301
|
Bertin N, Mendez M, Hasegawa A, Lizio M, Abugessaisa I, Severin J, Sakai-Ohno M, Lassmann T, Kasukawa T, Kawaji H, Hayashizaki Y, Forrest ARR, Carninci P, Plessy C. Linking FANTOM5 CAGE peaks to annotations with CAGEscan. Sci Data 2017; 4:170147. [PMID: 28972578 PMCID: PMC5625555 DOI: 10.1038/sdata.2017.147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/09/2017] [Indexed: 01/22/2023] Open
Abstract
The FANTOM5 expression atlas is a quantitative measurement of the activity of nearly 200,000 promoter regions across nearly 2,000 different human primary cells, tissue types and cell lines. Generation of this atlas was made possible by the use of CAGE, an experimental approach to localise transcription start sites at single-nucleotide resolution by sequencing the 5′ ends of capped RNAs after their conversion to cDNAs. While 50% of CAGE-defined promoter regions could be confidently associated to adjacent transcriptional units, nearly 100,000 promoter regions remained gene-orphan. To address this, we used the CAGEscan method, in which random-primed 5′-cDNAs are paired-end sequenced. Pairs starting in the same region are assembled in transcript models called CAGEscan clusters. Here, we present the production and quality control of CAGEscan libraries from 56 FANTOM5 RNA sources, which enhances the FANTOM5 expression atlas by providing experimental evidence associating core promoter regions with their cognate transcripts.
Collapse
Affiliation(s)
- Nicolas Bertin
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Mickaël Mendez
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Akira Hasegawa
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Marina Lizio
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Imad Abugessaisa
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Jessica Severin
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Mizuho Sakai-Ohno
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Timo Lassmann
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Takeya Kasukawa
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan
| | - Hideya Kawaji
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako 351-0198, Japan
| | - Yoshihide Hayashizaki
- RIKEN Omics Science Center, Yokohama 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako 351-0198, Japan
| | - Alistair R R Forrest
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako 351-0198, Japan
| | - Piero Carninci
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| | - Charles Plessy
- RIKEN Center for Life Science Technologies, Division of Genomics Technologies, Yokohama 230-0045, Japan.,RIKEN Omics Science Center, Yokohama 230-0045, Japan
| |
Collapse
|
302
|
Kelso TWR, Porter DK, Amaral ML, Shokhirev MN, Benner C, Hargreaves DC. Chromatin accessibility underlies synthetic lethality of SWI/SNF subunits in ARID1A-mutant cancers. eLife 2017; 6:30506. [PMID: 28967863 PMCID: PMC5643100 DOI: 10.7554/elife.30506] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/28/2017] [Indexed: 02/06/2023] Open
Abstract
ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, is frequently mutated in cancer. Deficiency in its homolog ARID1B is synthetically lethal with ARID1A mutation. However, the functional relationship between these homologs has not been explored. Here, we use ATAC-seq, genome-wide histone modification mapping, and expression analysis to examine colorectal cancer cells lacking one or both ARID proteins. We find that ARID1A has a dominant role in maintaining chromatin accessibility at enhancers, while the contribution of ARID1B is evident only in the context of ARID1A mutation. Changes in accessibility are predictive of changes in expression and correlate with loss of H3K4me and H3K27ac marks, nucleosome spacing, and transcription factor binding, particularly at growth pathway genes including MET. We find that ARID1B knockdown in ARID1A mutant ovarian cancer cells causes similar loss of enhancer architecture, suggesting that this is a conserved function underlying the synthetic lethality between ARID1A and ARID1B.
Collapse
Affiliation(s)
- Timothy W R Kelso
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, California, United States
| | - Devin K Porter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, California, United States
| | - Maria Luisa Amaral
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, California, United States
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, California, United States
| | - Christopher Benner
- Department of Medicine, University of California San Diego, California, United States
| | - Diana C Hargreaves
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, California, United States
| |
Collapse
|
303
|
Coons LA, Hewitt SC, Burkholder AB, McDonnell DP, Korach KS. DNA Sequence Constraints Define Functionally Active Steroid Nuclear Receptor Binding Sites in Chromatin. Endocrinology 2017; 158:3212-3234. [PMID: 28977594 PMCID: PMC5659708 DOI: 10.1210/en.2017-00468] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/30/2017] [Indexed: 12/17/2022]
Abstract
Gene regulatory programs are encoded in the sequence of the DNA. Since the completion of the Human Genome Project, millions of gene regulatory elements have been identified in the human genome. Understanding how each of those sites functionally contributes to gene regulation, however, remains a challenge for nearly every field of biology. Transcription factors influence cell function by interpreting information contained within cis-regulatory elements in chromatin. Whereas chromatin immunoprecipitation-sequencing has been used to identify and map transcription factor-DNA interactions, it has been difficult to assign functionality to the binding sites identified. Thus, in this study, we probed the transcriptional activity, DNA-binding competence, and functional activity of select nuclear receptor mutants in cellular and animal model systems and used this information to define the sequence constraints of functional steroid nuclear receptor cis-regulatory elements. Analysis of the architecture within sNR chromatin interacting sites revealed that only a small fraction of all sNR chromatin-interacting events is associated with transcriptional output and that this functionality is restricted to elements that vary from the consensus palindromic elements by one or two nucleotides. These findings define the transcriptional grammar necessary to predict functionality from regulatory sequences, with a multitude of future implications.
Collapse
Affiliation(s)
- Laurel A Coons
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Sylvia C Hewitt
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Adam B Burkholder
- Integrative Bioinformatics, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Kenneth S Korach
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27709
| |
Collapse
|
304
|
Yu NY, Bieder A, Raman A, Mileti E, Katayama S, Einarsdottir E, Fredholm BB, Falk A, Tapia-Páez I, Daub CO, Kere J. Acute doses of caffeine shift nervous system cell expression profiles toward promotion of neuronal projection growth. Sci Rep 2017; 7:11458. [PMID: 28904364 PMCID: PMC5597620 DOI: 10.1038/s41598-017-11574-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022] Open
Abstract
Caffeine is a widely consumed psychoactive substance, but little is known about the effects of caffeine stimulation on global gene expression changes in neurons. Here, we conducted gene expression profiling of human neuroepithelial stem cell-derived neurons, stimulated with normal consumption levels of caffeine (3 μM and 10 μM), over a period of 9 h. We found dosage-dependent activation of immediate early genes after 1 h. Neuronal projection development processes were up-regulated and negative regulation of axon extension processes were down-regulated at 3 h. In addition, genes involved in extracellular matrix organization, response for wound healing, and regulation of immune system processes were down-regulated by caffeine at 3 h. This study identified novel genes within the neuronal projection guidance pathways that respond to acute caffeine stimulation and suggests potential mechanisms for the effects of caffeine on neuronal cells.
Collapse
Affiliation(s)
- Nancy Y Yu
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden
| | - Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden
| | - Amitha Raman
- Department of Medicine (MedH), Karolinska Institutet, Huddinge, SE-141 86, Sweden
| | - Enrichetta Mileti
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden
| | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden
| | - Elisabet Einarsdottir
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden.,Folkhälsan Institute of Genetics, and Molecular Neurology Research Program, University of Helsinki, Helsinki, 00014, Finland
| | - Bertil B Fredholm
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, SE-171 77, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Solna, SE-171 77, Sweden
| | - Isabel Tapia-Páez
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden.,Department of Medicine/Center for Molecular Medicine, Karolinska University Hospital, Solna, SE-171 76, Sweden
| | - Carsten O Daub
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden.,Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, #230-0045, Japan
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83, Sweden. .,Folkhälsan Institute of Genetics, and Molecular Neurology Research Program, University of Helsinki, Helsinki, 00014, Finland. .,Department of Medical and Molecular Genetics, King's College London, London, SE1 9RT, United Kingdom.
| |
Collapse
|
305
|
Transcriptome Analysis Uncovers a Growth-Promoting Activity of Orosomucoid-1 on Hepatocytes. EBioMedicine 2017; 24:257-266. [PMID: 28927749 PMCID: PMC5652006 DOI: 10.1016/j.ebiom.2017.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 02/08/2023] Open
Abstract
The acute phase protein orosomucoid-1 (Orm1) is mainly expressed by hepatocytes (HPCs) under stress conditions. However, its specific function is not fully understood. Here, we report a role of Orm1 as an executer of HPC proliferation. Increases in serum levels of Orm1 were observed in patients after surgical resection for liver cancer and in mice undergone partial hepatectomy (PH). Transcriptome study showed that Orm1 became the most abundant in HPCs isolated from regenerating mouse liver tissues after PH. Both in vitro and in vivo siRNA-induced knockdown of Orm1 suppressed proliferation of mouse regenerating HPCs and human hepatic cells. Microarray analysis in regenerating mouse livers revealed that the signaling pathways controlling chromatin replication, especially the minichromosome maintenance protein complex genes were uniformly down-regulated following Orm1 knockdown. These data suggest that Orm1 is induced in response to hepatic injury and executes liver regeneration by activating cell cycle progression in HPCs. Serum Orm1 levels increased approximately 1.3- to 2.5-folds in both humans and mice after partial hepatectomy. Transcriptome analysis revealed that Orm1 mostly induced in hepatocytes as a regulator of mouse liver regeneration. Orm1 knockdown in mice impaired liver regeneration with poor hepatocyte growth and suppressed cell cycle signaling.
Orosomucoid-1 (Orm1) is an acute phase protein mainly expressed by hepatocytes under stress conditions. Beginning from the finding that Orm1 was induced after partial hepatectomy in humans and mice, we showed enrichment of Orm1 in regenerating hepatocytes of hepatectomized mice by transcriptome analysis and following culture and animal experiments. Knockdown of Orm1 in mice resulted in decreases in hepatocyte growth accompanying suppressed signaling in controlling chromatin replication. Therefore, Orm1 would be a potential therapeutic and prognostic biomarker for liver diseases, especially after surgical resection of cancer-bearing liver, through its newly found ability to stimulate the cell cycle in regenerating hepatocytes.
Collapse
|
306
|
Lizio M, Deviatiiarov R, Nagai H, Galan L, Arner E, Itoh M, Lassmann T, Kasukawa T, Hasegawa A, Ros MA, Hayashizaki Y, Carninci P, Forrest ARR, Kawaji H, Gusev O, Sheng G. Systematic analysis of transcription start sites in avian development. PLoS Biol 2017; 15:e2002887. [PMID: 28873399 PMCID: PMC5600399 DOI: 10.1371/journal.pbio.2002887] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/15/2017] [Accepted: 08/18/2017] [Indexed: 01/07/2023] Open
Abstract
Cap Analysis of Gene Expression (CAGE) in combination with single-molecule sequencing technology allows precision mapping of transcription start sites (TSSs) and genome-wide capture of promoter activities in differentiated and steady state cell populations. Much less is known about whether TSS profiling can characterize diverse and non-steady state cell populations, such as the approximately 400 transitory and heterogeneous cell types that arise during ontogeny of vertebrate animals. To gain such insight, we used the chick model and performed CAGE-based TSS analysis on embryonic samples covering the full 3-week developmental period. In total, 31,863 robust TSS peaks (>1 tag per million [TPM]) were mapped to the latest chicken genome assembly, of which 34% to 46% were active in any given developmental stage. ZENBU, a web-based, open-source platform, was used for interactive data exploration. TSSs of genes critical for lineage differentiation could be precisely mapped and their activities tracked throughout development, suggesting that non-steady state and heterogeneous cell populations are amenable to CAGE-based transcriptional analysis. Our study also uncovered a large set of extremely stable housekeeping TSSs and many novel stage-specific ones. We furthermore demonstrated that TSS mapping could expedite motif-based promoter analysis for regulatory modules associated with stage-specific and housekeeping genes. Finally, using Brachyury as an example, we provide evidence that precise TSS mapping in combination with Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-on technology enables us, for the first time, to efficiently target endogenous avian genes for transcriptional activation. Taken together, our results represent the first report of genome-wide TSS mapping in birds and the first systematic developmental TSS analysis in any amniote species (birds and mammals). By facilitating promoter-based molecular analysis and genetic manipulation, our work also underscores the value of avian models in unravelling the complex regulatory mechanism of cell lineage specification during amniote development.
Collapse
Affiliation(s)
- Marina Lizio
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
| | - Ruslan Deviatiiarov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Hiroki Nagai
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- RIKEN Center for Developmental Biology, Kobe, Japan
| | - Laura Galan
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria-SODERCAN), Santander, Spain
| | - Erik Arner
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
| | - Masayoshi Itoh
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Yokohama, Japan
| | - Timo Lassmann
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
| | - Takeya Kasukawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
| | - Akira Hasegawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
| | - Marian A. Ros
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC-Universidad de Cantabria-SODERCAN), Santander, Spain
| | - Yoshihide Hayashizaki
- RIKEN Omics Science Center (OSC), Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Yokohama, Japan
| | - Piero Carninci
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
| | - Alistair R. R. Forrest
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, Western Australia, Australia
| | - Hideya Kawaji
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Yokohama, Japan
- * E-mail: (GS); (HK); (OG)
| | - Oleg Gusev
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies (CLST), Yokohama, Japan
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Yokohama, Japan
- RIKEN Innovation Center, Wako, Japan
- * E-mail: (GS); (HK); (OG)
| | - Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- RIKEN Center for Developmental Biology, Kobe, Japan
- * E-mail: (GS); (HK); (OG)
| |
Collapse
|
307
|
The FANTOM5 collection, a data series underpinning mammalian transcriptome atlases in diverse cell types. Sci Data 2017; 4:170113. [PMID: 28850107 PMCID: PMC5574373 DOI: 10.1038/sdata.2017.113] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 07/14/2017] [Indexed: 11/11/2022] Open
Abstract
The latest project from the FANTOM consortium, an international collaborative effort initiated by RIKEN, generated atlases of transcriptomes, in particular promoters, transcribed enhancers, and long-noncoding RNAs, across a diverse set of mammalian cell types. Here, we introduce the FANTOM5 collection, bringing together data descriptors, articles and analyses of FANTOM5 data published across the Nature Research journals. Associated data are openly available for reuse by all.
