Triphosphate RNA Regulates RIG-I Sensitivity

Total Page:16

File Type:pdf, Size:1020Kb

Triphosphate RNA Regulates RIG-I Sensitivity Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press DUSP11-mediated control of 5′-triphosphate RNA regulates RIG-I sensitivity Joon H. Choi, James M. Burke,1 Kayla H. Szymanik, Upasana Nepal, Anna Battenhouse, Justin T. Lau, Aaron Stark, Victor Lam,2 and Christopher S. Sullivan Department of Molecular Biosciences, LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin Texas 78712, USA Deciphering the mechanisms that regulate the sensitivity of pathogen recognition receptors is imperative to un- derstanding infection and inflammation. Here we demonstrate that the RNA triphosphatase dual-specificity phos- phatase 11 (DUSP11) acts on both host and virus-derived 5′-triphosphate RNAs rendering them less active in inducing a RIG-I-mediated immune response. Reducing DUSP11 levels alters host triphosphate RNA packaged in extracellular vesicles and induces enhanced RIG-I activation in cells exposed to extracellular vesicles. Virus in- fection of cells lacking DUSP11 results in a higher proportion of triphosphorylated viral transcripts and attenuated virus replication, which is rescued by reducing RIG-I expression. Consistent with the activity of DUSP11 in the cellular RIG-I response, mice lacking DUSP11 display lower viral loads, greater sensitivity to triphosphorylated RNA, and a signature of enhanced interferon activity in select tissues. Our results reveal the importance of con- trolling 5′-triphosphate RNA levels to prevent aberrant RIG-I signaling and demonstrate DUSP11 as a key effector of this mechanism. [Keywords: DUSP11; RIG-I signaling; inflammation; innate immunity; noncoding RNA; tumor–stromal interaction; virus infection] Supplemental material is available for this article. Received June 18, 2020; revised version accepted September 28, 2020. Life is a great balancing act with overlapping and unique lar, many host transcripts generated by RNA polymerase molecular controls required to maintain homeostasis III (RNAP III) lack a 5′ capping mechanism (Dieci et al. (Kotas and Medzhitov 2015; tenOever 2016). Host detec- 2013). A series of recent studies indicate that some of tion of pathogens must be properly tuned to promote rapid these endogenous RNAP III transcripts can function as and effective responses when an actual pathogen is detect- damage-associated molecular patterns (DAMPs), which ed, but must not be so sensitive as to trigger an inflamma- are recognized by RIG-I (Boelens et al. 2014; Nabet et al. tory response when no pathogen is present (Medzhitov 2017; Chiang et al. 2018; Zhao et al. 2018). To avoid auto- and Janeway 2002; Roers et al. 2016). RIG-I is a pattern rec- inflammatory pathology, the RIG-I response must there- ognition receptor (PRR) that triggers the antiviral cyto- fore be carefully regulated. Indeed, several upstream and kine type I interferon (IFN) response (Yoneyama et al. downstream protein modulators of RIG-I signaling have 2004; Kato et al. 2005; Gack et al. 2007). Major patho- already been identified (Chiang et al. 2014). However, gen-associated molecular patterns (PAMPs) that activate host mechanisms that directly control the 5′-triphosphate RIG-I are structured or double-stranded RNAs that pos- proinflammatory capacity of PAMP or DAMP transcripts sess a 5′-triphosphate moiety, a hallmark of viral RNA-de- are not well understood. pendent RNA polymerase (RdRP) transcripts that are DUSP11 is an RNA triphosphatase that we and others expressed during the course of infection (Hornung et al. have previously demonstrated to be active on the 5′ ends 2006; Pichlmair et al. 2006). However, host transcripts, of structured host and viral transcripts (Deshpande et al. at least initially, also possess a 5′-triphosphate. In particu- 1999; Burke et al. 2016; Burke and Sullivan 2017; Kincaid et al. 2018; Vabret et al. 2019). The 5′-triphosphates of hep- atitis C virus (HCV) RNAs are converted to monophos- Present