A Role for Janus Kinases in Crosstalk Between Erbb3 and the Interferon-Alpha Signaling Complex in Myeloma Cells

Total Page:16

File Type:pdf, Size:1020Kb

A Role for Janus Kinases in Crosstalk Between Erbb3 and the Interferon-Alpha Signaling Complex in Myeloma Cells Oncogene (2004) 23, 1197–1205 & 2004 Nature Publishing Group All rights reserved 0950-9232/04 $25.00 www.nature.com/onc A role for Janus kinases in crosstalk between ErbB3 and the interferon-alpha signaling complex in myeloma cells Denise K Walters1 and Diane F Jelinek*,1 1Department of Immunology, Tumor Biology Program, Mayo Graduate and Medical Schools, Mayo Clinic/Foundation, Rochester, MN 55905, USA Receptor crosstalk is an emerging and recurrent theme in activation of a particular signaling cascade(s) and the cytokine and growth factor signaling; however,insight resulting biological response (Dumont et al., 2001). into the mechanism(s) underlying these interactions However, over the last decade it has become clear that remains limited. Recently,we reported that crosstalk this process may be much more complex than initially occurs between ErbB3 and the interferon alpha (IFN-a) anticipated. In fact, numerous reports have now shown signaling complex in the myeloma cell line KAS-6/1 and that biological responses may be brought about through that this crosstalk contributes to the regulation of cell receptor crosstalk or the transactivation of additional proliferation. In this study,we examined the mechanism and often unrelated receptor subtypes (reviewed by Hill, underlying the transactivation of ErbB3 in the IFN-a 1998). For example, in rat hepatocytes binding of growth-responsive KAS-6/1 cells. The examination of growth hormone (GH) to its receptor was shown not IFN-a receptor 1 and 2 (IFNAR1 and IFNAR2) levels only to result in the activation of the GH receptor but revealed that the KAS-6/1 cell line overexpresses also in the tyrosine phosphorylation of the epidermal IFNAR1 relative to other myeloma cell lines that are growth factor receptor (EGFR) (Yamauchi et al., 1997). growth arrested by IFN-a. Subsequent investigation of Interestingly, Jak2 appeared to play a role in the Tyk2,which is constitutively associated with IFNAR1, transactivation of the EGFR following GH stimulation. demonstrated that Tyk2 activation is uniquely sustained in In addition, the phosphorylation of the EGFR was also the KAS-6/1 cell line following IFN-a stimulation. shown to be required for GH-induced activation of the Interestingly,silencing of Tyk2 expression via siRNA MAP kinase pathway. resulted in attenuation of ErbB3 transactivation. How- During tumorigenesis and/or tumor progression, ever,inhibition of Jak1 expression also decreased IFN- a- malignant cells frequently overexpress or acquire induced tyrosine phosphorylation of ErbB3. Finally, atypical expression of one or more signaling receptors. siRNA downregulation of Tyk2 and Jak1 was found to Given the fact that a variety of receptors have now been decrease IFN-a-stimulated proliferation. These findings shown to be able to act in a nonlinear fashion, receptor validate our previous report of ErbB3 involvement in IFN- crosstalk has emerged as a potential mechanism by a-induced proliferation and further suggest that both which tumor cells may potentially amplify and/or Janus kinase members,Tyk2 and Jak1,play a role in the diversify their responses to growth factors/cytokines transactivation of ErbB3 in this model system. (Jelinek et al., 2003). To acquire an understanding of Oncogene (2004) 23, 1197–1205. doi:10.1038/sj.onc.1207203 growth and survival control in malignant cells, it is Published online 1 December 2003 therefore imperative to comprehend fully linear as well as potential nonlinear interactions in transformed Keywords: Jak1; Tyk2; ErbB3; interferon-alpha; multi- cells. ple myeloma In this regard, we have had a long-standing interest in the growth regulation of multiple myeloma (MM), a typically fatal plasma cell malignancy. Thus, we