IL-18 Drives ILC3 Proliferation and Promotes IL-22 Production Via NF-Κb

Total Page:16

File Type:pdf, Size:1020Kb

IL-18 Drives ILC3 Proliferation and Promotes IL-22 Production Via NF-Κb IL-18 Drives ILC3 Proliferation and Promotes IL-22 Production via NF- κB Aaron R. Victor, Ansel P. Nalin, Wenjuan Dong, Susan McClory, Min Wei, Charlene Mao, Raleigh D. Kladney, This information is current as Youssef Youssef, Wing Keung Chan, Edward L. Briercheck, of October 3, 2021. Tiffany Hughes, Steven D. Scoville, Jason R. Pitarresi, Charlie Chen, Sarah Manz, Lai-Chu Wu, Jianying Zhang, Michael C. Ostrowski, Aharon G. Freud, Gustavo W. Leone, Michael A. Caligiuri and Jianhua Yu J Immunol 2017; 199:2333-2342; Prepublished online 25 Downloaded from August 2017; doi: 10.4049/jimmunol.1601554 http://www.jimmunol.org/content/199/7/2333 http://www.jimmunol.org/ Supplementary http://www.jimmunol.org/content/suppl/2017/08/24/jimmunol.160155 Material 4.DCSupplemental References This article cites 76 articles, 31 of which you can access for free at: http://www.jimmunol.org/content/199/7/2333.full#ref-list-1 by guest on October 3, 2021 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 *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 IL-18 Drives ILC3 Proliferation and Promotes IL-22 Production via NF-kB Aaron R. Victor,*,1 Ansel P. Nalin,*,1 Wenjuan Dong,†,1 Susan McClory,*,† Min Wei,† Charlene Mao,† Raleigh D. Kladney,†,‡,x Youssef Youssef,{ Wing Keung Chan,† Edward L. Briercheck,*,† Tiffany Hughes,† Steven D. Scoville,*,† Jason R. Pitarresi,†,‡ Charlie Chen,† Sarah Manz,† Lai-Chu Wu,† Jianying Zhang,‖ Michael C. Ostrowski,†,‡ Aharon G. Freud,†,{ Gustavo W. Leone,†,‡,x Michael A. Caligiuri,†,# and Jianhua Yu†,# Group 3 innate lymphoid cells (ILC3s) are important regulators of the immune system, maintaining homeostasis in the presence of commensal bacteria, but activating immune defenses in response to microbial pathogens. ILC3s are a robust source of IL-22, a cytokine critical for stimulating the antimicrobial response. We sought to identify cytokines that can promote proliferation and induce or maintain IL-22 production by ILC3s and determine a molecular mechanism for this process. We identified IL-18 as Downloaded from a cytokine that cooperates with an ILC3 survival factor, IL-15, to induce proliferation of human ILC3s, as well as induce and maintain IL-22 production. To determine a mechanism of action, we examined the NF-kB pathway, which is activated by IL- 18 signaling. We found that the NF-kB complex signaling component, p65, binds to the proximal region of the IL22 promoter and promotes transcriptional activity. Finally, we observed that CD11c+ dendritic cells expressing IL-18 are found in close proximity to ILC3s in human tonsils in situ. Therefore, we identify a new mechanism by which human ILC3s proliferate and produce IL-22, and identify NF-kB as a potential therapeutic target to be considered in pathologic states characterized by overproduction of IL- http://www.jimmunol.org/ 18 and/or IL-22. The Journal of Immunology, 2017, 199: 2333–2342. nnate lymphoid cells (ILCs) play many roles in protective orphan receptor g isoform b (RORgt) and aryl hydrocarbon re- immunity and disease (1). There are several types of ILCs, ceptor, and they produce the cytokines IL-17A and IL-22 (3–7). I each of which produces a characteristic cytokine profile and ILC3s are enriched within human secondary lymphoid tissues is regulated by unique transcription factors (2). Group 3 ILCs (SLTs), such as the tonsils and lymph nodes (8–10). (ILC3s) are composed of ILC3s and lymphoid tissue–inducer ILC3s are critical regulators of homeostasis and immunity (9– cells. They are dependent