Differential and Overlapping Immune Programs Regulated by IRF3 and IRF5 in Plasmacytoid Dendritic Cells

Total Page:16

File Type:pdf, Size:1020Kb

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. We show that the protein kinases IKKa, IKKb, IKK«, and TANK-binding kinase 1 each confer IRF5 phosphorylation/dimerization, thus extending the family of IRF5 activator kinases. Among primary human immune cell subsets, we found that IRF5 is most abundant in plasmacytoid dendritic cells (pDCs). Flow cytometric cell imaging revealed that IRF5 is specifically activated by endosomal TLR signaling. Comparative analyses revealed that IRF3 is activated in pDCs uniquely through RIG-I–like receptor (RLR) signaling. Transcriptomic analyses of pDCs show that the partitioning of TLR7/IRF5 and RLR/IRF3 pathways confers differential gene expression and immune cytokine production in pDCs, linking IRF5 with immune Downloaded from regulatory and proinflammatory gene expression. Thus, TLR7/IRF5 and RLR–IRF3 partitioning serves to polarize pDC response outcome. Strategies to differentially engage IRF signaling pathways should be considered in the design of immunotherapeutic approaches to modulate or polarize the immune response for specific outcome. The Journal of Immunology, 2018, 201: 000–000. nterferon regulatory factors (IRFs) are transcription factors of these IRFs and translocation into the nucleus to induce gene that regulate the intricate gene networks essential for coor- expression (6). Stimulation of some TLRs also activates IRF3 and http://www.jimmunol.org/ I dinating an appropriate and effective immune response (1, 2). IRF7 to induce type I IFNs (7). In particular, IRF3 and IRF7 have been extensively studied and In contrast to IRF3 and IRF7, IRF5 regulation and function are shown to regulate the induction of type I IFNs and other cytokines less well characterized. Mouse studies revealed essential roles of in response to pattern recognition receptor (PRR) recognition of IRF5 in the production of IFN-b and proinflammatory mediators, pathogen-associated molecular patterns (PAMPs) during virus including IL-6, IL-12, and TNF-a (8–12). In humans, carriers of infection (3, 4). During RNA virus infection, viral PAMP RNA autoimmune risk haplotypes at the IRF5 locus exhibit elevated motifs are recognized by RIG-I–like receptors (RLRs), leading to levels of IFN-a (13–16), and dendritic cells (DCs) from these RLR signaling activation and interaction with the adaptor MAVS carriers produced elevated TNF-a and IL-12 upon TLR stimula- (5). MAVS recruits TANK-binding kinase 1 (TBK1), which tion (17, 18). In HEK293 cells overexpressing TLR7, the TLR7/8 by guest on September 28, 2021 phosphorylates IRF3 and IRF7, leading to the homodimerization agonist R848 induced activation of an IRF5 reporter, accompanied by the translocation of IRF5–GFP into the nucleus (19). TBK1 *Department of Immunology, Center for Innate Immunity and Immune Disease, was reported to phosphorylate IRF5 (19, 20), and a kinase-dead University of Washington, Seattle, WA 98109; †Department of Biomedical Sciences, mutant of TBK1 or the related IKKε inhibited the TLR7-dependent City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region; activation of a Gal4–IRF5 reporter (19). These results suggest that and ‡Laboratory of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata, Niigata Prefecture 950-2181, Japan TBK1 and IKKε activate both IRF5 and IRF3. Subsequent reports ORCIDs: 0000-0003-1012-0797 (K.T.C.); 0000-0003-2007-1183 (M.N.). identified IKKb as the activating kinase of IRF5 (21, 22). MAVS Received for publication February 15, 2018. Accepted for publication September 13, overexpression was also shown to induce IRF5 dimerization in 2018. HEK293T cells (21, 22), and RIG-I and IRF5 coexpression rescued This work was supported by the Croucher Foundation Postdoctoral Fellowship, a cytokine production defects in Irf3/5/7-deficient mouse cells (23), National Research Service Award F32 Postdoctoral Fellowship from the National suggesting that both TLR and RLR stimulation may activate IRF5. Institutes of Health (NIH) to K.T.C. (AI115935), and NIH Grants AI104002, AI100625, and AI083019. Of note is that these studies have typically relied on over- expression approaches often in epithelial cell lines to evaluate K.T.C. designed and performed experiments and wrote the manuscript; C.W. and R.G. performed bioinformatics analyses; Y.