Collapse
|
308
|
Noguchi S, Arakawa T, Fukuda S, Furuno M, Hasegawa A, Hori F, Ishikawa-Kato S, Kaida K, Kaiho A, Kanamori-Katayama M, Kawashima T, Kojima M, Kubosaki A, Manabe RI, Murata M, Nagao-Sato S, Nakazato K, Ninomiya N, Nishiyori-Sueki H, Noma S, Saijyo E, Saka A, Sakai M, Simon C, Suzuki N, Tagami M, Watanabe S, Yoshida S, Arner P, Axton RA, Babina M, Baillie JK, Barnett TC, Beckhouse AG, Blumenthal A, Bodega B, Bonetti A, Briggs J, Brombacher F, Carlisle AJ, Clevers HC, Davis CA, Detmar M, Dohi T, Edge AS, Edinger M, Ehrlund A, Ekwall K, Endoh M, Enomoto H, Eslami A, Fagiolini M, Fairbairn L, Farach-Carson MC, Faulkner GJ, Ferrai C, Fisher ME, Forrester LM, Fujita R, Furusawa JI, Geijtenbeek TB, Gingeras T, Goldowitz D, Guhl S, Guler R, Gustincich S, Ha TJ, Hamaguchi M, Hara M, Hasegawa Y, Herlyn M, Heutink P, Hitchens KJ, Hume DA, Ikawa T, Ishizu Y, Kai C, Kawamoto H, Kawamura YI, Kempfle JS, Kenna TJ, Kere J, Khachigian LM, Kitamura T, Klein S, Klinken SP, Knox AJ, Kojima S, Koseki H, Koyasu S, Lee W, Lennartsson A, Mackay-sim A, Mejhert N, Mizuno Y, Morikawa H, Morimoto M, Moro K, Morris KJ, Motohashi H, et alNoguchi S, Arakawa T, Fukuda S, Furuno M, Hasegawa A, Hori F, Ishikawa-Kato S, Kaida K, Kaiho A, Kanamori-Katayama M, Kawashima T, Kojima M, Kubosaki A, Manabe RI, Murata M, Nagao-Sato S, Nakazato K, Ninomiya N, Nishiyori-Sueki H, Noma S, Saijyo E, Saka A, Sakai M, Simon C, Suzuki N, Tagami M, Watanabe S, Yoshida S, Arner P, Axton RA, Babina M, Baillie JK, Barnett TC, Beckhouse AG, Blumenthal A, Bodega B, Bonetti A, Briggs J, Brombacher F, Carlisle AJ, Clevers HC, Davis CA, Detmar M, Dohi T, Edge AS, Edinger M, Ehrlund A, Ekwall K, Endoh M, Enomoto H, Eslami A, Fagiolini M, Fairbairn L, Farach-Carson MC, Faulkner GJ, Ferrai C, Fisher ME, Forrester LM, Fujita R, Furusawa JI, Geijtenbeek TB, Gingeras T, Goldowitz D, Guhl S, Guler R, Gustincich S, Ha TJ, Hamaguchi M, Hara M, Hasegawa Y, Herlyn M, Heutink P, Hitchens KJ, Hume DA, Ikawa T, Ishizu Y, Kai C, Kawamoto H, Kawamura YI, Kempfle JS, Kenna TJ, Kere J, Khachigian LM, Kitamura T, Klein S, Klinken SP, Knox AJ, Kojima S, Koseki H, Koyasu S, Lee W, Lennartsson A, Mackay-sim A, Mejhert N, Mizuno Y, Morikawa H, Morimoto M, Moro K, Morris KJ, Motohashi H, Mummery CL, Nakachi Y, Nakahara F, Nakamura T, Nakamura Y, Nozaki T, Ogishima S, Ohkura N, Ohno H, Ohshima M, Okada-Hatakeyama M, Okazaki Y, Orlando V, Ovchinnikov DA, Passier R, Patrikakis M, Pombo A, Pradhan-Bhatt S, Qin XY, Rehli M, Rizzu P, Roy S, Sajantila A, Sakaguchi S, Sato H, Satoh H, Savvi S, Saxena A, Schmidl C, Schneider C, Schulze-Tanzil GG, Schwegmann A, Sheng G, Shin JW, Sugiyama D, Sugiyama T, Summers KM, Takahashi N, Takai J, Tanaka H, Tatsukawa H, Tomoiu A, Toyoda H, van de Wetering M, van den Berg LM, Verardo R, Vijayan D, Wells CA, Winteringham LN, Wolvetang E, Yamaguchi Y, Yamamoto M, Yanagi-Mizuochi C, Yoneda M, Yonekura Y, Zhang PG, Zucchelli S, Abugessaisa I, Arner E, Harshbarger J, Kondo A, Lassmann T, Lizio M, Sahin S, Sengstag T, Severin J, Shimoji H, Suzuki M, Suzuki H, Kawai J, Kondo N, Itoh M, Daub CO, Kasukawa T, Kawaji H, Carninci P, Forrest AR, Hayashizaki Y. FANTOM5 CAGE profiles of human and mouse samples. Sci Data 2017; 4:170112. [PMID: 28850106 PMCID: PMC5574368 DOI: 10.1038/sdata.2017.112] [Show More Authors] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/25/2017] [Indexed: 01/22/2023] Open
Abstract
In the FANTOM5 project, transcription initiation events across the human and mouse genomes were mapped at a single base-pair resolution and their frequencies were monitored by CAGE (Cap Analysis of Gene Expression) coupled with single-molecule sequencing. Approximately three thousands of samples, consisting of a variety of primary cells, tissues, cell lines, and time series samples during cell activation and development, were subjected to a uniform pipeline of CAGE data production. The analysis pipeline started by measuring RNA extracts to assess their quality, and continued to CAGE library production by using a robotic or a manual workflow, single molecule sequencing, and computational processing to generate frequencies of transcription initiation. Resulting data represents the consequence of transcriptional regulation in each analyzed state of mammalian cells. Non-overlapping peaks over the CAGE profiles, approximately 200,000 and 150,000 peaks for the human and mouse genomes, were identified and annotated to provide precise location of known promoters as well as novel ones, and to quantify their activities.
Collapse
Affiliation(s)
- Shuhei Noguchi
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Takahiro Arakawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Shiro Fukuda
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Masaaki Furuno
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Akira Hasegawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Fumi Hori
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Sachi Ishikawa-Kato
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Kaoru Kaida
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Ai Kaiho
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | | | - Tsugumi Kawashima
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Miki Kojima
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | | | - Ri-ichiroh Manabe
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Mitsuyoshi Murata
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Sayaka Nagao-Sato
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | | | - Noriko Ninomiya
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Hiromi Nishiyori-Sueki
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Shohei Noma
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Eri Saijyo
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Akiko Saka
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Mizuho Sakai
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | | | - Naoko Suzuki
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Michihira Tagami
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Shoko Watanabe
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | | | - Peter Arner
- Department of Medicine, Karolinska Institutet, 141 86, Stockholm, Sweden
- Karolinska University Hospital, Center for Metabolism and Endocrinology, 141 86, Stockholm, Sweden
| | - Richard A. Axton
- Scottish Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Magda Babina
- Department of Dermatology and Allergy, Charite University Medicine Berlin, Charitéplatz 1, 10117 Berlin, German
| | - J. Kenneth Baillie
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Timothy C. Barnett
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | | | - Antje Blumenthal
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102 Australia
| | - Beatrice Bodega
- IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Alessandro Bonetti
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - James Briggs
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, St Lucia, QLD 4072, Australia
| | - Frank Brombacher
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Ailsa J. Carlisle
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Hans C. Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- University Medical Centre Utrecht, Postbus 85500, 3508 GA Utrecht, The Netherlands
| | - Carrie A. Davis
- Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11797, USA
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H 303, 8093 Zurich, Switzerland
| | - Taeko Dohi
- Gastroenterology, Research Center for Hepatitis and Immunology, Research Institute National Center for Global Health and Medicine, Ichikawa, Chiba 272-8516, Japan
| | - Albert S.B. Edge
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Matthias Edinger
- Department of Internal Medicine III, University Hospital Regensburg, F.-J.-Strauss Allee 11, D-93053 Regensburg, Germany
- RCI Regensburg Centre for Interventional Immunology, University Hospital Regensburg, F.-J.-Strauss Allee 11, D-93053 Regensburg, Germany
| | - Anna Ehrlund
- Department of Medicine, Karolinska Institutet, 141 86, Stockholm, Sweden
- Karolinska University Hospital, Center for Metabolism and Endocrinology, 141 86, Stockholm, Sweden
| | - Karl Ekwall
- Department of Biosciences and Nutrition, Karolinska Institutet, Halsovagen 7-9, SE-141 83 Huddinge, Sweden
| | - Mitsuhiro Endoh
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hideki Enomoto
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, Chuou-ku, Kobe 650-0047, Japan
| | - Afsaneh Eslami
- Department of Bioinformatics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Michela Fagiolini
- F.M. Kirby Neurobiology Center, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Lynsey Fairbairn
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Mary C. Farach-Carson
- The University of Texas Health Science Center at Houston, Houston, TX 77251-1892, USA
| | - Geoffrey J. Faulkner
- Cancer Biology Program, Mater Medical Research Institute, South Brisbane, Queensland 4101, Australia
| | - Carmelo Ferrai
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center, Robert Roessle Str.10, 13125 Berlin, Germany
| | - Malcolm E. Fisher
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Lesley M. Forrester
- Scottish Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Rie Fujita
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Jun-ichi Furusawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Teunis B. Geijtenbeek
- Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Thomas Gingeras
- Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11797, USA
| | - Daniel Goldowitz
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Sven Guhl
- Department of Dermatology and Allergy, Charite University Medicine Berlin, Charitéplatz 1, 10117 Berlin, German
| | - Reto Guler
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Stefano Gustincich
- Neuroscience, SISSA, Via Bonomea 265, 34136 Trieste, Italy
- Department of Neuroscience and Brian Technologies, Italian Istitute of Technology, Via Morego 30, Genova, Italy
| | - Thomas J. Ha
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Masahide Hamaguchi
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mitsuko Hara
- RIKEN Center for Life Science Technologies, Wako, Saitama 351-0198, Japan
| | - Yuki Hasegawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Meenhard Herlyn
- Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania 19104, USA
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Otfried Müller Straße 23, 72076 Tübingen, Germany
| | - Kelly J. Hitchens
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, St Lucia, QLD 4072, Australia
| | - David A. Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Tomokatsu Ikawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Yuri Ishizu
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Chieko Kai
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Hiroshi Kawamoto
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Yuki I. Kawamura
- Gastroenterology, Research Center for Hepatitis and Immunology, Research Institute National Center for Global Health and Medicine, Ichikawa, Chiba 272-8516, Japan
| | - Judith S. Kempfle
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Tony J. Kenna
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Halsovagen 7-9, SE-141 83 Huddinge, Sweden
- Department of Genetics and Molecular Medicine, King's College London, Guy’s St Thomas Street, London, UK
| | - Levon M. Khachigian
- Centre for Vascular Research, University of New South Wales, Sydney, New South Wales 2052, Australia
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Toshio Kitamura
- Division of Cellular Therapy and Division of Stem Cell Signaling, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Sarah Klein
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H 303, 8093 Zurich, Switzerland
| | - S. Peter Klinken
- Harry Perkins Institute of Medical Research, Perth, WA 6009, Australia
| | - Alan J. Knox
- Respiratory Medicine, University of Nottingham, Hucknall Road, Nottingham NG5 1PB, UK
| | - Soichi Kojima
- RIKEN Center for Life Science Technologies, Wako, Saitama 351-0198, Japan
| | - Haruhiko Koseki
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Shigeo Koyasu
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Weonju Lee
- Dermatology, School of Medicine Kyungpook National University, Jung-gu, Daegu 41944, Korea
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Halsovagen 7-9, SE-141 83 Huddinge, Sweden
| | | | - Niklas Mejhert
- Department of Medicine, Karolinska Institutet, 141 86, Stockholm, Sweden
- Karolinska University Hospital, Center for Metabolism and Endocrinology, 141 86, Stockholm, Sweden
| | - Yosuke Mizuno
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan
| | - Hiromasa Morikawa
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mitsuru Morimoto
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, Chuou-ku, Kobe 650-0047, Japan
| | - Kazuyo Moro
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Kelly J. Morris
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center, Robert Roessle Str.10, 13125 Berlin, Germany
| | - Hozumi Motohashi
- Center for Radioisotope Sciences, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Christine L. Mummery
- Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Yutaka Nakachi
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan
| | - Fumio Nakahara
- Division of Cellular Therapy and Division of Stem Cell Signaling, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Toshiyuki Nakamura
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074, Japan
| | - Tadasuke Nozaki
- Department of Clinical Molecular Genetics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Soichi Ogishima
- Department of Bioclinical Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Mitsuhiro Ohshima
- Department of Biochemistry, Ohu University School of Pharmaceutical Sciences, Koriyama, Fukushima 963-8611 Japan
| | - Mariko Okada-Hatakeyama
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Insitute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasushi Okazaki
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama 350-1241, Japan
| | - Valerio Orlando
- IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Environmental Epigenetics Program, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Dmitry A. Ovchinnikov
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, St Lucia, QLD 4072, Australia
| | - Robert Passier
- Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Margaret Patrikakis
- Centre for Vascular Research, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Ana Pombo
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center, Robert Roessle Str.10, 13125 Berlin, Germany
| | | | - Xian-Yang Qin
- RIKEN Center for Life Science Technologies, Wako, Saitama 351-0198, Japan
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg, F.-J.-Strauss Allee 11, D-93053 Regensburg, Germany
- RCI Regensburg Centre for Interventional Immunology, University Hospital Regensburg, F.-J.-Strauss Allee 11, D-93053 Regensburg, Germany
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Otfried Müller Straße 23, 72076 Tübingen, Germany
| | - Sugata Roy
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Antti Sajantila
- Hjelt Institute, Department of Forensic Medicine, University of Helsinki, Kytosuontie 11, 003000 Helsinki, Finland
| | - Shimon Sakaguchi
- Department of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroki Sato
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Hironori Satoh
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Suzana Savvi
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Alka Saxena
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Christian Schmidl