on the transcription factors RAR-related 12). ILC3s are the primary steady-state source of IL-22, a cytokine by guest on October 3, 2021 critical for tissue regeneration and for maintenance of barrier func- tion in the gut, skin, oral mucosa, and lung (3, 13). IL-22 signaling *Medical Scientist Training Program, Ohio State University, Columbus, OH 43210; †The James Cancer Hospital and Solove Research Institute, The Ohio State Univer- in epithelial cells drives genes involved in proliferation and wound sity Comprehensive Cancer Center, Columbus, OH 43210; ‡Department of Molecular healing (14, 15). In addition to bolstering the physical barrier of the Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210; xDepartment of Molecular Genetics, College of Biological Sciences, The epithelium, IL-22 stimulates epithelial cells to produce antimicro- Ohio State University, Columbus, OH 43210; {Department of Pathology, The Ohio bial peptides necessary for barrier maintenance and prevention of ‖ State University, Columbus, OH 43210; Center for Biostatistics, Department of infection by commensal bacteria (16, 17). ILC3s are also necessary Bioinformatics, The Ohio State University, Columbus, OH 43210; and #Division of Hematology, Department of Internal Medicine, The Ohio State University, Colum- for the formation of cryptopatches and isolated lymphoid follicles in bus, OH 43210 the intestinal lamina propria (4) as well as repair of lymph nodes 1A.R.V., A.P.N., and W.D. contributed equally to this work. following infection (18). IL-22 and IL-18 have recently been shown ORCIDs: 0000-0003-4118-3088 (A.R.V.); 0000-0002-5517-7183 (C.M.); 0000- to cooperatively contribute to murine intestinal immunity to mul- 0002-5257-1521 (W.K.C.); 0000-0003-1224-470X (T.H.); 0000-0002-1798- tiple infectious agents. IL-18 induces IL-22 during Toxoplasma 9113 (C.C.); 0000-0002-0489-9645 (S.M.); 0000-0002-1348-269X (L.-C.W.). gondii infection in the murine ileum, whereas IL-22 induces IL-18 Received for publication September 20, 2016. Accepted for publication July 27, 2017. during Citrobacter rodentium infection (17). A combination of This work was supported by National Institutes of Health Grants AI129582, IL-18 and IL-22 was shown to be critical for clearance of rotavirus CA095426, CA185301, and CA097189, American Cancer Society Research Scholar in infected mice (19). Grant RSG-14-243-01-LIB, a grant from Gabrielle’s Angel Foundation for Cancer In this study, we identified IL-18 as a cytokine that can induce Research, and by a grant from the Leukemia and Lymphoma Society. proliferation of and sustain IL-22 production by human ILC3s. Address correspondence and reprint requests to Dr. Michael A. Caligiuri and Dr. Jianhua Yu, Division of Hematology, Department of Internal Medicine, College of IL-18 signals through the IL-18 receptor to activate NF-kBsignaling, Medicine, Ohio State University, 320 West 10th Avenue, Columbus, OH 43210. E-mail which acts at the IL22 promoter. In the tonsil, ILC3s reside in close addresses: [email protected] (M.A.C.) and [email protected] (J.Y.) proximity to dendritic cells (DCs), a source of IL-18. Taken to- The online version of this article contains supplemental material. gether, these data support the hypothesis that DC-derived IL-18 Abbreviations used in this article: DC, dendritic cell; EdU, 5-ethynyl-29-deoxyuri- stimulates ILC3 function by maintaining the population through dine; HPC, hematopoietic progenitor cell; IHC, immunohistochemistry; ILC, innate lymphoid cell; ILC3, group 3 ILC; MCT, mast cell tryptase; PI, propidium iodide; proliferation and by sustaining production of IL-22 through an NF- RORgt, RAR–related orphan receptor g isoform b; SLT, secondary