H3K27 Methylation Dynamics During CD4 T Cell Activation: Regulation of JAK/STAT and IL12RB2 Expression by JMJD3

Total Page:16

File Type:pdf, Size:1020Kb

H3K27 Methylation Dynamics During CD4 T Cell Activation: Regulation of JAK/STAT and IL12RB2 Expression by JMJD3 H3K27 Methylation Dynamics during CD4 T Cell Activation: Regulation of JAK/STAT and IL12RB2 Expression by JMJD3 This information is current as Sarah A. LaMere, Ryan C. Thompson, Xiangzhi Meng, H. of September 24, 2021. Kiyomi Komori, Adam Mark and Daniel R. Salomon J Immunol published online 25 September 2017 http://www.jimmunol.org/content/early/2017/09/23/jimmun ol.1700475 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2017/09/23/jimmunol.170047 Material 5.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 24, 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 © 2017 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published September 25, 2017, doi:10.4049/jimmunol.1700475 The Journal of Immunology H3K27 Methylation Dynamics during CD4 T Cell Activation: Regulation of JAK/STAT and IL12RB2 Expression by JMJD3 Sarah A. LaMere,1 Ryan C. Thompson, Xiangzhi Meng, H. Kiyomi Komori,2 Adam Mark,1 and Daniel R. Salomon3 The changes to the epigenetic landscape in response to Ag during CD4 T cell activation have not been well characterized. Although CD4 T cell subsets have been mapped globally for numerous epigenetic marks, little has been done to study their dynamics early after activation. We have studied changes to promoter H3K27me3 during activation of human naive and memory CD4 T cells. Our results show that these changes occur relatively early (1 d) after activation of naive and memory cells and that demethylation is the predominant change to H3K27me3 at this time point, reinforcing high expression of target genes. Additionally, inhibition of the H3K27 demethylase JMJD3 in naive CD4 T cells demonstrates how critically important molecules required for T cell differen- tiation, such as JAK2 and IL12RB2, are regulated by H3K27me3. Our results show that H3K27me3 is a dynamic and important Downloaded from epigenetic modification during CD4 T cell activation and that JMJD3-driven H3K27 demethylation is critical for CD4 T cell function. The Journal of Immunology, 2017, 199: 000–000. D4 T cells are an integral component of the adaptive which confer another layer of complexity to this already enigmatic immune response, facilitating Ag-specific “memory” via system. Epigenetic studies in T cells have been more limited and http://www.jimmunol.org/ C adaptation following exposure to pathogens and other in this early phase of investigation are still largely descriptive, foreign invaders. The molecular mechanisms responsible for the although clear correlations between epigenetic modifications and transition to memory are still poorly understood and likely stem T cell differentiation have been illustrated with genome-wide from multiple epigenetic processes in the cell following activation studies (6–13). (1). Numerous epigenetic marks have been explored in cancer and H3K27me3, in particular, is a conventionally “repressive” his- embryonic stem (ES) cell differentiation, including DNA methyl- tone modification (9, 14) that plays a role in CD4 T cell differ- ation, histone modifications, and chromatin regulators, all of which entiation (15, 16). However, its dynamics during CD4 T cell appear to play roles in modulating gene expression (2–5). These activation and early differentiation has not been explored, and marks can target multiple regulatory regions, including promoters, its role in early differentiation is still poorly characterized. One by guest on September 24, 2021 enhancers, superenhancers, gene bodies, and intergenic regions, study performed global mapping of H3K27me3 after in vitro polarization of murine CD4 T cell differentiation to reveal that the presence of H3K27me3 in Th-related genes corresponded to Department of Molecular and Experimental Medicine, The Scripps Research Insti- tute, La Jolla, CA 92037 silencing of those genes in their opposing lineages (7). A study of 1Current address: University of California, San Diego, La Jolla, CA. murine CD8 T cell dynamics after viral infection also demon- 2Current address: Synthetic Genomics, La Jolla, CA. strated a profound loss of H3K27me3 following activation, sup- porting the role of repressive H3K27me3 marks in naive CD8 3Daniel R. Salomon is deceased. T cells to maintain a state of restraint during rest (8). ORCIDs: 0000-0002-4612-0425 (S.A.L.); 0000-0002-0450-8181 (R.C.T.); 0000- 0002-4743-9972 (X.M.). Two demethylases, JMJD3 and UTX, are known to catalyze Received for publication March 31, 2017. Accepted for publication August 21, 2017. H3K27me3 demethylation. Recently an exploration into the role of This work was supported by National Institutes of Health