The Impact of Vitamin D on Innate Immune Responsiveness to Pattern Recognition Receptor Stimulation in Humans

Total Page:16

File Type:pdf, Size:1020Kb

The Impact of Vitamin D on Innate Immune Responsiveness to Pattern Recognition Receptor Stimulation in Humans The Impact of Vitamin D on Innate Immune Responsiveness to Pattern Recognition Receptor Stimulation in Humans by Natascha Fitch A Thesis submitted to the Faculty of Graduate Studies of The University of Manitoba in partial fulfillment of the requirements of the degree of MASTER OF SCIENCE Department of Immunology University of Manitoba Winnipeg Copyright © 2013 by Natascha Fitch Abstract Objective: Study the effects of vitamin D on viral driven innate immune responses, by looking at differences in cytokine production, receptor expression, and endogenous vitamin D levels. Methods: Primary peripheral blood mononuclear cells (PBMC) and epithelial cells (EC) were cultured in the presence of viral ligands and vitamin D. Enzyme-linked immunosorbent assay (ELISA) and real-time polymerase chain reaction (RT-PCR) were used to determine cytokine production and mRNA expression. Results: PBMC stimulated with toll-like receptor 4 ligand (TLR4L), but not viral TLR8L, led to decreased pro- and anti-inflammatory cytokine production in the presence of 1,25(OH)2D3. RIG-like receptor (RLR) activation, on the other hand, in primary EC exhibited decreased pro-inflammatory cytokine production in the presence of vitamin D. Conclusions: Our findings are among the first to show differences between bacterial and viral driven innate immune responses in the presence of vitamin D. As responsiveness in RLR activated primary EC was altered in the presence of vitamin D, our data reveal the importance of studying the immune system as a whole. i Acknowledgments I would like to thank my supervisor Dr. Kent HayGlass for his guidance and support throughout my program. In particular, I have benefitted from his extensive knowledge, and constructive analysis, but also his ability to present science in a logical and meaningful manner. As a result, completing my Master’s Degree under his supervision has been both enjoyable and productive. I would also like to thank all the members of the HayGlass lab for providing a positive training environment and for being wonderful colleagues. A special thanks to Larisa Lotoski for always making the time to answer my questions and for being a great mentor. I am especially grateful to Rishma Chooniedass and the rest of the MICH Allergy Lab/CHILD team for their immense contribution towards the research and data sample collection upon which my thesis has been developed. To my committee members, Dr. Allan Becker, Dr. Neeloffer Mookerjee and Dr. Carla Taylor, thank you for your support and constructive feedback over the last three years and for reviewing my thesis. Lastly, I would like to acknowledge MHRC, NSERC and Mindel and Tom Olenick for providing financial support to me throughout my degree, as well as CIHR for supporting the research conducted in our lab. To all the members of the department, thank you for making this degree both fun and challenging and for always providing great feedback. ii Dedications I would like to dedicate this thesis to my husband, parents and sister. Thank-you all for you continuous support, especially Jaron who listened to countless research presentations and always provided positive feedback. I love you all! You too Rupes! iii Table of Contents List of Tables ...................................................................................................................... viii List of Figures ....................................................................................................................... ix List of Abbreviations ........................................................................................................ xii Section 1: Pattern Recognition Receptors ................................ 1 1.1 Overview of Innate Immunity ................................................................................... 1 1.2 TLRs ................................................................................................................................... 4 1.21 Cell Surface TLRs .............................................................................................................. 5 1.22 Endosomal TLRs ............................................................................................................... 6 1.3 RLRs ................................................................................................................................... 8 Section 2: Vitamin D ....................................................................... 11 2.1 Mechanism of Action ................................................................................................. 11 2.2 Vitamin D is Regulated by Several Vitamin D Associated Proteins .......... 12 2.21 CYP24a1 and CYP27b1 ................................................................................................... 12 2.22 Vitamin D Binding Protein .............................................................................................. 13 2.23 Vitamin D Receptor and Vitamin D Response Elements ................................................ 15 2.24 PTH, Calcium and FGF23 ................................................................................................ 15 2.3 Vitamin D and Its Impact on Innate Immunity ................................................ 16 2.31 Vitamin D and Antimicrobial Peptide Production .......................................................... 16 2.32 Vitamin D and Cytokine Production ............................................................................... 17 2.33 Vitamin D Influences Intracellular Signalling Pathways: From Receptor Expression to Transcription Factor Function ................................................................................................ 