lymphoid tissue; kB–dependent mechanism. Our study further clarifies the role of TPCK, N-tosyl-L-phenylalanine chloromethyl ketone. DCs in ILC3 function and identifies NF-kB as a potential target for Copyright Ó 2017 by The American Association of Immunologists, Inc. 0022-1767/17/$35.00 future therapies against IL-22–mediated diseases. www.jimmunol.org/cgi/doi/10.4049/jimmunol.1601554 2334 REGULATION OF ILC3s BY IL-18 Materials and Methods EMSA and Ab supershift assays Isolation of human ILC3s and developmental precursors Nuclear extracts were isolated using a nuclear extraction kit (Active Motif). Complementary oligonucleotides—probe 1 (2248 to 2216 bp, referred to All procedures were performed with approval of the Ohio State Uni- 2 2 versity Institutional Review Board. Normal human pediatric tonsils were as transcription start site) and probe 2 ( 191 to 163 bp)—containing a obtained following routine tonsillectomy from Nationwide Children’s putative NF-kB binding site from the human IL22 promoter were syn- thesized. EMSAs were performed as described previously (23). In brief, Hospital (Columbus, OH). ILC3s and developmental precursors were 32 isolated as previously described (20). Briefly, total mononuclear cells probes 1 and 2 were P labeled and incubated with the nuclear extracts were depleted of CD19+ and/or CD3+ cells via magnetic negative se- (2 mg) in the presence of poly(deoxyinosinic-deoxycytidylic) acid (1 mg). lection (Miltenyi Biotec). For some experiments, B and/or T cell– For the Ab gel supershift assays, nuclear extracts were incubated with Abs depleted mononuclear cells were used immediately for flow cytometric for p65 (Rockland Immunochemicals) or p50 (Millipore) overnight at 4˚C analysis. Alternatively, ILC3s were sorted directly from the depleted fraction before the addition of the IL22 promoter probes. 2 2 2 2 2 2 2 + by gating on CD3 CD14 CD19 CD20 CD34 CD16 CD94 CD117 Transient transfection and luciferase assay events on a FACSAria II cell sorter (BD Biosciences).
Recommended publications
  • DS Human IL-18
    ® MSD Human IL-18 Kit For quantitative determination in human serum, plasma, and tissue culture supernatants Alzheimer’s Disease IL-18 BioProcess Cardiac Cell Signaling Clinical Immunology Cytokines Growth Factors Hypoxia Immunogenicity Inflammation Metabolic Oncology Interleukin-18 (IL-18) is an 18 kDa cytokine and a co-stimulatory factor that is produced in Kuppfer cells, activated macrophages, Toxicology keratinocytes, and intestinal epithelial cells.1 One of the main functions of IL-18 is to promote the production of IFN-γ from T and NK Vascular cells, particularly in the presence of IL-12p70. IL-18 also promotes the secretion of other proinflammatory cytokines like TNF-α, IL-1β, 2 and GM-CSF that enhance the migration and activtion of neutrophils during microbial infections. IL-18 enhances cytotoxic activity and 2,3 Catalog Numbers proliferation of CD8+ T and NK cells and has been shown to stimulate the production of IL-13 and other Th2 cytokines. Dysregulation of IL-18 may therefore contribute to inflammatory-associated disorders, unchecked infections, autoimmune diseases such as 2,3 Human IL-18 Kit rheumatoid arthritis, acute and chronic kidney injury, cancer, and pathogenic conditions related to metabolic syndrome. Kit size The MSD Human IL - 18 assay is available on 96-well 4-spot plates. This datasheet outlines the performance of the assay. 1 plate K151MCD-1 5 plates K151MCD-2 Assay Sensitivity 25 plates K151MCD-4 IL-18 LLOD (pg/mL) 0.71 The lower limit of detection (LLOD) is a calculated concentration based on a signal Ordering information 2.5 standard deviations above the background (zero calibrator blank).
