Non-Coding Genetic Analysis Implicates Interleukin 18 Receptor Accessory Protein 3'UTR in Amyotrophic Lateral Sclerosis

Total Page:16

File Type:pdf, Size:1020Kb

Non-Coding Genetic Analysis Implicates Interleukin 18 Receptor Accessory Protein 3'UTR in Amyotrophic Lateral Sclerosis bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Non-Coding Genetic Analysis Implicates Interleukin 18 Receptor Accessory Protein 3′UTR in Amyotrophic Lateral Sclerosis Authors: Chen Eitan1, Elad Barkan2, Tsviya Olender1, Kristel R. van Eijk3, Matthieu Moisse4,5, Sali M. K. Farhan6,7, Aviad Siany1, Shu-Ting Hung8-10, Nancy Yacovzada1,2, Johnathan Cooper-Knock11, Kevin P. Kenna3, Rick A. A. van der Spek3, William Sproviero12, Ahmad Al Khleifat12, Alfredo Iacoangeli12, Aleksey Shatunov12, Ashley R. Jones12, Elik Chapnik1, Daphna Rothschild2,13,14, Omer Weissbrod2, Sebastian Werneburg15, Dorothy P. Schafer15, Robert H. Brown Jr16, Pamela J. Shaw11, Philip Van Damme4,5,17, Leonard H. van den Berg3, Hemali P. Phatnani18, Eran Segal2, Justin K. Ichida8-10, Ammar Al-Chalabi12,19, Jan H. Veldink3, Project MinE ALS Sequencing Consortium20, NYGC ALS Consortium20 and Eran Hornstein1* Affiliations: 1Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel. 2Department of Computer Science And Applied Math, Weizmann Institute of Science, Rehovot 7610001, Israel. 3Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CG, The Netherlands. 4KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, B-3000 Leuven, Belgium. 5VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium. 6Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. 7Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA. 8Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA. 9Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA. 10Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA. 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 11Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK. 12King's College London, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, United Kingdom. 13Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA 14Department of Genetics, Stanford University, Stanford, CA 94305, USA 15Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA. 16Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA. 17University Hospitals Leuven, Department of Neurology, Leuven, Belgium. 18Center for Genomics of Neurodegenerative Disease, New York Genome Center. 19King's College Hospital, Denmark Hill, London, SE5 9RS, United Kingdom. 20A list of Consortiums PIs and affiliations appears in the Supplementary Information. *Corresponding author. Tel: +972 89346215; Fax: +972 89342108; E-mail: [email protected] 2 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract: The non-coding genome is substantially larger than the protein-coding genome, but the lack of appropriate methodologies for identifying functional variants limits genetic association studies. Here, we developed analytical tools to identify rare variants in pre-miRNAs, miRNA recognition elements in 3′UTRs, and miRNA-target networks. Region-based burden analysis of >23,000 variants in 6,139 amyotrophic lateral sclerosis (ALS) whole-genomes and 70,403 non-ALS controls identified Interleukin-18 Receptor Accessory Protein (IL18RAP) 3′UTR variants significantly enriched in non-ALS genomes, replicate in an independent cohort and associate with a five-fold reduced risk of developing ALS. IL18RAP 3′UTR variants modify NF-κB signaling, provide survival advantage for cultured ALS motor neurons and ALS patients, and reveal direct genetic evidence and therapeutic targets for neuro-inflammation. This systematic analysis of the non-coding genome and specifically miRNA-networks will increase the power of genetic association studies and uncover mechanisms of neurodegeneration. 3 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. One Sentence Summary: Non-coding genetics demonstrate rare variants in IL-18 receptor 3’UTR that modifies ALS risk and progression. 4 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 [Main Text:] 2 Introduction 3 Genomic sequencing technologies facilitate identification of variants in open reading frames (ORFs). 4 Although allelic variants in non-coding regions are expected to be numerous 1, 2 they are largely 5 overlooked because current analytical approaches do not adequately prioritize variants that are likely to 6 be functional, over neutral background variation. 7 Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative syndrome, primarily affecting the human 8 motor neuron system with a strong genetic predisposing component 3, 4. Thus, mutations in approximately 9 25 protein-coding genes have been associated with ALS 3, 5, 6 and a hexanucleotide repeat expansion in an 10 intronic sequence of the C9orf72 gene is the most common genetic cause of ALS 7-9. However, non-coding 11 nucleotide variants in ALS have yet to be systematically explored. 