addresses: 1Department of Biochemistry, University of Colorado, Boulder, CO 80309, USA; 2Department of Pharmaceutical Chemistry, phates rendering them susceptible to exonuclease XRN- University of California at San Francisco, San Francisco, CA 94158, USA. mediated attack (Amador-Cañizares et al. 2018; Kincaid Corresponding author: [email protected] Article published online ahead of print. Article and publication date are online at http://www.genesdev.org/cgi/doi/10.1101/gad.340604.120. Free- © 2020 Choi et al. This article, published in Genes & Development,is ly available online through the Genes & Development Open Access available under a Creative Commons License (Attribution 4.0 Internation- option. al), as described at http://creativecommons.org/licenses/by/4.0/. GENES & DEVELOPMENT 34:1–16 Published by Cold Spring Harbor Laboratory Press; ISSN 0890-9369/20; www.genesdev.org 1 Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press Choi et al. et al. 2018). A different outcome arises when triphos- (Supplemental Fig. S1A,B). Combined with our previous phorylated RNAP III viral precursor microRNAs (miR- work (Burke et al. 2016), these data do not support a back- NAs) are processed by DUSP11. These viral miRNAs ground of strong antiviral response in cultured cells with require DUSP11 for full silencing activity via stable load- reduced DUSP11, but are consistent with a subtle en- ing of 5′-monophosphate miRNAs into the RNA-induced hanced predisposition of these cells to undergo RIG-I silencing complex (RISC) (Burke et al. 2016). Additionally, signaling. the 5′-triphosphate status of several host RNAP III non- We next studied the effects of DUSP11 on exogenously coding RNA transcripts including vault RNAs (vtRNAs), introduced in vitro transcribed RNAs (Fig. 1A). We used Alu SINE RNAs, Y RNAs, and RMRP RNA are also mod- the 5′ untranslated region (UTR) of the HCV RNA, as its ulated by DUSP11 (Burke et al. 2016; Zhao et al. 2018; expression is immunostimulatory (Saito et al. 2007). Addi- Vabret et al. 2019). The ability of DUSP11 to convert the tionally, we and others had previously demonstrated tri- 5′ end of diverse structured and partially double-stranded phosphatase activity of DUSP11 on HCV RNAs during host and viral transcripts into monophosphates opens up infection (Amador-Cañizares et al. 2018; Kincaid et al. the possibility that DUSP11 can alter the immunostimu- 2018). Consistent with RIG-I induction, cationic-lipid-me- latory activity of endogenous and exogenous triphos- diated transfection of HCV 5′-triphosphate RNA (referred phorylated transcripts (Burke and Sullivan 2017). to here as “5′-ppp-RNA”) into the A549 adenocarcinoma Although DUSP11 can convert structured triphosphate human lung epithelial cell line resulted in the induction RNAs into monophosphates (Burke and Sullivan 2017), of both IFNB1 cytokine and ISG15 mRNA transcripts and DUSP11 has been implicated as being reduced during (Fig. 1B). Under conditions where DUSP11 protein levels infection by some viruses (Zhao et al. 2018; Vabret et al. are abolished due to CRISPR-mediated knockout, trans- 2019), the physiological role of DUSP11 in the innate im- fection of 5′-ppp-RNA results in enhanced induction of mune response is poorly understood. Here we test the hy- IFNB1 and ISG15 mRNA (Fig. 1B). Consistent with RIG- pothesis that DUSP11 is a modulator of the RNA-induced I being an interferon-stimulated gene (Matsumiya and innate immune response. Our data demonstrate that tun- Stafforini 2010), RIG-I protein levels were also further in- ing the 5′-triphosphate levels of both endogenous host and duced in cells with reduced DUSP11 (Supplemental Fig. exogenous viral transcripts is a regulator of RIG-I signal- S1C). To determine whether this regulation was depen- ing and that DUSP11 is a key mediator of 5′-triphosphate dent on the catalytic triphosphatase activity of DUSP11, balance. we used A549 DUSP11 knockout cells stably reconstitut- ed with a control empty vector (vector), wild-type DUSP11 (D11), or the catalytically inactive mutant DUSP11 Results (Cys152Ser, D11-CM) (Fig. 1A; Yuan et al. 1998; Desh- pande et al. 1999; Burke et al. 2016). Upon transfection of DUSP11 modulates RIG-I signaling sensitivity ′ 5 -ppp-RNA, expression of wild-type DUSP11, but not to liposomal 5′-triphosphate RNAs the empty vector or catalytic mutant DUSP11, reduced Among the host defense machineries that sense invading ISG15 mRNA induction (Fig. 1C). Consistent with pathogens, RIG-I is the key PRR that recognizes the 5′-tri- DUSP11 directly reducing the visibility of RNAs to RIG- phosphate or diphosphate moiety of double-stranded I, pretreatment of 5′-ppp-RNA with calf intestinal phos- RNA transcripts and in turn activates the antiviral im- phatase (CIP) or the purified catalytic core of DUSP11 mune defense signaling pathway (Hornung et al. 2006). repeatably resulted in reduced induction of IFNB1 and Previous work demonstrated that DUSP11 sequentially ISG15 transcripts (Fig. 1D). To test the requirement of removes the γ and β phosphates of triphosphorylated RIG-I for ISG induction, we used siRNA-mediated knock- structured host and viral transcripts leaving 5′-monophos- down of RIG-I. Cells with reduced RIG-I displayed signifi- phates (Deshpande et al. 1999; Burke et al. 2016; Kincaid cantly reduced induction of ISG15 transcripts (Fig. 1E). et al. 2018). This led us to hypothesize that DUSP11 can Together, these results indicate that DUSP11 modulates modulate the immunogenicity of triphosphorylated tran- RIG-I signaling sensitivity via its catalytic phosphatase ac- scripts (Burke and Sullivan 2017) and alter the sensitivity tivity on 5′-triphosphate PAMP RNA. of these transcripts to detection by RIG-I. To begin to test this hypothesis, we first looked for evidence of enhanced DUSP11 alleviates the interferon response mediated interferon-mediated gene expression. Although our previ- by tumor–fibroblast cell interaction ous RNA-seq experiments (Burke et al. 2016) did not dis- play a strong signal of interferon response in HEK293T We next wanted to determine whether DUSP11 alters cel- or A549 cells lacking DUSP11, we re-examined this possi- lular susceptibility to endogenous RIG-I DAMP RNAs.
Recommended publications
  • Supporting Information
    Supporting Information Celhar et al. 10.1073/pnas.1507052112 SI Materials and Methods using a Nanodrop spectrophotometer (Thermo Fisher Scien- Proteinuria. Proteinuria was assessed using Albustix (Bayer). Al- tific). A TaqMan RNA-to-CT 1-Step Kit (Applied Biosystems) bumin levels in urine were assayed using an Albumin Mouse was used to perform the reverse transcription and quantitative ELISA Kit (Abcam) according to the manufacturer’s instructions; PCR reactions according to the manufacturer’s instructions samples were assayed at a dilution of 1:400. Samples were nor- using TaqMan gene expression assays (Applied Biosystems) to malized for creatinine using a Creatinine (urinary) Colorimetric either Tlr7 (Mm00446590) or the B2m housekeeping gene Assay Kit (Cayman Chemical) according to the manufacturer’s (Mm00437762). Real-time PCR was performed on the 7900H instructions; initial sample dilution of 1:10. fast real-time PCR system and analyzed using SDS 2.4 (Applied Biosystems). Relative mRNA expression was calculated using the Cell Sorting, RNA Isolation, and RT-PCR. Splenic B cells were comparative C method. + − + + t sorted as live CD45 Gr1 B220 CD19 , splenic T cells as live + − + + CD45 Gr1 CD3 CD5 and peritoneal macrophages as live Imaging. Kidney sections from OCT embedded tissue were fixed + − CD45 Gr1 CD11bhiF4/80hi. Sorted cells were centrifuged, re- with 4% paraformaldehyde before permeabilization with acetone suspended in TRIzol (Life Technologies) and stored at −80°. RNA and stained with Phalloidin (AF647) and anti-CD3d (unlabeled was extracted by TRIzol/chloroform and purified with the Qiagen Ab followed by secondary staining with donkey anti-goat Dylight RNeasy Mini purification kit according to the manufacturer’s 550).