    [Show full text]
  • Role of Interleukin 36Γ in Host Defense Against Tuberculosis Fadhil Ahsan,1,2,A Pedro Moura-Alves,1,A Ute Guhlich-Bornhof,1 Marion Klemm,1 Stefan H
    The Journal of Infectious Diseases MAJOR ARTICLE Role of Interleukin 36γ in Host Defense Against Tuberculosis Fadhil Ahsan,1,2,a Pedro Moura-Alves,1,a Ute Guhlich-Bornhof,1 Marion Klemm,1 Stefan H. E. Kaufmann,1 and Jeroen Maertzdorf1 Downloaded from https://academic.oup.com/jid/article-abstract/214/3/464/2577344 by Deutsches Rheumaforschungs Zentrum user on 26 June 2019 1Department of Immunology, Max Planck Institute for Infection Biology, and 2ZIBI Graduate School Berlin, Germany Tuberculosis remains a major killer worldwide, not the least because of our incomplete knowledge of protective and pathogenic immune mechanism. The roles of the interleukin 1 (IL-1) and interleukin 18 pathways in host defense are well established, as are their regulation through the inflammasome complex. In contrast, the regulation of interleukin 36γ (IL-36γ), a recently described member of the IL-1 family, and its immunological relevance in host defense remain largely unknown. Here we show that Myco- bacterium tuberculosis infection of macrophages induces IL-36γ production in a 2-stage-regulated fashion. In the first stage, mi- crobial ligands trigger host Toll-like receptor and MyD88-dependent pathways, leading to IL-36γ secretion. In the second stage, endogenous IL-1β and interleukin 18 further amplify IL-36γ synthesis. The relevance of this cytokine in the control of M. tuber- culosis is demonstrated by IL-36γ–induced antimicrobial peptides and IL-36 receptor–dependent restriction of M. tuberculosis growth. Thus, we provide first insight into the induction and regulation of the proinflammatory cytokine IL-36γ during tuberculosis. Keywords. IL-36γ; Mycobacterium tuberculosis; TLR; inflammasome; antimicrobial peptide.
    [Show full text]
  • The Role of Interleukin-18 in the Metabolic Syndrome Marius Trøseid1*, Ingebjørg Seljeflot1,2, Harald Arnesen1,2
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Springer - Publisher Connector Trøseid et al. Cardiovascular Diabetology 2010, 9:11 CARDIO http://www.cardiab.com/content/9/1/11 VASCULAR DIABETOLOGY REVIEW Open Access The role of interleukin-18 in the metabolic syndrome Marius Trøseid1*, Ingebjørg Seljeflot1,2, Harald Arnesen1,2 Abstract The metabolic syndrome is thought to be associated with a chronic low-grade inflammation, and a growing body of evidence suggests that interleukin-18 (IL-18) might be closely related to the metabolic syndrome and its conse- quences. Circulating levels of IL-18 have been reported to be elevated in subjects with the metabolic syndrome, to be closely associated with the components of the syndrome, to predict cardiovascular events and mortality in populations with the metabolic syndrome and to precede the development of type 2 diabetes. IL-18 is found in the unstable atherosclerotic plaque, in adipose tissue and in muscle tissue, and is subject to several regulatory steps including cleavage by caspase-1, inactivation by IL-18 binding protein and the influence of other cytokines in modulating its interaction with the IL-18 receptor. The purpose of this review is to outline the role of IL-18 in the metabolic syndrome, with particular emphasis on cardiovascular risk and the potential effect of life style interventions. Introduction developing CVD is approximately doubled in the meta- The metabolic syndrome is a cluster of risk factors that bolic syndrome [10]. In a meta-analysis including identifies a population with increased risk for developing 43 cohorts, the relative risk for cardiovascular events type 2 diabetes mellitus and cardiovascular disease and death was 1.78, with the highest risk in women [11].
