Profiling DNA Methylation Differences Between Inbred Mouse Strains

Total Page:16

File Type:pdf, Size:1020Kb

Profiling DNA Methylation Differences Between Inbred Mouse Strains bioRxiv preprint doi: https://doi.org/10.1101/194464; this version posted September 27, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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. 1 Profiling DNA Methylation Differences Between Inbred Mouse Strains 2 on the Illumina Human Infinium MethylationEPIC Microarray 3 Short title: Cross-species utility of human DNA methylation microarray 4 Hemant Gujar1, Jane W. Liang1, Nicholas C. Wong2, Khyobeni Mozhui1,3* 5 6 1 Department of Preventive Medicine, University of Tennessee Health Science Center, 7 Memphis, Tennessee 38163, USA 8 2 Monash Bioinformatics Platform, Monash University, Clayton VIC, Australia 9 3 Department of Genetics, Genomics and Informatics, University of Tennessee Health 10 Science Centre, Memphis, Tennessee 38163, USA 11 * Corresponding author 12 Khyobeni Mozhui 13 Department of Preventive Medicine, College of Medicine 14 University of Tennessee Health Science Center 15 66 N. Pauline Street, #631 16 Memphis, TN 38163 17 Phone no. 901-448-7334 18 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/194464; this version posted September 27, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.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. 19 Abstract 20 The Illumina Infinium MethylationEPIC provides an efficient platform for profiling DNA 21 methylation in humans at over 850,000 CpGs. Model organisms such as mice do not 22 currently benefit from an equivalent array. Here we used this array to measure DNA 23 methylation in mice. We defined probes targeting conserved regions and performed a 24 comparison between the array-based assay and affinity-based DNA sequencing of 25 methyl-CpGs (MBD-seq). Mouse samples consisted of 11 liver DNA from two strains, 26 C57BL/6J (B6) and DBA/2J (D2), that varied widely in age. Linear regression was 27 applied to detect differential methylation. In total, 13,665 probes (1.6% of total probes) 28 aligned to conserved CpGs. Beta-values (β-value) for these probes showed a 29 distribution similar to that in humans. Overall, there was high concordance in 30 methylation signal between the EPIC array and MBD-seq (Pearson correlation r = 0.70, 31 p-value < 0.0001). However, the EPIC probes had higher quantitative sensitivity at 32 CpGs that are hypo- (β-value < 0.3) or hypermethylated (β-value > 0.7). In terms of 33 differential methylation, no EPIC probe detected significant difference between age 34 groups at a Benjamini-Hochberg threshold of 10%, and the MBD-seq performed better 35 at detecting age-dependent change in methylation. However, the top most significant 36 probe for age (cg13269407; uncorrected p-value = 1.8 x 10-5) is part of the clock CpGs 37 used to estimate the human epigenetic age. For strain, 219 Infinium probes detected 38 significant differential methylation (FDR cutoff 10%) with ~80% CpGs associated with 39 higher methylation in D2. This higher methylation profile in D2 compared to B6 was also 40 replicated by the MBD-seq data. To summarize, we found only a small subset of EPIC 41 probes that target conserved sites. However, for this small subset the array provides a 2 bioRxiv preprint doi: https://doi.org/10.1101/194464; this version posted September 27, 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. 42 reliable assay of DNA methylation and can be effectively used to measure differential 43 methylation in mice. 44 Keywords: DNA methylation, epigenetics, microarray, cross-species comparison 45 3 bioRxiv preprint doi: https://doi.org/10.1101/194464; this version posted September 27, 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. 46 Introduction 47 There has been a surge in large-scale epigenetic studies in recent years. In particular, 48 epigenome-wide association studies (EWAS) of DNA methylation have shown 49 association with physiological traits [1,2], diseases [3-5], environmental exposures [6,7], 50 aging [8], and even socioeconomic [9] and emotional experiences [10]. The 51 development of robust and reliable methylation microarrays has been an important 52 driving force. In particular, the Illumina Human Methylation BeadChips have made it 53 both convenient and cost-effective to incorporate an epigenetic arm to large 54 epidemiological studies [11,12]. The latest version, the Illumina Infinium 55 MethylationEPIC BeadChip (EPIC), provides an efficient high throughput platform to 56 quantify methylation at 866,836 CpG sites on the human genome [13,14]. A remarkable 57 biological insight that has emerged from these array-based studies is the definition of 58 the methylation-based “epigenetic clock,” a biomarker of human age and aging (i.e., the 59 epigenetic clock) that is defined using specific probes represented on these arrays [8]. 