- Department of Internal Medicine III, University Hospital Regensburg, F.-J.-Strauss Allee 11, D-93053 Regensburg, Germany
| | | | - Gundula G. Schulze-Tanzil
- Department of Orthopedic, Trauma and Reconstructive Surgery, Charite Universitatsmedizin Berlin, Charitéplatz 1, 10117 Berlin, German
| | - Anita Schwegmann
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC), University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Chuo-ku, Kumamoto 860-0811, Japan
| | - Jay W. Shin
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Daisuke Sugiyama
- Department of Clinical Study, Center for Advanced Medical Innovation, Kyushu University, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Takaaki Sugiyama
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Kim M. Summers
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Naoko Takahashi
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Jun Takai
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Hiroshi Tanaka
- Department of Bioinformatics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hideki Tatsukawa
- Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Andru Tomoiu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian EH25 9RG, UK
| | - Hiroo Toyoda
- Center for Radioisotope Sciences, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Marc van de Wetering
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Linda M. van den Berg
- Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Roberto Verardo
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie (LNCIB), Padriciano 99, 34149 Trieste, Italy
| | - Dipti Vijayan
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Christine A. Wells
- Centre for Stem Cell Systems, Department of Anatomy and Neuroscience, MDHS, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Ernst Wolvetang
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, St Lucia, QLD 4072, Australia
| | - Yoko Yamaguchi
- Department of Biochemistry, Nihon University School of Dentistry, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Chiyo Yanagi-Mizuochi
- Center for Clinical and Translational Reseach, Kyushu University Hospital, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Misako Yoneda
- Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Yohei Yonekura
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, Chuou-ku, Kobe 650-0047, Japan
| | - Peter G. Zhang
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | | | - Imad Abugessaisa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Erik Arner
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Jayson Harshbarger
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Atsushi Kondo
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Timo Lassmann
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- Telethon Kids Institute, the University of Western Australia, Perth, WA, Australia
| | - Marina Lizio
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Serkan Sahin
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | | | - Jessica Severin
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Hisashi Shimoji
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- Preventive medicine and applied genomics unit, RIKEN Advanced Center for Computing and Communication, Yokohama, Kanagawa 230-0045, Japan
| | - Masanori Suzuki
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Harukazu Suzuki
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Jun Kawai
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
| | - Naoto Kondo
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Masayoshi Itoh
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
| | - Carsten O. Daub
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- Department of Biosciences and Nutrition, Karolinska Institutet, Halsovagen 7-9, SE-141 83 Huddinge, Sweden
| | - Takeya Kasukawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Hideya Kawaji
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- Preventive medicine and applied genomics unit, RIKEN Advanced Center for Computing and Communication, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
| | - Piero Carninci
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
| | - Alistair R.R. Forrest
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- Harry Perkins Institute of Medical Research, Perth, WA 6009, Australia
| | - Yoshihide Hayashizaki
- RIKEN Omics Science Center, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
| |
Collapse
|
309
|
Dieterich LC, Ducoli L, Shin JW, Detmar M. Distinct transcriptional responses of lymphatic endothelial cells to VEGFR-3 and VEGFR-2 stimulation. Sci Data 2017; 4:170106. [PMID: 28850122 PMCID: PMC5574372 DOI: 10.1038/sdata.2017.106] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/31/2017] [Indexed: 12/25/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors play crucial roles in the formation of blood and lymphatic vessels during embryogenesis, and also under pathologic conditions in the adult. Despite intensive efforts over the last decades to elucidate the precise functions of VEGFs, transcriptional responses to VEGF receptor stimulation are still not fully characterized. To investigate the specific transcriptional effects of VEGFR-2 and VEGFR-3 activation, we performed a correlation analysis of previously published CAGE sequencing and microarray data of human lymphatic endothelial cells (LECs) stimulated with distinct VEGFs acting through either VEGFR-2 or VEGFR-3. We identified that specific activation of VEGFR-3 by VEGF-C156S results in the downregulation of many genes involved in immune regulation and inflammation, suggesting that VEGFR-3 stimulation has direct anti-inflammatory effects. Comparing CAGE and microarray data sets, we furthermore identified a small number of genes that showed a receptor-dependent response in LECs, demonstrating that these receptors, despite activating very similar signaling pathways, fulfill overlapping but not identical functions within the same cell type (LECs).
Collapse
Affiliation(s)
- Lothar C. Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Jay W. Shin
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
310
|
FANTOM5 CAGE profiles of human and mouse reprocessed for GRCh38 and GRCm38 genome assemblies. Sci Data 2017; 4:170107. [PMID: 28850105 PMCID: PMC5574367 DOI: 10.1038/sdata.2017.107] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/09/2017] [Indexed: 01/17/2023] Open
Abstract
The FANTOM5 consortium described the promoter-level expression atlas of human and mouse by using CAGE (Cap Analysis of Gene Expression) with single molecule sequencing. In the original publications, GRCh37/hg19 and NCBI37/mm9 assemblies were used as the reference genomes of human and mouse respectively; later, the Genome Reference Consortium released newer genome assemblies GRCh38/hg38 and GRCm38/mm10. To increase the utility of the atlas in forthcoming researches, we reprocessed the data to make them available on the recent genome assemblies. The data include observed frequencies of transcription starting sites (TSSs) based on the realignment of CAGE reads, and TSS peaks that are converted from those based on the previous reference. Annotations of the peak names were also updated based on the latest public databases. The reprocessed results enable us to examine frequencies of transcription initiations on the recent genome assemblies and to refer promoters with updated information across the genome assemblies consistently.
Collapse
|
311
|
An integrated expression atlas of miRNAs and their promoters in human and mouse. Nat Biotechnol 2017; 35:872-878. [PMID: 28829439 DOI: 10.1038/nbt.3947] [Citation(s) in RCA: 383] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/25/2017] [Indexed: 12/26/2022]
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs with key roles in cellular regulation. As part of the fifth edition of the Functional Annotation of Mammalian Genome (FANTOM5) project, we created an integrated expression atlas of miRNAs and their promoters by deep-sequencing 492 short RNA (sRNA) libraries, with matching Cap Analysis Gene Expression (CAGE) data, from 396 human and 47 mouse RNA samples. Promoters were identified for 1,357 human and 804 mouse miRNAs and showed strong sequence conservation between species. We also found that primary and mature miRNA expression levels were correlated, allowing us to use the primary miRNA measurements as a proxy for mature miRNA levels in a total of 1,829 human and 1,029 mouse CAGE libraries. We thus provide a broad atlas of miRNA expression and promoters in primary mammalian cells, establishing a foundation for detailed analysis of miRNA expression patterns and transcriptional control regions.
Collapse
|
312
|
A Novel and Multivalent Role of Pax6 in Cerebellar Development. J Neurosci 2017; 36:9057-69. [PMID: 27581449 DOI: 10.1523/jneurosci.4385-15.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 07/12/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Pax6 is a prominent gene in brain development. The deletion of Pax6 results in devastated development of eye, olfactory bulb, and cortex. However, it has been reported that the Pax6-null Sey cerebellum only has minor defects involving granule cells despite Pax6 being expressed throughout cerebellar development. The present work has uncovered a requirement of Pax6 in the development of all rhombic lip (RL) lineages. A significant downregulation of Tbr1 and Tbr2 expression is found in the Sey cerebellum, these are cell-specific markers of cerebellar nuclear (CN) neurons and unipolar brush cells (UBCs), respectively. The examination of Tbr1 and Lmx1a immunolabeling and Nissl staining confirmed the loss of CN neurons from the Sey cerebellum. CN neuron progenitors are produced in the mutant but there is an enhanced death of these neurons as shown by increased presence of caspase-3-positive cells. These data indicate that Pax6 regulates the survival of CN neuron progenitors. Furthermore, the analysis of experimental mouse chimeras suggests a cell-extrinsic role of Pax6 in CN neuron survival. For UBCs, using Tbr2 immunolabeling, these cells are significantly reduced in the Sey cerebellum. The loss of UBCs in the mutant is due partly to cell death in the RL and also to the reduced production of progenitors from the RL. These results demonstrate a critical role for Pax6 in regulating the generation and survival of UBCs. This and previous work from our laboratory demonstrate a seminal role of Pax6 in the development of all cerebellar glutamatergic neurons. SIGNIFICANCE STATEMENT Pax6 is a key molecule in development. Pax6 is best known as the master control gene in eye development with mutations causing aniridia in humans. Pax6 also plays important developmental roles in the cortex and olfactory bulb. During cerebellar development, Pax6 is robustly expressed in the germinal zone of all glutamatergic neurons [cerebellar nuclear (CN) neurons, granule cells, and unipolar brush cells (UBCs)]. Past work has not found abnormalities in the CN and UBC populations. Our study reveals that the Pax6-null mutation dramatically affects these cells and identifies Pax6 as a key regulator of cell survival in CN neurons and of cell production in UBCs. The present study shows how Pax6 is key to the development of glutamatergic cells in the cerebellum.
Collapse
|
313
|
Vihervaara A, Mahat DB, Guertin MJ, Chu T, Danko CG, Lis JT, Sistonen L. Transcriptional response to stress is pre-wired by promoter and enhancer architecture. Nat Commun 2017; 8:255. [PMID: 28811569 PMCID: PMC5557961 DOI: 10.1038/s41467-017-00151-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/05/2017] [Indexed: 12/29/2022] Open
Abstract
Programs of gene expression are executed by a battery of transcription factors that coordinate divergent transcription from a pair of tightly linked core initiation regions of promoters and enhancers. Here, to investigate how divergent transcription is reprogrammed upon stress, we measured nascent RNA synthesis at nucleotide-resolution, and profiled histone H4 acetylation in human cells. Our results globally show that the release of promoter-proximal paused RNA polymerase into elongation functions as a critical switch at which a gene's response to stress is determined. Highly transcribed and highly inducible genes display strong transcriptional directionality and selective assembly of general transcription factors on the core sense promoter. Heat-induced transcription at enhancers, instead, correlates with prior binding of cell-type, sequence-specific transcription factors. Activated Heat Shock Factor 1 (HSF1) binds to transcription-primed promoters and enhancers, and CTCF-occupied, non-transcribed chromatin. These results reveal chromatin architectural features that orient transcription at divergent regulatory elements and prime transcriptional responses genome-wide.Heat Shock Factor 1 (HSF1) is a regulator of stress-induced transcription. Here, the authors investigate changes to transcription and chromatin organization upon stress and find that activated HSF1 binds to transcription-primed promoters and enhancers, and to CTCF occupied, untranscribed chromatin.
Collapse
Affiliation(s)
- Anniina Vihervaara
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, 20520, Finland
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, 14853, USA
| | - Dig Bijay Mahat
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, 14853, USA
| | - Michael J Guertin
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Tinyi Chu
- Department of Biomedical Sciences, The Baker Institute for Animal Health, Cornell University, Ithaca, New York, 14853, USA
- Graduate Field of Computational Biology, Cornell University, Ithaca, New York, 14853, USA
| | - Charles G Danko
- Department of Biomedical Sciences, The Baker Institute for Animal Health, Cornell University, Ithaca, New York, 14853, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, 14853, USA.
| | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, 20520, Finland.
| |
Collapse
|
314
|
Rahman S, Zorca CE, Traboulsi T, Noutahi E, Krause MR, Mader S, Zenklusen D. Single-cell profiling reveals that eRNA accumulation at enhancer-promoter loops is not required to sustain transcription. Nucleic Acids Res 2017; 45:3017-3030. [PMID: 27932455 PMCID: PMC5389544 DOI: 10.1093/nar/gkw1220] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/23/2016] [Indexed: 02/06/2023] Open
Abstract
Enhancers are intergenic DNA elements that regulate the transcription of target genes in response to signaling pathways by interacting with promoters over large genomic distances. Recent studies have revealed that enhancers are bi-directionally transcribed into enhancer RNAs (eRNAs). Using single-molecule fluorescence in situ hybridization (smFISH), we investigated the eRNA-mediated regulation of transcription during estrogen induction in MCF-7 cells. We demonstrate that eRNAs are localized exclusively in the nucleus and are induced with similar kinetics as target mRNAs. However, eRNAs are mostly nascent at enhancers and their steady-state levels remain lower than those of their cognate mRNAs. Surprisingly, at the single-allele level, eRNAs are rarely co-expressed with their target loci, demonstrating that active gene transcription does not require the continuous transcription of eRNAs or their accumulation at enhancers. When co-expressed, sub-diffraction distance measurements between nascent mRNA and eRNA signals reveal that co-transcription of eRNAs and mRNAs rarely occurs within closed enhancer–promoter loops. Lastly, basal eRNA transcription at enhancers, but not E2-induced transcription, is maintained upon depletion of MLL1 and ERα, suggesting some degree of chromatin accessibility prior to signal-dependent activation of transcription. Together, our findings suggest that eRNA accumulation at enhancer–promoter loops is not required to sustain target gene transcription.