    [Show full text]
  • IL-33 Can Promote Survival, Adhesion and Cytokine Production in Human
    Laboratory Investigation (2007) 87, 971–978 & 2007 USCAP, Inc All rights reserved 0023-6837/07 $30.00 IL-33 can promote survival, adhesion and cytokine production in human mast cells Motoyasu Iikura1,2, Hajime Suto1,3, Naoki Kajiwara4, Keisuke Oboki5, Tatsukuni Ohno5, Yoshimichi Okayama4, Hirohisa Saito3,5, Stephen J Galli1 and Susumu Nakae1,3,5 IL-33 is a recently identified member of the IL-1 family of molecules, which also includes IL-1 and IL-18. IL-33 binds to the receptor, T1/ST2/IL-1R4, and can promote cytokine secretion by Th2 cells and NF-kB phosphorylation in mouse mast cells. However, the effects of these molecules, especially IL-33, in human mast cells are poorly understood. Expression of the receptors for IL-1 family molecules, specifically, IL-1R1, IL-18R and T1/ST2, was detectable intracellularly in human umbilical cord blood-derived mast cells (HUCBMCs) by flow cytometry, but was scarcely detectable on the cells’ surface. However, IL-1b, IL-18 or IL-33 induced phosphorylation of Erk, p38 and JNK in naı¨ve HUCBMCs, and IL-33 or IL-1b, but not IL-18, enhanced the survival of naive HUCBMCs and promoted their adhesion to fibronectin. IL-33 or IL-1b also induced IL-8 and IL-13 production in naı¨ve HUCBMCs, and enhanced production of these cytokines in IgE/anti-IgE-stimulated HUCBMCs, without enhancing secretion of either PGD2 or histamine. Moreover, IL-33-mediated IL-8 production by HUCBMCs was markedly reduced by the p38 MAPK inhibitor, SB203580. In contrast to findings with mouse mast cells, IL-18 neither induced nor enhanced secretion of the mediators PGD2 or histamine by HUCBMCs.
    [Show full text]
  • Interleukin-18 in Health and Disease
    International Journal of Molecular Sciences Review Interleukin-18 in Health and Disease Koubun Yasuda 1 , Kenji Nakanishi 1,* and Hiroko Tsutsui 2 1 Department of Immunology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan; [email protected] 2 Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan; [email protected] * Correspondence: [email protected]; Tel.: +81-798-45-6573 Received: 21 December 2018; Accepted: 29 January 2019; Published: 2 February 2019 Abstract: Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4+ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells.
    [Show full text]
  • Comprehensive Association Study of Genetic Variants in the IL-1 Gene Family in Systemic Juvenile Idiopathic Arthritis
    Genes and Immunity (2008) 9, 349–357 & 2008 Nature Publishing Group All rights reserved 1466-4879/08 $30.00 www.nature.com/gene ORIGINAL ARTICLE Comprehensive association study of genetic variants in the IL-1 gene family in systemic juvenile idiopathic arthritis CJW Stock1, EM Ogilvie1, JM Samuel1, M Fife1, CM Lewis2 and P Woo1 1Centre for Paediatric and Adolescent Rheumatology, Windeyer Institute for Medical Sciences, University College London, London, UK and 2Guy’s, Kings and St Thomas’ School of Medicine, London, UK Patients with systemic juvenile idiopathic arthritis (sJIA) have a characteristic daily spiking fever and elevated levels of inflammatory cytokines. Members of the interleukin-1 (IL-1) gene family have been implicated in various inflammatory and autoimmune diseases, and treatment with the IL-1 receptor antagonist, Anakinra, shows remarkable improvement in some patients. This work describes the most comprehensive investigation to date of the involvement of the IL-1 gene family in sJIA. A two-stage case–control association study was performed to investigate the two clusters of IL-1 family genes using a tagging single nucleotide polymorphism (SNP) approach. Genotyping data of 130 sJIA patients and 151 controls from stage 1 highlighted eight SNPs in the IL1 ligand cluster region and two SNPs in the IL1 receptor cluster region as showing a significant frequency difference between the populations. These 10 SNPs were typed in an additional 105 sJIA patients and 184 controls in stage 2. Meta-analysis of the genotypes from both stages showed that three IL1 ligand cluster SNPs (rs6712572, rs2071374 and rs1688075) and one IL1 receptor cluster SNP (rs12712122) show evidence of significant association with sJIA.