have been actively investigating the ability of the tumor cells Introduction in this disease to signal in a nonlinear fashion. More specifically we have had a particular interest in Signal transduction pathways have classically been determining if receptor crosstalk plays a role in the described as a linear sequence of events that involve ability of IFN-a to modulate myeloma cell growth in a the interaction of a ligand with a specific receptor, variable manner (Brenning et al., 1985; Jourdan et al., 1991; Westendorf et al., 1996; Jelinek et al., 1997). Recently, we reported detection of ErbB3, a member of *Correspondence: DF Jelinek, 200 First Street SW, Rochester, the ErbB family member, on the MM cell line KAS-6/1, MN 55905, USA; E-mail: [email protected] which is growth responsive to IFN-a. Based on the fact This work was supported by National Institutes of Health Grants CA62242 and CA62228 that ErbB family members are known to play an Received 28July 2003; revised 8September 2003; accepted 9 September important role in several cancers (i.e. breast, prostate) 2003 and that various ErbB members have previously been Jak kinases mediate crosstalk in myeloma cells DK Walters and DF Jelinek 1198 suggested to be potentially involved in the development Materials and methods and progression of a subset of B-lymphoblastic leuke- mias and MM (Buhring et al., 1995; Klein, B., 2003, Cell lines, culture medium and reagents personal communication), we were interested in deter- The myeloma cell lines ANBL-6, DP-6, KAS-6/1, and KP-6 mining if ErbB3 may play a role in the growth were derived from primary patient myeloma cells and all have regulation of the KAS-6/1 cell line. Interestingly, we been described previously (Westendorf et al., 1996; Jelinek discovered that crosstalk occurs between the atypically et al., 1997). These cell lines were maintained as previously expressed ErbB3 molecule and the IFN-a receptor in described (Arora et al., 1999). Recombinant purified IFN-a2b these cells. Furthermore, we were also able to demon- (Schering, Kenilworth, NJ) was used at 10 000 U/ml in strate through siRNA-mediated downregulation of immunoprecipitation experiments and at 1000 U/ml in DNA synthesis assays. Interleukin-6 (IL-6) (Novartis Pharma, Basel, ErbB3 that this crosstalk does indeed contribute to the Switzerland) was used at 1 ng/ml in DNA synthesis assays. IFN-a-induced proliferative ability of these cells (Wal- ters et al., 2003). Immunofluorescence analysis Despite the abundance of evidence supporting the incidence and importance of receptor crosstalk, few Analysis of cell surface protein expression was determined by studies have thoroughly examined the mechanism by immunofluorescence. Briefly, 1 Â 106 cells were incubated with which receptor crosstalk is facilitated. However, based mAbs to IFNAR1 (R&D Systems Inc., Minneapolis, MN, on our knowledge of receptor signaling and previous USA), IFNAR2 (Research Diagnostics Inc., Flanders, NY, USA), or ErbB3 (Neomarkers, Fremont, CA, USA) for 30 min receptor crosstalk reports, we hypothesized that there on ice in PBS/2% FCS, washed and then incubated with PE- are at least two mechanisms by which receptor crosstalk conjugated goat anti-mouse IgG for 30 min (Biosource, most likely occurs. These mechanisms include direct Camarillo, CA, USA). Cells were then washed, fixed in 1% physical association of the receptors and/or lateral paraformaldehyde and analysed for immunofluorescence on a signaling by nonreceptor tyrosine kinases. As we FACSCAN flow cytometer (Becton Dickinson, Mountain were previously unable to detect a physical association View, CA, USA). Control Abs included mouse IgG1 and between ErbB3 and the IFN-a receptor (Walters IgG2a. Data were analysed using WinMDI 2.5 software. et al., 2003), we hypothesized that crosstalk must be mediated via lateral signaling by nonreceptor tyrosine Immunoprecipitation and immunoblotting kinases. Cells were cultured in IL-6-free medium supplemented with Similar