-M.L. and M.K. performed experiments; IRF5 functions. In human plasmacytoid DC (pDC) cell lines, and M.G. directed the research and edited the manuscript. RNA interference–mediated IRF5 knockdown experiments have The microarray data presented in this article have been submitted to the Gene established IRF5 as a crucial mediator type I IFN induction Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) under accession number (21, 24). However, the relevant regulatory pathways, endogenous GSE108526. steps of IRF5 activation, and IRF5 transcriptional signatures Address correspondence and reprint requests to Dr. Michael Gale, Jr. and Dr. Yueh- Ming Loo, Department of Immunology, University of Washington, Office E383, Box have not been defined nor directly compared with signaling from 358059, 750 Republican Street, Seattle, WA 981909-4766. E-mail addresses: other IRFs. [email protected] (M.G.) and [email protected] (Y.-M.L.) In this study, we evaluated the IRF5 activation process and The online version of this article contains supplemental material. defined the IRF5 transcriptome. Our study shows that each IKK Abbreviations used in this article: DC, dendritic cell; DE, differentially expressed; kinase can direct IRF5 phosphorylation for dimerization/activation GO, gene ontology; HAU, hemagglutination unit; HCV, hepatitis C virus; IRF, IFN regulatory factor; PAMP, pathogen-associated molecular pattern; pDC, plasmacytoid and that IRF5 is variably present in different primary immune cell DC; PRR, pattern recognition receptor; RLR, RIG-I–like receptor; RNA-seq, RNA subsets wherein it is highly abundant in pDCs. Using robust assays sequencing; SeV, Sendai virus; siRNA, small interfering RNA; SS, similarity score; to assess and compare endogenous IRF5 and IRF3 activation in TBK1, TANK-binding kinase 1; xRNA, X region of the HCV genome. pDCs, we show that IRF5 is activated upon endosomal TLR7/8 Copyright Ó 2018 by The American Association of Immunologists, Inc. 0022-1767/18/$37.50 stimulation, whereas RLR stimulation leads to IRF3 activation. www.jimmunol.org/cgi/doi/10.4049/jimmunol.1800221 2 PARTITIONED SIGNALING OF IRF5 AND IRF3 High throughput transcriptomic analyses reveal that triggering pDCs and anti-IRF3 (D83B9; Cell Signaling Technology). AF680- or AF790- through TLR7/IRF5 and RLR/IRF3 pathways leads to a response conjugated secondary Abs were purchased from Jackson ImmunoResearch. that induces specific gene expression profiles and the production of ImageStream imaging flow cytometry distinct sets of cytokines for differential immune activation. This unique partitioning of TLR7/IRF5 and RLR/IRF3 signaling serves to PBMCs or enriched pDCs were stained with the following cell surface markers: CD3-FITC (UCHT1; BD Biosciences), CD14-AF488 (M5E2; BD drive expanded proinflammatory, immune regulatory, and antiviral Biosciences), CD19-FITC (HIB19; BD Biosciences), CD56-AF488 (B159; actions of pDCs. BD Biosciences), CD11c-FITC, HLA-DR-PE (L243; BioLegend), BCDA2- PE (AC144; Miltenyi Biotec), and FITC-conjugated lineage mixture panel 1 (BD Biosciences). Cells were fixed and permeabilized using the Cytofix/ Materials and Methods Cytoperm kit (Becton Dickinson) and stained with IRF5-AF647 Ab Cells (EPR6094; Abcam) and DAPI (Life Technologies). Stained cells were 3 CAL-1 was gifted by Dr. T. Maeda (Nagasaki University). PMDC05 cells acquired on an ImageStreamX Mark II imaging flow cytometer at 60 were from M. Narita
Recommended publications
  • Imm Catalog.Pdf