12 MicroRNAs (miRNAs) are endogenous posttranscriptional repressors that silence mRNA expression 13 through sequence complementarity. miRNA dysregulation has been implicated in ALS pathogenesis, and 14 ALS-associated RNA-binding proteins, TARDBP/TDP-43 and FUS, regulate miRNA biogenesis 10-21. 15 miRNA primarily act on 3′ untranslated regions (3′UTRs) 22, which are non-coding parts of messenger 16 RNAs (mRNAs) and often regulate degradation and translation 23. 17 Burden analysis is a genetics approach that is based on the rationale that different rare variants in the same 18 gene may have a cumulative contribution 24. Therefore, burden analysis allows the identification of genes 19 containing an excess of rare and presumably functional variation in cases relative to controls. Although 20 de novo mutations in non-coding regions were recently shown in family-based autism studies 25, variants 21 in non-coding regions are not routinely included in rare-variant burden association studies. The application 22 of burden analysis to non-coding regulatory variation is constrained by the availability of whole genome 23 sequencing (WGS) data, and the ability to recognize functional variants in non-coding regulatory regions, 24 which is currently far less effective than for protein-coding genes. 25 To effectively apply burden analyses to regulatory variation, appropriate ways to determine what are the 26 relevant (qualifying) variants should be determined. In the case of miRNAs and miRNA recognition 27 elements such a framework can be achieved with relatively high confidence because of miRNA high 5 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 28 conservation and wealth of mechanistic insight about sequence impact on function. Therefore, miRNA 29 regulatory networks make an excellent gene set to explore. 30 Here, we developed tools that identify, or call, qualifying variants in miRNAs and 3′UTR of mRNAs, and 31 performed collapsed genetic analysis 26, to test if these regulatory RNAs are associated with ALS. We 32 discovered an enrichment of rare variants in the IL18RAP 3′UTR, implicating the IL-18 pathway in ALS. 33 Non-coding variants analysis in miRNA networks may impact research of human traits, increase the power 34 of rare-variant association methods and encourage systematic exploration of non-coding regions, to 35 uncover genetic mechanisms of disease. 6 bioRxiv preprint doi: https://doi.org/10.1101/2021.06.03.446863; this version posted June 5, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 36 Results 37 To test whether genetic variations in non-coding regulatory regions are associated with ALS, we analyzed 38 regions of interest WGS data from the Project MinE ALS sequencing consortium
Recommended publications
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [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]
  • The Interleukin-18 Receptor Is Differentially Expressed in Whole
    1 The interleukin-18 receptor is differentially expressed in the blood during sepsis. 2 3 Shahan Mamoor, MS1 4 [email protected] East Islip, NY, USA 5 6 We probed published and public microarray datasets1,2 to discover the most significant gene expression changes in the blood of patients with sepsis. We found significant induction 7 expression of IL18RAP and IL18R1, genes encoding subunits of the interleukin-18 receptor, in 8 whole blood from patients with sepsis. 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 Keywords: sepsis, septic shock, interleukin-18, IL18R1, IL18RAP, systems biology of septic 26 shock. 27 28 PAGE 1 OF 13 1 Septic shock is a leading cause of mortality in the United States and worldwide3. We 2 used published and public microarray datasets1,2 to identify differentially expressed genes in the 3 4 blood of patients with sepsis. We identified IL18R1 and IL18RAP as among the genes most 5 differentially expressed in blood in the septic state. 6 7 Methods 8 9 We utilized microarray datasets GSE1001591 and GSE264402 for this differential gene 10 11 expression analysis of blood cells during sepsis. GSE100509 was generated with whole blood 12 using Illumina HumanWG-6 v3.0 expression beadchip technology with n=12 whole blood from 13 control subjects and n=33 whole blood from sepsis patients. GSE26440 was generated using 14 15 Affymetrix Human Genome U133 Plus 2.0 Array technology with n=32 control subjects and 16 n=98 sepsis patients. The Benjamini and Hochberg method of p-value adjustment was used for 17 ranking of differential expression but raw p-values were used for assessment of statistical 18 19 significance of global differential expression.