60 Currently there is no equivalent microarray platform for model organisms and work in 61 experimental species have largely relied on high-throughput sequencing. For instance, 62 while the human DNA methylation age can be calculated from a few hundred probes on 63 the Illumina BeadChips, a similar effort in mice required a more extensive sequencing of 64 the mouse methylome [15]. However, CpG islands (CGIs) are largely conserved 65 between mice and humans and the two species share similar numbers of CGIs and 66 similar proportions of CGIs in promoter regions of genes [16]. Considering that these 67 CpGs and CGIs are highly conserved in gene regulatory regions, it is feasible that 4 bioRxiv preprint doi: https://doi.org/10.1101/194464; this version posted September 27, 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. 68 probes on the human microarrays that target these sites may have some application in 69 research using rodent models. This was previously evaluated for the two older versions 70 of the Illumina HumanMethylation BeadChips [17]. The work by Wong et al. 71 demonstrated that a subset of the probes targeting highly conserved sites provide 72 reliable measures of DNA methylation in mice, and could be feasibly used to evaluate 73 tissue specific methylation and in cancer related studies using the mouse as a model 74 system. 75 In the present work, we extend the conservation analysis to the EPIC platform, and 76 evaluate the capacity of these probes to detect differential methylation. We begin by 77 defining the conserved probes and the key features of the corresponding CpG sites in 78 the context of the larger mouse and human genome. We also compare the methylation 79 signal detected by the conserved probes with affinity-based methyl-CpG enriched DNA 80 sequence (MBD-seq) data from the same samples and evaluate if the conserved 81 probes are informative of age and strain differences in mice. 82 Materials and Methods 83 Defining Conserved EPIC probes 84 Sequences for the 866,836 CpG probes were obtained from Illumina 85 (http://www.illumina.com/). The probe sequences were aligned to the mouse genome 86 (mm10) using bowtie2 (version 2.2.6) with standard default parameters. A total of 87 34,981 probes aligned to the mouse genome of varying alignment quality. Conserved 88 probes were then defined based on quality of alignment. For this, we filtered out all 5 bioRxiv preprint doi: https://doi.org/10.1101/194464; this version posted September 27, 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. 89 sequences with a low mapping quality (MAPQ) of less than 60 (15,717 excluded) and 90 those that contain more than two non-matching base pairs (1,092). To retain only the 91 high quality probes, we further filtered probes based on confidence in DNA methylation 92 signal and based on this, 4,507 probes with detection p-values > 0.0001 were removed. 93 This generated a list of 13,665 high quality probes that are conserved sequences and 94 provide reliable methylation assays in mice (these are listed in Supplementary Data 95 S1). CpG island annotations [18] for the respective genome were downloaded from 96 UCSC Genome Browser (http://genome.ucsc.edu) and distribution of conserved probes 97 and positions of CGIs were plotted to the human (GRCh37) and mouse (mm10) 98 genomes using CIRCOS [19]. 99 For conserved sequences, there is high correspondence in functional and genomic 100 features between mouse and human genomes and we referred to the human probe 101 annotations provided by Illumina to define the location of conserved probes with respect 102 to gene features and CpG context (i.e., islands, shores, shelves) (Supplementary 103 Data S1). To evaluate if the conserved set is enriched in specific features relative to the 104 full background set, we performed a hypergeometric test using the phyper function in R. 105 Animals and sample preparation 106 Tissues samples were derived from mice that were part of an aging cohort maintained 107 at the University of Tennessee Health Science Center (PI: Robert W.