19 Section 3: Vitamin D and Disease .............................................. 23 3.1 Vitamin D Deficiency in the Population ............................................................. 23 3.2 Vitamin D Deficiency and Risk of Respiratory Tract Infections ................ 26 3.21 Epidemiological Evidence ............................................................................................... 26 3.22 Vitamin D Lowers Risk of Infection by Respiratory Syncytial Virus ................................ 27 3.3 Vitamin D Deficiency and Increased Risk of Autoimmunity ....................... 29 3.21 Asthma and Allergic Disease .......................................................................................... 29 iv 3.22 Crohn’s Disease and Diabetes ........................................................................................ 31 Thesis Overview ................................................................................................................ 32 Materials and Methods ................................................................. 34 Volunteers ........................................................................................................................... 34 Peripheral Blood Mononuclear Cell (PBMC) Isolation ........................................ 35 Preparation of Tissue Culture Reagents ................................................................... 35 Preparation of 1,25(OH)2D3 .................................................................................................... 35 Preparation of 25(OH)D3 ........................................................................................................ 36 Preparation of Respiratory Syncytial Virus and Reovirus ....................................................... 37 Primary Cell Culture ........................................................................................................ 37 Human Cytokine Enzyme-Linked Immuno Sorbent Assay (ELISA) ................. 39 RNA Isolation and RT-PCR ............................................................................................. 40 Mass Spectrometry ........................................................................................................... 43 Statistical Analysis ............................................................................................................ 43 CHAPTER 1 ........................................................................................ 44 Abstract ................................................................................................................................ 45 Results ................................................................................................................................... 47 Vitamin D Decreases Pro-inflammatory and Anti-inflammatory Cytokine Production in Adult PBMC Stimulated with LPS ..................................................................................................... 47 Vitamin D Fails to Alter Innate Cytokine Responses to TLR8 Stimulation in Primary PBMC .. 49 Vitamin D Fails to Reduce Pro-inflammatory and Anti-inflammatory Production in the Presence of Intact Infectious Virus RSV .................................................................................. 51 Cathelicidin Gene hCAP-18 is Downregulated in the Presence of LPS and 3M 002 ............... 54 Vitamin D Inhibits TLR4 and TLR8 Receptor Expression in PBMC in the Presence or Absence of Stimulation ......................................................................................................................... 56 Vitamin D Activating and Deactivating Enzyme Expression Levels Differ in TLR8L Compared to TLR4L Stimulated PBMC ..................................................................................................... 58 TLR8L Downregulates VDR
Recommended publications
  • Activated Peripheral-Blood-Derived Mononuclear Cells
    Transcription factor expression in lipopolysaccharide- activated peripheral-blood-derived mononuclear cells Jared C. Roach*†, Kelly D. Smith*‡, Katie L. Strobe*, Stephanie M. Nissen*, Christian D. Haudenschild§, Daixing Zhou§, Thomas J. Vasicek¶, G. A. Heldʈ, Gustavo A. Stolovitzkyʈ, Leroy E. Hood*†, and Alan Aderem* *Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103; ‡Department of Pathology, University of Washington, Seattle, WA 98195; §Illumina, 25861 Industrial Boulevard, Hayward, CA 94545; ¶Medtronic, 710 Medtronic Parkway, Minneapolis, MN 55432; and ʈIBM Computational Biology Center, P.O. Box 218, Yorktown Heights, NY 10598 Contributed by Leroy E. Hood, August 21, 2007 (sent for review January 7, 2007) Transcription factors play a key role in integrating and modulating system. In this model system, we activated peripheral-blood-derived biological information. In this study, we comprehensively measured mononuclear cells, which can be loosely termed ‘‘macrophages,’’ the changing abundances of mRNAs over a time course of activation with lipopolysaccharide (LPS). We focused on the precise mea- of human peripheral-blood-derived mononuclear cells (‘‘macro- surement of mRNA concentrations. There is currently no high- phages’’) with lipopolysaccharide. Global and dynamic analysis of throughput technology that can precisely and sensitively measure all transcription factors in response to a physiological stimulus has yet to mRNAs in a system, although such technologies are likely to be be achieved in a human system, and our efforts significantly available in the near future. To demonstrate the potential utility of advanced this goal. We used multiple global high-throughput tech- such technologies, and to motivate their development and encour- nologies for measuring mRNA levels, including massively parallel age their use, we produced data from a combination of two distinct signature sequencing and GeneChip microarrays.
