004), CADM1/TSLC1 at 11q23.1 (Lung revealed IRF8 silencing in 5 of 6 NPC cell lines with et al., 2006); THY1/CD90 at 11q22.3 (Lung et al., 2005), weak expression in C666-1, but not for the other two CDH1 at 16q22.1 (Chang et al., 2003), ADAMTS18 and genes COX4NB and COX4I1, suggesting that IRF8 is CDH13 at 16q23 (Sun et al., 2007; Jin et al., 2007b). the candidate TSG located at the 16q24.1 deletion These limited findings suggest that additional cancer- (Figure 1b). related genes are yet to be identified in NPC in the reported regions, or other unidentified loci. Frequent downregulation of IRF8 in multiple carcinomas In this study, we have refined a 16q24.1 deletion as We further determined IRF8 expression in a panel of one of the critical tumor suppressor loci in NPC cell normal adult and fetal tissues and multiple cell lines by lines using high-resolution, 1-Mb array comparative semi-quantitative RT–PCR. IRF8 expression was de- genomic hybridization (aCGH) analysis (Ying et al., tected in all normal tissues including nasopharynx and 2006). Loss of heterozygosity (LOH) at 16q24 is also esophagus (Figure 1d), as well as immortalized normal frequently present in multiple other solid tumors, such nasopharyngeal (NP69) and esophageal epithelial (NE1 as breast, prostate, hepatocellular and Wilm’s tumor and NE3) cell lines (Figure 2a). In contrast, IRF8 was (Lo et al., 2000; Paige et al., 2000; Jin et al., 2007b), frequently downregulated or silenced in carcinoma cell suggesting the presence of critical TSG(s) at this locus. lines of nasopharyngeal, esophageal, breast, lung, Several functional candidate TSGs have been identified cervical and colorectal carcinomas (Figure 2a), but less at this locus including CDH13 (Toyooka et al., 2001). frequently in gastric and seldom in hepatocellular Thus, it is needed to search thoroughly for more carcinoma cell lines (Supplementary Figure). Mean- TSGs at this locus and define their functions in while, expression of the IRF8 protein was confirmed by tumors. western blot in normal esophageal epithelial tissues and Among the genes resided at the 16q24.1 deletion, normal lymphoid cell line LCL-CCL256.1 (Figure 2b). silencing of interferon regulatory factor 8 (IRF8) was However, IRF8 was absent in HONE1 and HCT116 identified in several NPC cell lines. IRF8 is also known cell lines that are completely methylated and silenced for as interferon consensus sequence-binding protein this gene. (ICSBP), which belongs to the IRF family of transcrip- tion factors (Nguyen et al., 1997). IRF8 has been characterized as a central element of the interferon Methylation of the IRF8 promoter correlates (IFN)-g-signaling pathway, which modulates immune with its transcriptional silencing response and also regulates cell growth and differentia- We then examined the possible genetic/epigenetic tion (Tamura and Ozato, 2002). IRF8-deficient mice mechanisms of IRF8 downregulation. As predicted by developed a human chronic myelogenous leukemia online CpG Island Searcher (http://cpgislands.usc.edu/), (CML)-like disease (Holtschke et al., 1996). In patients there is a CGI spanning the transcription start site of with CML and acute myelogenous leukemia, IRF8 IRF8 (Figure 1c). Methylation-specific PCR (MSP) was expression was dramatically decreased, suggesting that thus performed to study the methylation status of this IRF8 could be a candidate TSG (Schmidt et al., 1998). CGI. Results showed that IRF8 was frequently methy- Here, we report the frequent epigenetic inactivation of lated in cell lines with downregulation (6 of 6 IRF8 in NPC, as well as other common carcinomas, nasopharyngeal, 14 of 16 esophageal, 7 of 9 breast, 3 such as esophageal, breast, cervical, lung and colorectal of 4 colorectal, 5 of 7 lung, 4 of 6 cervical and 3 of 17 carcinomas. Methylation of the IRF8 promoter gastric carcinoma cell lines) (Figure 2a, Supplementary abolishes its response to IFN-g. Moreover, ectopic Figure, Table 1), whereas no methylation was detected expression of IRF8 in silenced tumor cells strongly in the immortalized epithelial cell lines (NP69, NE1 and inhibited their clonogenicity, indicating that IRF8 is a NE3), indicating that methylation is tumor specific functional TSG. (Figure 2a). The presence of either unmethylated or methylated promoter alleles in virtually all cell lines including silenced ones