Recommended publications
  • MARCH5 Requires MTCH2 to Coordinate Proteasomal Turnover of the MCL1:NOXA Complex
    Cell Death & Differentiation (2020) 27:2484–2499 https://doi.org/10.1038/s41418-020-0517-0 ARTICLE MARCH5 requires MTCH2 to coordinate proteasomal turnover of the MCL1:NOXA complex 1,2 1,2,5 1,2 1,2 1,2,3 1,2 Tirta Mario Djajawi ● Lei Liu ● Jia-nan Gong ● Allan Shuai Huang ● Ming-jie Luo ● Zhen Xu ● 4 1,2 1,2 1,2 Toru Okamoto ● Melissa J. Call ● David C. S. Huang ● Mark F. van Delft Received: 3 July 2019 / Revised: 6 February 2020 / Accepted: 7 February 2020 / Published online: 24 February 2020 © The Author(s) 2020. This article is published with open access Abstract MCL1, a BCL2 relative, is critical for the survival of many cells. Its turnover is often tightly controlled through both ubiquitin-dependent and -independent mechanisms of proteasomal degradation. Several cell stress signals, including DNA damage and cell cycle arrest, are known to elicit distinct E3 ligases to ubiquitinate and degrade MCL1. Another trigger that drives MCL1 degradation is engagement by NOXA, one of its BH3-only protein ligands, but the mechanism responsible has remained unclear. From an unbiased genome-wide CRISPR-Cas9 screen, we discovered that the ubiquitin E3 ligase MARCH5, the ubiquitin E2 conjugating enzyme UBE2K, and the mitochondrial outer membrane protein MTCH2 co- — fi 1234567890();,: 1234567890();,: operate to mark MCL1 for degradation by the proteasome speci cally when MCL1 is engaged by NOXA. This mechanism of degradation also required the MCL1 transmembrane domain and distinct MCL1 lysine residues to proceed, suggesting that the components likely act on the MCL1:NOXA complex by associating with it in a specific orientation within the mitochondrial outer membrane.
    [Show full text]
  • Molecular Profiling of Peripheral Blood Is Associated with Circulating Tumor Cells Content and Poor Survival in Metastatic Castration-Resistant Prostate Cancer
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No. 12 Molecular profiling of peripheral blood is associated with circulating tumor cells content and poor survival in metastatic castration-resistant prostate cancer Mercedes Marín-Aguilera1, Òscar Reig1,2, Juan José Lozano3, Natalia Jiménez1, Susana García-Recio1,4, Nadina Erill5, Lydia Gaba2, Andrea Tagliapietra2, Vanesa Ortega2, Gemma Carrera6, Anna Colomer5, Pedro Gascón4 and Begoña Mellado1,2 1 Translational Genomics Group and Targeted Therapeutics in Solid Tumors Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain 2 Medical Oncology Department, Hospital Clínic, Barcelona, Spain 3 Bioinformatics Platform Department, Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBEREHD), Hospital Clínic, Barcelona, Spain 4 Laboratory of Translational Oncology, Fundació Clínic per a la Recerca Biomèdica, Barcelona, Spain 5 Althia, Barcelona, Spain 6 Medical Oncology Department, Hospital Plató, Barcelona, Spain Correspondence to: Begoña Mellado, email: [email protected] Keywords: circulating tumor cells, peripheral blood, microarrays, cell search system Received: January 22, 2015 Accepted: February 14, 2015 Published: March 12, 2015 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT The enumeration of circulating
    [Show full text]
  • The Ire1a-XBP1 Pathway Promotes T Helper Cell Differentiation by Resolving Secretory Stress and Accelerating Proliferation
    bioRxiv preprint doi: https://doi.org/10.1101/235010; this version posted December 15, 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-NC-ND 4.0 International license. The IRE1a-XBP1 pathway promotes T helper cell differentiation by resolving secretory stress and accelerating proliferation Jhuma Pramanik1, Xi Chen1, Gozde Kar1,2, Tomás Gomes1, Johan Henriksson1, Zhichao Miao1,2, Kedar Natarajan1, Andrew N. J. McKenzie3, Bidesh Mahata1,2*, Sarah A. Teichmann1,2,4* 1. Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom 2. EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom 3. MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 OQH, United Kingdom 4. Theory of Condensed Matter, Cavendish Laboratory, 19 JJ Thomson Ave, Cambridge CB3 0HE, United Kingdom. *To whom correspondence should be addressed: [email protected] and [email protected] Keywords: Th2 lymphocyte, XBP1, Genome wide XBP1 occupancy, Th2 lymphocyte proliferation, ChIP-seq, RNA-seq, Th2 transcriptome Summary The IRE1a-XBP1 pathway, a conserved adaptive mediator of the unfolded protein response, is indispensable for the development of secretory cells. It maintains endoplasmic reticulum homeostasis by facilitating protein folding and enhancing secretory capacity of the cells. Its role in immune cells is emerging. It is involved in dendritic cell, plasma cell and eosinophil development and differentiation. Using genome-wide approaches, integrating ChIPmentation and mRNA-sequencing data, we have elucidated the regulatory circuitry governed by the IRE1a-XBP1 pathway in type-2 T helper cells (Th2).