to other cytokine receptors, the type I IFN 0.5% BSA prior to IFN-a addition. Cells (1.0–2.0 Â 107) were receptor is devoid of any intrinsic kinase activity lysed using RIPA lysis buffer (50 mM Tris pH 7.4, 150 mM (Novick et al., 1994). However, this deficiency is NaCl, 1% NP-40, 0.5% DOC, 0.1% SDS, 1 mM EDTA, overcome through utilization of the Jak-STAT pathway, 15 mM sodium molybdate, 1 mM NaF) or Brij lysis buffer which is comprised of a family of cytoplasmic non- (10 mM Tris pH 7.4, 150 mM NaCl, 0.5 mM CaCl2, 0.5 mM receptor tyrosine kinases called the Janus kinases (also MgCl2, 1% Brij-97) supplemented with protease inhibitors known as Jaks) and a family of SH2 domain containing (10 mg/ml leupeptin, 10 mg/ml aprotinin, 10 mg/ml pepstatin, proteins called signal transducers and activators of 2mM Na3VO4, and 1 mM PMSF). Lysates were cleared of insoluble material by centrifugation for 10 min at 14 000 r.p.m. transcription (STATs) (reviewed in Boundy and Kovar- Following centrifugation, 1 mg/ml of protein-specific antibody ik, 2002). Briefly, ligand binding induces transpho- (Jak1 or Tyk2; BD Biosciences, San Jose, CA, USA; ErbB3 or sphorylation and activation of Janus kinase family IFNAR1; Santa Cruz, CA, USA) bound to protein-A or members Tyk2 and Jak1, which are constitutively gammabind-G sepharose beads was added to lysates contain- associated with the a and b subunits of the IFN ing equal amounts of protein. The lysate and immune- signaling complex, respectively. Activated Tyk2 and complexes were then incubated with agitation for 2–4 h at Jak1 then rapidly phosphorylate specific tyrosine 41C. The resulting immunoprecipitates were then washed three residues on the receptor subunits allowing these times and resuspended in 25 mlof2Â SDS loading buffer, phosphorylated residues to serve as docking sites heated to 1001C for 5 min, resolved by SDS–PAGE and for members of the STAT family. Following docking transferred to Immobilon-P membranes (Millipore Corp., Bedford, MA, USA) for immunoblotting. Membranes were to receptors, the STAT proteins are phosphorylated blocked in 25 mM Tris-HCl (pH 7.2), 150 mM NaCl, 0.2% by Tyk2 and Jak1 allowing homo- or heterodimeriza- Tween (TBST) supplemented with 0.2% BSA for 1 h.
Recommended publications
  • Promoter Janus Kinase 3 Proximal Characterization and Analysis Of
    The Journal of Immunology Characterization and Analysis of the Proximal Janus Kinase 3 Promoter1 Martin Aringer,2*† Sigrun R. Hofmann,2* David M. Frucht,* Min Chen,* Michael Centola,* Akio Morinobu,* Roberta Visconti,* Daniel L. Kastner,* Josef S. Smolen,† and John J. O’Shea3* Janus kinase 3 (Jak3) is a nonreceptor tyrosine kinase essential for signaling via cytokine receptors that comprise the common ␥-chain (␥c), i.e., the receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Jak3 is preferentially expressed in hemopoietic cells and is up-regulated upon cell differentiation and activation. Despite the importance of Jak3 in lymphoid development and immune function, the mechanisms that govern its expression have not been defined. To gain insight into this issue, we set out to characterize the Jak3 promoter. The 5؅-untranslated region of the Jak3 gene is interrupted by a 3515-bp intron. Upstream of this intron and the transcription initiation site, we identified an ϳ1-kb segment that exhibited lymphoid-specific promoter activity and was responsive to TCR signals. Truncation of this fragment revealed that core promoter activity resided in a 267-bp fragment that contains putative Sp-1, AP-1, Ets, Stat, and other binding sites. Mutation of the AP-1 sites significantly diminished, whereas mutation of the Ets sites abolished, the inducibility of the promoter construct. Chromatin immunoprecipitation assays showed that histone acetylation correlates with mRNA expression and that Ets-1/2 binds this region. Thus, transcription factors that bind these sites, especially Ets family members, are likely to be important regulators of Jak3 expression.