Grants U19 AI063603 (to Jmjd3 in mice upon the regulation of CD4 T cell differentiation D.R.S.), 1TL1 TR001113-01 (to S.A.L.), and T32 DK007022-30 (to H.K.K.). H.K.K. found that a conditional knockout of Jmjd3 resulted in skewing to was the recipient of a Juvenile Diabetes Research Foundation Postdoctoral Fellow- Th2 and Th17 differentiation (15). Both demethylases are required ship, X.M. received support from the Predoctoral Mendez National Institute of Trans- plantation’s Fund, and Verna Harrah Research Funds provided support to the for in vivo thymocyte differentiation in mice (17). In ES cells, Salomon laboratory. JMJD3 appears to delocalize polycomb repressive complex (PRC) The raw data for ChIP and RNA sequencing presented in this article have been proteins, which is essential for further development. Additionally, submitted to the Gene Expression Omnibus under accession number GSE73214 UTX is a component of the MLL complex, strongly suggesting (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE73214). that H3K27 demethylation can be coupled with the “activating” Address correspondence and reprint requests to Dr. Sarah A. LaMere at the current address: Department of Pathology, University of California, San Diego, 9500 Gilman methylation of H3K4 by MLL (18). UTX is ubiquitously Drive, La Jolla, CA 92093. E-mail address: [email protected] expressed in tissues and is also important for embryonic cell de- The online version of this article contains supplemental material. velopment (19). In contrast, JMJD3 is commonly induced during Abbreviations used in this article: B2M, b-2 microglobulin; CGI, CpG island; ChIP, inflammation or upon exposure to antigenic or oncogenic stimuli chromatin immunoprecipitation; ChIP-seq, ChIP sequencing; CPM, counts per mil- (18, 20, 21). JMJD3 inhibits somatic cell reprogramming in in- lion; ES, embryonic stem; FDR, false discovery rate; FPKM, fragments per kilobase per million fragments sequenced; IDR, Irreproducible Discovery Rate; IPA, Ingenu- ducible pluripotent stem cells, whereas UTX is essential for it, ity Pathway Analysis; PRC, polycomb repressive complex; qPCR, quantitative PCR; suggesting contrasting roles for these two enzymes (22, 23). The qRT-PCR, quantitative real time PCR; RNA-seq, RNA sequencing; siRNA, small two enzymes also play contrasting roles in acute lymphoblastic interfering RNA; TSS, transcription start site. leukemia, with JMJD3 inducing the neoplastic process and UTX Copyright Ó 2017 by The American Association of Immunologists, Inc. 0022-1767/17/$35.00 acting as a tumor suppressor (24). www.jimmunol.org/cgi/doi/10.4049/jimmunol.1700475 2 H3K27 METHYLATION OF CD4 T CELLS In the current study, we have examined the dynamics of added to each ChIP assay and incubated at 4˚C with rotation for 2 h. Beads promoter-associated H3K27me3 upon activation of human naive were washed three times in low-salt wash buffer and then two times with and memory CD4 T cells. We find that, in both subsets, profound high-salt wash buffer (0.1% SDS, 1.0% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl [pH 8.1], 500 mM NaCl) with 5 min of rotation at 4˚C for demethylation of H3K27 is observed by 1 d after activation, which each wash. Beads were resuspended in 150 ml of elution buffer (1% SDS, is in contrast to H3K4 methylation, for which changes are not ob- 0.1 M NaHCO3) and incubated in a ThermoMixer at 65˚C for 30 min at served until days later (25). Mapping specific states of H3K27me3 1200 rpm to reverse cross-linking. Two microliters of Proteinase K to known immune pathways demonstrates that loss of H3K27me3 (Invitrogen) was added to each sample, and 6 ml of 5 M NaCl was added for a total concentration of 200 mM. Samples were then incubated in a early in activation corresponds to pathways crucial to T cell func- ThermoMixer at 65˚C overnight at 1200 rpm. Eluted samples were re- tion, including T cell activation and the JAK/STAT pathways. moved from the beads and purified using the QIAGEN QIAquick PCR Mechanistic studies by perturbation of H3K27 demethylation with a purification kit, per the manufacturer’s instructions. small-molecule inhibitor (GSK-J4) and small interfering RNA Preparation of sequencing libraries for ChIP-seq and deep (siRNA) knockdown of the two H3K27 demethylases confirms that RNA-seq H3K27 demethylation by JMJD3 is important for key members of early differentiation–related pathways. Altogether, these data con- RNA for RNA-seq was isolated from purified cells using an AllPrep kit firm that H3K27 is a highly dynamic epigenetic modification in (QIAGEN), following the manufacturer’s instructions, and purified total CD4 T cells during early activation, and the nature of these dynamic RNA was converted to cDNA using Ovation RNA-seq (NuGEN), followed by S1 endonuclease digestion (Promega), as previously described (26).