    [Show full text]
  • Conserved Transcriptomic Profile Between Mouse and Human Colitis
    ARTICLE https://doi.org/10.1038/s41467-019-10769-x OPEN Conserved transcriptomic profile between mouse and human colitis allows unsupervised patient stratification Paulo Czarnewski1, Sara M. Parigi1, Chiara Sorini 1, Oscar E. Diaz 1, Srustidhar Das1, Nicola Gagliani1,2,3 & Eduardo J. Villablanca 1,3 1234567890():,; Clinical manifestations and response to therapies in ulcerative colitis (UC) are hetero- geneous, yet patient classification criteria for tailored therapies are currently lacking. Here, we present an unsupervised molecular classification of UC patients, concordant with response to therapy in independent retrospective cohorts. We show that classical clustering of UC patient tissue transcriptomic data sets does not identify clinically relevant profiles, likely due to associated covariates. To overcome this, we compare cross-sectional human data sets with a newly generated longitudinal transcriptome profile of murine DSS-induced colitis. We show that the majority of colitis risk-associated gene expression peaks during the inflammatory rather than the recovery phase. Moreover, we achieve UC patient clustering into two distinct transcriptomic profiles, differing in neutrophil-related gene activation. Notably, 87% of patients in UC1 cluster are unresponsive to two most widely used biological therapies. These results demonstrate that cross-species comparison enables stratification of patients undistinguishable by other molecular approaches. 1 Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, 17176 Stockholm, Sweden. 2 Department of Medicine and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany. 3These authors jointly supervised this work: Nicola Gagliani, Eduardo J. Villablanca. Correspondence and requests for materials should be addressed to E.J.V.
    [Show full text]
  • Nº Ref Uniprot Proteína Péptidos Identificados Por MS/MS 1 P01024
    Document downloaded from http://www.elsevier.es, day 26/09/2021. This copy is for personal use. Any transmission of this document by any media or format is strictly prohibited. Nº Ref Uniprot Proteína Péptidos identificados 1 P01024 CO3_HUMAN Complement C3 OS=Homo sapiens GN=C3 PE=1 SV=2 por 162MS/MS 2 P02751 FINC_HUMAN Fibronectin OS=Homo sapiens GN=FN1 PE=1 SV=4 131 3 P01023 A2MG_HUMAN Alpha-2-macroglobulin OS=Homo sapiens GN=A2M PE=1 SV=3 128 4 P0C0L4 CO4A_HUMAN Complement C4-A OS=Homo sapiens GN=C4A PE=1 SV=1 95 5 P04275 VWF_HUMAN von Willebrand factor OS=Homo