    [Show full text]
  • Molecular Mechanisms Involved Involved in the Interaction Effects of HCV and Ethanol on Liver Cirrhosis
    Virginia Commonwealth University VCU Scholars Compass Theses and Dissertations Graduate School 2010 Molecular Mechanisms Involved Involved in the Interaction Effects of HCV and Ethanol on Liver Cirrhosis Ryan Fassnacht Virginia Commonwealth University Follow this and additional works at: https://scholarscompass.vcu.edu/etd Part of the Physiology Commons © The Author Downloaded from https://scholarscompass.vcu.edu/etd/2246 This Thesis is brought to you for free and open access by the Graduate School at VCU Scholars Compass. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of VCU Scholars Compass. For more information, please contact [email protected]. Ryan C. Fassnacht 2010 All Rights Reserved Molecular Mechanisms Involved in the Interaction Effects of HCV and Ethanol on Liver Cirrhosis A thesis submitted in partial fulfillment of the requirements for the degree of Master of Science at Virginia Commonwealth University. by Ryan Christopher Fassnacht, B.S. Hampden Sydney University, 2005 M.S. Virginia Commonwealth University, 2010 Director: Valeria Mas, Ph.D., Associate Professor of Surgery and Pathology Division of Transplant Department of Surgery Virginia Commonwealth University Richmond, Virginia July 9, 2010 Acknowledgement The Author wishes to thank his family and close friends for their support. He would also like to thank the members of the molecular transplant team for their help and advice. This project would not have been possible with out the help of Dr. Valeria Mas and her endearing
    [Show full text]
  • A Functional C-Terminal TRAF3-Binding Site in MAVS Participates in Positive and Negative Regulation of the IFN Antiviral Response
    Cell Research (2011) 21:895-910. © 2011 IBCB, SIBS, CAS All rights reserved 1001-0602/11 $ 32.00 npg ORIGINAL ARTICLE www.nature.com/cr A functional C-terminal TRAF3-binding site in MAVS participates in positive and negative regulation of the IFN antiviral response Suzanne Paz1, 2, Myriam Vilasco3, Steven J Werden1, Meztli Arguello1, Deshanthe Joseph-Pillai1, 2, Tiejun Zhao1, Thi Lien-Anh Nguyen1, Qiang Sun1, Eliane F Meurs3, Rongtuan Lin1, 4, John Hiscott1, 2, 4 1Terry Fox Molecular Oncology Group, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec H3T1E2, Canada; 2Department of Microbiology and Immunology, McGill University Montreal, Quebec H3A 2B4, Canada; 3Department of Virology, Unit of Hepacivirus and Innate Immunity, Pasteur Institute, Paris 75724 France; 4Department of Medicine, McGill University Montreal, Quebec H3A 2B4, Canada Recognition of viral RNA structures by the cytosolic sensor retinoic acid-inducible gene-I (RIG-I) results in the activation of signaling cascades that culminate with the generation of the type I interferon (IFN) antiviral response. Onset of antiviral and inflammatory responses to viral pathogens necessitates the regulated spatiotemporal recruitment of signaling adapters, kinases and transcriptional proteins to the mitochondrial antiviral signaling protein (MAVS). We previously demonstrated that the serine/threonine kinase IKKε is recruited to the C-terminal region of MAVS following Sendai or vesicular stomatitis virus (VSV) infection, mediated by Lys63-linked polyubiquitination of MAVS at Lys500, resulting in inhibition of downstream IFN signaling (Paz et al, Mol Cell Biol, 2009). In this study, we demonstrate that C-terminus of MAVS harbors a novel TRAF3-binding site in the aa450- 468 region of MAVS.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Human Ifnα4 / Ifna4 / Interferon Alpha-4 Protein (Fc Tag)
    Human IFNα4 / IFNa4 / Interferon alpha-4 Protein (Fc Tag) Catalog Number: 10336-H01H General Information SDS-PAGE: Gene Name Synonym: IFN-alpha4a; INFA4; MGC142200 Protein Construction: A DNA sequence encoding the mature form of human IFN-α4a (NP_066546.1) (Cys 23-Asp 189) was expressed with the N-terminal fused Fc region of human IgG1. Source: Human Expression Host: HEK293 Cells QC Testing Purity: > 97 % as determined by SDS-PAGE Bio Activity: Protein Description Measured in antiviral assays using WISH human amnion cells infected Interferon, alpha 4 (IFNA4) belongs to the alpha/beta interferon family. Two with vesicular stomatitis virus (VSV). The EC50 for this effect is 0.2-1 variants of IFNA4 (IFNA4a and IFNA4b) are known, which differ from each ng/mL. other by changes in their coding regions at nucleotide positions 220 and 410 and can be distinguished by selective restriction enzyme analysis. Endotoxin: Interferons are produced by macrophages, IFN-alpha have antiviral activities. Interferon stimulates the production of two enzymes: a protein < 1.0 EU per μg of the protein as determined by the LAL method kinase and an oligoadenylate synthetase. IFN-alpha, the first cytokine to be produced by recombinant DNA technology, has emerged as an important Stability: regulator of growth and differentiation, affecting cellular communication and Samples are stable for up to twelve months from date of receipt at -70 ℃ signal transduction pathways as well as immunological control. Originally discovered as an antiviral substance, the efficacy of IFN-alpha in malignant, Predicted N terminal: Glu viral, immunological, angiogenic, inflammatory, and fibrotic diseases suggests a spectrum of interrelated pathophysiologies.