Recommended publications
  • A Novel Ve-Zinc Nger-Related-Gene Signature Related to Tumor Immune
    A novel ve-zinc nger-related-gene signature related to tumor immune microenvironment allows for treatment stratication and predicts prognosis in clear cell renal cell carcinoma patients Feilong Zhang Beijing Chaoyang Hospital Jiyue Wu Beijing Chaoyang Hospital Jiandong Zhang Beijing Chaoyang Hospital Peng Cao Beijing Chaoyang Hospital Zejia Sun Beijing Chaoyang Hospital Wei Wang ( [email protected] ) Beijing Chaoyang Hospital https://orcid.org/0000-0003-2642-3338 Research Keywords: Posted Date: February 23rd, 2021 DOI: https://doi.org/10.21203/rs.3.rs-207677/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/25 Abstract Background Clear cell renal cell carcinoma (ccRCC) is one of the most prevalent renal malignant tumors, which survival rate and quality of life of ccRCC patients are not satisfactory. Therefore, identication of prognostic biomarkers of ccRCC patients will contribute to early and accurate clinical intervention and treatment, and then improve their prognosis. Methods We downloaded the original expression data of mRNAs from The Cancer Genome Atlas database and the zinc nger(ZNF)-related genes (ZRGs) from UniProt online database. Differentially expressed ZRGs (DE- ZRGs) was screened from tumor and adjacent nontumor tissues and functional enrichment analysis was conducted out. A ve-ZRG signature were constructed by univariate Cox regression, least absolute shrinkage and selection operator and multivariate Cox regression. Furthermore, we screened out independent prognosis-related factors to build a nomogram by univariate and multivariate Cox regression. Potential biological pathways of ve ZRGs were analyzed by Gene Set Enrichment Analysis (GSEA). Then, we further quantitatively analyze immune inltration and evaluate tumor microenvironment by single sample GSEA.
    [Show full text]
  • PCDHGC3 (NM 032403) Human Tagged ORF Clone Lentiviral Particle – RC211429L4V | Origene
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RC211429L4V PCDHGC3 (NM_032403) Human Tagged ORF Clone Lentiviral Particle Product data: Product Type: Lentiviral Particles Product Name: PCDHGC3 (NM_032403) Human Tagged ORF Clone Lentiviral Particle Symbol: PCDHGC3 Synonyms: PC43; PCDH-GAMMA-C3; PCDH2 Vector: pLenti-C-mGFP-P2A-Puro (PS100093) ACCN: NM_032403 ORF Size: 402 bp ORF Nucleotide The ORF insert of this clone is exactly the same as(RC211429). Sequence: OTI Disclaimer: The molecular sequence of this clone aligns with the gene accession number as a point of reference only. However, individual transcript sequences of the same gene can differ through naturally occurring variations (e.g. polymorphisms), each with its own valid existence. This clone is substantially in agreement with the reference, but a complete review of all prevailing variants is recommended prior to use. More info OTI Annotation: This clone was engineered to express the complete ORF with an expression tag. Expression varies depending on the nature of the gene. RefSeq: NM_032403.1 RefSeq Size: 2326 bp RefSeq ORF: 405 bp Locus ID: 5098 UniProt ID: Q9BR81 Protein Families: Transmembrane MW: 13.9 kDa This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 2 PCDHGC3 (NM_032403) Human Tagged ORF Clone Lentiviral Particle – RC211429L4V Gene Summary: This gene is a member of the protocadherin gamma gene cluster, one of three related clusters tandemly linked on chromosome five.