Collapse
Affiliation(s)
- Samir Rahman
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Cornelia E Zorca
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Tatiana Traboulsi
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada.,Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Emmanuel Noutahi
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Matthew R Krause
- Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Sylvie Mader
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada.,Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Daniel Zenklusen
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
315
|
Initial high-resolution microscopic mapping of active and inactive regulatory sequences proves non-random 3D arrangements in chromatin domain clusters. Epigenetics Chromatin 2017; 10:39. [PMID: 28784182 PMCID: PMC5547466 DOI: 10.1186/s13072-017-0146-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The association of active transcription regulatory elements (TREs) with DNAse I hypersensitivity (DHS[+]) and an 'open' local chromatin configuration has long been known. However, the 3D topography of TREs within the nuclear landscape of individual cells in relation to their active or inactive status has remained elusive. Here, we explored the 3D nuclear topography of active and inactive TREs in the context of a recently proposed model for a functionally defined nuclear architecture, where an active and an inactive nuclear compartment (ANC-INC) form two spatially co-aligned and functionally interacting networks. RESULTS Using 3D structured illumination microscopy, we performed 3D FISH with differently labeled DNA probe sets targeting either sites with DHS[+], apparently active TREs, or DHS[-] sites harboring inactive TREs. Using an in-house image analysis tool, DNA targets were quantitatively mapped on chromatin compaction shaped 3D nuclear landscapes. Our analyses present evidence for a radial 3D organization of chromatin domain clusters (CDCs) with layers of increasing chromatin compaction from the periphery to the CDC core. Segments harboring active TREs are significantly enriched at the decondensed periphery of CDCs with loops penetrating into interchromatin compartment channels, constituting the ANC. In contrast, segments lacking active TREs (DHS[-]) are enriched toward the compacted interior of CDCs (INC). CONCLUSIONS Our results add further evidence in support of the ANC-INC network model. The different 3D topographies of DHS[+] and DHS[-] sites suggest positional changes of TREs between the ANC and INC depending on their functional state, which might provide additional protection against an inappropriate activation. Our finding of a structural organization of CDCs based on radially arranged layers of different chromatin compaction levels indicates a complex higher-order chromatin organization beyond a dichotomic classification of chromatin into an 'open,' active and 'closed,' inactive state.
Collapse
|
316
|
Pan RY, Liu P, Zhou HT, Sun WX, Song J, Shu J, Cui GJ, Yang ZJ, Jia EZ. Circular RNAs promote TRPM3 expression by inhibiting hsa-miR-130a-3p in coronary artery disease patients. Oncotarget 2017; 8:60280-60290. [PMID: 28947970 PMCID: PMC5601138 DOI: 10.18632/oncotarget.19941] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022] Open
Abstract
We investigated the differential expression of circular RNAs (circRNAs) in plasma samples from three coronary artery disease (CAD) patients to identify putative therapeutic targets. We identified 24 differentially expressed circRNAs (18 up-regulated and 6 down-regulated) and 7 differentially expressed mRNAs (6 up-regulated and 1 down-regulated) in CAD patients based on competing endogenous RNA (ceRNA) microarray analysis. MiR-221(p = 0.001), miR-155(p = 0.049), and miR-130a (p = 0.001) were downregulated in CAD patients based on qRT-PCR analysis of another independent population of 932 study subjects (648 CAD subjects and 284 controls). We constructed a hsa-miR-130a-3p-mediated circRNA-mRNA ceRNA network using the miRanda database. This included 9 circRNAs (hsa_circ_0089378, hsa_circ_0083357, hsa_circ_0082824, hsa_circ_0068942, hsa_circ_0057576, hsa_circ_0054537, hsa_circ_0051172, hsa_circ_0032970, and hsa_circ_0006323) and 1 mRNA (transient receptor potential cation channel subfamily M member 3 [TRPM3]). We have shown that 9 circRNAs promote TRPM3 expression by inhibiting hsa-miR-130a-3p in CAD patients.
Collapse
Affiliation(s)
- Ren-You Pan
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Ping Liu
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Hai-Tang Zhou
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Wei-Xin Sun
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Jun Song
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Jiang Shu
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Guo-Jing Cui
- Department of Cardiovascular Medicine, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng 224000, Jiangsu Province, China
| | - Zhi-Jian Yang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - En-Zhi Jia
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
317
|
Gillinder KR, Tuckey H, Bell CC, Magor GW, Huang S, Ilsley MD, Perkins AC. Direct targets of pSTAT5 signalling in erythropoiesis. PLoS One 2017; 12:e0180922. [PMID: 28732065 PMCID: PMC5521770 DOI: 10.1371/journal.pone.0180922] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/25/2017] [Indexed: 12/29/2022] Open
Abstract
Erythropoietin (EPO) acts through the dimeric erythropoietin receptor to stimulate proliferation, survival, differentiation and enucleation of erythroid progenitor cells. We undertook two complimentary approaches to find EPO-dependent pSTAT5 target genes in murine erythroid cells: RNA-seq of newly transcribed (4sU-labelled) RNA, and ChIP-seq for pSTAT5 30 minutes after EPO stimulation. We found 302 pSTAT5-occupied sites: ~15% of these reside in promoters while the rest reside within intronic enhancers or intergenic regions, some >100kb from the nearest TSS. The majority of pSTAT5 peaks contain a central palindromic GAS element, TTCYXRGAA. There was significant enrichment for GATA motifs and CACCC-box motifs within the neighbourhood of pSTAT5-bound peaks, and GATA1 and/or KLF1 co-occupancy at many sites. Using 4sU-RNA-seq we determined the EPO-induced transcriptome and validated differentially expressed genes using dynamic CAGE data and qRT-PCR. We identified known direct pSTAT5 target genes such as Bcl2l1, Pim1 and Cish, and many new targets likely to be involved in driving erythroid cell differentiation including those involved in mRNA splicing (Rbm25), epigenetic regulation (Suv420h2), and EpoR turnover (Clint1/EpsinR). Some of these new EpoR-JAK2-pSTAT5 target genes could be used as biomarkers for monitoring disease activity in polycythaemia vera, and for monitoring responses to JAK inhibitors.
Collapse
Affiliation(s)
- Kevin R. Gillinder
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - Hugh Tuckey
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Charles C. Bell
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - Graham W. Magor
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - Stephen Huang
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Melissa D. Ilsley
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Andrew C. Perkins
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
318
|
Hutchins AP, Yang Z, Li Y, He F, Fu X, Wang X, Li D, Liu K, He J, Wang Y, Chen J, Esteban MA, Pei D. Models of global gene expression define major domains of cell type and tissue identity. Nucleic Acids Res 2017; 45:2354-2367. [PMID: 28426095 PMCID: PMC5389706 DOI: 10.1093/nar/gkx054] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/22/2017] [Indexed: 01/22/2023] Open
Abstract
The current classification of cells in an organism is largely based on their anatomic and developmental origin. Cells types and tissues are traditionally classified into those that arise from the three embryonic germ layers, the ectoderm, mesoderm and endoderm, but this model does not take into account the organization of cell type-specific patterns of gene expression. Here, we present computational models for cell type and tissue specification derived from a collection of 921 RNA-sequencing samples from 272 distinct mouse cell types or tissues. In an unbiased fashion, this analysis accurately predicts the three known germ layers. Unexpectedly, this analysis also suggests that in total there are eight major domains of cell type-specification, corresponding to the neurectoderm, neural crest, surface ectoderm, endoderm, mesoderm, blood mesoderm, germ cells and the embryonic domain. Further, we identify putative genes responsible for specifying the domain and the cell type. This model has implications for understanding trans-lineage differentiation for stem cells, developmental cell biology and regenerative medicine.
Collapse
Affiliation(s)
- Andrew P Hutchins
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China.,Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Zhongzhou Yang
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yuhao Li
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Fangfang He
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Xiuling Fu
- Department of Biology, Southern University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Xiaoshan Wang
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Dongwei Li
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Kairong Liu
- Academy of Mathematics and Systems Science, National Center for Mathematics and Interdisciplinary Sciences, Chinese Academy of Sciences, Beijing 100080, China.,Beihang University, Beijing 100191, China
| | - Jiangping He
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Yong Wang
- Academy of Mathematics and Systems Science, National Center for Mathematics and Interdisciplinary Sciences, Chinese Academy of Sciences, Beijing 100080, China
| | - Jiekai Chen
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Miguel A Esteban
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China.,Laboratory of RNA, Chromatin and Human disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| |
Collapse
|
319
|
Seixas AI, Loureiro JR, Costa C, Ordóñez-Ugalde A, Marcelino H, Oliveira CL, Loureiro JL, Dhingra A, Brandão E, Cruz VT, Timóteo A, Quintáns B, Rouleau GA, Rizzu P, Carracedo Á, Bessa J, Heutink P, Sequeiros J, Sobrido MJ, Coutinho P, Silveira I. A Pentanucleotide ATTTC Repeat Insertion in the Non-coding Region of DAB1, Mapping to SCA37, Causes Spinocerebellar Ataxia. Am J Hum Genet 2017; 101:87-103. [PMID: 28686858 DOI: 10.1016/j.ajhg.2017.06.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/13/2017] [Indexed: 01/01/2023] Open
Abstract
Advances in human genetics in recent years have largely been driven by next-generation sequencing (NGS); however, the discovery of disease-related gene mutations has been biased toward the exome because the large and very repetitive regions that characterize the non-coding genome remain difficult to reach by that technology. For autosomal-dominant spinocerebellar ataxias (SCAs), 28 genes have been identified, but only five SCAs originate from non-coding mutations. Over half of SCA-affected families, however, remain without a genetic diagnosis. We used genome-wide linkage analysis, NGS, and repeat analysis to identify an (ATTTC)n insertion in a polymorphic ATTTT repeat in DAB1 in chromosomal region 1p32.2 as the cause of autosomal-dominant SCA; this region has been previously linked to SCA37. The non-pathogenic and pathogenic alleles have the configurations [(ATTTT)7-400] and [(ATTTT)60-79(ATTTC)31-75(ATTTT)58-90], respectively. (ATTTC)n insertions are present on a distinct haplotype and show an inverse correlation between size and age of onset. In the DAB1-oriented strand, (ATTTC)n is located in 5' UTR introns of cerebellar-specific transcripts arising mostly during human fetal brain development from the usage of alternative promoters, but it is maintained in the adult cerebellum. Overexpression of the transfected (ATTTC)58 insertion, but not (ATTTT)n, leads to abnormal nuclear RNA accumulation. Zebrafish embryos injected with RNA of the (AUUUC)58 insertion, but not (AUUUU)n, showed lethal developmental malformations. Together, these results establish an unstable repeat insertion in DAB1 as a cause of cerebellar degeneration; on the basis of the genetic and phenotypic evidence, we propose this mutation as the molecular basis for SCA37.
Collapse
|
320
|
Robson MI, de Las Heras JI, Czapiewski R, Sivakumar A, Kerr ARW, Schirmer EC. Constrained release of lamina-associated enhancers and genes from the nuclear envelope during T-cell activation facilitates their association in chromosome compartments. Genome Res 2017; 27:1126-1138. [PMID: 28424353 PMCID: PMC5495065 DOI: 10.1101/gr.212308.116] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 04/12/2017] [Indexed: 01/15/2023]
Abstract
The 3D organization of the genome changes concomitantly with expression changes during hematopoiesis and immune activation. Studies have focused either on lamina-associated domains (LADs) or on topologically associated domains (TADs), defined by preferential local chromatin interactions, and chromosome compartments, defined as higher-order interactions between TADs sharing functionally similar states. However, few studies have investigated how these affect one another. To address this, we mapped LADs using Lamin B1-DamID during Jurkat T-cell activation, finding significant genome reorganization at the nuclear periphery dominated by release of loci frequently important for T-cell function. To assess how these changes at the nuclear periphery influence wider genome organization, our DamID data sets were contrasted with TADs and compartments. Features of specific repositioning events were then tested by fluorescence in situ hybridization during T-cell activation. First, considerable overlap between TADs and LADs was observed with the TAD repositioning as a unit. Second, A1 and A2 subcompartments are segregated in 3D space through differences in proximity to LADs along chromosomes. Third, genes and a putative enhancer in LADs that were released from the periphery during T-cell activation became preferentially associated with A2 subcompartments and were constrained to the relative proximity of the lamina. Thus, lamina associations influence internal nuclear organization, and changes in LADs during T-cell activation may provide an important additional mode of gene regulation.