    [Show full text]
  • Type I Interferons and the Development of Impaired Vascular Function and Repair in Human and Murine Lupus
    Type I Interferons and the Development of Impaired Vascular Function and Repair in Human and Murine Lupus by Seth G Thacker A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Immunology) in The University of Michigan 2011 Doctoral Committee: Associate Professor Mariana J. Kaplan, Chair Professor David A. Fox Professor Alisa E. Koch Professor Matthias Kretzler Professor Nicholas W. Lukacs Associate Professor Daniel T. Eitzman © Seth G Thacker 2011 Sharon, this work is dedicated to you. This achievement is as much yours as it is mine. Your support through all six years of this Ph.D. process has been incredible. You put up with my countless miscalculations on when I would finish experiments, and still managed to make me and our kids feel loved and special. Without you this would have no meaning. Sharon, you are the safe harbor in my life. ii Acknowledgments I have been exceptionally fortunate in my time here at the University of Michigan. I have been able to interact with so many supportive people over the years. I would like to express my thanks and admiration for my mentor. Mariana has taught me so much about writing, experimental design and being a successful scientist in general. I could never have made it here without her help. I would also like to thank Mike Denny. He had a hand in the beginning of all of my projects in one way or another, and was always quick and eager to help in whatever way he could. He really made my first year in the lab successful.
    [Show full text]
  • IFN-Ε Protects Primary Macrophages Against HIV Infection
    RESEARCH ARTICLE IFN-ε protects primary macrophages against HIV infection Carley Tasker,1 Selvakumar Subbian,2 Pan Gao,3 Jennifer Couret,1 Carly Levine,2 Saleena Ghanny,1 Patricia Soteropoulos,1 Xilin Zhao,1,2 Nathaniel Landau,4 Wuyuan Lu,3 and Theresa L. Chang1,2 1Department of Microbiology, Biochemistry and Molecular Genetics and 2Public Health Research Institute, Rutgers University, New Jersey Medical School, Newark, New Jersey, USA.3Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA.4Department of Microbiology, New York University School of Medicine, New York, New York, USA. IFN-ε is a unique type I IFN that is not induced by pattern recognition response elements. IFN-ε is constitutively expressed in mucosal tissues, including the female genital mucosa. Although the direct antiviral activity of IFN-ε was thought to be weak compared with IFN-α, IFN-ε controls Chlamydia muridarum and herpes simplex virus 2 in mice, possibly through modulation of immune response. We show here that IFN-ε induces an antiviral state in human macrophages that blocks HIV-1 replication. IFN-ε had little or no protective effect in activated CD4+ T cells or transformed cell lines unless activated CD4+ T cells were infected with replication-competent HIV-1 at a low MOI. The block to HIV infection of macrophages was maximal after 24 hours of treatment and was reversible. IFN-ε acted on early stages of the HIV life cycle, including viral entry, reverse transcription, and nuclear import. The protection did not appear to operate through known type I IFN-induced HIV host restriction factors, such as APOBEC3A and SAMHD1.