    $ Gene Symbol A B 3 C 4 D 9 E 10 F 11 G 12 H 13 I 14 J. K 17 L 18 M 19 N 20 O. P 22 R 26 S 27 T 30 U 32 V. W. X. Y. Z 33 A ® ® Gene Symbol Gene ID Antibody Monoclonal Antibody Polyclonal MaxPab Full-length Protein Partial-length Protein Antibody Pair KIt siRNA/Chimera Gene Symbol Gene ID Antibody Monoclonal Antibody Polyclonal MaxPab Full-length Protein Partial-length Protein Antibody Pair KIt siRNA/Chimera A1CF 29974 ● ● ADAMTS13 11093 ● ● ● ● ● A2M 2 ● ● ● ● ● ● ADAMTS20 80070 ● AACS 65985 ● ● ● ADAMTS5 11096 ● ● ● AANAT 15 ● ● ADAMTS8 11095 ● ● ● ● AATF 26574 ● ● ● ● ● ADAMTSL2 9719 ● AATK 9625 ● ● ● ● ADAMTSL4 54507 ● ● ABCA1 19 ● ● ● ● ● ADAR 103 ● ● ABCA5 23461 ● ● ADARB1 104 ● ● ● ● ABCA7 10347 ● ADARB2 105 ● ABCB9 23457 ● ● ● ● ● ADAT1 23536 ● ● ABCC4 10257 ● ● ● ● ADAT2 134637 ● ● ABCC5 10057 ● ● ● ● ● ADAT3 113179 ● ● ● ABCC8 6833 ● ● ● ● ADCY10 55811 ● ● ABCD2 225 ● ADD1 118 ● ● ● ● ● ● ABCD4 5826 ● ● ● ADD3 120 ● ● ● ABCG1 9619 ● ● ● ● ● ADH5 128 ● ● ● ● ● ● ABL1 25 ● ● ADIPOQ 9370 ● ● ● ● ● ABL2 27 ● ● ● ● ● ADK 132 ● ● ● ● ● ABO 28 ● ● ADM 133 ● ● ● ABP1 26 ● ● ● ● ● ADNP 23394 ● ● ● ● ABR 29 ● ● ● ● ● ADORA1 134 ● ● ACAA2 10449 ● ● ● ● ADORA2A 135 ● ● ● ● ● ● ● ACAN 176 ● ● ● ● ● ● ADORA2B 136 ● ● ACE 1636 ● ● ● ● ADRA1A 148 ● ● ● ● ACE2 59272 ● ● ADRA1B 147 ● ● ACER2 340485 ● ADRA2A 150 ● ● ACHE 43 ● ● ● ● ● ● ADRB1 153 ● ● ACIN1 22985 ● ● ● ADRB2 154 ● ● ● ● ● ACOX1 51 ● ● ● ● ● ADRB3 155 ● ● ● ● ACP5 54 ● ● ● ● ● ● ● ADRBK1 156 ● ● ● ● ACSF2 80221 ● ● ADRM1 11047 ● ● ● ● ACSF3 197322 ● ● AEBP1 165 ● ● ● ● ACSL4 2182 ●
    [Show full text]
  • 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]
  • Supplementary Figure S1. Intracellular Ca2+ Levels Following Decursin Treatment in F11 Cells in the Presence of Menthol
    Supplementary Figure S1. Intracellular Ca2+ levels following decursin treatment in F11 cells in the presence of menthol (A) Intracellular Ca2+ levels after treatment with decursin every 3 s. The red arrow indicates the duration of treatment with 200 μM of menthol and decursin. NC: The negative control treated with DMSO only; PC: The positive control treated with 200 μM menthol without decursin. (B) Average intracellular Ca2+ levels after treatment with decursin. The average was quantified from the normalized Δ340/380 ratio for 10 cycles after treatment with the decursin solution at the 10th cycle, as shown in Fig. 1A. The normalized Δ340/380 ratio was calculated using the following for- mula: [ratio of fluorescence intensity at 510 nm (emission) to that at 340 nm (excitation)]/[ratio of fluorescence intensity at 510 nm (emission) to that at a wavelength of 380 nm (excitation)]. Cells 2021, 10, 547. https://doi.org/10.3390/cells10030547 www.mdpi.com/journal/cells Cells 2021, 10, 547 2 of 5 Table S1. List of protein targets of decursin detected by the SwissTargetPrediction web tool Common Target Uniprot ID ChEMBL ID Target Class Probability name Poly [ADP-ribose] polymerase-1 PARP1 P09874 CHEMBL3105 Enzyme 0.104671941 N-acylsphingosine-amidohydro- NAAA Q02083 CHEMBL4349 Enzyme 0.104671941 lase Acid ceramidase ASAH1 Q13510 CHEMBL5463 Enzyme 0.104671941 Family A G protein- Neuropeptide Y receptor type 5 NPY5R Q15761 CHEMBL4561 0.104671941 coupled receptor Family A G protein- Melatonin receptor 1A MTNR1A P48039 CHEMBL1945 0.104671941 coupled
    [Show full text]
  • Significant Shortest Paths for the Detection of Putative Disease Modules
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.01.019844; this version posted April 2, 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-ND 4.0 International license. SIGNIFICANT SHORTEST PATHS FOR THE DETECTION OF PUTATIVE DISEASE MODULES Daniele Pepe1 1Department of Oncology, KU Leuven, LKI–Leuven Cancer Institute, Leuven, Belgium Email address: DP: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.04.01.019844; this version posted April 2, 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-ND 4.0 International license. Keywords Structural equation modeling, significant shortest paths, pathway analysis, disease modules. Abstract Background The characterization of diseases in terms of perturbated gene modules was recently introduced for the analysis of gene expression data. Some approaches were proposed in literature, but many times they are inductive approaches. This means that starting directly from data, they try to infer key gene networks potentially associated to the biological phenomenon studied. However they ignore the biological information already available to characterize the gene modules. Here we propose the detection of perturbed gene modules using the combination of data driven and hypothesis-driven approaches relying on biological metabolic pathways and significant shortest paths tested by structural equation modeling.
