Summary Human Genetics Mechanistic Insight Relevance To

Total Page:16

File Type:pdf, Size:1020Kb

Summary Human Genetics Mechanistic Insight Relevance To Non-Coding Genetic Analysis Implicates IL18RAP 3′UTR in ALS Chen Eitan1, Elad Barkan1, Tsviya Olender1, Aviad Siany1, Natalia Rivkin1, Hagai Marmor-Kollet1, Kristel R. van Eijk2, Matthieu Moisse3, Sali M. K. Farhan4, Shu-Ting Hung5, Yael Alon1, Nancy Yacovzada1, Johnathan Cooper-Knock6, Kevin P. Kenna2, Rick A. A. van der Spek2, William Sproviero7, Ahmad Al Khleifat7, Alfredo Iacoangeli7, Elik Chapnik1, Daphna Rothschild1, Omer Weissbrod1, Sebastian Werneburg8, Dorothy P. Schafer8, Robert H. Brown Jr8, Pamela J. Shaw7, Philip Van Damme3, Leonard H. van den Berg2, Hemali P. Phatnani9, Eran Segal1, Justin K. Ichida5, Ammar Al-Chalabi7, Jan H. Veldink2, Project MinE ALS Sequencing Consortium, NYGC ALS Consortium and Eran Hornstein1* 1Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel; 2Department of Neurology, UMC Utrecht Brain Center, Utrecht, The Netherlands; 3KU Leuven - University of Leuven, Department of Neurosciences, Leuven, Belgium; 4Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; 5Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Los Angeles, CA, USA; 6Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, UK; 7King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK; 8Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA; 9Center for Genomics of Neurodegenerative Disease, New York Genome Center. 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. Human Genetics Mechanistic Insight Relevance to ALS Region-based rare-variant association analysis identified IL18RAP 3′UTR variants Functional validation of IL18RAP Variants IL18RAP 3'UTR variant ameliorates *** disease in C9orf72 patient iMNs significantly enriched in non-ALS genomes ** ** ** and in patients with ALS 3.5 *** ** ** *** 1.5 3.0 2.5 1.0 2.0 1.5 1.0 0.5 0.5 IL18RAP/Tubulin [a.u] IL18RAP/Tubulin 0.0 0.0 Relative IL18RAP expression IL18RAP Relative sALS sALS sALS sALS Control Control Control Control IL18RAP 3'UTR IL18RAP 3'UTR variants - + variants - + IL18RAP 3’UTR protective variants significantly reduce IL18RAP expression IL18RAP 3’UTR variant attenuates IL-18 / NF-κB signaling Mutant IL18RAP 3’UTR is protective in human patients with ALS (A) Region-based rare-variant association analysis was performed on rare (MAF ≤0.01) qualifying variants in: (i) 295 candidate protein-coding genes encoding for ALS-relevant proteins or proteins associated with miRNA biogenesis/activity. Variants were included if predicted to be deleterious; (ii) variants abrogating or gaining miRNA recognition elements in 3′UTRs of the 295 genes; (iii) all known pre-miRNA genes in the human genome; and (iv) predicted networks, comprised of aggregated variants detected in a specific mature miRNA sequence and its cognate down-stream 3’UTR targets. (B) QQ plot of obtained and expected P-values for the burden of rare variants (log scale) gained by collapsed region-based association analysis of all genomic regions described in (A). Survival tracking microscopy of Hb9::RFP+ iMNs, transduced with IL18RAP lentiviruses, after neurotrophic factors withdrawal and introduction of IL-18 (A). Kaplan-Meier survival plots for control Genome-wide analysis of all known human 3’UTRs Rare IL18RAP 3′UTR variants are depleted in an (CTRL) and C9orf72 patient (C9-ALS) iMNs, transduced with IL18RAP lentiviruses (WT/V3/3CDS). Traces of iMNs from 2 donors per genotype. Log-rank test for the entire survival time course (B-D). independent ALS cohort NF-κB reporter assay in U2OS cell line transfected with wild-type (WT) IL18RAP, IL18RAP harboring IL18RAP variant is associated with delayed age of death (+6.1 years, 9/4263) (E) and age of diagnosis variants in the 3’UTR (V3, V1) or an IL18RAP dominant negative coding mutant (3CDS) vectors. (+6.2 years, 8/4216) (F), relative to the mean age of all Project MinE and NYGC ALS patients. Summary IL18RAP 3′UTR is the most significant We devised a method for identifying rare qualifying variants in non-coding genomic regions. 3’UTR associated with ALS We performed an unbiased genetic study of miRNAs and 3’UTRs using Project MinE’s large-scale WGS data. Region-based burden analysis identified rare IL18RAP 3′UTR variants significantly enriched in non-ALS genomes and associate with a five-fold reduced risk of developing ALS. Carriers have 5 times less ALS risk IL18RAP 3′UTR variants modify NF-κB signaling, provide survival advantage for cultured ALS motor neurons and ALS patients, and highlight its potential protective value..