    [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]
  • A Clinicopathological and Molecular Genetic Analysis of Low-Grade Glioma in Adults
    A CLINICOPATHOLOGICAL AND MOLECULAR GENETIC ANALYSIS OF LOW-GRADE GLIOMA IN ADULTS Presented by ANUSHREE SINGH MSc A thesis submitted in partial fulfilment of the requirements of the University of Wolverhampton for the degree of Doctor of Philosophy Brain Tumour Research Centre Research Institute in Healthcare Sciences Faculty of Science and Engineering University of Wolverhampton November 2014 i DECLARATION This work or any part thereof has not previously been presented in any form to the University or to any other body whether for the purposes of assessment, publication or for any other purpose (unless otherwise indicated). Save for any express acknowledgments, references and/or bibliographies cited in the work, I confirm that the intellectual content of the work is the result of my own efforts and of no other person. The right of Anushree Singh to be identified as author of this work is asserted in accordance with ss.77 and 78 of the Copyright, Designs and Patents Act 1988. At this date copyright is owned by the author. Signature: Anushree Date: 30th November 2014 ii ABSTRACT The aim of the study was to identify molecular markers that can determine progression of low grade glioma. This was done using various approaches such as IDH1 and IDH2 mutation analysis, MGMT methylation analysis, copy number analysis using array comparative genomic hybridisation and identification of differentially expressed miRNAs using miRNA microarray analysis. IDH1 mutation was present at a frequency of 71% in low grade glioma and was identified as an independent marker for improved OS in a multivariate analysis, which confirms the previous findings in low grade glioma studies.
    [Show full text]
  • PCDHGC3 (NM 032402) Human Tagged ORF Clone Lentiviral Particle Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RC211379L3V PCDHGC3 (NM_032402) Human Tagged ORF Clone Lentiviral Particle Product data: Product Type: Lentiviral Particles Product Name: PCDHGC3 (NM_032402) Human Tagged ORF Clone Lentiviral Particle Symbol: PCDHGC3 Synonyms: PC43; PCDH-GAMMA-C3; PCDH2 Vector: pLenti-C-Myc-DDK-P2A-Puro (PS100092) ACCN: NM_032402 ORF Size: 2589 bp ORF Nucleotide The ORF insert of this clone is exactly the same as(RC211379). Sequence: OTI Disclaimer: The molecular sequence of this clone aligns with the gene accession number as a point of reference only. However, individual transcript sequences of the same gene can differ through naturally occurring variations (e.g. polymorphisms), each with its own valid existence. This clone is substantially in agreement with the reference, but a complete review of all prevailing variants is recommended prior to use. More info OTI Annotation: This clone was engineered to express the complete ORF with an expression tag. Expression varies depending on the nature of the gene. RefSeq: NM_032402.1 RefSeq Size: 2794 bp RefSeq ORF: 2592 bp Locus ID: 5098 UniProt ID: Q9UN70, Q9BR81 Domains: CA Protein Families: Transmembrane MW: 91.2 kDa This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 2 PCDHGC3 (NM_032402) Human Tagged ORF Clone Lentiviral Particle – RC211379L3V Gene Summary: This gene is a member of the protocadherin gamma gene cluster, one of three related clusters tandemly linked on chromosome five.
    [Show full text]
  • Tepzz¥ 6Z54za T
    (19) TZZ¥ ZZ_T (11) EP 3 260 540 A1 (12) EUROPEAN PATENT APPLICATION (43) Date of publication: (51) Int Cl.: 27.12.2017 Bulletin 2017/52 C12N 15/113 (2010.01) A61K 9/127 (2006.01) A61K 31/713 (2006.01) C12Q 1/68 (2006.01) (21) Application number: 17000579.7 (22) Date of filing: 12.11.2011 (84) Designated Contracting States: • Sarma, Kavitha AL AT BE BG CH CY CZ DE DK EE ES FI FR GB Philadelphia, PA 19146 (US) GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO • Borowsky, Mark PL PT RO RS SE SI SK SM TR Needham, MA 02494 (US) • Ohsumi, Toshiro Kendrick (30) Priority: 12.11.2010 US 412862 P Cambridge, MA 02141 (US) 20.12.2010 US 201061425174 P 28.07.2011 US 201161512754 P (74) Representative: Clegg, Richard Ian et al Mewburn Ellis LLP (62) Document number(s) of the earlier application(s) in City Tower accordance with Art. 76 EPC: 40 Basinghall Street 11840099.3 / 2 638 163 London EC2V 5DE (GB) (71) Applicant: The General Hospital Corporation Remarks: Boston, MA 02114 (US) •Thecomplete document including Reference Tables and the Sequence Listing can be downloaded from (72) Inventors: the EPO website • Lee, Jeannie T •This application was filed on 05-04-2017 as a Boston, MA 02114 (US) divisional application to the application mentioned • Zhao, Jing under INID code 62. San Diego, CA 92122 (US) •Claims filed after the date of receipt of the divisional application (Rule 68(4) EPC). (54) POLYCOMB-ASSOCIATED NON-CODING RNAS (57) This invention relates to long non-coding RNAs (IncRNAs), libraries of those ncRNAs that bind chromatin modifiers, such as Polycomb Repressive Complex 2, inhibitory nucleic acids and methods and compositions for targeting IncRNAs.