    [Show full text]
  • Biomarker Testing in Non- Small Cell Lung Cancer (NSCLC)
    The biopharma business of Merck KGaA, Darmstadt, Germany operates as EMD Serono in the U.S. and Canada. Biomarker testing in non- small cell lung cancer (NSCLC) Copyright © 2020 EMD Serono, Inc. All rights reserved. US/TEP/1119/0018(1) Lung cancer in the US: Incidence, mortality, and survival Lung cancer is the second most common cancer diagnosed annually and the leading cause of mortality in the US.2 228,820 20.5% 57% Estimated newly 5-year Advanced or 1 survival rate1 metastatic at diagnosed cases in 2020 diagnosis1 5.8% 5-year relative 80-85% 2 135,720 survival with NSCLC distant disease1 Estimated deaths in 20201 2 NSCLC, non-small cell lung cancer; US, United States. 1. National Institutes of Health (NIH), National Cancer Institute. Cancer Stat Facts: Lung and Bronchus Cancer website. www.seer.cancer.gov/statfacts/html/lungb.html. Accessed May 20, 2020. 2. American Cancer Society. What is Lung Cancer? website. https://www.cancer.org/cancer/non-small-cell-lung-cancer/about/what-is-non-small-cell-lung-cancer.html. Accessed May 20, 2020. NSCLC is both histologically and genetically diverse 1-3 NSCLC distribution by histology Prevalence of genetic alterations in NSCLC4 PTEN 10% DDR2 3% OTHER 25% PIK3CA 12% LARGE CELL CARCINOMA 10% FGFR1 20% SQUAMOUS CELL CARCINOMA 25% Oncogenic drivers in adenocarcinoma Other or ADENOCARCINOMA HER2 1.9% 40% KRAS 25.5% wild type RET 0.7% 55% NTRK1 1.7% ROS1 1.7% Oncogenic drivers in 0% 20% 40% 60% RIT1 2.2% squamous cell carcinoma Adenocarcinoma DDR2 2.9% Squamous cell carcinoma NRG1 3.2% Large cell carcinoma
    [Show full text]
  • Kinase Inhibitors: an Introduction
    David Peters Baran Group Meeting Kinase Inhibitors: An Introduction 2/2/19 INTRODUCTION SIGNAL TRANSDUCTION KINASES ARE IMPORTANT IN HUMAN BIOLOGY/DISEASE: The process describing how a signal (chemical/physical) is transmitted through a - Kinases are a superfamily of proteins (5th largest in humans) cell ultimately resulting in a cellular response - 518 genes and 106 pseudogenes - Diverse in size, subunit structure, and cellular location RECEPTOR TRANSDUCERS EFFECTORS - ~260 residues make up their conserved catalytic core - dysregulation of kinases occurs in many diseases (cancer, inflamatory, degenerative, and autoimmune diseases) - signals can originate inside or outside the cell - 244 of the 518 map to disease loci - signals generally passed through a series of steps (signal transduction pathway) often consisting of multiple enzymes and messengers protein O - pathways open possibility for signal aplification (>1x106) kinase ATP + PROTEIN OH ADP + PROTEIN O P O - Extracellular signals transduced by RTKs, GPCR’s, guanlyl cyclases, or ligand-gated Ion channels O - Phosphorylation is the most prominent covalent modification/signal in KINASES INHIBITORS ARE IMPORTANT IN DISEASE THERAPY/RESEARCH: cellular regulation - currently 51 FDA approved small molecule kinase inhibitors - “converter enzymes” (protein kinases and phosphoprotein phosphorlyases) - 4 FDA approved antibody kinase inhibitors are regulated; conserve ATP/maintain desired target protein state - thousands of known inhibitors spanning the kinome - pathway ends by affecting biomolecule
    [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]
  • Type I Interferon Suppresses Tumor Growth Through Activating the STAT3-Granzyme B Pathway in Tumor-Infiltrating Cytotoxic T Lymphocytes Chunwan Lu1,2,3*, John D
    Lu et al. Journal for ImmunoTherapy of Cancer (2019) 7:157 https://doi.org/10.1186/s40425-019-0635-8 RESEARCHARTICLE Open Access Type I interferon suppresses tumor growth through activating the STAT3-granzyme B pathway in tumor-infiltrating cytotoxic T lymphocytes Chunwan Lu1,2,3*, John D. Klement1,2,3, Mohammed L. Ibrahim1,2,3, Wei Xiao1,2, Priscilla S. Redd1,2,3, Asha Nayak-Kapoor2, Gang Zhou2 and Kebin Liu1,2,3* Abstract Background: Type I interferons (IFN-I) have recently emerged as key regulators of tumor response to chemotherapy and immunotherapy. However, IFN-I function in cytotoxic T lymphocytes (CTLs) in the tumor microenvironment is largely unknown. Methods: Tumor tissues and CTLs of human colorectal cancer patients were analyzed for interferon (alpha and beta) receptor 1 (IFNAR1) expression. IFNAR1 knock out (IFNAR-KO), mixed wild type (WT) and IFNAR1-KO bone marrow chimera mice, and mice with IFNAR1 deficiency only in T cells (IFNAR1-TKO) were used to determine IFN-I function in T cells in tumor suppression. IFN-I target genes in tumor-infiltrating and antigen-specific CTLs were identified and functionally analyzed. Results: IFNAR1 expression level is significantly lower in human colorectal carcinoma tissue than in normal colon tissue. IFNAR1 protein is also significantly lower on CTLs from colorectal cancer patients than those from healthy donors. Although IFNAR1-KO mice exhibited increased susceptibility to methylcholanthrene-induced sarcoma, IFNAR1-sufficient tumors also grow significantly faster in IFNAR1-KO mice and in mice with IFNAR1 deficiency only in T cells (IFNAR1-TKO), suggesting that IFN-I functions in T cells to enhance host cancer immunosurveillance.