    [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]
  • Comprehensive Study of Nuclear Receptor DNA Binding Provides a Revised Framework for Understanding Receptor Specificity
    ARTICLE https://doi.org/10.1038/s41467-019-10264-3 OPEN Comprehensive study of nuclear receptor DNA binding provides a revised framework for understanding receptor specificity Ashley Penvose 1,2,4, Jessica L. Keenan 2,3,4, David Bray2,3, Vijendra Ramlall 1,2 & Trevor Siggers 1,2,3 The type II nuclear receptors (NRs) function as heterodimeric transcription factors with the retinoid X receptor (RXR) to regulate diverse biological processes in response to endogenous 1234567890():,; ligands and therapeutic drugs. DNA-binding specificity has been proposed as a primary mechanism for NR gene regulatory specificity. Here we use protein-binding microarrays (PBMs) to comprehensively analyze the DNA binding of 12 NR:RXRα dimers. We find more promiscuous NR-DNA binding than has been reported, challenging the view that NR binding specificity is defined by half-site spacing. We show that NRs bind DNA using two distinct modes, explaining widespread NR binding to half-sites in vivo. Finally, we show that the current models of NR specificity better reflect binding-site activity rather than binding-site affinity. Our rich dataset and revised NR binding models provide a framework for under- standing NR regulatory specificity and will facilitate more accurate analyses of genomic datasets. 1 Department of Biology, Boston University, Boston, MA 02215, USA. 2 Biological Design Center, Boston University, Boston, MA 02215, USA. 3 Bioinformatics Program, Boston University, Boston, MA 02215, USA. 4These authors contributed equally: Ashley Penvose, Jessica L. Keenan. Correspondence
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • IRF5 Gene Interferon Regulatory Factor 5
    IRF5 gene interferon regulatory factor 5 Normal Function The protein produced from the IRF5 gene, called interferon regulatory factor 5 (IRF5), acts as a transcription factor, which means that it attaches (binds) to specific regions of DNA and helps control the activity of certain genes. When a virus is recognized in the cell, the IRF5 gene is turned on (activated), which leads to the production of IRF5 protein. The protein binds to specific regions of DNA that regulate the activity of genes that produce interferons and other cytokines. Cytokines are proteins that help fight infection by promoting inflammation and regulating the activity of immune system cells. In particular, interferons control the activity of genes that help block the replication of viruses, and they stimulate the activity of certain immune system cells known as natural killer cells. Health Conditions Related to Genetic Changes Systemic scleroderma Several normal variations in the IRF5 gene have been associated with an increased risk of developing systemic scleroderma, which is an autoimmune disorder characterized by the buildup of scar tissue (fibrosis) in the skin and internal organs. Although the IRF5 gene is known to stimulate the immune system in response to viruses, it is unknown how the gene variations contribute to the increased risk of systemic scleroderma. Researchers believe that a combination of genetic and environmental factors may play a role in development of the condition. Rheumatoid arthritis MedlinePlus Genetics provides information about Rheumatoid arthritis Systemic lupus erythematosus MedlinePlus Genetics provides information about Systemic lupus erythematosus Ulcerative colitis MedlinePlus Genetics provides information about Ulcerative colitis Reprinted from MedlinePlus Genetics (https://medlineplus.gov/genetics/) 1 Autoimmune disorders Studies have associated normal variations in the IRF5 gene with an increased risk of several autoimmune disorders.