    [Show full text]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • Learning from Cadherin Structures and Sequences: Affinity Determinants and Protein Architecture
    Learning from cadherin structures and sequences: affinity determinants and protein architecture Klára Fels ıvályi 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 Klara Felsovalyi All rights reserved ABSTRACT Learning from cadherin structures and sequences: affinity determinants and protein architecture Klara Felsovalyi Cadherins are a family of cell-surface proteins mediating adhesion that are important in development and maintenance of tissues. The family is defined by the repeating cadherin domain (EC) in their extracellular region, but they are diverse in terms of protein size, architecture and cellular function. The best-understood subfamily is the type I classical cadherins, which are found in vertebrates and have five EC domains. Among the five different type I classical cadherins, the binding interactions are highly specific in their homo- and heterophilic binding affinities, though their sequences are very similar. As previously shown, E- and N-cadherins, two prototypic members of the subfamily, differ in their homophilic K D by about an order of magnitude, while their heterophilic affinity is intermediate. To examine the source of the binding affinity differences among type I cadherins, we used crystal structures, analytical ultracentrifugation (AUC), surface plasmon resonance (SPR), and electron paramagnetic resonance (EPR) studies. Phylogenetic analysis and binding affinity behavior show that the type I cadherins can be further divided into two subgroups, with E- and N-cadherin representing each. In addition to the affinity differences in their wild-type binding through the strand-swapped interface, a second interface also shows an affinity difference between E- and N-cadherin.
    [Show full text]
  • Metastatic Adrenocortical Carcinoma Displays Higher Mutation Rate and Tumor Heterogeneity Than Primary Tumors
    ARTICLE DOI: 10.1038/s41467-018-06366-z OPEN Metastatic adrenocortical carcinoma displays higher mutation rate and tumor heterogeneity than primary tumors Sudheer Kumar Gara1, Justin Lack2, Lisa Zhang1, Emerson Harris1, Margaret Cam2 & Electron Kebebew1,3 Adrenocortical cancer (ACC) is a rare cancer with poor prognosis and high mortality due to metastatic disease. All reported genetic alterations have been in primary ACC, and it is 1234567890():,; unknown if there is molecular heterogeneity in ACC. Here, we report the genetic changes associated with metastatic ACC compared to primary ACCs and tumor heterogeneity. We performed whole-exome sequencing of 33 metastatic tumors. The overall mutation rate (per megabase) in metastatic tumors was 2.8-fold higher than primary ACC tumor samples. We found tumor heterogeneity among different metastatic sites in ACC and discovered recurrent mutations in several novel genes. We observed 37–57% overlap in genes that are mutated among different metastatic sites within the same patient. We also identified new therapeutic targets in recurrent and metastatic ACC not previously described in primary ACCs. 1 Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA. 2 Center for Cancer Research, Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA. 3 Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA. Correspondence and requests for materials should be addressed to E.K. (email: [email protected]) NATURE COMMUNICATIONS | (2018) 9:4172 | DOI: 10.1038/s41467-018-06366-z | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-06366-z drenocortical carcinoma (ACC) is a rare malignancy with types including primary ACC from the TCGA to understand our A0.7–2 cases per million per year1,2.