    [Show full text]
  • Chemotherapy Followed by Anti-CD137 Mab Immunotherapy Improves Disease Control in a Mouse Myeloma Model
    Chemotherapy followed by anti-CD137 mAb immunotherapy improves disease control in a mouse myeloma model Camille Guillerey, … , Ludovic Martinet, Mark J. Smyth JCI Insight. 2019;4(14):e125932. https://doi.org/10.1172/jci.insight.125932. Research Article Immunology Immunotherapy holds promise for patients with multiple myeloma (MM), but little is known about how MM-induced immunosuppression influences response to therapy. Here, we investigated the impact of disease progression on immunotherapy efficacy in the Vk*MYC mouse model. Treatment with agonistic anti-CD137 (4-1BB) mAbs efficiently protected mice when administered early but failed to contain MM growth when delayed more than 3 weeks after Vk*MYC tumor cell challenge. The quality of the CD8+ T cell response to CD137 stimulation was not altered by the presence of MM, but CD8+ T cell numbers were profoundly reduced at the time of treatment. Our data suggest that an insufficient ratio of CD8+ T cells to MM cells (CD8/MM ratio) accounts for the loss of anti-CD137 mAb efficacy. We established serum M- protein levels prior to therapy as a predictive factor of response. Moreover, we developed an in silico model to capture the dynamic interactions between CD8+ T cells and MM cells. Finally, we explored two methods to improve the CD8/MM ratio: anti-CD137 mAb immunotherapy combined with Treg depletion or administered after chemotherapy treatment with cyclophosphamide or melphalan efficiently reduced MM burden and prolonged survival. Together, our data indicate that consolidation treatment with anti-CD137 mAbs might prevent MM relapse. Find the latest version: https://jci.me/125932/pdf RESEARCH ARTICLE Chemotherapy followed by anti-CD137 mAb immunotherapy improves disease control in a mouse myeloma model Camille Guillerey,1,2,3 Kyohei Nakamura,1 Andrea C.
    [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]
  • The Role of Signaling Pathways in the Development and Treatment of Hepatocellular Carcinoma
    Oncogene (2010) 29, 4989–5005 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 www.nature.com/onc REVIEW The role of signaling pathways in the development and treatment of hepatocellular carcinoma S Whittaker1,2, R Marais3 and AX Zhu4 1Dana-Farber Cancer Institute, Boston, MA, USA; 2The Broad Institute, Cambridge, MA, USA; 3Institute of Cancer Research, London, UK and 4Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA Hepatocellular carcinoma (HCC) is a highly prevalent, malignancy in adults (Pons-Renedo and Llovet, 2003). treatment-resistant malignancy with a multifaceted mole- For the vast majority of patients, HCC is a late cular pathogenesis. Current evidence indicates that during complication of chronic liver disease, and as such, is hepatocarcinogenesis, two main pathogenic mechanisms often associated with cirrhosis. The main risk factors for prevail: (1) cirrhosis associated with hepatic regeneration the development of HCC include infection with hepatitis after tissue damage caused by hepatitis infection, toxins B virus (HBV) or hepatitis C virus (HCV). Hepatitis (for example, alcohol or aflatoxin) or metabolic influ- infection is believed to be the main etiologic factor in ences, and (2) mutations occurring in single or multiple 480% of cases (Anzola, 2004). Other risk factors oncogenes or tumor suppressor genes. Both mechanisms include excessive alcohol consumption, nonalcoholic have been linked with alterations in several important steatohepatitis, autoimmune hepatitis, primary biliary cellular signaling pathways. These pathways are of cirrhosis, exposure to environmental carcinogens (parti- interest from a therapeutic perspective, because targeting cularly aflatoxin B) and the presence of various genetic them may help to reverse, delay or prevent tumorigenesis.