    [Show full text]
  • Proquest Dissertations
    INFORMATION TO USERS This manuscript has been reproduced from the microfilm master. UMI films the text directly from the original or copy submitted. Thus, some thesis and dissertation copies are in typewriter face, while others may be from any type of computer printer. The quality of this reproduction is dependent upon the quality of the copy submitted. Broken or indistinct print, colored or poor quality illustrations and photographs, print bleedthrough, substandard margins, and improper alignment can adversely affect reproduction. In the unlikely event that the author did not send UMI a complete manuscript and there are missing pages, these will be noted. Also, if unauthorized copyright material had to be removed, a note will indicate the deletion. Oversize materials (e.g., maps, drawings, charts) are reproduced by sectioning the original, beginning at the upper left-hand comer and continuing from left to right in equal sections with small overlaps. Each original is also photographed in one exposure and is included in reduced form at the back of the book. Photographs included in the original manuscript have been reproduced xerographically in this copy. Higher quality 6” x 9” black and white photographic prints are available for any photographs or illustrations appearing in this copy for an additional charge. Contact UMI directly to order. UMI Bell & Howell Information and Learning 300 North Zeeb Road, Ann Arbor, Ml 48106-1346 USA 800-521-0600 NOTE TO USERS Page(s) missing in number only; text follows. Microfilmed as received. 222-229 This reproduction is the best copy available. UMI MOLECULAR GENETIC AND BIOCHEMICAL STUDIES OF THE HUMAN AND MOUSE MHC COMPLEMENT GENE CLUSTERS DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Zhenyu Yang, M.S.
    [Show full text]
  • Aneuploidy: Using Genetic Instability to Preserve a Haploid Genome?
    Health Science Campus FINAL APPROVAL OF DISSERTATION Doctor of Philosophy in Biomedical Science (Cancer Biology) Aneuploidy: Using genetic instability to preserve a haploid genome? Submitted by: Ramona Ramdath In partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biomedical Science Examination Committee Signature/Date Major Advisor: David Allison, M.D., Ph.D. Academic James Trempe, Ph.D. Advisory Committee: David Giovanucci, Ph.D. Randall Ruch, Ph.D. Ronald Mellgren, Ph.D. Senior Associate Dean College of Graduate Studies Michael S. Bisesi, Ph.D. Date of Defense: April 10, 2009 Aneuploidy: Using genetic instability to preserve a haploid genome? Ramona Ramdath University of Toledo, Health Science Campus 2009 Dedication I dedicate this dissertation to my grandfather who died of lung cancer two years ago, but who always instilled in us the value and importance of education. And to my mom and sister, both of whom have been pillars of support and stimulating conversations. To my sister, Rehanna, especially- I hope this inspires you to achieve all that you want to in life, academically and otherwise. ii Acknowledgements As we go through these academic journeys, there are so many along the way that make an impact not only on our work, but on our lives as well, and I would like to say a heartfelt thank you to all of those people: My Committee members- Dr. James Trempe, Dr. David Giovanucchi, Dr. Ronald Mellgren and Dr. Randall Ruch for their guidance, suggestions, support and confidence in me. My major advisor- Dr. David Allison, for his constructive criticism and positive reinforcement.