    [Show full text]
  • A Molecular Switch from STAT2-IRF9 to ISGF3 Underlies Interferon-Induced Gene Transcription
    ARTICLE https://doi.org/10.1038/s41467-019-10970-y OPEN A molecular switch from STAT2-IRF9 to ISGF3 underlies interferon-induced gene transcription Ekaterini Platanitis 1, Duygu Demiroz1,5, Anja Schneller1,5, Katrin Fischer1, Christophe Capelle1, Markus Hartl 1, Thomas Gossenreiter 1, Mathias Müller2, Maria Novatchkova3,4 & Thomas Decker 1 Cells maintain the balance between homeostasis and inflammation by adapting and inte- grating the activity of intracellular signaling cascades, including the JAK-STAT pathway. Our 1234567890():,; understanding of how a tailored switch from homeostasis to a strong receptor-dependent response is coordinated remains limited. Here, we use an integrated transcriptomic and proteomic approach to analyze transcription-factor binding, gene expression and in vivo proximity-dependent labelling of proteins in living cells under homeostatic and interferon (IFN)-induced conditions. We show that interferons (IFN) switch murine macrophages from resting-state to induced gene expression by alternating subunits of transcription factor ISGF3. Whereas preformed STAT2-IRF9 complexes control basal expression of IFN-induced genes (ISG), both type I IFN and IFN-γ cause promoter binding of a complete ISGF3 complex containing STAT1, STAT2 and IRF9. In contrast to the dogmatic view of ISGF3 formation in the cytoplasm, our results suggest a model wherein the assembly of the ISGF3 complex occurs on DNA. 1 Max Perutz Labs (MPL), University of Vienna, Vienna 1030, Austria. 2 Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna 1210, Austria. 3 Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria. 4 Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna 1030, Austria.
    [Show full text]
  • Inactivation of Type I IFN Jak-STAT Pathway in EBV Latency
    Ning S and Wang L, J Cancer Biol Treat 2016, 3: 009 DOI: 10.24966/CBT-7546/100009 HSOA Journal of Cancer Biology and Treatment Research Article defense system, among which IRF7 is the “master” regulator of type I Inactivation of Type I IFN Interferon (IFN-I) response to pathogenic infection [4]. Robust IFN-I response during infection requires a positive feedback loop between Jak-STAT Pathway in EBV IRF7 and IFN-I [5-6]. Aberrant production of IFN-I, however, is Latency associated with autoimmune disorders and malignancies [7-9]. Thus, tight regulation of IRF7 is important in balancing the appropriate Shunbin Ning and Ling Wang* immune response to clear invading pathogens while preventing Department of Internal Medicine, Center of Excellence for Inflammation, immune-mediated pathogenesis [10]. Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, USA IFNs exert their functions through induction of IFN-Stimulated Genes (ISGs) via Jak-STAT pathways [11-13]. The IFN-I Jak-STAT pathway comprises of IFNAR1/2, Jak1, Tyk2, STAT1/2, and IRF9 [14]. Following IFN-I binding to IFNARs, signaling via protein kinases leads to tyrosine phosphorylation and activation of IFNAR1 Abstract at Y466 [15], Tyk2(Y1054/Y1055) and Jak1(Y1034/Y1035), and then to that Epstein-Barr Virus (EBV) latent infection is associated with a of STAT1(Y701) and STAT2(Y690 and S287) [16]. The phosphorylated variety of lymphomas and carcinomas. Interferon (IFN) Regulatory STAT1/2 then dimerize and associate with IRF9 to form a complex Factors (IRFs) are a family of transcription factors, among which termed interferon-stimulated gene factor 3 (ISGF3).
    [Show full text]
  • Beyond Viral Interferon Regulatory Factors: Immune Evasion Strategies Jinjong Myoung1, Shin-Ae Lee2, and Hye-Ra Lee3*
    J. Microbiol. Biotechnol. (2019), 29(12), 1873–1881 https://doi.org/10.4014/jmb.1910.10004 Research Article Review jmb Beyond Viral Interferon Regulatory Factors: Immune Evasion Strategies Jinjong Myoung1, Shin-Ae Lee2, and Hye-Ra Lee3* 1Korea Zoonosis Research Institute, Genetic Engineering Research Institute and Department of Bioactive Material Science, College of Natural Science, Jeonbuk National University, Jeonju 54531, Republic of Korea 2Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA 3Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong 30019, Republic of Korea Received: October 8, 2019 Revised: October 8, 2019 The innate immune response serves as a first-line-of-defense mechanism for a host against Accepted: October 24, 2019 viral infection. Viruses must therefore subvert this anti-viral response in order to establish an First published online: efficient life cycle. In line with this fact, Kaposi’s sarcoma-associated herpesvirus (KSHV) October 25, 2019 encodes numerous genes that function as immunomodulatory proteins to antagonize the host *Corresponding author immune system. One such mechanism through which KSHV evades the host immunity is by Phone: +82-44-860-1831 encoding a viral homolog of cellular interferon (IFN) regulatory factors (IRFs), known as Fax: +82-44-860-1598 E-mail: [email protected] vIRFs. Herein, we summarize recent advances in the study of the immunomodulatory strategies of KSHV vIRFs and their effects on KSHV-associated pathogenesis. pISSN 1017-7825, eISSN 1738-8872 Copyright© 2019 by Keywords: KSHV, viral interferon regulatory factor, immune evasion strategy, PRR-mediated The Korean Society for Microbiology signaling pathway, apoptosis pathway and Biotechnology Introduction various transcription factors.