    [Show full text]
  • HIC1 Modulates Prostate Cancer Progression by Epigenetic Modification
    Author Manuscript Published OnlineFirst on January 22, 2013; DOI: 10.1158/1078-0432.CCR-12-2888 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. HIC1 modulates prostate cancer progression by epigenetic modification Jianghua Zheng#1, 2, Jinglong Wang#1, Xueqing Sun#1,Mingang Hao1,Tao Ding3, Dan Xiong 1, Xiumin Wang1, Yu Zhu 4, Gang Xiao 1, Guangcun Cheng1, Meizhong Zhao5, Jian Zhang6, Jianhua Wang1* 1 Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. 2 Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China. 3 Department of Urology, Shanghai Putuo Hospital, Shanghai Traditional Chinese Medicine University, Shanghai, 200062, China. 4 Department of Urology, Shanghai Ruijin Hospital, Shanghai, 200025, China. 5 Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025, China. 6 Guangxi Medical University, Nanning, 530021, China. # J Zheng, J Wang and X Sun contributed equally to this work. *Corresponding author: Jianhua Wang, PhD, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. Phone: 021-54660871; Fax: 021-63842157; E-mail:[email protected]. No potential conflicts of interest were disclosed 1 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 22, 2013; DOI: 10.1158/1078-0432.CCR-12-2888 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Statement of Translational Relevance This study aimed to further our understanding of the role that hypermethylatioted in cancer 1 (HIC1) plays in prostate cancer (PCa) progression.
    [Show full text]
  • Methods in and Applications of the Sequencing of Short Non-Coding Rnas" (2013)
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2013 Methods in and Applications of the Sequencing of Short Non- Coding RNAs Paul Ryvkin University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Bioinformatics Commons, Genetics Commons, and the Molecular Biology Commons Recommended Citation Ryvkin, Paul, "Methods in and Applications of the Sequencing of Short Non-Coding RNAs" (2013). Publicly Accessible Penn Dissertations. 922. https://repository.upenn.edu/edissertations/922 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/922 For more information, please contact [email protected]. Methods in and Applications of the Sequencing of Short Non-Coding RNAs Abstract Short non-coding RNAs are important for all domains of life. With the advent of modern molecular biology their applicability to medicine has become apparent in settings ranging from diagonistic biomarkers to therapeutics and fields angingr from oncology to neurology. In addition, a critical, recent technological development is high-throughput sequencing of nucleic acids. The convergence of modern biotechnology with developments in RNA biology presents opportunities in both basic research and medical settings. Here I present two novel methods for leveraging high-throughput sequencing in the study of short non- coding RNAs, as well as a study in which they are applied to Alzheimer's Disease (AD). The computational methods presented here include High-throughput Annotation of Modified Ribonucleotides (HAMR), which enables researchers to detect post-transcriptional covalent modifications ot RNAs in a high-throughput manner. In addition, I describe Classification of RNAs by Analysis of Length (CoRAL), a computational method that allows researchers to characterize the pathways responsible for short non-coding RNA biogenesis.