    [Show full text]
  • IFN-Α Wakes up Sleeping Hematopoietic Stem Cells
    NEWS AND VIEWS calcium consumption does not seem to have without increasing the risk of coronary heart 7. Aoki, K. et al. Ann. Epidemiol. 15, 598–606 (2005). such an effect. How to optimize calcium sup- disease, which has been associated with cal- 8. Langhans, N. et al. Gastroenterology 112, 280–286 13 plements for human health deserves further cium supplementation . (1997). investigation. 9. Recker, R.R. N. Engl. J. Med. 313, 70–73 (1985). 10. Yang, Y.-X. et al. J. Am. Med. Assoc. , 2947– Maybe it’s time to determine whether low- 1. Zaidi, M. Nat. Med. 13, 791–801 (2007). 296 2. Del Fattore, A. et al. Bone 42, 19–29 (2008). 2953 (2006). fat milk–based drinks taken as an alternative 3. Sobacchi, C. et al. Hum. Mol. Genet. 10, 1767–1773 11. Collazo-Clavell, M.L., Jimenez, A., Hodgson, S.F. & to the soda so frequently consumed, especially (2001). Sarr, M.G. Endocr. Pract. 10, 195–198 (2004). 4. Amling, M. et al. Nat. Med. 15, 674–681 (2009). 12. Bo-Linn, G.W. et al. J. Clin. Invest. 73, 640–647 during the early adolescent years, can attain 5. Straub, D.A. Nutr. Clin. Pract. 22, 286–296 (2007). (1984). the noble goal of keeping bones healthy— 6. Teitelbaum, S.L. Science 289, 1504–1508 (2000). 13. Bolland, M.J. BMJ 336, 262–266 (2008). IFN-α wakes up sleeping hematopoietic stem cells Emmanuelle Passegué & Patricia Ernst The cytokine interferon-α stimulates the turnover and proliferation of hematopoietic cells in vivo (pages 696–700).
    [Show full text]
  • Circrna LRIG3 Knockdown Inhibits Hepatocellular Carcinoma Progression by Regulating Mir-223-3P and MAPK/ERK Pathway
    CircRNA LRIG3 knockdown inhibits hepatocellular carcinoma progression by regulating miR-223-3p and MAPK/ERK pathway Type Research paper Keywords hepatocellular carcinoma, miR-223-3p, circ_LRIG3, MAP2K6, MAPK/ERK pathway Abstract Introduction Emerging evidence suggests that circular RNAs (circRNAs) play critical roles in tumorigenesis. However, the roles and molecular mechanisms of circRNA leucine-rich repeat immunoglobulin domain-containing protein 3 (circ_LRIG3) in hepatocellular carcinoma (HCC) has not been investigated. Material and methods The expression levels of circ_LRIG3, miR-223-3p, and mitogen-activated protein kinase kinase 6 (MAP2K6) were determined by qRT-PCR. Flow cytometry was applied to determine the cell cycle distribution and apoptosis. Cell proliferation, migration and invasion were assessed by MTT, colony formation, and transwell assays. Western blot assay was employed to measure the protein levels of the snail, E-cadherin, MAP2K6, mitogen-activated protein kinase (MAPK), phospho-MAPK (p- MAPK), extracellular signal-regulated kinases (ERKs), and phospho-ERKs (p- ERKs). The relationship between miR-223-3p and circ_LRIG3 or MAP2K6 was predicted by bioinformatics tools and verified by dual-luciferase reporter assay. A xenograft tumor model was established to confirm the functions of circ_LRIG3 in vivo. Results Preprint Circ_LRIG3 and MAP2K6 expression were enhanced while miR-223-3p abundance was reduced in HCC tissues and cells. Knockdown of circ_LRIG3 inhibited cell proliferation, metastasis, and increasing apoptosis. MiR-223-3p was a target of circ_LRIG3, and its downregulation reversed the inhibitory effect of circ_LRIG3 knockdown on the progression of HCC cells. Moreover, MAP2K6 could bind to miR-223-3p, and MAP2K6 upregulation also abolished the suppressive impact of circ_LRIG3 interference on progression of HCC cells.