    [Show full text]
  • Herpes Simplex Virus 1 Targets IRF7 Via ICP0 to Limit Type I IFN Induction David Shahnazaryan1,2, Rana Khalil3, Claire Wynne4, Caroline A
    www.nature.com/scientificreports OPEN Herpes simplex virus 1 targets IRF7 via ICP0 to limit type I IFN induction David Shahnazaryan1,2, Rana Khalil3, Claire Wynne4, Caroline A. Jeferies5,6, Joan Ní Gabhann‑Dromgoole1,3,6* & Conor C. Murphy1,2,6 Herpes simplex keratitis (HSK), caused by herpes simplex virus type 1 (HSV‑1) infection, is the commonest cause of infectious blindness in the developed world. Following infection the virus is initially suspended in the tear flm, where it encounters a multi‑pronged immune response comprising enzymes, complement, immunoglobulins and crucially, a range of anti‑viral and pro‑infammatory cytokines. However, given that HSV‑1 can overcome innate immune responses to establish lifelong latency throughout a susceptible individual’s lifetime, there is signifcant interest in understanding the mechanisms employed by HSV‑1 to downregulate the anti‑viral type I interferon (IFN) mediated immune responses. This study aimed to investigate the interactions between infected cell protein (ICP)0 and key elements of the IFN pathway to identify possible novel targets that contribute to viral immune evasion. Reporter gene assays demonstrated the ability of ICP0 to inhibit type I IFN activity downstream of pathogen recognition receptors (PRRs) which are known to be involved in host antiviral defences. Further experiments identifed interferon regulatory factor (IRF)7, a driver of type I IFN, as a potential target for ICP0. These fndings increase our understanding of the pathogenesis of HSK and suggest IRF7 as a potential therapeutic target. Herpes simplex keratitis (HSK) is the commonest cause of infectious corneal blindness in the western world1. Tere are over 500,000 afected individuals in the United States (US) alone2.
    [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]
  • Type I Interferon/IRF7 Axis Instigates Chemotherapy-Induced Immunological Dormancy in Breast Cancer
    Oncogene https://doi.org/10.1038/s41388-018-0624-2 ARTICLE Type I interferon/IRF7 axis instigates chemotherapy-induced immunological dormancy in breast cancer 1,2,8 1 1 1,2 3 4,5 Qiang Lan ● Sanam Peyvandi ● Nathalie Duffey ● Yu-Ting Huang ● David Barras ● Werner Held ● 6 3,4,5 6 6 1,2 François Richard ● Mauro Delorenzi ● Christos Sotiriou ● Christine Desmedt ● Girieca Lorusso ● Curzio Rüegg1,2,7 Received: 18 April 2018 / Revised: 27 September 2018 / Accepted: 6 November 2018 © The Author(s) 2018. This article is published with open access Abstract Neoadjuvant and adjuvant chemotherapies provide survival benefits to breast cancer patients, in particular in estrogen receptor negative (ER−) cancers, by reducing rates of recurrences. It is assumed that the benefits of (neo)adjuvant chemotherapy are due to the killing of disseminated, residual cancer cells, however, there is no formal evidence for it. Here, we provide experimental evidence that ER− breast cancer cells that survived high-dose Doxorubicin and Methotrexate based chemotherapies elicit a state of immunological dormancy. Hallmark of this dormant phenotype is the sustained activation of β 1234567890();,: 1234567890();,: the IRF7/IFN- /IFNAR axis subsisting beyond chemotherapy treatment. Upregulation of IRF7 in treated cancer cells promoted resistance to chemotherapy, reduced cell growth and induced switching of the response from a myeloid derived suppressor cell-dominated immune response to a CD4+/CD8+ T cell-dependent anti-tumor response. IRF7 silencing in tumor cells or systemic blocking of IFNAR reversed the state of dormancy, while spontaneous escape from dormancy was associated with loss of IFN-β production. Presence of IFN-β in the circulation of ER− breast cancer patients treated with neoadjuvant Epirubicin chemotherapy correlated with a significantly longer distant metastasis-free survival.