Collapse
Affiliation(s)
- Michael I Robson
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Jose I de Las Heras
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Rafal Czapiewski
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Aishwarya Sivakumar
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Alastair R W Kerr
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Eric C Schirmer
- The Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| |
Collapse
|
321
|
Ilsley MD, Gillinder KR, Magor GW, Huang S, Bailey TL, Crossley M, Perkins AC. Krüppel-like factors compete for promoters and enhancers to fine-tune transcription. Nucleic Acids Res 2017; 45:6572-6588. [PMID: 28541545 PMCID: PMC5499887 DOI: 10.1093/nar/gkx441] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
Krüppel-like factors (KLFs) are a family of 17 transcription factors characterized by a conserved DNA-binding domain of three zinc fingers and a variable N-terminal domain responsible for recruiting cofactors. KLFs have diverse functions in stem cell biology, embryo patterning, and tissue homoeostasis. KLF1 and related family members function as transcriptional activators via recruitment of co-activators such as EP300, whereas KLF3 and related members act as transcriptional repressors via recruitment of C-terminal Binding Proteins. KLF1 directly activates the Klf3 gene via an erythroid-specific promoter. Herein, we show KLF1 and KLF3 bind common as well as unique sites within the erythroid cell genome by ChIP-seq. We show KLF3 can displace KLF1 from key erythroid gene promoters and enhancers in vivo. Using 4sU RNA labelling and RNA-seq, we show this competition results in reciprocal transcriptional outputs for >50 important genes. Furthermore, Klf3-/- mice displayed exaggerated recovery from anemic stress and persistent cell cycling consistent with a role for KLF3 in dampening KLF1-driven proliferation. We suggest this study provides a paradigm for how KLFs work in incoherent feed-forward loops or networks to fine-tune transcription and thereby control diverse biological processes such as cell proliferation.
Collapse
Affiliation(s)
- Melissa D. Ilsley
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
| | - Kevin R. Gillinder
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
| | - Graham W. Magor
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
| | - Stephen Huang
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
| | | | | | - Andrew C. Perkins
- Mater Research Institute, Translational Research Institute, University of Queensland, Brisbane 4102, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane 4072, Australia
- The Princess Alexandra Hospital, Brisbane 4102, Australia
| |
Collapse
|
322
|
Chromatin-enriched lncRNAs can act as cell-type specific activators of proximal gene transcription. Nat Struct Mol Biol 2017. [PMID: 28628087 PMCID: PMC5682930 DOI: 10.1038/nsmb.3424] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We recently described a new class of long noncoding RNA defined by especially tight chromatin association, whose presence is strongly correlated with expression of nearby genes in HEK293 cells. Here we critically examine the generality and cis-enhancer mechanism of this class of chromatin enriched RNA (cheRNA). CheRNA are largely cell-type specific, and remain the most effective chromatin signature for predicting cis-gene transcription in all cell types examined. Targeted depletion of three cheRNAs decreases gene expression of their neighbors, indicating potential co-activator function. Single-molecule FISH of one cheRNA-distal target gene pair suggests spatial overlap consistent with a role in chromosome looping. In another example, the cheRNA HIDALGO stimulates the fetal hemoglobin HBG1 gene during erythroid differentiation by promoting contacts to a downstream enhancer. Our results suggest that many cheRNAs activate proximal, lineage-specific gene transcription.
Collapse
|
323
|
Relatively frequent switching of transcription start sites during cerebellar development. BMC Genomics 2017; 18:461. [PMID: 28610618 PMCID: PMC5470264 DOI: 10.1186/s12864-017-3834-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/31/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alternative transcription start site (TSS) usage plays important roles in transcriptional control of mammalian gene expression. The growing interest in alternative TSSs and their role in genome diversification spawned many single-gene studies on differential usages of tissue-specific or temporal-specific alternative TSSs. However, exploration of the switching usage of alternative TSS usage on a genomic level, especially in the central nervous system, is largely lacking. RESULTS In this study, We have prepared a unique set of time-course data for the developing cerebellum, as part of the FANTOM5 consortium ( http://fantom.gsc.riken.jp/5/ ) that uses their innovative capturing of 5' ends of all transcripts followed by Helicos next generation sequencing. We analyzed the usage of all transcription start sites (TSSs) at each time point during cerebellar development that provided information on multiple RNA isoforms that emerged from the same gene. We developed a mathematical method that systematically compares the expression of different TSSs of a gene to identify temporal crossover and non-crossover switching events. We identified 48,489 novel TSS switching events in 5433 genes during cerebellar development. This includes 9767 crossover TSS switching events in 1511 genes, where the dominant TSS shifts over time. CONCLUSIONS We observed a relatively high prevalence of TSS switching in cerebellar development where the resulting temporally-specific gene transcripts and protein products can play important regulatory and functional roles.
Collapse
|
324
|
Sawyer IA, Dundr M. Chromatin loops and causality loops: the influence of RNA upon spatial nuclear architecture. Chromosoma 2017; 126:541-557. [PMID: 28593374 DOI: 10.1007/s00412-017-0632-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/17/2017] [Accepted: 05/23/2017] [Indexed: 01/18/2023]
Abstract
An intrinsic and essential trait exhibited by cells is the properly coordinated and integrated regulation of an astoundingly large number of simultaneous molecular decisions and reactions to maintain biochemical homeostasis. This is especially true inside the cell nucleus, where the recognition of DNA and RNA by a vast range of nucleic acid-interacting proteins organizes gene expression patterns. However, this dynamic system is not regulated by simple "on" or "off" signals. Instead, transcription factor and RNA polymerase recruitment to DNA are influenced by the local chromatin and epigenetic environment, a gene's relative position within the nucleus and the action of noncoding RNAs. In addition, major phase-separated structural features of the nucleus, such as nucleoli and paraspeckles, assemble in direct response to specific transcriptional activities and, in turn, influence global genomic function. Currently, the interpretation of these data is trapped in a causality dilemma reminiscent of the "chicken and the egg" paradox as it is unclear whether changes in nuclear architecture promote RNA function or vice versa. Here, we review recent advances that suggest a complex and interdependent interaction network between gene expression, chromatin topology, and noncoding RNA function. We also discuss the functional links between these essential nuclear processes from the nanoscale (gene looping) to the macroscale (sub-nuclear gene positioning and nuclear body function) and briefly highlight some of the challenges that researchers may encounter when studying these phenomena.
Collapse
Affiliation(s)
- Iain A Sawyer
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Miroslav Dundr
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
325
|
Yeung J, Goldowitz D. Wls expression in the rhombic lip orchestrates the embryonic development of the mouse cerebellum. Neuroscience 2017; 354:30-42. [DOI: 10.1016/j.neuroscience.2017.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/17/2017] [Accepted: 04/14/2017] [Indexed: 01/19/2023]
|
326
|
Gallone G, Haerty W, Disanto G, Ramagopalan SV, Ponting CP, Berlanga-Taylor AJ. Identification of genetic variants affecting vitamin D receptor binding and associations with autoimmune disease. Hum Mol Genet 2017; 26:2164-2176. [PMID: 28335003 PMCID: PMC5886188 DOI: 10.1093/hmg/ddx092] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 01/24/2023] Open
Abstract
Large numbers of statistically significant associations between sentinel SNPs and case-control status have been replicated by genome-wide association studies. Nevertheless, few underlying molecular mechanisms of complex disease are currently known. We investigated whether variation in binding of a transcription factor, the vitamin D receptor (VDR), whose activating ligand vitamin D has been proposed as a modifiable factor in multiple disorders, could explain any of these associations. VDR modifies gene expression by binding DNA as a heterodimer with the Retinoid X receptor (RXR). We identified 43,332 genetic variants significantly associated with altered VDR binding affinity (VDR-BVs) using a high-resolution (ChIP-exo) genome-wide analysis of 27 HapMap lymphoblastoid cell lines. VDR-BVs are enriched in consensus RXR::VDR binding motifs, yet most fell outside of these motifs, implying that genetic variation often affects the binding affinity only indirectly. Finally, we compared 341 VDR-BVs replicating by position in multiple individuals against background sets of variants lying within VDR-binding regions that had been matched in allele frequency and were independent with respect to linkage disequilibrium. In this stringent test, these replicated VDR-BVs were significantly (q < 0.1) and substantially (>2-fold) enriched in genomic intervals associated with autoimmune and other diseases, including inflammatory bowel disease, Crohn's disease and rheumatoid arthritis. The approach's validity is underscored by RXR::VDR motif sequence being predictive of binding strength and being evolutionarily constrained. Our findings are consistent with altered RXR::VDR binding contributing to immunity-related diseases. Replicated VDR-BVs associated with these disorders could represent causal disease risk alleles whose effect may be modifiable by vitamin D levels.
Collapse
Affiliation(s)
- Giuseppe Gallone
- MRC Functional Genomics Unit
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Wilfried Haerty
- MRC Functional Genomics Unit
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Giulio Disanto
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | | | - Chris P. Ponting
- MRC Functional Genomics Unit
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
- MRC Human Genetics Unit, The Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Antonio J. Berlanga-Taylor
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7BN, UK
- CGAT, MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- MRC-PHE Centre for Environment and Health, Department of Epidemiology & Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, St Mary’s Campus, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
327
|
Seemann SE, Mirza AH, Hansen C, Bang-Berthelsen CH, Garde C, Christensen-Dalsgaard M, Torarinsson E, Yao Z, Workman CT, Pociot F, Nielsen H, Tommerup N, Ruzzo WL, Gorodkin J. The identification and functional annotation of RNA structures conserved in vertebrates. Genome Res 2017; 27:1371-1383. [PMID: 28487280 PMCID: PMC5538553 DOI: 10.1101/gr.208652.116] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/04/2017] [Indexed: 01/15/2023]
Abstract
Structured elements of RNA molecules are essential in, e.g., RNA stabilization, localization, and protein interaction, and their conservation across species suggests a common functional role. We computationally screened vertebrate genomes for conserved RNA structures (CRSs), leveraging structure-based, rather than sequence-based, alignments. After careful correction for sequence identity and GC content, we predict ∼516,000 human genomic regions containing CRSs. We find that a substantial fraction of human–mouse CRS regions (1) colocalize consistently with binding sites of the same RNA binding proteins (RBPs) or (2) are transcribed in corresponding tissues. Additionally, a CaptureSeq experiment revealed expression of many of our CRS regions in human fetal brain, including 662 novel ones. For selected human and mouse candidate pairs, qRT-PCR and in vitro RNA structure probing supported both shared expression and shared structure despite low abundance and low sequence identity. About 30,000 CRS regions are located near coding or long noncoding RNA genes or within enhancers. Structured (CRS overlapping) enhancer RNAs and extended 3′ ends have significantly increased expression levels over their nonstructured counterparts. Our findings of transcribed uncharacterized regulatory regions that contain CRSs support their RNA-mediated functionality.
Collapse
Affiliation(s)
- Stefan E Seemann
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Aashiq H Mirza
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Claus Hansen
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Cellular and Molecular Medicine (ICMM), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Claus H Bang-Berthelsen
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Obesity Biology and Department of Molecular Genetics, Novo Nordisk A/S, DK-2880 Bagsværd, Denmark
| | - Christian Garde
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Mikkel Christensen-Dalsgaard
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Cellular and Molecular Medicine (ICMM), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Elfar Torarinsson
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Christopher T Workman
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Flemming Pociot
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev University Hospital, DK-2730 Herlev, Denmark
| | - Henrik Nielsen
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Cellular and Molecular Medicine (ICMM), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Niels Tommerup
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Cellular and Molecular Medicine (ICMM), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Walter L Ruzzo
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,School of Computer Science and Engineering and Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA.,Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Jan Gorodkin
- Center for non-coding RNA in Technology and Health (RTH), University of Copenhagen, DK-1870 Frederiksberg, Denmark.,Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| |
Collapse
|
328
|
Trinh LA, Chong-Morrison V, Gavriouchkina D, Hochgreb-Hägele T, Senanayake U, Fraser SE, Sauka-Spengler T. Biotagging of Specific Cell Populations in Zebrafish Reveals Gene Regulatory Logic Encoded in the Nuclear Transcriptome. Cell Rep 2017; 19:425-440. [PMID: 28402863 PMCID: PMC5400779 DOI: 10.1016/j.celrep.2017.03.045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 12/21/2016] [Accepted: 03/13/2017] [Indexed: 11/22/2022] Open
Abstract
Interrogation of gene regulatory circuits in complex organisms requires precise tools for the selection of individual cell types and robust methods for biochemical profiling of target proteins. We have developed a versatile, tissue-specific binary in vivo biotinylation system in zebrafish termed biotagging that uses genetically encoded components to biotinylate target proteins, enabling in-depth genome-wide analyses of their molecular interactions. Using tissue-specific drivers and cell-compartment-specific effector lines, we demonstrate the specificity of the biotagging toolkit at the biochemical, cellular, and transcriptional levels. We use biotagging to characterize the in vivo transcriptional landscape of migratory neural crest and myocardial cells in different cellular compartments (ribosomes and nucleus). These analyses reveal a comprehensive network of coding and non-coding RNAs and cis-regulatory modules, demonstrating that tissue-specific identity is embedded in the nuclear transcriptomes. By eliminating background inherent to complex embryonic environments, biotagging allows analyses of molecular interactions at high resolution. Biotagging enables cell- and compartment-specific in vivo biotinylation in zebrafish Technique yields comprehensive nuclear transcriptional analysis of cardiomyocytes Biotagging finds bidirectionally transcribed neural crest cis-regulatory modules System reveals tissue-specific regulation of noncoding RNA species
Collapse
Affiliation(s)
- Le A Trinh
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Vanessa Chong-Morrison
- Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Daria Gavriouchkina
- Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Tatiana Hochgreb-Hägele
- Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Upeka Senanayake
- Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Scott E Fraser
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Tatjana Sauka-Spengler
- Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
329
|
Enhancers and chromatin structures: regulatory hubs in gene expression and diseases. Biosci Rep 2017; 37:BSR20160183. [PMID: 28351896 PMCID: PMC5408663 DOI: 10.1042/bsr20160183] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/23/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Gene expression requires successful communication between enhancer and promoter regions, whose activities are regulated by a variety of factors and associated with distinct chromatin structures; in addition, functionally related genes and their regulatory repertoire tend to be arranged in the same subchromosomal regulatory domains. In this review, we discuss the importance of enhancers, especially clusters of enhancers (such as super-enhancers), as key regulatory hubs to integrate environmental cues and encode spatiotemporal instructions for genome expression, which are critical for a variety of biological processes governing mammalian development. Furthermore, we emphasize that the enhancer–promoter interaction landscape provides a critical context to understand the aetiologies and mechanisms behind numerous complex human diseases and provides new avenues for effective transcription-based interventions.