sapiens GN=VWF PE=1 SV=4 81 6 P02675 FIBB_HUMAN Fibrinogen beta chain OS=Homo sapiens GN=FGB PE=1 SV=2 78 7 P01031 CO5_HUMAN Complement C5 OS=Homo sapiens GN=C5 PE=1 SV=4 66 8 P02768 ALBU_HUMAN Serum albumin OS=Homo sapiens GN=ALB PE=1 SV=2 66 9 P00450 CERU_HUMAN Ceruloplasmin OS=Homo sapiens GN=CP PE=1 SV=1 64 10 P02671 FIBA_HUMAN Fibrinogen alpha chain OS=Homo sapiens GN=FGA PE=1 SV=2 58 11 P08603 CFAH_HUMAN Complement factor H OS=Homo sapiens GN=CFH PE=1 SV=4 56 12 P02787 TRFE_HUMAN Serotransferrin OS=Homo sapiens GN=TF PE=1 SV=3 54 13 P00747 PLMN_HUMAN Plasminogen OS=Homo sapiens GN=PLG PE=1 SV=2 48 14 P02679 FIBG_HUMAN Fibrinogen gamma chain OS=Homo sapiens GN=FGG PE=1 SV=3 47 15 P01871 IGHM_HUMAN Ig mu chain C region OS=Homo sapiens GN=IGHM PE=1 SV=3 41 16 P04003 C4BPA_HUMAN C4b-binding protein alpha chain OS=Homo sapiens GN=C4BPA PE=1 SV=2 37 17 Q9Y6R7 FCGBP_HUMAN IgGFc-binding protein OS=Homo sapiens GN=FCGBP PE=1 SV=3 30 18 O43866 CD5L_HUMAN CD5 antigen-like OS=Homo
    [Show full text]
  • IL-17RA-Signaling Modulates CD8+ T Cell Survival and Exhaustion During 2 Trypanosoma Cruzi Infection
    bioRxiv preprint doi: https://doi.org/10.1101/314336; this version posted May 11, 2018. 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-ND 4.0 International license. 1 IL-17RA-signaling modulates CD8+ T cell survival and exhaustion during 2 Trypanosoma cruzi infection 3 Jimena Tosello Boari1,2, Cintia L. Araujo Furlan1,2, Facundo Fiocca Vernengo1,2, Constanza 4 Rodriguez1,2, María C. Ramello1,2, María C. Amezcua Vesely1,2, Melisa Gorosito Serrán1,2, 5 Nicolás G. Nuñez3,4, Wilfrid Richer3,4, Eliane Piaggio3,4, Carolina L. Montes1,2, Adriana 6 Gruppi1,2, Eva V. Acosta Rodríguez1,2*. 7 1 Departamento de Bioquímica Clínica. Facultad de Ciencias Químicas, Universidad 8 Nacional de Córdoba, Córdoba, X5000HUA. Argentina. 9 2 Centro de Investigaciones en Bioquímica Clínica e Inmunología. CONICET. Córdoba, 10 X5000HUA. Argentina. 11 3 SiRIC TransImm «Translational Immunotherapy Team», Translational Research 12 Department, Research Center, PSL Research University, INSERM U932, Institut Curie, 13 Paris, 75005. France. 14 4 Centre d’Investigation Clinique Biothérapie CICBT 1428, Institut Curie, Paris, 75005. 15 France. 16 Running title: IL-17RA signaling regulates CD8+ T cell responses to T. cruzi 17 *Corresponding autor: 18 Eva V. Acosta Rodríguez 19 [email protected] 20 1 bioRxiv preprint doi: https://doi.org/10.1101/314336; this version posted May 11, 2018. 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.