    [Show full text]
  • Identification of Gene Modules Associated with Warfarin Dosage by a Genome-Wide DNA Methylation Study
    ORIGINAL ARTICLES Department of Clinical Pharmacology1, Xiangya Hospital, Institute of Clinical Pharmacology2, Central South University, Hunan Key Laboratory of Pharmacogenetics; Changsha; Department of Cardio-Thoracic Surgery3, the Second Xiangya Hospital of Central South University, Changsha; Key Laboratory of Bio-resources and Eco-environment4 (Ministry of Education), College of Life Science, Sichuan University, Chengdu, China Identification of gene modules associated with warfarin dosage by a genome-wide DNA methylation study ZHIYING LUO1, 2, RONG LIU1, 2, BAO SUN1, 2, XINMING ZHOU3, ZHAOQIAN LIU1, 2, HONGHAO ZHOU1, 2, HENG XU4, XI LI1,2,*,#, WEI ZHANG1,2,*,# Received December 12, 2017, accepted January 10, 2018 *Corresponding authors: Prof. Wei Zhang, Xi Li, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, Changsha, Hunan 410008, China [email protected]; [email protected] #Wei Zhang and Xi Li contributed equally to this study. Pharmazie 73: 288–293 (2018) doi: 10.1691/ph.2018.7319 Objective: To identify warfarin dose-associated DNA methylation changes, we conducted the first genomewide DNA methylation association study. Method: A total of 22 patients who required an extreme warfarin dosage from VKORC1 -1639AA & CYP2C9*1*1 genotype group were enrolled in this study. The Illumina Infinium Human- Methylation450 platform was used to perform genome-scale DNA methylation profiling, identifying differentially methylated CpG sites by a nonparametric test. WGCNA was used to analyze the association between gene modules and extreme warfarin dosage. Results: For a total of 378,313 CpG sites that passed the quality control processes, we identified eight differentially methylated CpG probes (p<0.05) showing altered DNA methylation level (>20%) between two extreme dose groups.
    [Show full text]
  • The Conserved DNMT1-Dependent Methylation Regions in Human Cells
    Freeman et al. Epigenetics & Chromatin (2020) 13:17 https://doi.org/10.1186/s13072-020-00338-8 Epigenetics & Chromatin RESEARCH Open Access The conserved DNMT1-dependent methylation regions in human cells are vulnerable to neurotoxicant rotenone exposure Dana M. Freeman1 , Dan Lou1, Yanqiang Li1, Suzanne N. Martos1 and Zhibin Wang1,2,3* Abstract Background: Allele-specifc DNA methylation (ASM) describes genomic loci that maintain CpG methylation at only one inherited allele rather than having coordinated methylation across both alleles. The most prominent of these regions are germline ASMs (gASMs) that control the expression of imprinted genes in a parent of origin-dependent manner and are associated with disease. However, our recent report reveals numerous ASMs at non-imprinted genes. These non-germline ASMs are dependent on DNA methyltransferase 1 (DNMT1) and strikingly show the feature of random, switchable monoallelic methylation patterns in the mouse genome. The signifcance of these ASMs to human health has not been explored. Due to their shared allelicity with gASMs, herein, we propose that non-tradi- tional ASMs are sensitive to exposures in association with human disease. Results: We frst explore their conservancy in the human genome. Our data show that our putative non-germline ASMs were in conserved regions of the human genome and located adjacent to genes vital for neuronal develop- ment and maturation. We next tested the hypothesized vulnerability of these regions by exposing human embryonic kidney cell HEK293 with the neurotoxicant rotenone for 24 h. Indeed,14 genes adjacent to our identifed regions were diferentially expressed from RNA-sequencing. We analyzed the base-resolution methylation patterns of the predicted non-germline ASMs at two neurological genes, HCN2 and NEFM, with potential to increase the risk of neurodegenera- tion.
    [Show full text]
  • Advances in Prognostic Methylation Biomarkers for Prostate Cancer
    cancers Review Advances in Prognostic Methylation Biomarkers for Prostate Cancer 1 1,2 1,2, 1,2, , Dilys Lam , Susan Clark , Clare Stirzaker y and Ruth Pidsley * y 1 Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; [email protected] (D.L.); [email protected] (S.C.); [email protected] (C.S.) 2 St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia * Correspondence: [email protected]; Tel.: +61-2-92958315 These authors have contributed equally. y Received: 22 September 2020; Accepted: 13 October 2020; Published: 15 October 2020 Simple Summary: Prostate cancer is a major cause of cancer-related death in men worldwide. There is an urgent clinical need for improved prognostic biomarkers to better predict the likely outcome and course of the disease and thus inform the clinical management of these patients. Currently, clinically recognised prognostic markers lack sensitivity and specificity in distinguishing aggressive from indolent disease, particularly in patients with localised, intermediate grade prostate cancer. Thus, there is major interest in identifying new molecular biomarkers to complement existing standard clinicopathological markers. DNA methylation is a frequent alteration in the cancer genome and offers potential as a reliable and robust biomarker. In this review, we provide a comprehensive overview of the current state of DNA methylation biomarker studies in prostate cancer prognosis. We highlight advances in this field that have enabled the discovery of novel prognostic genes and discuss the potential of methylation biomarkers for noninvasive liquid-biopsy testing.
    [Show full text]