Collapse
|
330
|
Bornholdt J, Saber AT, Lilje B, Boyd M, Jørgensen M, Chen Y, Vitezic M, Jacobsen NR, Poulsen SS, Berthing T, Bressendorff S, Vitting-Seerup K, Andersson R, Hougaard KS, Yauk CL, Halappanavar S, Wallin H, Vogel U, Sandelin A. Identification of Gene Transcription Start Sites and Enhancers Responding to Pulmonary Carbon Nanotube Exposure in Vivo. ACS NANO 2017; 11:3597-3613. [PMID: 28345861 DOI: 10.1021/acsnano.6b07533] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Increased use of nanomaterials in industry, medicine, and consumer products has raised concerns over their toxicity. To ensure safe use of nanomaterials, understanding their biological effects at the molecular level is crucial. In particular, the regulatory mechanisms responsible for the cascade of genes activated by nanomaterial exposure are not well-characterized. To this end, we profiled the genome-wide usage of gene transcription start sites and linked active enhancer regions in lungs of C57BL/6 mice 24 h after intratracheal instillation of a single dose of the multiwalled carbon nanotube (MWCNT) Mitsui-7. Our results revealed a massive gene regulatory response, where expression of key inflammatory genes (e.g., Csf3, Il24, and Fgf23) was increased >100-fold 24 h after Mitsui-7 exposure. Many of the Mitsui-7-responsive transcription start sites were alternative transcription start sites for known genes, and the number of alternative transcription start sites used in a given gene was correlated with overall Mitsui-7 response. Strikingly, genes that were up-regulated after Mitsui-7 exposure only through their main annotated transcription start site were linked to inflammatory and defense responses, while genes up-regulated only through alternative transcription start sites were functionally heterogeneous and not inflammation-associated. Furthermore, we identified almost 12 000 active enhancers, many of which were Mitsui-7-responsive, and we identified similarly responding putative target genes. Overall, our study provides the location and activity of Mitsui-7-induced enhancers and transcription start sites, providing a useful resource for targeted experiments elucidating the biological effects of nanomaterials and the identification of biomarkers for early detection of MWCNT-induced inflammation.
Collapse
Affiliation(s)
- Jette Bornholdt
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | | | - Berit Lilje
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Mette Boyd
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Mette Jørgensen
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Yun Chen
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Morana Vitezic
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | | | - Sarah Søs Poulsen
- National Research Centre for the Working Environment , 2100 Copenhagen, Denmark
| | - Trine Berthing
- National Research Centre for the Working Environment , 2100 Copenhagen, Denmark
| | - Simon Bressendorff
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
| | - Kristoffer Vitting-Seerup
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Robin Andersson
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
| | | | - Carole L Yauk
- Environmental and Radiation Health Sciences Directorate, Health Canada , Ottawa, Ontario K1A 0K9, Canada
| | - Sabina Halappanavar
- Environmental and Radiation Health Sciences Directorate, Health Canada , Ottawa, Ontario K1A 0K9, Canada
| | - Håkan Wallin
- National Research Centre for the Working Environment , 2100 Copenhagen, Denmark
- Department of Public Health, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment , 2100 Copenhagen, Denmark
- Department of Micro and Nanotechnology, Technical University of Denmark , 2800 Kongens Lyngby, Denmark
| | - Albin Sandelin
- The Bioinformatics Centre, Department of Biology University of Copenhagen , 2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen , 2200 Copenhagen, Denmark
| |
Collapse
|
331
|
Takamochi K, Mogushi K, Kawaji H, Imashimizu K, Fukui M, Oh S, Itoh M, Hayashizaki Y, Ko W, Akeboshi M, Suzuki K. Correlation of EGFR or KRAS mutation status with 18F-FDG uptake on PET-CT scan in lung adenocarcinoma. PLoS One 2017; 12:e0175622. [PMID: 28422979 PMCID: PMC5396974 DOI: 10.1371/journal.pone.0175622] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/27/2017] [Indexed: 12/02/2022] Open
Abstract
Background 18F-fluoro-2-deoxy-glucose (18F-FDG) positron emission tomography (PET) is a functional imaging modality based on glucose metabolism. The correlation between EGFR or KRAS mutation status and the standardized uptake value (SUV) of 18F-FDG PET scanning has not been fully elucidated. Methods Correlations between EGFR or KRAS mutation status and clinicopathological factors including SUVmax were statistically analyzed in 734 surgically resected lung adenocarcinoma patients. Molecular causal relationships between EGFR or KRAS mutation status and glucose metabolism were then elucidated in 62 lung adenocarcinomas using cap analysis of gene expression (CAGE), a method to determine and quantify the transcription initiation activities of mRNA across the genome. Results EGFR and KRAS mutations were detected in 334 (46%) and 83 (11%) of the 734 lung adenocarcinomas, respectively. The remaining 317 (43%) patients had wild-type tumors for both genes. EGFR mutations were more frequent in tumors with lower SUVmax. In contrast, no relationship was noted between KRAS mutation status and SUVmax. CAGE revealed that 4 genes associated with glucose metabolism (GPI, G6PD, PKM2, and GAPDH) and 5 associated with the cell cycle (ANLN, PTTG1, CIT, KPNA2, and CDC25A) were positively correlated with SUVmax, although expression levels were lower in EGFR-mutated than in wild-type tumors. No similar relationships were noted with KRAS mutations. Conclusions EGFR-mutated adenocarcinomas are biologically indolent with potentially lower levels of glucose metabolism than wild-type tumors. Several genes associated with glucose metabolism and the cell cycle were specifically down-regulated in EGFR-mutated adenocarcinomas.
Collapse
Affiliation(s)
- Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| | - Kaoru Mogushi
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Hideya Kawaji
- Preventive Medicine and Applied Genomics Unit, RIKEN Advanced Center for Computing and Communication, Yokohama, Kanagawa, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama, Japan
| | - Kota Imashimizu
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Mariko Fukui
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Shiaki Oh
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Masayoshi Itoh
- Preventive Medicine and Applied Genomics Unit, RIKEN Advanced Center for Computing and Communication, Yokohama, Kanagawa, Japan
| | - Yoshihide Hayashizaki
- Preventive Medicine and Applied Genomics Unit, RIKEN Advanced Center for Computing and Communication, Yokohama, Kanagawa, Japan
| | - Weijey Ko
- Diagnostic Imaging Center, Yotsuya Medical Cube, Tokyo, Japan
| | - Masao Akeboshi
- Diagnostic Imaging Center, Yotsuya Medical Cube, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
332
|
Sengupta S, George RE. Super-Enhancer-Driven Transcriptional Dependencies in Cancer. Trends Cancer 2017; 3:269-281. [PMID: 28718439 DOI: 10.1016/j.trecan.2017.03.006] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 11/24/2022]
Abstract
Transcriptional deregulation is one of the core tenets of cancer biology and is underpinned by alterations in both protein-coding genes and noncoding regulatory elements. Large regulatory elements, so-called super-enhancers (SEs), are central to the maintenance of cancer cell identity and promote oncogenic transcription to which cancer cells become highly addicted. Such dependence on SE-driven transcription for proliferation and survival offers an Achilles heel for the therapeutic targeting of cancer cells. Indeed, inhibition of the cellular machinery required for the assembly and maintenance of SEs dampens oncogenic transcription and inhibits tumor growth. In this article, we review the organization, function, and regulation of oncogenic SEs and their contribution to the cancer cell state.
Collapse
Affiliation(s)
- Satyaki Sengupta
- Department of Pediatric Hematology and Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Rani E George
- Department of Pediatric Hematology and Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
333
|
Pradeepa MM, McKenna F, Taylor GCA, Bengani H, Grimes GR, Wood AJ, Bhatia S, Bickmore WA. Psip1/p52 regulates posterior Hoxa genes through activation of lncRNA Hottip. PLoS Genet 2017; 13:e1006677. [PMID: 28384324 PMCID: PMC5383017 DOI: 10.1371/journal.pgen.1006677] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/09/2017] [Indexed: 02/07/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been implicated in various biological functions including the regulation of gene expression, however, the functionality of lncRNAs is not clearly understood and conflicting conclusions have often been reached when comparing different methods to investigate them. Moreover, little is known about the upstream regulation of lncRNAs. Here we show that the short isoform (p52) of a transcriptional co-activator-PC4 and SF2 interacting protein (Psip1), which is known to be involved in linking transcription to RNA processing, specifically regulates the expression of the lncRNA Hottip-located at the 5' end of the Hoxa locus. Using both knockdown and knockout approaches we show that Hottip expression is required for activation of the 5' Hoxa genes (Hoxa13 and Hoxa10/11) and for retaining Mll1 at the 5' end of Hoxa. Moreover, we demonstrate that artificially inducing Hottip expression is sufficient to activate the 5' Hoxa genes and that Hottip RNA binds to the 5' end of Hoxa. By engineering premature transcription termination, we show that it is the Hottip lncRNA molecule itself, not just Hottip transcription that is required to maintains active expression of posterior Hox genes. Our data show a direct role for a lncRNA molecule in regulating the expression of developmentally-regulated mRNA genes in cis.
Collapse
Affiliation(s)
- Madapura M. Pradeepa
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
- School of biological sciences, University of Essex, Colchester, United Kingdom
| | - Fionnuala McKenna
- School of biological sciences, University of Essex, Colchester, United Kingdom
| | - Gillian C. A. Taylor
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
| | - Hemant Bengani
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
| | - Graeme R. Grimes
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J. Wood
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
| | - Shipra Bhatia
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
| | - Wendy A. Bickmore
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
334
|
Frank-Bertoncelj M, Klein K, Gay S. Interplay between genetic and epigenetic mechanisms in rheumatoid arthritis. Epigenomics 2017; 9:493-504. [PMID: 28322583 DOI: 10.2217/epi-2016-0142] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Genetic and environmental factors contribute to the risk for rheumatoid arthritis (RA), with epigenetics serving as a possible interface through which risk factors contribute to RA. High-throughput technologies for interrogating genome and epigenome, and the availability of genetic and epigenetic datasets across a diversity of cell types, enable the identification of candidate causal genetic variants for RA to study their function in core RA processes. To date, RA risk variants were studied in the immune cells but not joint resident cells, for example, synovial fibroblasts. Synovial fibroblasts from different joints are distinct, anatomically specialized cells, defined by joint-specific transcriptomes, epigenomes and phenotypes. Cell type-specific analysis of epigenetic changes, together with genetic fine mapping and interrogation of chromatin 3D interactions may identify new disease relevant pathways, potential therapeutic targets and biomarkers for RA progression or therapy response.
Collapse
Affiliation(s)
| | - Kerstin Klein
- Center of Experimental Rheumatology, University Hospital Zurich, Switzerland
| | - Steffen Gay
- Center of Experimental Rheumatology, University Hospital Zurich, Switzerland
| |
Collapse
|
335
|
Michel M, Demel C, Zacher B, Schwalb B, Krebs S, Blum H, Gagneur J, Cramer P. TT-seq captures enhancer landscapes immediately after T-cell stimulation. Mol Syst Biol 2017; 13:920. [PMID: 28270558 PMCID: PMC5371733 DOI: 10.15252/msb.20167507] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
To monitor transcriptional regulation in human cells, rapid changes in enhancer and promoter activity must be captured with high sensitivity and temporal resolution. Here, we show that the recently established protocol TT-seq ("transient transcriptome sequencing") can monitor rapid changes in transcription from enhancers and promoters during the immediate response of T cells to ionomycin and phorbol 12-myristate 13-acetate (PMA). TT-seq maps eRNAs and mRNAs every 5 min after T-cell stimulation with high sensitivity and identifies many new primary response genes. TT-seq reveals that the synthesis of 1,601 eRNAs and 650 mRNAs changes significantly within only 15 min after stimulation, when standard RNA-seq does not detect differentially expressed genes. Transcription of enhancers that are primed for activation by nucleosome depletion can occur immediately and simultaneously with transcription of target gene promoters. Our results indicate that enhancer transcription is a good proxy for enhancer regulatory activity in target gene activation, and establish TT-seq as a tool for monitoring the dynamics of enhancer landscapes and transcription programs during cellular responses and differentiation.