    [Show full text]
  • Single Cell Transcriptomics of Regulatory T Cells Reveals Trajectories of Tissue Adaptation ​ ​ ​ ​ ​ ​
    bioRxiv preprint doi: https://doi.org/10.1101/217489; this version posted November 22, 2017. 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 4.0 International license. Single cell transcriptomics of regulatory T cells reveals trajectories of tissue adaptation ​ ​ ​ ​ ​ ​ Authors: 1,2,a 1,a 3,4 7 Ricardo J Miragaia ,​ Tomás Gomes ,​ Agnieszka Chomka ,​ Laura Jardine ,​ Angela ​ ​ ​ ​ 8 3,4,5 1,9 1 3,4,6 Riedel ,​ Ahmed N. Hegazy ,​ Ida Lindeman ,​ Guy Emerton ,​ Thomas Krausgruber ,​ ​ ​ ​ ​ ​ 8 7 3,4 1,10,11,* Jacqueline Shields ,​ Muzlifah Haniffa ,​ Fiona Powrie ,​ Sarah A. Teichmann ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ Affiliations: 1 Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ 2 Centre of Biological Engineering, University of Minho, Braga, Portugal ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ 3 Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK ​ ​ ​ ​ ​ ​ ​ ​ 4 Translational Gastroenterology Unit, Experimental Medicine Division Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK ​ ​ ​ ​ ​ ​ ​ ​ ​ ​ 5 Current affiliation: Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health,
    [Show full text]
  • The Multiple Sclerosis Susceptibility Genes TAGAP and IL2RA Are Regulated by Vitamin D in CD4+ T Cells
    Genes and Immunity (2016) 17, 118–127 OPEN © 2016 Macmillan Publishers Limited All rights reserved 1466-4879/16 www.nature.com/gene ORIGINAL ARTICLE The multiple sclerosis susceptibility genes TAGAP and IL2RA are regulated by vitamin D in CD4+ T cells T Berge1, IS Leikfoss1,2,7, IS Brorson1,2,7, SD Bos1,2, CM Page1,2, MW Gustavsen1,2, A Bjølgerud1,2, T Holmøy2,3, EG Celius1, J Damoiseaux4, J Smolders5, HF Harbo1,2 and A Spurkland6 Multiple sclerosis (MS) is an inflammatory, demyelinating disorder of the central nervous system that develops in genetically susceptible individuals. The majority of the MS-associated gene variants are located in genetic regions with importance for T-cell differentiation. Vitamin D is a potent immunomodulator, and vitamin D deficiency has been suggested to be associated with increased MS disease susceptibility and activity. In CD4+ T cells, we have analyzed in vitro vitamin D responsiveness of genes that contain an MS-associated single-nucleotide polymorphism (SNP) and with one or more vitamin D response elements in their regulatory regions. We identify IL2RA and TAGAP as novel vitamin D target genes. The vitamin D response is observed in samples from both MS patients and controls, and is not dependent on the genotype of MS-associated SNPs in the respective genes. Genes and Immunity (2016) 17, 118–127; doi:10.1038/gene.2015.61; published online 14 January 2016 INTRODUCTION located in non-coding regions of the genome.6,8 Interestingly, the Multiple sclerosis (MS) is an inflammatory, demyelinating disorder MS-associated SNPs are enriched in DNase I hypersensitive sites, of the central nervous system leading to various degrees of indicating a functional role in regulation of gene transcription.9 physical and cognitive disability.
    [Show full text]
  • Expansions of Adaptive-Like NK Cells with a Tissue-Resident Phenotype in Human Lung and Blood
    Expansions of adaptive-like NK cells with a tissue-resident phenotype in human lung and blood Demi Brownliea,1, Marlena Scharenberga,1, Jeff E. Moldb, Joanna Hårdb, Eliisa Kekäläinenc,d,e, Marcus Buggerta, Son Nguyenf,g, Jennifer N. Wilsona, Mamdoh Al-Amerih, Hans-Gustaf Ljunggrena, Nicole Marquardta,2,3, and Jakob Michaëlssona,2 aCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14152 Stockholm, Sweden; bDepartment of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden; cTranslational Immunology Research Program, University of Helsinki, 00014 Helsinki, Finland; dDepartment of Bacteriology and Immunology, University of Helsinki, 00014 Helsinki, Finland; eHelsinki University Central Hospital Laboratory, Division of Clinical Microbiology, Helsinki University Hospital, 00290 Helsinki, Finland; fDepartment of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; gInstitute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and hThoracic Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, 171 76 Stockholm, Sweden Edited by Marco Colonna, Washington University in St. Louis School of Medicine, St. Louis, MO, and approved January 27, 2021 (received for review August 18, 2020) Human adaptive-like “memory” CD56dimCD16+ natural killer (NK) We and others recently identified a subset of tissue-resident − cells in peripheral blood from cytomegalovirus-seropositive indi- CD49a+CD56brightCD16 NK cells in the human lung (14, 15). viduals have been extensively investigated in recent years and are The human lung is a frequent site of infection with viruses such currently explored as a treatment strategy for hematological can- as influenza virus and HCMV, as well as a reservoir for latent cers.