    [Show full text]
  • Type I Interferons in Anticancer Immunity
    REVIEWS Type I interferons in anticancer immunity Laurence Zitvogel1–4*, Lorenzo Galluzzi1,5–8*, Oliver Kepp5–9, Mark J. Smyth10,11 and Guido Kroemer5–9,12 Abstract | Type I interferons (IFNs) are known for their key role in antiviral immune responses. In this Review, we discuss accumulating evidence indicating that type I IFNs produced by malignant cells or tumour-infiltrating dendritic cells also control the autocrine or paracrine circuits that underlie cancer immunosurveillance. Many conventional chemotherapeutics, targeted anticancer agents, immunological adjuvants and oncolytic 1Gustave Roussy Cancer Campus, F-94800 Villejuif, viruses are only fully efficient in the presence of intact type I IFN signalling. Moreover, the France. intratumoural expression levels of type I IFNs or of IFN-stimulated genes correlate with 2INSERM, U1015, F-94800 Villejuif, France. favourable disease outcome in several cohorts of patients with cancer. Finally, new 3Université Paris Sud/Paris XI, anticancer immunotherapies are being developed that are based on recombinant type I IFNs, Faculté de Médecine, F-94270 Le Kremlin Bicêtre, France. type I IFN-encoding vectors and type I IFN-expressing cells. 4Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, F-94800 Villejuif, France. Type I interferons (IFNs) were first discovered more than Type I IFNs in cancer immunosurveillance 5Equipe 11 labellisée par la half a century ago as the factors underlying viral inter­ Type I IFNs are known to mediate antineoplastic effects Ligue Nationale contre le ference — that is, the ability of a primary viral infection against several malignancies, which is a clinically rel­ Cancer, Centre de Recherche 1 des Cordeliers, F-75006 Paris, to render cells resistant to a second distinct virus .
    [Show full text]
  • Detecting the Stable Point of Therapeutic Effect of Chronic Myeloid
    Xu et al. BMC Bioinformatics 2019, 20(Suppl 7):202 https://doi.org/10.1186/s12859-019-2738-0 RESEARCH Open Access Detecting the stable point of therapeutic effect of chronic myeloid leukemia based on dynamic network biomarkers Junhua Xu1,MinWu1, Shanshan Zhu1,JinzhiLei2 and Jie Gao1* From The 12th International Conference on Computational Systems Biology (ISB 2018) Guiyang, China. 18-21 August 2018 Abstract Background: Most researches of chronic myeloid leukemia (CML) are currently focused on the treatment methods, while there are relatively few researches on the progress of patients’ condition after drug treatment. Traditional biomarkers of disease can only distinguish normal state from disease state, and cannot recognize the pre-stable state after drug treatment. Results: A therapeutic effect recognition strategy based on dynamic network biomarkers (DNB) is provided for CML patients’ gene expression data. With the DNB criteria, the DNB with 250 genes is selected and the therapeutic effect index (TEI) is constructed for the detection of individual disease. The pre-stable state before the disease condition becomes stable is 1 month. Through functional analysis for the DNB, some genes are confirmed as key genes to affect the progress of CML patients’ condition. Conclusions: The results provide a certain theoretical direction and theoretical basis for medical personnel in the treatment of CML patients, and find new therapeutic targets in the future. The biomarkers of CML can help patients to be treated promptly and minimize drug resistance, treatment failure and relapse, which reduce the mortality of CML significantly. Keywords: Chronic myeloid leukemia (CML), Dynamic network biomarkers (DNB), Differentially expressed genes (DEGs), Therapeutic effect index (TEI), Pre-stable state, Treatment time Introduction At present, the use of ABL kinase inhibitors (e.g.