    [Show full text]
  • Mechanisms Controlling Cell Life and Death Decisions
    Department of Biological Regulation echanisms controlling cell Atan Gross Yehudit Zaltsman, Mlife and death decisions Natalie Yivgi-Ohana, Iris Kamer, Galia Oberkovitz, Liat Shachnai, Maria Maryanovich Programmed cell death or apoptosis in the extraembryonic (ExEm) region of is essential for both the development E7.5 wild type embryos, and this region and maintenance of tissue homeostasis is largely impaired in the Mtch2/Mimp-/- in multicellular organisms. The BCL-2 embryos. Thus, Mtch2/Mimp might play family members are critical regulators a critical role in the formation of the 972 8 934 3656 of the apoptotic program, whereas ExEm region. To study the connection the caspase proteases are the major between Mtch2/Mimp and apoptosis FAX 972 8 934 4116 executioners of this program. Members we generated Mtch2/Mimp-/- stable [email protected] of the BCL-2 family include both anti- and embryonic stem (ES) cell lines carrying www.weizmann.ac.il/ pro-apoptotic proteins. The BH3-only either an empty vector or Mtch2/Mimp. Biological_Regulation/gross proteins (e.g., BID) are an important Using these lines we demonstrated that subset of the pro-apoptotic proteins the presence of Mtch2/Mimp sensitizes that act as sentinels of intercellular cells to tBID-induced MOMP. Thus, we damage. In our laboratory, we are discovered that Mtch2/Mimp is critical S phase and inhibition of apoptosis focused on elucidating the mechanisms for normal embryonic development, following DNA damage. This surprising that balance between cell life and and is an important positive regulator and new pro-survival function of BID death, and BID is one of our primary of tBID-induced apoptosis at the is regulated by its phosphorylation tools to study these mechanisms.
    [Show full text]
  • SND1, a Component of RNA-Induced Silencing Complex, Is Up-Regulated in Human Colon Cancers and Implicated in Early Stage Colon Carcinogenesis
    Research Article SND1, a Component of RNA-Induced Silencing Complex, Is Up-regulated in Human Colon Cancers and Implicated in Early Stage Colon Carcinogenesis Naoto Tsuchiya, Masako Ochiai, Katsuhiko Nakashima, Tsuneyuki Ubagai, Takashi Sugimura, and Hitoshi Nakagama Biochemistry Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan Abstract transcription factors is well known to be a causative event involved Colon cancers have been shown to develop after accumulation in malignant transformation, for example, with functional inacti- vation or intragenic mutation of p53 (3), and gene amplification or of multiple genetic and epigenetic alterations with changes in global gene expression profiles, contributing to the establish- overexpression of c-myc (4). ment of widely diverse phenotypes. Transcriptional and Recently, posttranscriptional regulation, especially translational posttranscriptional regulation of gene expression by small control, of gene expression has become a major interest in the field of cancer research, because accumulating evidence strongly RNA species, such as the small interfering RNA and microRNA suggests its biological relevance for cancer development and and the RNA-induced silencing complex (RISC), is currently drawing major interest with regard to cancer development. progression (5). Some oncogenes and tumor suppressor genes, including c-myc (6), Mdm-2(7), and p53 (8), have been shown to be SND1, also called Tudor-SN and p100 and recently reported to regulated at the translational level. Overexpression of the transla- be a component of RISC, is among the list of highly expressed tion initiation factor eIF4E promotes malignant transformation of genes in human colon cancers. In the present study, we normal fibroblasts (9) and cooperates with c-Myc to drive B-cell showed remarkable up-regulation of SND1 mRNA in human lymphomatogenesis in eIF4E transgenic mice (10).