    [Show full text]
  • Interactions Between the Parasite Philasterides Dicentrarchi and the Immune System of the Turbot Scophthalmus Maximus.A Transcriptomic Analysis
    biology Article Interactions between the Parasite Philasterides dicentrarchi and the Immune System of the Turbot Scophthalmus maximus.A Transcriptomic Analysis Alejandra Valle 1 , José Manuel Leiro 2 , Patricia Pereiro 3 , Antonio Figueras 3 , Beatriz Novoa 3, Ron P. H. Dirks 4 and Jesús Lamas 1,* 1 Department of Fundamental Biology, Institute of Aquaculture, Campus Vida, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; [email protected] 2 Department of Microbiology and Parasitology, Laboratory of Parasitology, Institute of Research on Chemical and Biological Analysis, Campus Vida, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; [email protected] 3 Institute of Marine Research, Consejo Superior de Investigaciones Científicas-CSIC, 36208 Vigo, Spain; [email protected] (P.P.); antoniofi[email protected] (A.F.); [email protected] (B.N.) 4 Future Genomics Technologies, Leiden BioScience Park, 2333 BE Leiden, The Netherlands; [email protected] * Correspondence: [email protected]; Tel.: +34-88-181-6951; Fax: +34-88-159-6904 Received: 4 September 2020; Accepted: 14 October 2020; Published: 15 October 2020 Simple Summary: Philasterides dicentrarchi is a free-living ciliate that causes high mortality in marine cultured fish, particularly flatfish, and in fish kept in aquaria. At present, there is still no clear picture of what makes this ciliate a fish pathogen and what makes fish resistant to this ciliate. In the present study, we used transcriptomic techniques to evaluate the interactions between P. dicentrarchi and turbot leucocytes during the early stages of infection. The findings enabled us to identify some parasite genes/proteins that may be involved in virulence and host resistance, some of which may be good candidates for inclusion in fish vaccines.
    [Show full text]
  • Age, DNA Methylation and the Malignant Potential of the Serrated Neoplasia Pathway Lochlan John Fennell B
    Age, DNA Methylation and the Malignant Potential of the Serrated Neoplasia Pathway Lochlan John Fennell B. Biomed Sci A thesis submitted for the degree of Doctor of Philosophy at The University of Queensland in 2020 Faculty of Medicine ORC ID: 0000-0003-3214-3527 1 Abstract Colorectal cancer is the third most common cancer in Australia and is responsible for the death of over four thousand Australians each year. There are two overarching molecular pathways leading to colorectal cancer. The conventional pathway, which is responsible for ~75% of colorectal cancer diagnoses, occurs in a step-wise manner and is the consequence of a series of genetic alterations including mutations of tumour suppressor genes and gross chromosomal abnormalities. This pathway has been extensively studied over the past three decades. The serrated neoplasia pathway is responsible for the remaining colorectal cancers. This pathway is triggered by oncogenic BRAF mutation and these cancers accumulate epigenetic alterations while progressing to invasive cancer. DNA methylation is important in serrated neoplasia, however the extent and role of DNA methylation on the initiation and progression of serrated lesions is not clear. DNA methylation accumulates in tissues with age, and advanced serrated lesions and cancers occur almost exclusively in elderly patients. How this methylation affects serrated lesions is unknown. In this thesis I set out to address three key research questions related to DNA methylation, age and serrated colorectal neoplasia. First, what is the extent of DNA methylation in colorectal cancers?; Second, Does age-related hypermethylation, and namely that occurring at the loci encoding tumour suppressor genes, increase the risk of serrated colorectal neoplasia?; and if true, how can we reconcile this with the existence of early onset serrated colorectal cancer? In the first chapter of this thesis, I examine the DNA methylation and transcriptional architecture of 216 colorectal cancer samples collected consecutively at the Royal Brisbane and Women’s hospital.
    [Show full text]