    [Show full text]
  • Mechanisms of Type-I Ifn Inhibition: Equine Herpesvirus-1 Escape from the Antiviral Effect of Type-1 Interferon Response in Host Cell
    University of Kentucky UKnowledge Theses and Dissertations--Veterinary Science Veterinary Science 2019 MECHANISMS OF TYPE-I IFN INHIBITION: EQUINE HERPESVIRUS-1 ESCAPE FROM THE ANTIVIRAL EFFECT OF TYPE-1 INTERFERON RESPONSE IN HOST CELL Fatai S. Oladunni University of Kentucky, [email protected] Author ORCID Identifier: https://orcid.org/0000-0001-5050-0183 Digital Object Identifier: https://doi.org/10.13023/etd.2019.374 Right click to open a feedback form in a new tab to let us know how this document benefits ou.y Recommended Citation Oladunni, Fatai S., "MECHANISMS OF TYPE-I IFN INHIBITION: EQUINE HERPESVIRUS-1 ESCAPE FROM THE ANTIVIRAL EFFECT OF TYPE-1 INTERFERON RESPONSE IN HOST CELL" (2019). Theses and Dissertations--Veterinary Science. 43. https://uknowledge.uky.edu/gluck_etds/43 This Doctoral Dissertation is brought to you for free and open access by the Veterinary Science at UKnowledge. It has been accepted for inclusion in Theses and Dissertations--Veterinary Science by an authorized administrator of UKnowledge. For more information, please contact [email protected]. STUDENT AGREEMENT: I represent that my thesis or dissertation and abstract are my original work. Proper attribution has been given to all outside sources. I understand that I am solely responsible for obtaining any needed copyright permissions. I have obtained needed written permission statement(s) from the owner(s) of each third-party copyrighted matter to be included in my work, allowing electronic distribution (if such use is not permitted by the fair use doctrine) which will be submitted to UKnowledge as Additional File. I hereby grant to The University of Kentucky and its agents the irrevocable, non-exclusive, and royalty-free license to archive and make accessible my work in whole or in part in all forms of media, now or hereafter known.
    [Show full text]
  • Evolutionary Divergence and Functions of the Human Interleukin (IL) Gene Family Chad Brocker,1 David Thompson,2 Akiko Matsumoto,1 Daniel W
    UPDATE ON GENE COMPLETIONS AND ANNOTATIONS Evolutionary divergence and functions of the human interleukin (IL) gene family Chad Brocker,1 David Thompson,2 Akiko Matsumoto,1 Daniel W. Nebert3* and Vasilis Vasiliou1 1Molecular Toxicology and Environmental Health Sciences Program, Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO 80045, USA 2Department of Clinical Pharmacy, University of Colorado Denver, Aurora, CO 80045, USA 3Department of Environmental Health and Center for Environmental Genetics (CEG), University of Cincinnati Medical Center, Cincinnati, OH 45267–0056, USA *Correspondence to: Tel: þ1 513 821 4664; Fax: þ1 513 558 0925; E-mail: [email protected]; [email protected] Date received (in revised form): 22nd September 2010 Abstract Cytokines play a very important role in nearly all aspects of inflammation and immunity. The term ‘interleukin’ (IL) has been used to describe a group of cytokines with complex immunomodulatory functions — including cell proliferation, maturation, migration and adhesion. These cytokines also play an important role in immune cell differentiation and activation. Determining the exact function of a particular cytokine is complicated by the influence of the producing cell type, the responding cell type and the phase of the immune response. ILs can also have pro- and anti-inflammatory effects, further complicating their characterisation. These molecules are under constant pressure to evolve due to continual competition between the host’s immune system and infecting organisms; as such, ILs have undergone significant evolution. This has resulted in little amino acid conservation between orthologous proteins, which further complicates the gene family organisation. Within the literature there are a number of overlapping nomenclature and classification systems derived from biological function, receptor-binding properties and originating cell type.