Collapse
Affiliation(s)
- Margaux Michel
- Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Carina Demel
- Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Benedikt Zacher
- Gene Center Munich, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Björn Schwalb
- Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Stefan Krebs
- Gene Center Munich, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Helmut Blum
- Gene Center Munich, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julien Gagneur
- Department of Informatics, Technische Universität München, Garching, Germany
| | - Patrick Cramer
- Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
336
|
Analysis of the human monocyte-derived macrophage transcriptome and response to lipopolysaccharide provides new insights into genetic aetiology of inflammatory bowel disease. PLoS Genet 2017; 13:e1006641. [PMID: 28263993 PMCID: PMC5358891 DOI: 10.1371/journal.pgen.1006641] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 03/20/2017] [Accepted: 02/17/2017] [Indexed: 12/15/2022] Open
Abstract
The FANTOM5 consortium utilised cap analysis of gene expression (CAGE) to provide an unprecedented insight into transcriptional regulation in human cells and tissues. In the current study, we have used CAGE-based transcriptional profiling on an extended dense time course of the response of human monocyte-derived macrophages grown in macrophage colony-stimulating factor (CSF1) to bacterial lipopolysaccharide (LPS). We propose that this system provides a model for the differentiation and adaptation of monocytes entering the intestinal lamina propria. The response to LPS is shown to be a cascade of successive waves of transient gene expression extending over at least 48 hours, with hundreds of positive and negative regulatory loops. Promoter analysis using motif activity response analysis (MARA) identified some of the transcription factors likely to be responsible for the temporal profile of transcriptional activation. Each LPS-inducible locus was associated with multiple inducible enhancers, and in each case, transient eRNA transcription at multiple sites detected by CAGE preceded the appearance of promoter-associated transcripts. LPS-inducible long non-coding RNAs were commonly associated with clusters of inducible enhancers. We used these data to re-examine the hundreds of loci associated with susceptibility to inflammatory bowel disease (IBD) in genome-wide association studies. Loci associated with IBD were strongly and specifically (relative to rheumatoid arthritis and unrelated traits) enriched for promoters that were regulated in monocyte differentiation or activation. Amongst previously-identified IBD susceptibility loci, the vast majority contained at least one promoter that was regulated in CSF1-dependent monocyte-macrophage transitions and/or in response to LPS. On this basis, we concluded that IBD loci are strongly-enriched for monocyte-specific genes, and identified at least 134 additional candidate genes associated with IBD susceptibility from reanalysis of published GWA studies. We propose that dysregulation of monocyte adaptation to the environment of the gastrointestinal mucosa is the key process leading to inflammatory bowel disease. Macrophages are immune cells that form the first line of defense against pathogens, but also mediate tissue damage in inflammatory disease. Macrophages initiate inflammation by recognising and responding to components of bacterial cells. Macrophages of the wall of the gut are constantly replenished from the blood. Upon entering the intestine, newly-arrived cells modulate their response to stimuli derived from the bacteria in the wall of the gut. This process fails in chronic inflammatory bowel diseases (IBD). Both the major forms of IBD, Crohn’s disease and ulcerative colitis, run in families. The inheritance is complex, involving more than 200 different regions of the genome. We hypothesised that the genetic risk of IBD is associated specifically with altered regulation of genes that control the development of macrophages. In this study, we used the comprehensive transcriptome dataset produced by the FANTOM5 consortium to identify the sets of promoters and enhancers that are involved in adaptation of macrophages to the gut wall, their response to bacterial stimuli, and how their functions are integrated. A reanalysis of published genome-wide association data based upon regulated genes in monocytes as candidates strongly supports the view that susceptibility to IBD arises from a primary defect in macrophage differentiation.
Collapse
|
337
|
An atlas of human long non-coding RNAs with accurate 5' ends. Nature 2017; 543:199-204. [PMID: 28241135 DOI: 10.1038/nature21374] [Citation(s) in RCA: 726] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/08/2017] [Indexed: 12/15/2022]
Abstract
Long non-coding RNAs (lncRNAs) are largely heterogeneous and functionally uncharacterized. Here, using FANTOM5 cap analysis of gene expression (CAGE) data, we integrate multiple transcript collections to generate a comprehensive atlas of 27,919 human lncRNA genes with high-confidence 5' ends and expression profiles across 1,829 samples from the major human primary cell types and tissues. Genomic and epigenomic classification of these lncRNAs reveals that most intergenic lncRNAs originate from enhancers rather than from promoters. Incorporating genetic and expression data, we show that lncRNAs overlapping trait-associated single nucleotide polymorphisms are specifically expressed in cell types relevant to the traits, implicating these lncRNAs in multiple diseases. We further demonstrate that lncRNAs overlapping expression quantitative trait loci (eQTL)-associated single nucleotide polymorphisms of messenger RNAs are co-expressed with the corresponding messenger RNAs, suggesting their potential roles in transcriptional regulation. Combining these findings with conservation data, we identify 19,175 potentially functional lncRNAs in the human genome.
Collapse
|
338
|
Kleftogiannis D, Kalnis P, Arner E, Bajic VB. Discriminative identification of transcriptional responses of promoters and enhancers after stimulus. Nucleic Acids Res 2017; 45:e25. [PMID: 27789687 PMCID: PMC5389464 DOI: 10.1093/nar/gkw1015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 10/17/2016] [Indexed: 01/19/2023] Open
Abstract
Promoters and enhancers regulate the initiation of gene expression and maintenance of expression levels in spatial and temporal manner. Recent findings stemming from the Cap Analysis of Gene Expression (CAGE) demonstrate that promoters and enhancers, based on their expression profiles after stimulus, belong to different transcription response subclasses. One of the most promising biological features that might explain the difference in transcriptional response between subclasses is the local chromatin environment. We introduce a novel computational framework, PEDAL, for distinguishing effectively transcriptional profiles of promoters and enhancers using solely histone modification marks, chromatin accessibility and binding sites of transcription factors and co-activators. A case study on data from MCF-7 cell-line reveals that PEDAL can identify successfully the transcription response subclasses of promoters and enhancers from two different stimulations. Moreover, we report subsets of input markers that discriminate with minimized classification error MCF-7 promoter and enhancer transcription response subclasses. Our work provides a general computational approach for identifying effectively cell-specific and stimulation-specific promoter and enhancer transcriptional profiles, and thus, contributes to improve our understanding of transcriptional activation in human.
Collapse
Affiliation(s)
- Dimitrios Kleftogiannis
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.,The Institute of Cancer Research (ICR), London, SW7 3RP, UK
| | - Panos Kalnis
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Erik Arner
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies) (CLST (DGT)), Yokohama, Kanagawa 230-0045, Japan
| | - Vladimir B Bajic
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| |
Collapse
|
339
|
Retinoic Acid Negatively Impacts Proliferation and MC TC Specific Attributes of Human Skin Derived Mast Cells, but Reinforces Allergic Stimulability. Int J Mol Sci 2017; 18:ijms18030525. [PMID: 28264498 PMCID: PMC5372541 DOI: 10.3390/ijms18030525] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 01/27/2023] Open
Abstract
The Vitamin-A-metabolite retinoic acid (RA) acts as a master regulator of cellular programs. Mast cells (MCs) are primary effector cells of type-I-allergic reactions. We recently uncovered that human cutaneous MCs are enriched with RA network components over other skin cells. Yet, direct experimental evidence on the significance of the RA-MC axis is limited. Here, skin-derived cultured MCs were exposed to RA for seven days and investigated by flow-cytometry (BrdU incorporation, Annexin/PI, FcεRI), microscopy, RT-qPCR, histamine quantitation, protease activity, and degranulation assays. We found that while MC size and granularity remained unchanged, RA potently interfered with MC proliferation. Conversely, a modest survival-promoting effect from RA was noted. The granule constituents, histamine and tryptase, remained unaffected, while RA had a striking impact on MC chymase, whose expression dropped by gene and by peptidase activity. The newly uncovered MRGPRX2 performed similarly to chymase. Intriguingly, RA fostered allergic MC degranulation, in a way completely uncoupled from FcεRI expression, but it simultaneously restricted MRGPRX2-triggered histamine release in agreement with the reduced receptor expression. Vitamin-A-derived hormones thus re-shape skin-derived MCs numerically, phenotypically, and functionally. A general theme emerges, implying RA to skew MCs towards processes associated with (allergic) inflammation, while driving them away from the skin-imprinted MCTC (“MCs containing tryptase and chymase”) signature (chymase, MRGPRX2). Collectively, MCs are substantial targets of the skin retinoid network.
Collapse
|
340
|
Le Gras S, Keime C, Anthony A, Lotz C, De Longprez L, Brouillet E, Cassel JC, Boutillier AL, Merienne K. Altered enhancer transcription underlies Huntington's disease striatal transcriptional signature. Sci Rep 2017; 7:42875. [PMID: 28225006 PMCID: PMC5320509 DOI: 10.1038/srep42875] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/16/2017] [Indexed: 01/09/2023] Open
Abstract
Epigenetic and transcriptional alterations are both implicated in Huntington’s disease (HD), a progressive neurodegenerative disease resulting in degeneration of striatal neurons in the brain. However, how impaired epigenetic regulation leads to transcriptional dysregulation in HD is unclear. Here, we investigated enhancer RNAs (eRNAs), a class of long non-coding RNAs transcribed from active enhancers. We found that eRNAs are expressed from many enhancers of mouse striatum and showed that a subset of those eRNAs are deregulated in HD vs control mouse striatum. Enhancer regions producing eRNAs decreased in HD mouse striatum were associated with genes involved in striatal neuron identity. Consistently, they were enriched in striatal super-enhancers. Moreover, decreased eRNA expression in HD mouse striatum correlated with down-regulation of associated genes. Additionally, a significant number of RNA Polymerase II (RNAPII) binding sites were lost within enhancers associated with decreased eRNAs in HD vs control mouse striatum. Together, this indicates that loss of RNAPII at HD mouse enhancers contributes to reduced transcription of eRNAs, resulting in down-regulation of target genes. Thus, our data support the view that eRNA dysregulation in HD striatum is a key mechanism leading to altered transcription of striatal neuron identity genes, through reduced recruitment of RNAPII at super-enhancers.
Collapse
Affiliation(s)
- Stéphanie Le Gras
- GenomeEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/University of Strasbourg-UMR 7104, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Céline Keime
- GenomeEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/University of Strasbourg-UMR 7104, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Anne Anthony
- University of Strasbourg, Laboratory of Cognitive and Adaptive Neurosciences (LNCA), 12 rue Goethe, 67000 Strasbourg, France.,CNRS, LNCA UMR 7364, 12 rue Goethe, 67000 Strasbourg, France
| | - Caroline Lotz
- University of Strasbourg, Laboratory of Cognitive and Adaptive Neurosciences (LNCA), 12 rue Goethe, 67000 Strasbourg, France.,CNRS, LNCA UMR 7364, 12 rue Goethe, 67000 Strasbourg, France
| | - Lucie De Longprez
- Commissariat à l'Energie Atomique (CEA), Département de Recherches Fondamentales (DRF), Institut d'Imagerie Biomédicale (I2BM), Molecular Imaging Research Center (MIRCen), F-92260 Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, F-92260 Fontenay-aux-Roses, France
| | - Emmanuel Brouillet
- Commissariat à l'Energie Atomique (CEA), Département de Recherches Fondamentales (DRF), Institut d'Imagerie Biomédicale (I2BM), Molecular Imaging Research Center (MIRCen), F-92260 Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, F-92260 Fontenay-aux-Roses, France
| | - Jean-Christophe Cassel
- University of Strasbourg, Laboratory of Cognitive and Adaptive Neurosciences (LNCA), 12 rue Goethe, 67000 Strasbourg, France.,CNRS, LNCA UMR 7364, 12 rue Goethe, 67000 Strasbourg, France
| | - Anne-Laurence Boutillier
- University of Strasbourg, Laboratory of Cognitive and Adaptive Neurosciences (LNCA), 12 rue Goethe, 67000 Strasbourg, France.,CNRS, LNCA UMR 7364, 12 rue Goethe, 67000 Strasbourg, France
| | - Karine Merienne
- University of Strasbourg, Laboratory of Cognitive and Adaptive Neurosciences (LNCA), 12 rue Goethe, 67000 Strasbourg, France.,CNRS, LNCA UMR 7364, 12 rue Goethe, 67000 Strasbourg, France
| |
Collapse
|
341
|
Kitagawa Y, Ohkura N, Kidani Y, Vandenbon A, Hirota K, Kawakami R, Yasuda K, Motooka D, Nakamura S, Kondo M, Taniuchi I, Kohwi-Shigematsu T, Sakaguchi S. Guidance of regulatory T cell development by Satb1-dependent super-enhancer establishment. Nat Immunol 2017; 18:173-183. [PMID: 27992401 PMCID: PMC5582804 DOI: 10.1038/ni.3646] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.