Recommended publications
  • 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]
  • 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]
  • 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]
  • Epithelial-Derived IL-18 Regulates Th17 Cell Differentiation and Foxp3 Þ Treg Cell Function in the Intestine
    ARTICLES nature publishing group Epithelial-derived IL-18 regulates Th17 cell differentiation and Foxp3 þ Treg cell function in the intestine OJ Harrison1, N Srinivasan1, J Pott1, C Schiering2, T Krausgruber2, NE Ilott2 and KJ Maloy1 Elevated levels of interleukin-18 (IL-18) are found in many chronic inflammatory disorders, including inflammatory bowel disease (IBD), and polymorphisms in the IL18R1–IL18RAP locus are associated with IBD susceptibility. IL-18 is an IL-1 family cytokine that has been proposed to promote barrier function in the intestine, but the effects of IL-18 on intestinal CD4 þ T cells are poorly understood. Here we demonstrate that IL-18R1 expression is enhanced on both effector and regulatory CD4 þ T cells in the intestinal lamina propria, with T helper type 17 (Th17) cells exhibiting particularly high levels. We further show that, during steady state, intestinal epithelial cells constitutively secrete IL-18 that acts directly on IL-18R1-expressing CD4 þ Tcells to limit colonic Th17 cell differentiation, in part by antagonizing IL-1R1 signaling. In addition, although IL-18R1 is not required for colonic Foxp3 þ regulatory T (Treg) cell differentiation, we found that IL-18R1 signaling was critical for Foxp3 þ Treg cell–mediated control of intestinal inflammation, where it promoted the expression of key Treg effector molecules. Thus IL-18 is a key epithelial-derived cytokine that differentially regulates distinct subsets of intestinal CD4 þ Tcells during both homeostatic and inflammatory conditions, a finding with potential
    [Show full text]
  • Assessment and Diagnostic Relevance of Novel Serum Biomarkers for Early Decision of ST-Elevation Myocardial Infarction
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No. 15 Assessment and diagnostic relevance of novel serum biomarkers for early decision of ST-elevation myocardial infarction Hun-Jun Park1,*, Ji Heon Noh2,3,*, Jung Woo Eun2,3,*, Yoon-Seok Koh1, Suk Min Seo1, Won Sang Park2, Jung Young Lee2, Kiyuk Chang1, Ki Bae Seung1, Pum-Joon Kim1, Suk Woo Nam2,3,4 1Department of Cardiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea 2Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea 3 Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea 4Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea *These authors have contributed equally to this work Correspondence to: Suk Woo Nam, e-mail: [email protected] Pum-Joon Kim, e-mail: [email protected] Keywords: ST-segment-elevation myocardial infarction, peripheral blood transcriptome, molecular signature, diagnostic biomarkers Received: March 25, 2015 Accepted: May 04, 2015 Published: May 18, 2015 ABSTRACT Blood transcriptome reflects the status of diseases, and characteristic molecular signature provides a novel window on gene expression preceding acute coronary events. We aim to determine blood transcriptome-based molecular signature of acute coronary syndrome (ACS), and to identify novel serum biomarkers for early stage ST-segment-elevation myocardial infarction (STEMI). We obtained peripheral blood from the patients with ACS who visited emergency department within 4 hours after the onset of chest pain: STEMI (n = 10), Non-ST-segment-elevation MI (NSTEMI, n = 10) and unstable angina (UA, n = 11).