    [Show full text]
  • Interferon-Alpha-Based Immunotherapies in the Treatment Of
    Zhang et al. Exp Hematol Oncol (2017) 6:20 DOI 10.1186/s40164-017-0081-6 Experimental Hematology & Oncology REVIEW Open Access Interferon‑alpha‑based immunotherapies in the treatment of B cell‑derived hematologic neoplasms in today’s treat‑to‑target era Li Zhang1,2, Yu‑Tzu Tai1*, Matthew Zhi Guang Ho1,3,4, Lugui Qiu5 and Kenneth C. Anderson1* Abstract B cell lymphoma and multiple myeloma (MM) are the most common hematological malignancies which beneft from therapeutic monoclonal antibodies (mAbs)-based immunotherapies. Despite signifcant improvement on patient outcome following the use of novel therapies for the past decades, curative treatment is unavailable for the major‑ ity of patients. For example, the 5-year survival of MM is currently less than 50%. In the 1980s, interferon-α was used as monotherapy in newly diagnosed or previously treated MM with an overall response rate of 15–20%. Noticeably, a small subset of patients who responded to long-term interferon-α further achieved sustained complete remission. Since 1990, interferon-α-containing regimens have been used as a central maintenance strategy for patients with MM. However, the systemic administration of interferon-α was ultimately limited by its pronounced toxicity. To address this, the selective mAb-mediated delivery of interferon-α has been developed to enhance specifc killing of MM and B-cell malignant cells. As such, targeted interferon-α therapy may improve therapeutic window and sustain responses, while further overcoming suppressive microenvironment. This review aims to reinforce the role of interferon-α by consolidating our current understanding of targeting interferon-α with tumor-specifc mAbs for B cell lymphoma and myeloma.
    [Show full text]
  • Wright1260921490.Pdf (1.84
    A POTENTIAL STRATEGY TO MAINTAIN HSV-1 IN A LATENT STATE: USE OF IMMUNOREGULATORY PEPTIDE MIMETICS A thesis submitted in partial fulfillment of the requirements for the degree of Master of Science By NASRIN MAJIDI 2009 Wright State University WRIGHT STATE UNIVERSITY SCHOOL OF GRADUATE STUDIES December 7, 2009 I HEREBY RECOMMEND THAT THE THESIS PREPARED UNDER MY SUPERVISION BY Nasrin Majidi ENTITLED. A potential strategy to maintain HSV-1 in a latent state: use of immunoregulatory peptide mimetics. BE ACCEPTED IN PARTIAL FULFILLMENT OF THE REQUIRMENTS FOR THE DEGREE OF Master of Science. ___________________________ Nancy J. Bigley, Ph.D. Thesis Director ___________________________ Barbara E. Hull, Ph.D. Program Director Committee on Final Examination _______________________ Nancy J. Bigley, Ph.D. _______________________ Barbara E. Hull, Ph.D. _______________________ Cheryl L. Conley, Ph.D. _______________________ Dean Joseph F. Thomas, Jr., Ph.D. Dean, School of Graduate Studies ABSTRACT Majidi, Nasrin. M.S., Department of Biological Sciences, Microbiology and Immunology Program, Wright State University, 2009. A potential strategy to maintain HSV-1 in a latent state: use of immunoregulatory peptide mimetics. This study reviews the role of interferon-gamma (IFN-γ) in HSV-1 latency. CD8+ T cells inhibit the reactivation of HSV-1 in trigeminal ganglia (TG) by production of IFN-γ. Although CD8+ T cells include all the cytotoxic apparatus for cytotoxicity, latently infected neuronal cells are not killed by CD8+ T cells. The CD94-NKG2a molecule on CD8+ T cells, binds to Qa-1b (a MHC class I like molecule) present on neuronal cell to inhibit CD8+ T cells cytotoxicity.
    [Show full text]