    [Show full text]
  • Epigenetic Gene Regulation by Janus Kinase 1 in Diffuse Large B-Cell Lymphoma
    Epigenetic gene regulation by Janus kinase 1 in diffuse large B-cell lymphoma Lixin Ruia,b,c,1,2, Amanda C. Drennanb,c,1, Michele Ceribellia,1, Fen Zhub,c, George W. Wrightd, Da Wei Huanga, Wenming Xiaoe, Yangguang Lib,c, Kreg M. Grindleb,c,LiLub,c, Daniel J. Hodsona, Arthur L. Shaffera, Hong Zhaoa, Weihong Xua, Yandan Yanga, and Louis M. Staudta,2 aLymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892; bDepartment of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705; cCarbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705; dBiometric Research Branch, DCTD, National Cancer Institute, NIH, Bethesda, MD 20892; and eDivision of Bioinformatics and Biostatistics, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079 Contributed by Louis M. Staudt, September 29, 2016 (sent for review July 22, 2016; reviewed by Anthony R. Green and Ross L. Levine) Janus kinases (JAKs) classically signal by activating STAT transcription promote STAT dimerization, nuclear translocation, and binding factors but can also regulate gene expression by epigenetically to cis-regulatory elements to regulate transcription (15, 17). This phosphorylating histone H3 on tyrosine 41 (H3Y41-P). In diffuse large canonical JAK/STAT pathway is deregulated in several hemato- B-cell lymphomas (DLBCLs), JAK signaling is a feature of the activated logic malignancies (16). In DLBCL, STAT3 is activated in the B-cell (ABC) subtype and is triggered by autocrine production of IL-6 ABC subtype and regulates gene expression to promote the sur- and IL-10.
    [Show full text]
  • Open Chen-Thesis Finalv5.Pdf
    The Pennsylvania State University The Graduate School Department of Neural and Behavioral Sciences EPIGENETIC ANALYSIS OF IMMUNE ASSOCIATED SIGNALING MOLECULES DURING MOUSE RETINA DEVELOPMENT A Thesis in Anatomy by Chen Yang © 2013 Chen Yang Submitted in Partial Fulfillment of the Requirements for the Degree of Master of Science May 2013 The thesis of Chen Yang was reviewed and approved* by the following: Samuel Shao-Min Zhang Assistant Professor of Neural and Behavioral Sciences Thesis Advisor Colin J. Barnstable Department Head of Neural and Behavioral Sciences Professor of Neural and Behavioral Sciences Patricia J. McLaughlin Professor of Neural and Behavioral Sciences Director of Graduate Program in Anatomy *Signatures are on file in the Graduate School. ii ABSTRACT The retina is an immune-privileged organ. Many autoimmune diseases, such as AMD, glaucoma, and diabetic retinopathy, are caused by excessive inflammatory responses targeting self-tissue. The physiological functions of extracellular and intracellular signaling molecules of immune responses have been well characterized. The epigenetic aspects of these molecules in the retina, however, have not been well elucidated. In this study, we examined the expression of selected immune-related genes, and their transcriptional accessibility via epigenetic mapping, cluster analysis, and RT-PCR. Among these genes, interleukin receptor related genes and intracellular signaling molecules exhibit higher transcriptional accessibility. Epigenetic mapping of the toll-like receptor (TLR) family revealed that 3 out of 13 TLRs exhibit H3K4me2 accumulation during retina development, suggesting that TLR2, TLR3, and TLR9 are the only TLR members expressed in the retina. Most of the NF-κB signaling molecules exhibited transcriptional accessibility, implying their essential roles in inflammatory regulation during retina maturation.
    [Show full text]