Recommended publications
  • 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]
  • 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]
  • 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]
  • 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]
  • 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]
  • 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]
  • 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]
  • Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function
    REVIEW published: 10 February 2017 doi: 10.3389/fimmu.2017.00108 Estrogen Receptor-Dependent Regulation of Dendritic Cell Development and Function Sophie Laffont1*, Cyril Seillet2,3 and Jean-Charles Guéry1* 1 Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France, 2 Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia, 3 Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia Autoimmunity, infectious diseases and cancer affect women and men differently. Because they tend to develop more vigorous adaptive immune responses than men, women are less susceptible to some infectious diseases but also at higher risk of autoimmunity. The regulation of immune responses by sex-dependent factors probably involves several non-redundant mechanisms. A privileged area of study, however, concerns the role of sex steroid hormones in the biology of innate immune cells, especially dendritic cells (DCs). In recent years, our understanding of the lineage origin of DC populations has expanded, Edited by: and the lineage-committing transcription factors shaping peripheral DC subsets have Manfred B. Lutz, been identified. Both progenitor cells and mature DC subsets express estrogen receptors University of Würzburg, Germany (ERs), which are ligand-dependent transcription factors. This suggests that estrogens Reviewed by: may contribute to the reported sex differences in immunity by regulating DC biology. Meredith O’Keeffe, Monash University, Australia Here, we review the recent literature and highlight evidence that estrogen-dependent Pieter J. M. Leenen, activation of ERα regulates the development or the functional responses of particular DC Erasmus University Rotterdam, + Netherlands subsets.
    [Show full text]
  • Stromal Cell Signature Associated with Response to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer
    cells Article Stromal Cell Signature Associated with Response to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer 1, 2, 3 Maria Lucia Hirata Katayama y, René Aloísio da Costa Vieira y, Victor Piana Andrade , Rosimeire Aparecida Roela 1, Luiz Guilherme Cernaglia Aureliano Lima 3, Ligia Maria Kerr 4, Adriano Polpo de Campos 5,6, Carlos Alberto de Bragança Pereira 7, Pedro Adolpho de Menezes Pacheco Serio 1, Giselly Encinas 1, Simone Maistro 1, Matheus de Almeida Leite Petroni 1, Maria Mitzi Brentani 1 and Maria Aparecida Azevedo Koike Folgueira 1,* 1 Departamento de Radiologia e Oncologia, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-000, SP, Brazil; [email protected] (M.L.H.K.); [email protected] (R.A.R.); [email protected] (P.A.d.M.P.S.); [email protected] (G.E.); [email protected] (S.M.); [email protected] (M.d.A.L.P.); [email protected] (M.M.B.) 2 Departamento de Mastologia, Hospital de Câncer de Barretos, Barretos 14.784-400, SP, Brazil; [email protected] 3 A.C. Camargo Cancer Center, Sao Paulo 01525-001, SP, Brazil; [email protected] (V.P.A.); [email protected] (L.G.C.A.L.) 4 Departamento de Patologia, Hospital de Câncer de Barretos, Barretos 14.784-400, SP, Brazil; [email protected] 5 Departamento de Estatística, Centro de Ciências Exatas e de Tecnologia, Universidade Federal de São Carlos, Sao Carlos 13565-905, SP, Brazil; [email protected] 6 Department of Mathematics and Statistics, The University of Western Australia, M019, 35 Stirling Highway, 6009 Crawley, WA, Australia 7 Departamento de Estatística, Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo 05508-090, SP, Brazil; [email protected] * Correspondence: [email protected] These authors participated equally in this work.
    [Show full text]