    [Show full text]
  • Mouse Gpatch8 Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Gpatch8 Knockout Project (CRISPR/Cas9) Objective: To create a Gpatch8 knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Gpatch8 gene (NCBI Reference Sequence: NM_001159492 ; Ensembl: ENSMUSG00000034621 ) is located on Mouse chromosome 11. 8 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 8 (Transcript: ENSMUST00000143842). Exon 4~6 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 4 starts from about 4.3% of the coding region. Exon 4~6 covers 6.62% of the coding region. The size of effective KO region: ~7409 bp. The KO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 4 5 6 8 Legends Exon of mouse Gpatch8 Knockout region Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section upstream of Exon 4 is aligned with itself to determine if there are tandem repeats. No significant tandem repeat is found in the dot plot matrix. So this region is suitable for PCR screening or sequencing analysis. Overview of the Dot Plot (down) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section downstream of Exon 6 is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Primary Chicken and Duck Endothelial Cells Display a Differential Response to Infection with Highly Pathogenic Avian Influenza Virus
    G C A T T A C G G C A T genes Article Primary Chicken and Duck Endothelial Cells Display a Differential Response to Infection with Highly Pathogenic Avian Influenza Virus Zhen Wei Marcus Tong 1,†, Anjana C. Karawita 1,2,† , Colin Kern 3, Huaijun Zhou 3 , Jane E. Sinclair 1 , Limin Yan 1, Keng Yih Chew 1, Sue Lowther 2, Lee Trinidad 2, Arjun Challagulla 2 , Karel A. Schat 4 , Michelle L. Baker 2 and Kirsty R. Short 1,5,* 1 School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia; [email protected] (Z.W.M.T.); [email protected] (A.C.K.); [email protected] (J.E.S.); [email protected] (L.Y.); [email protected] (K.Y.C.) 2 CSIRO, Australian Centre for Disease Preparedness, Health, and Biosecurity Business Unit, Geelong 3219, Australia; [email protected] (S.L.); [email protected] (L.T.); [email protected] (A.C.); [email protected] (M.L.B.) 3 Department of Animal Science, University of California, Davis, CA 95616, USA; [email protected] (C.K.); [email protected] (H.Z.) 4 Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA; [email protected] 5 Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane 4072, Australia * Correspondence: [email protected] † These authors contributed equally to this work. Citation: Tong, Z.W.M.; Karawita, A.C.; Kern, C.; Zhou, H.; Sinclair, J.E.; Abstract: Highly pathogenic avian influenza viruses (HPAIVs) in gallinaceous poultry are associated Yan, L.; Chew, K.Y.; Lowther, S.; Trinidad, L.; Challagulla, A.; et al.
    [Show full text]
  • Differentiation During Th1 and Th2 Il18r1 Remodeling of Transcription
    Transcription Factor-Dependent Chromatin Remodeling of Il18r1 during Th1 and Th2 Differentiation This information is current as Qing Yu, Hua-Chen Chang, Ayele-Nati N. Ahyi and Mark of October 2, 2021. H. Kaplan J Immunol 2008; 181:3346-3352; ; doi: 10.4049/jimmunol.181.5.3346 http://www.jimmunol.org/content/181/5/3346 Downloaded from References This article cites 26 articles, 13 of which you can access for free at: http://www.jimmunol.org/content/181/5/3346.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on October 2, 2021 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 © 2008 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Transcription Factor-Dependent Chromatin Remodeling of Il18r1 during Th1 and Th2 Differentiation1 Qing Yu, Hua-Chen Chang, Ayele-Nati N. Ahyi, and Mark H. Kaplan2 The IL-18R␣-chain is expressed on Th1 but not Th2 cells. We have recently shown that Stat4 is an important component of programming the Il18r1 locus (encoding IL-18R␣) for maximal expression in Th1 cells.
    [Show full text]