    [Show full text]
  • Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association Between BMI and Adult-Onset Non- Atopic
    Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association between BMI and Adult-Onset Non- Atopic Asthma Ayoung Jeong 1,2, Medea Imboden 1,2, Akram Ghantous 3, Alexei Novoloaca 3, Anne-Elie Carsin 4,5,6, Manolis Kogevinas 4,5,6, Christian Schindler 1,2, Gianfranco Lovison 7, Zdenko Herceg 3, Cyrille Cuenin 3, Roel Vermeulen 8, Deborah Jarvis 9, André F. S. Amaral 9, Florian Kronenberg 10, Paolo Vineis 11,12 and Nicole Probst-Hensch 1,2,* 1 Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; [email protected] (A.J.); [email protected] (M.I.); [email protected] (C.S.) 2 Department of Public Health, University of Basel, 4001 Basel, Switzerland 3 International Agency for Research on Cancer, 69372 Lyon, France; [email protected] (A.G.); [email protected] (A.N.); [email protected] (Z.H.); [email protected] (C.C.) 4 ISGlobal, Barcelona Institute for Global Health, 08003 Barcelona, Spain; [email protected] (A.-E.C.); [email protected] (M.K.) 5 Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain 6 CIBER Epidemiología y Salud Pública (CIBERESP), 08005 Barcelona, Spain 7 Department of Economics, Business and Statistics, University of Palermo, 90128 Palermo, Italy; [email protected] 8 Environmental Epidemiology Division, Utrecht University, Institute for Risk Assessment Sciences, 3584CM Utrecht, Netherlands; [email protected] 9 Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College, SW3 6LR London, UK; [email protected] (D.J.); [email protected] (A.F.S.A.) 10 Division of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; [email protected] 11 MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, W2 1PG London, UK; [email protected] 12 Italian Institute for Genomic Medicine (IIGM), 10126 Turin, Italy * Correspondence: [email protected]; Tel.: +41-61-284-8378 Int.
    [Show full text]
  • Table S1. Complete Gene List. Genbank Refseq and Description of Each Gene Were Provided By
    Document downloaded from http://www.elsevier.es, day 24/09/2021. This copy is for personal use. Any transmission of this document by any media or format is strictly prohibited. Table S1. Complete gene list. GenBank RefSeq and description of each gene were provided by the array supplier. Unigene GeneBank Symbol Description Gene Name/s Rn.11422 NM_033230 Akt1 V-akt murine thymoma viral oncogene homolog 1 Akt Rn.2104 NM_019288 App Amyloid beta (A4) precursor protein - Rn.23323 NM_001034933 Arsa Arylsulfatase A MGC125207 Rn.94004 NM_033443 Arsb Arylsulfatase B - Rn.6224 NM_001038495 Atg12 ATG12 autophagy related 12 Apg12l, MGC125080 Rn.101734NM_001108809 Atg16l1 ATG16 autophagy related 16-like 1 Apg16l, Wdr30 Rn.104199NM_001191560 Atg16l2 ATG16 autophagy related 16-like 2 RGD1311400 Rn.3084 NM_134394 Atg3 ATG3 autophagy related 3 Apg3l, PIG-1, Pig1 Rn.163086NM_001025711 Atg4b ATG4 autophagy related 4 homolog B Apg4b, MGC112887 Rn.23378 NM_001107948 Atg4c ATG4 autophagy related 4 homolog C - Rn.98385 NM_001014250 Atg5 ATG5 autophagy related 5 - Rn.162765NM_001012097 Atg7 ATG7 autophagy related 7 Apg7l Rn.35248 NM_001014218 Atg9a ATG9 autophagy related 9 homolog A MGC105908, RGD1310450 Rn.36696 NM_022698 Bad BCL2-associated agonist of cell death MGC72439 Rn.14598 NM_053812 Bak1 BCL2-antagonist/killer 1 MGC108627 Rn.10668 NM_017059 Bax Bcl2-associated X protein - Rn.9996 NM_016993 Bcl2 B-cell CLL/lymphoma 2 Bcl-2 Rn.10323 NM_031535 Bcl2l1 Bcl2-like 1 Bcl-xl, Bcl2l, Bclx, bcl-X Rn.2776 NM_053739 Becn1 Beclin 1, autophagy related - Rn.31142 NM_022684
    [Show full text]
  • Datasheet Blank Template
    SAN TA C RUZ BI OTEC HNOL OG Y, INC . DOM3Z (B-12): sc-393141 BACKGROUND APPLICATIONS DOM3Z (dom-3 homolog Z), also known as NG6 or DOM3L, is a 396 amino DOM3Z (B-12) is recommended for detection of DOM3Z of mouse, rat and acid ubiquitously expressed protein belonging to the DOM3Z family. The gene human origin by Western Blotting (starting dilution 1:100, dilution range encoding DOM3Z maps to human chromosome 6 in the major histocompati - 1:100-1:1000), immunoprecipitation [1-2 µg per 100-500 µg of total protein bility complex (MHC) class III region. Chromosome 6 contains 170 million base (1 ml of cell lysate)], immunofluorescence (starting dilution 1:50, dilution pairs and comprises nearly 6% of the human genome. Deletion of a portion range 1:50-1:500), immunohistochemistry (including paraffin-embedded of the q arm of chromosome 6 is associated with early onset intestinal can - sections) (starting dilution 1:50, dilution range 1:50-1:500) and solid phase cer, suggesting the presence of a cancer susceptibility locus. Additionally, ELISA (starting dilution 1:30, dilution range 1:30-1:3000). Porphyria cutanea tarda, Parkinson’s disease and Stickler syndrome are all Suitable for use as control antibody for DOM3Z siRNA (h): sc-95321, DOM3Z associated with genes that map to chromosome 6. siRNA (m): sc-143143, DOM3Z shRNA Plasmid (h): sc-95321-SH, DOM3Z shRNA Plasmid (m): sc-143143-SH, DOM3Z shRNA (h) Lentiviral Particles: REFERENCES sc-95321-V and DOM3Z shRNA (m) Lentiviral Particles: sc-143143-V. 1. Brunner, H.G., et al .