    [Show full text]
  • Unr Defines a Novel Class of Nucleoplasmic Reticulum Involved In
    © 2017. Published by The Company of Biologists Ltd | Journal of Cell Science (2017) 130, 1796-1808 doi:10.1242/jcs.198697 RESEARCH ARTICLE Unr defines a novel class of nucleoplasmic reticulum involved in mRNA translation Frédéric Saltel1,2,*,§, Alban Giese1,2,*, Lamia Azzi1,2,3,*, Habiba Elatmani1,2, Pierre Costet4, Zakaria Ezzoukhry1,2,‡, Nathalie Dugot-Senant2, Lucile Miquerol5,Oréda Boussadia6, Harald Wodrich2,7, Pierre Dubus1,2,3,# and Hélenè Jacquemin-Sablon1,2,#,§ ABSTRACT gene expression (Mihailovich et al., 2010; Wolffe, 1994). The Unr (officially known as CSDE1) is a cytoplasmic RNA-binding mammalian Unr proteins, composed of five CSDs, are highly similar protein with roles in the regulation of mRNA stability and translation. In by sharing >90% amino acid identity. Unr is a cytoplasmic RNA- in vitro this study, we identified a novel function for Unr, which acts as a binding protein that, , interacts preferentially with purine-rich positive regulator of placental development. Unr expression studies in motifs located in RNA loops (Jacquemin-Sablon et al., 1994; the developing placenta revealed the presence of Unr-rich foci that Triqueneaux et al., 1999). Unr has been characterized as a regulator of are apparently located in the nuclei of trophoblast giant cells (TGCs). mRNA turnover (Grosset et al., 2000) and translation. During We determined that what we initially thought to be foci, were actually cross translation, Unr acts as a positive or negative regulator of specific sections of a network of double-wall nuclear membrane invaginations transcripts; it either stimulates or represses the translation driven by that contain a cytoplasmic core related to the nucleoplasmic reticulum internal ribosome entry sites (IRESs) (Boussadia et al., 2003; (NR).
    [Show full text]
  • Cops5 Safeguards Genomic Stability of Embryonic Stem Cells Through Regulating Cellular Metabolism and DNA Repair
    Cops5 safeguards genomic stability of embryonic stem cells through regulating cellular metabolism and DNA repair Peng Lia, Lulu Gaoa, Tongxi Cuia, Weiyu Zhanga, Zixin Zhaoa, and Lingyi Chena,1 aState Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Collaborative Innovation Center for Biotherapy, Tianjin Key Laboratory of Protein Sciences, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, 300071 Tianjin, China Edited by Janet Rossant, Hospital for Sick Children, University of Toronto, Toronto, Canada, and approved December 24, 2019 (received for review August 29, 2019) The highly conserved COP9 signalosome (CSN), composed of 8 transiently expressed in about 5% of ESCs at a given time, subunits (Cops1 to Cops8), has been implicated in pluripotency promotes rapid telomere elongation by telomere recombination maintenance of human embryonic stem cells (ESCs). Yet, the mech- and regulates genomic stability (11). Induced by genotoxic stress, anism for the CSN to regulate pluripotency remains elusive. We Filia stimulates the PARP1 activity and relocates from centro- previously showed that Cops2, independent of the CSN, is essential somes to DNA damage sites and mitochondria to regulate DDR for the pluripotency maintenance of mouse ESCs. In this study, we and apoptosis (12). Sall4, a pluripotency transcription factor, set out to investigate how Cops5 and Cops8 regulate ESC differ- facilitates the ataxia telangiectasia-mutated activation in re- entiation and tried to establish Cops5 and Cops8 knockout (KO) sponse to DSBs (13). To minimize the ROS-induced genomic ESC lines by CRISPR/Cas9. To our surprise, no Cops5 KO ESC clones DNA damage, ESCs produce lower levels of mitochondrial ROS were identified out of 127 clones, while three Cops8 KO ESC lines and express higher levels of antioxidants than differentiated cells were established out of 70 clones.
    [Show full text]
  • Insights Into SND1 Oncogene Promoter Regulation
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Archivo Digital para la Docencia y la Investigación REVIEW published: 11 December 2018 doi: 10.3389/fonc.2018.00606 Insights Into SND1 Oncogene Promoter Regulation Begoña Ochoa, Yolanda Chico and María José Martínez* Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain The staphylococcal nuclease and Tudor domain containing 1 gene (SND1), also known as Tudor-SN, TSN or p100, encodes an evolutionarily conserved protein with invariant domain composition. SND1 contains four repeated staphylococcal nuclease domains and a single Tudor domain, which confer it endonuclease activity and extraordinary capacity for interacting with nucleic acids, individual proteins and protein complexes. Originally described as a transcriptional coactivator, SND1 plays fundamental roles in the regulation of gene expression, including RNA splicing, interference, stability, and editing, as well as in the regulation of protein and lipid homeostasis. Recently, SND1 has gained attention as a potential disease biomarker due to its positive correlation with cancer progression and metastatic spread. Such functional diversity of SND1 marks this gene as interesting for further analysis in relation with the multiple levels of regulation of SND1 protein production. In this review, we summarize the SND1 genomic region Edited by: and promoter architecture, the set of transcription factors that can bind the proximal Markus A. N. Hartl, promoter, and the evidence supporting transactivation of SND1 promoter by a number of University of Innsbruck, Austria signal transduction pathways operating in different cell types and conditions. Unraveling Reviewed by: John Strouboulis, the mechanisms responsible for SND1 promoter regulation is of utmost interest to King’s College London, decipher the SND1 contribution in the realm of both normal and abnormal physiology.