    [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]
  • Cysteine‑Rich 61‑Associated Gene Expression Profile Alterations in Human Glioma Cells
    MOLECULAR MEDICINE REPORTS 16: 5561-5567, 2017 Cysteine‑rich 61‑associated gene expression profile alterations in human glioma cells RUI WANG1, BO WEI2, JUN WEI3, YU TIAN2 and CHAO DU2 Departments of 1Radiology, 2Neurosurgery and 3Science and Education Section, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China Received January 28, 2016; Accepted February 20, 2017 DOI: 10.3892/mmr.2017.7216 Abstract. The present study aimed to investigate gene be critical for maintaining the role of CYR61 during cancer expression profile alterations associated with cysteine‑rich 61 progression. (CYR61) expression in human glioma cells. The GSE29384 dataset, downloaded from the Gene Expression Omnibus, Introduction includes three LN229 human glioma cell samples expressing CYR61 induced by doxycycline (Dox group), and three Cysteine‑rich 61 (CYR61) is a secreted, cysteine‑rich, control samples not exposed to doxycycline (Nodox group). heparin‑binding protein (1) involved in a variety of cellular Differentially expressed genes (DEGs) between the Dox and functions including adhesion, migration and proliferation (2). Nodox groups were identified with cutoffs of |log2 fold change Previously, Xie et al (3) reported that CYR61 was overexpressed (FC)|>0.5 and P<0.05. Gene ontology and Kyoto Encyclopedia in 66 primary gliomas compared with healthy brain samples, of Genes and Genomes pathway enrichment analyses for and that CYR61 expression was significantly correlated with DEGs were performed. Protein‑protein interaction (PPI) tumor grade and patient survival (3). CYR61‑overexpressing network and module analyses were performed to identify glioma cells were observed to have an increased proliferation the most important genes.
    [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]
  • In Vitro Targeting of Transcription Factors to Control the Cytokine Release Syndrome in 2 COVID-19 3
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.29.424728; this version posted December 30, 2020. 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 4.0 International license. 1 In vitro Targeting of Transcription Factors to Control the Cytokine Release Syndrome in 2 COVID-19 3 4 Clarissa S. Santoso1, Zhaorong Li2, Jaice T. Rottenberg1, Xing Liu1, Vivian X. Shen1, Juan I. 5 Fuxman Bass1,2 6 7 1Department of Biology, Boston University, Boston, MA 02215, USA; 2Bioinformatics Program, 8 Boston University, Boston, MA 02215, USA 9 10 Corresponding author: 11 Juan I. Fuxman Bass 12 Boston University 13 5 Cummington Mall 14 Boston, MA 02215 15 Email: [email protected] 16 Phone: 617-353-2448 17 18 Classification: Biological Sciences 19 20 Keywords: COVID-19, cytokine release syndrome, cytokine storm, drug repurposing, 21 transcriptional regulators 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.12.29.424728; this version posted December 30, 2020. 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 4.0 International license. 22 Abstract 23 Treatment of the cytokine release syndrome (CRS) has become an important part of rescuing 24 hospitalized COVID-19 patients. Here, we systematically explored the transcriptional regulators 25 of inflammatory cytokines involved in the COVID-19 CRS to identify candidate transcription 26 factors (TFs) for therapeutic targeting using approved drugs.
    [Show full text]
  • Differential and Overlapping Immune Programs Regulated by IRF3 and IRF5 in Plasmacytoid Dendritic Cells
    Differential and Overlapping Immune Programs Regulated by IRF3 and IRF5 in Plasmacytoid Dendritic Cells This information is current as Kwan T. Chow, Courtney Wilkins, Miwako Narita, Richard of September 28, 2021. Green, Megan Knoll, Yueh-Ming Loo and Michael Gale, Jr. J Immunol published online 8 October 2018 http://www.jimmunol.org/content/early/2018/10/05/jimmun ol.1800221 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2018/10/05/jimmunol.180022 Material 1.DCSupplemental 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 28, 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 © 2018 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published October 8, 2018, doi:10.4049/jimmunol.1800221 The Journal of Immunology Differential and Overlapping Immune Programs Regulated by IRF3 and IRF5 in Plasmacytoid Dendritic Cells Kwan T. Chow,*,† Courtney Wilkins,* Miwako Narita,‡ Richard Green,* Megan Knoll,* Yueh-Ming Loo,* and Michael Gale, Jr.* We examined the signaling pathways and cell type–specific responses of IFN regulatory factor (IRF) 5, an immune-regulatory transcription factor.
    [Show full text]