    [Show full text]
  • DNA Methylation Alterations in Blood Cells of Toddlers with Down Syndrome
    G C A T T A C G G C A T genes Article DNA Methylation Alterations in Blood Cells of Toddlers with Down Syndrome Oxana Yu. Naumova 1,2,* , Rebecca Lipschutz 2, Sergey Yu. Rychkov 1, Olga V. Zhukova 1 and Elena L. Grigorenko 2,3,4,* 1 Vavilov Institute of General Genetics RAS, 119991 Moscow, Russia; [email protected] (S.Y.R.); [email protected] (O.V.Z.) 2 Department of Psychology, University of Houston, Houston, TX 77204, USA; [email protected] 3 Department of Psychology, Saint-Petersburg State University, 199034 Saint Petersburg, Russia 4 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA * Correspondence: [email protected] or [email protected] (O.Y.N.); [email protected] (E.L.G.) Abstract: Recent research has provided evidence on genome-wide alterations in DNA methylation patterns due to trisomy 21, which have been detected in various tissues of individuals with Down syndrome (DS) across different developmental stages. Here, we report new data on the systematic genome-wide DNA methylation perturbations in blood cells of individuals with DS from a previously understudied age group—young children. We show that the study findings are highly consistent with those from the prior literature. In addition, utilizing relevant published data from two other developmental stages, neonatal and adult, we track a quasi-longitudinal trend in the DS-associated DNA methylation patterns as a systematic epigenomic destabilization with age. Citation: Naumova, O.Y.; Lipschutz, R.; Rychkov, S.Y.; Keywords: Down syndrome; infants and toddlers; trisomy 21; DNA methylation; Illumina 450K Zhukova, O.V.; Grigorenko, E.L.
    [Show full text]
  • Interferonsource.Com What's Inside New Products Research Tools Protocols & Tips Product Citations What's Your IFN-Α
    Interferon Matters A quarterly newsletter by PBL InterferonSource interferonsource.com May 2007, Issue 2 u What’s Inside What’s Your IFN-a Type? New Products Are all Type I IFNs created equal? Of Mice and Men page 4 & 5 Type I Interferons were the first cytokines to be The biological significance of the expression of discovered. They are a family of homologous cytokines so many different IFN-a subtypes has yet to be Rhesus/Cynomolgus originally identified for their antiviral activity but have determined. However, reports suggest that they show IFN-a2 Subtype also been shown to have both anti-proliferative and quantitatively distinct anti-viral, anti-proliferative, immunomodulatory activities. In humans and mice and killer cell-stimulatory activities. It is therefore Mouse IFN-a4 Subtype they are encoded by an intronless multigene family of importance for researchers to characterize and clustered on human chromosome 9 and murine study these IFN-a subtypes and their possible post- chromosome 4 respectively. translational modifications. Research Tools Also in both species there are multiple Interferon- The common models for IFN-a subtypes study are page 6 & 7 Alpha (IFN-a) genes and one Interferon-Beta (IFN-b) human and mouse. In 1998, Lawson et al showed that Human IFN-a gene. Other Type I Interferon genes described include there are in vivo differences in the antiviral activity of the Interferon-Omega (IFN-w) gene, murine limitin the mouse IFN-a subtypes.1 Using an experimental Multi-Subtype ELISA Kit gene, as well as the human/murine Interferon-Epilson, mouse model of IFN transgene expression in vivo, TM Interferon-Kappa, and Interferon-Tau (IFN-e, IFN-k, and the authors showed that mouse IFN-a1 has a greater iLite Human IFN-a Kit IFN-t).
    [Show full text]