    [Show full text]
  • Mechanisms Underlying Phenotypic Heterogeneity in Simplex Autism Spectrum Disorders
    Mechanisms Underlying Phenotypic Heterogeneity in Simplex Autism Spectrum Disorders Andrew H. Chiang Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2021 © 2021 Andrew H. Chiang All Rights Reserved Abstract Mechanisms Underlying Phenotypic Heterogeneity in Simplex Autism Spectrum Disorders Andrew H. Chiang Autism spectrum disorders (ASD) are a group of related neurodevelopmental diseases displaying significant genetic and phenotypic heterogeneity. Despite recent progress in ASD genetics, the nature of phenotypic heterogeneity across probands is not well understood. Notably, likely gene- disrupting (LGD) de novo mutations affecting the same gene often result in substantially different ASD phenotypes. We find that truncating mutations in a gene can result in a range of relatively mild decreases (15-30%) in gene expression due to nonsense-mediated decay (NMD), and show that more severe autism phenotypes are associated with greater decreases in expression. We also find that each gene with recurrent ASD mutations can be described by a parameter, phenotype dosage sensitivity (PDS), which characteriZes the relationship between changes in a gene’s dosage and changes in a given phenotype. Using simple linear models, we show that changes in gene dosage account for a substantial fraction of phenotypic variability in ASD. We further observe that LGD mutations affecting the same exon frequently lead to strikingly similar phenotypes in unrelated ASD probands. These patterns are observed for two independent proband cohorts and multiple important ASD-associated phenotypes. The observed phenotypic similarities are likely mediated by similar changes in gene dosage and similar perturbations to the relative expression of splicing isoforms.
    [Show full text]
  • Downloaded As a Non-Linear Data Structure Containing Ordered Pairs of 68 Proteins and All the Other Proteins with Which They Interact
    bioRxiv preprint doi: https://doi.org/10.1101/854695; this version posted November 27, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Network potential identifies therapeutic miRNA cocktails in Ewings Sarcoma Davis T. Weaver1, 2, *, Kathleen I. Pishas3,*, Drew Williamson4,*, Jessica Scarborough1, 2, Stephen L. Lessnick3, Andrew Dhawan2, 5, †, and Jacob G. Scott1, 2, 6, † 1Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA 2Translational Hematology Oncology Research, Cleveland Clinic, Cleveland OH, 44106, USA 3Nationwide Children’s Hospital, Columbus, Ohio, 43205 4Department of Pathology, Massachusetts General Hospital, Boston, MA, 02144 5Division of Neurology, Cleveland Clinic, Cleveland Ohio, 44195 6Department of Physics, Case Western Reserve University, Cleveland, OH, 44106, USA *contributed equally †[email protected], [email protected] ABSTRACT Introduction: Micro-RNA (miRNA)-based therapies are an emerging class of cancer therapies with many potential applications in the field owing to their ability to repress multiple, predictable targets and cause widespread changes in a cell signaling network. New miRNA-based oligonucleotide drugs have have shown significant promise for the treatment of cancer in pre-clinical studies. Because of the broad effects miRNAs can have on different cells and tissues, a network science-based approach is well-equipped to evaluate and identify miRNA candidates and combinations of candidates for the repression of key oncogenic targets. Methods: In this work, we present a novel network science-based approach for identification of potential miRNA therapies, using Ewings Sarcoma as a model system.
    [Show full text]