    [Show full text]
  • Identification of IL18RAP/IL18R1 and IL12B As Leprosy Risk Genes
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector REPORT Identification of IL18RAP/IL18R1 and IL12B as Leprosy Risk Genes Demonstrates Shared Pathogenesis between Inflammation and Infectious Diseases Hong Liu,1,2,3,4,11 Astrid Irwanto,5,6,11 Hongqing Tian,1,4,11 Xi’an Fu,2,3 Yongxiang Yu,2,3 Gongqi Yu,2,3 Huiqi Low,5 Tongsheng Chu,2,3 Yi Li,5 Benqing Shi,1,4 Mingfei Chen,2,3 Yonghu Sun,2,3 Chunying Yuan,2,3 Nan Lu,2,3 Jiabao You,2,3 Fangfang Bao,2,3 Jinghui Li,2,3 Jian Liu,2,3 Huaxu Liu,2,3 Dianchang Liu,2,3 Xiulu Yu,2,3 Lin Zhang,2,3 Qing Yang,2,3 Na Wang,2,3 Guiye Niu,2,3 Shanshan Ma,2,3 Yan Zhou,2,3 Chuan Wang,2,3 Shumin Chen,2,3 Xuejun Zhang,7,8 Jianjun Liu,2,5,9,12 and Furen Zhang1,2,3,4,10,12,* Of eight leprosy susceptibility loci identified by genome-wide association studies, five have been implicated in Crohn disease, suggesting a common genetic fingerprint between leprosy and inflammatory bowel disease (IBD). Here, we conducted a multiple-stage genetic asso- ciation study of 133 IBD susceptibility loci in multiple leprosy samples (totaling 4,971 leprosy cases and 5,503 controls) from a Chinese À population and discovered two associations at rs2058660 on 2q12.1 (p ¼ 4.57 3 10 19; odds ratio [OR] ¼ 1.30) and rs6871626 on 5q33.3 À (p ¼ 3.95 3 10 18;OR¼ 0.75), implicating IL18RAP/IL18R1 and IL12B as susceptibility genes for leprosy.
    [Show full text]
  • A Systems Immunology Approach Identifies the Collective Impact of Five Mirs in Th2 Inflammation Supplementary Information
    A systems immunology approach identifies the collective impact of five miRs in Th2 inflammation Supplementary information Ayşe Kılıç, Marc Santolini, Taiji Nakano, Matthias Schiller, Mizue Teranishi,Pascal Gellert, Yuliya Ponomareva, Thomas Braun, Shizuka Uchida, Scott T. Weiss, Amitabh Sharma and Harald Renz Corresponding Author: Harald Renz, MD Institute of Laboratory Medicine Philipps University Marburg 35043 Marburg, Germany Phone +49 6421 586 6235 [email protected] 1 Kılıç A et. al. 2018 A B C D PBS OVA chronic PAS PAS (cm H2O*s/ml (cm Raw Sirius Red Sirius Supplementary figure 1: Characteristic phenotype of the acute and chronic allergic airway inflammatory response in the OVA-induced mouse model. (a) Total and differential cell counts determined in BAL and cytospins show a dominant influx of eosinophils. (b) Serum titers of OVA-specific immunoglobulins are elevated. (c) Representative measurement of airway reactivity to increasing methacholine (Mch) responsiveness measured by head-out body plethysmography. (d) Representative lung section staining depicting airway inflammation and mucus production (PAS, original magnification x 10) and airway remodeling (SiriusRed; original magnification x 20). Data are presented as mean ± SEM and are representative for at least 3 independent experiments with n=8-10 animals per group. Statistical analyses have been performed with one-way ANOVA and Tukey’s post-test and show *p<0.05, **p<0.01 and ***p<0.001. 2 Kılıç A et. al. 2018 lymphocytes A lung activated CD4+-cells CD4+-T-cell-subpopulations SSC FSC ST2 live CD69 CD4 CXCR3 B spleen Naive CD4+- T cells DAPI FSC single cells SSC CD62L CD4 CD44 FSC FSC-W Supplementary figure 2.
    [Show full text]