Collapse
Affiliation(s)
- Yohko Kitagawa
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Laboratory of Experimental Immunology, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Yujiro Kidani
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Alexis Vandenbon
- Immuno-Genomics Research Unit, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Keiji Hirota
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Laboratory of Integrative Biological Science, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ryoji Kawakami
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Keiko Yasuda
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Laboratory of Experimental Immunology, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shota Nakamura
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Motonari Kondo
- Department of Molecular Immunology, School of Medicine, Toho University, Tokyo, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | | | - Shimon Sakaguchi
- Department of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
- Laboratory of Experimental Immunology, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
342
|
Navratilova P, Danks GB, Long A, Butcher S, Manak JR, Thompson EM. Sex-specific chromatin landscapes in an ultra-compact chordate genome. Epigenetics Chromatin 2017; 10:3. [PMID: 28115992 PMCID: PMC5240408 DOI: 10.1186/s13072-016-0110-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/23/2016] [Indexed: 12/15/2022] Open
Abstract
Background In multicellular organisms, epigenome dynamics are associated with transitions in the cell cycle, development, germline specification, gametogenesis and inheritance. Evolutionarily, regulatory space has increased in complex metazoans to accommodate these functions. In tunicates, the sister lineage to vertebrates, we examine epigenome adaptations to strong secondary genome compaction, sex chromosome evolution and cell cycle modes. Results Across the 70 MB Oikopleura dioica genome, we profiled 19 histone modifications, and RNA polymerase II, CTCF and p300 occupancies, to define chromatin states within two homogeneous tissues with distinct cell cycle modes: ovarian endocycling nurse nuclei and mitotically proliferating germ nuclei in testes. Nurse nuclei had active chromatin states similar to other metazoan epigenomes, with large domains of operon-associated transcription, a general lack of heterochromatin, and a possible role of Polycomb PRC2 in dosage compensation. Testis chromatin states reflected transcriptional activity linked to spermatogenesis and epigenetic marks that have been associated with establishment of transgenerational inheritance in other organisms. We also uncovered an unusual chromatin state specific to the Y-chromosome, which combined active and heterochromatic histone modifications on specific transposable elements classes, perhaps involved in regulating their activity. Conclusions Compacted regulatory space in this tunicate genome is accompanied by reduced heterochromatin and chromatin state domain widths. Enhancers, promoters and protein-coding genes have conserved epigenomic features, with adaptations to the organization of a proportion of genes in operon units. We further identified features specific to sex chromosomes, cell cycle modes, germline identity and dosage compensation, and unusual combinations of histone PTMs with opposing consensus functions. Electronic supplementary material The online version of this article (doi:10.1186/s13072-016-0110-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pavla Navratilova
- Sars International Centre for Marine Molecular Biology, University of Bergen, 5008 Bergen, Norway
| | - Gemma Barbara Danks
- Sars International Centre for Marine Molecular Biology, University of Bergen, 5008 Bergen, Norway
| | - Abby Long
- Departments of Biology and Pediatrics and the Roy J. Carver Center for Genomics, 459 Biology Building, University of Iowa, Iowa City, IA 52242 USA
| | - Stephen Butcher
- Departments of Biology and Pediatrics and the Roy J. Carver Center for Genomics, 459 Biology Building, University of Iowa, Iowa City, IA 52242 USA
| | - John Robert Manak
- Departments of Biology and Pediatrics and the Roy J. Carver Center for Genomics, 459 Biology Building, University of Iowa, Iowa City, IA 52242 USA
| | - Eric M Thompson
- Sars International Centre for Marine Molecular Biology, University of Bergen, 5008 Bergen, Norway.,Department of Biology, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
343
|
Smyk M, Akdemir KC, Stankiewicz P. SOX9 chromatin folding domains correlate with its real and putative distant cis-regulatory elements. Nucleus 2017; 8:182-187. [PMID: 28085555 DOI: 10.1080/19491034.2017.1279776] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Evolutionary conserved transcription factor SOX9, encoded by the dosage sensitive SOX9 gene on chromosome 17q24.3, plays an important role in development of multiple organs, including bones and testes. Heterozygous point mutations and genomic copy-number variant (CNV) deletions involving SOX9 have been reported in patients with campomelic dysplasia (CD), a skeletal malformation syndrome often associated with male-to-female sex reversal. Balanced and unbalanced structural genomic variants with breakpoints mapping up to 1.3 Mb up- and downstream to SOX9 have been described in patients with milder phenotypes, including acampomelic campomelic dysplasia, sex reversal, and Pierre Robin sequence. Based on the localization of breakpoints of genomic rearrangements causing different phenotypes, 5 genomic intervals mapping upstream to SOX9 have been defined. We have analyzed the publically available database of high-throughput chromosome conformation capture (Hi-C) in multiple cell lines in the genomic regions flanking SOX9. Consistent with the literature data, chromatin domain boundaries in the SOX9 locus exhibit conservation across species and remain largely constant across multiple cell types. Interestingly, we have found that chromatin folding domains in the SOX9 locus associate with the genomic intervals harboring real and putative regulatory elements of SOX9, implicating that variation in intra-domain interactions may be critical for dynamic regulation of SOX9 expression in a cell type-specific fashion. We propose that tissue-specific enhancers for other transcription factor genes may similarly utilize chromatin folding sub-domains in gene regulation.
Collapse
Affiliation(s)
- Marta Smyk
- a Department of Medical Genetics , Institute of Mother and Child , Warsaw , Poland
| | - Kadir Caner Akdemir
- b Genomic Medicine Department , MD Anderson Cancer Center , Houston , TX , USA
| | - Paweł Stankiewicz
- c Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
344
|
De Luca M, Pels K, Moleirinho S, 1 Department of Biomedical and Clinical Sciences, L. Sacco, University of Milano, Milano, 20157, Italy, Curtale G. The epigenetic landscape of innate immunity. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.1.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
345
|
Ehrlund A, Mejhert N, Björk C, Andersson R, Kulyté A, Åström G, Itoh M, Kawaji H, Lassmann T, Daub CO, Carninci P, Forrest ARR, Hayashizaki Y, Sandelin A, Ingelsson E, Rydén M, Laurencikiene J, Arner P, Arner E. Transcriptional Dynamics During Human Adipogenesis and Its Link to Adipose Morphology and Distribution. Diabetes 2017; 66:218-230. [PMID: 27803022 PMCID: PMC5860264 DOI: 10.2337/db16-0631] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022]
Abstract
White adipose tissue (WAT) can develop into several phenotypes with different pathophysiological impact on type 2 diabetes. To better understand the adipogenic process, the transcriptional events that occur during in vitro differentiation of human adipocytes were investigated and the findings linked to WAT phenotypes. Single-molecule transcriptional profiling provided a detailed map of the expressional changes of genes, enhancers, and long noncoding RNAs, where different types of transcripts share common dynamics during differentiation. Common signatures include early downregulated, transient, and late induced transcripts, all of which are linked to distinct developmental processes during adipogenesis. Enhancers expressed during adipogenesis overlap significantly with genetic variants associated with WAT distribution. Transiently expressed and late induced genes are associated with hypertrophic WAT (few but large fat cells), a phenotype closely linked to insulin resistance and type 2 diabetes. Transcription factors that are expressed early or transiently affect differentiation and adipocyte function and are controlled by several well-known upstream regulators such as glucocorticosteroids, insulin, cAMP, and thyroid hormones. Taken together, our results suggest a complex but highly coordinated regulation of adipogenesis.
Collapse
Affiliation(s)
- Anna Ehrlund
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Christel Björk
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Robin Andersson
- The Bioinformatics Centre, Department of Biology, and Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Agné Kulyté
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Gaby Åström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Masayoshi Itoh
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Preventive Medicine & Diagnosis Innovation Program, Wakō, Saitama, Japan
| | - Hideya Kawaji
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Preventive Medicine & Diagnosis Innovation Program, Wakō, Saitama, Japan
| | - Timo Lassmann
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
- Telethon Kids Institute and The University of Western Australia, Perth, Western Australia, Australia
| | - Carsten O Daub
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- Department of Biosciences and Nutrition and Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Piero Carninci
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
| | - Alistair R R Forrest
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
| | - Yoshihide Hayashizaki
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Preventive Medicine & Diagnosis Innovation Program, Wakō, Saitama, Japan
| | - Albin Sandelin
- The Bioinformatics Centre, Department of Biology, and Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Erik Ingelsson
- Molecular Epidemiology, Department of Medical Sciences, and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Mikael Rydén
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Peter Arner
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Erik Arner
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Tsurumi, Yokohama, Kanagawa, Japan
- RIKEN Omics Science Center, Tsurumi, Yokohama, Kanagawa, Japan
| |
Collapse
|
346
|
Deviatiiarov R, Lizio M, Gusev O. Application of a CAGE Method to an Avian Development Study. Methods Mol Biol 2017; 1650:101-109. [PMID: 28809016 DOI: 10.1007/978-1-4939-7216-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cap analysis of gene expression (CAGE) is a convenient approach for genome-wide identification of promoter regions at single base-pair resolution level and accurate expression estimation of the corresponding transcripts. Depending on the initial biomaterial amount and sequencing technology, different computational pipelines for data processing are available, as well as variations of the CAGE protocol that improve sensitivity and accuracy. Therefore, this chapter elucidates the key steps of sample preparation, sequencing, and data analysis via an example of a promoter expression estimation study in chicken development. We also describe the applicability of this approach for studying other avian and reptilian species.
Collapse
Affiliation(s)
- Ruslan Deviatiiarov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Marina Lizio
- Division of Genomic Technologies, Center for Life Science Technologies (CLST), RIKEN, Yokohama, Japan.,Omics Science Center (OSC), RIKEN, Yokohama, Japan
| | - Oleg Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia. .,Division of Genomic Technologies, Center for Life Science Technologies (CLST), RIKEN, Yokohama, Japan. .,Preventive Medicine and Diagnosis Innovation Program, RIKEN, Yokohama, Japan. .,RIKEN Innovation Center, Wako, Japan.
| |
Collapse
|
347
|
The FANTOM5 Computation Ecosystem: Genomic Information Hub for Promoters and Active Enhancers. Methods Mol Biol 2017; 1611:199-217. [PMID: 28451981 DOI: 10.1007/978-1-4939-7015-5_15] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Functional Annotation of the Mammalian Genome 5 (FANTOM5) project conducted transcriptome analysis of various mammalian cell types and provided a comprehensive resource to understand transcriptome and transcriptional regulation in individual cellular states encoded in the genome.FANTOM5 used cap analysis of gene expression (CAGE) with single-molecule sequencing to map transcription start sites (TSS) and measured their expression in a diverse range of samples. The main results from FANTOM5 were published as a promoter-level mammalian expression atlas and an atlas of active enhancers across human cell types. The FANTOM5 dataset is composed of raw experimental data and the results of bioinformatics analyses. In this chapter, we give a detailed description of the content of the FANTOM5 dataset and elaborate on different computing applications developed to publish the data and enable reproducibility and discovery of new findings. We present use cases in which the FANTOM5 dataset has been reused, leading to new findings.
Collapse
|
348
|
Ienasescu H, Li K, Andersson R, Vitezic M, Rennie S, Chen Y, Vitting-Seerup K, Lagoni E, Boyd M, Bornholdt J, de Hoon MJL, Kawaji H, Lassmann T, Hayashizaki Y, Forrest ARR, Carninci P, Sandelin A. On-the-fly selection of cell-specific enhancers, genes, miRNAs and proteins across the human body using SlideBase. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2016; 2016:baw144. [PMID: 28025337 PMCID: PMC5199134 DOI: 10.1093/database/baw144] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 12/19/2022]
Abstract
Genomics consortia have produced large datasets profiling the expression of genes, micro-RNAs, enhancers and more across human tissues or cells. There is a need for intuitive tools to select subsets of such data that is the most relevant for specific studies. To this end, we present SlideBase, a web tool which offers a new way of selecting genes, promoters, enhancers and microRNAs that are preferentially expressed/used in a specified set of cells/tissues, based on the use of interactive sliders. With the help of sliders, SlideBase enables users to define custom expression thresholds for individual cell types/tissues, producing sets of genes, enhancers etc. which satisfy these constraints. Changes in slider settings result in simultaneous changes in the selected sets, updated in real time. SlideBase is linked to major databases from genomics consortia, including FANTOM, GTEx, The Human Protein Atlas and BioGPS.Database URL: http://slidebase.binf.ku.dk.
Collapse
Affiliation(s)
- Hans Ienasescu
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Kang Li
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Department of Mathematical Sciences, University of Copenhagen, Universitetsparken 5, Copenhagen Ø, DK2100, Denmark
| | - Robin Andersson
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Morana Vitezic
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Sarah Rennie
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Yun Chen
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Kristoffer Vitting-Seerup
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Emil Lagoni
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Mette Boyd
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Jette Bornholdt
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark.,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| | - Michiel J L de Hoon
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Hideya Kawaji
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Timo Lassmann
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.,Telethon Kids Institute, The University of Western Australia, 100 Roberts Road, Subiaco, 6008, Australia Western Australia
| | | | - Yoshihide Hayashizaki
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Alistair R R Forrest
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.,Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, Western Australia, Australia
| | - Piero Carninci
- RIKEN Center for Life Science Technologies (Division of Genomic Technologies), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Albin Sandelin
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark .,Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen N, DK2200, Denmark
| |
Collapse
|
349
|
van Arensbergen J, FitzPatrick VD, de Haas M, Pagie L, Sluimer J, Bussemaker HJ, van Steensel B. Genome-wide mapping of autonomous promoter activity in human cells. Nat Biotechnol 2016; 35:145-153. [PMID: 28024146 DOI: 10.1038/nbt.3754] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 12/01/2016] [Indexed: 12/20/2022]
Abstract
Previous methods to systematically characterize sequence-intrinsic activity of promoters have been limited by relatively low throughput and the length of the sequences that could be tested. Here we present 'survey of regulatory elements' (SuRE), a method that assays more than 108 DNA fragments, each 0.2-2 kb in size, for their ability to drive transcription autonomously. In SuRE, a plasmid library of random genomic fragments upstream of a 20-bp barcode is constructed, and decoded by paired-end sequencing. This library is used to transfect cells, and barcodes in transcribed RNA are quantified by high-throughput sequencing. When applied to the human genome, we achieve 55-fold genome coverage, allowing us to map autonomous promoter activity genome-wide in K562 cells. By computational modeling we delineate subregions within promoters that are relevant for their activity. We show that antisense promoter transcription is generally dependent on the sense core promoter sequences, and that most enhancers and several families of repetitive elements act as autonomous transcription initiation sites.
Collapse
Affiliation(s)
- Joris van Arensbergen
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vincent D FitzPatrick
- Department of Biological Sciences, Columbia University, New York, New York, USA.,Department of Systems Biology, Columbia University Medical Center, New York, New York, USA
| | - Marcel de Haas
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ludo Pagie
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jasper Sluimer
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Harmen J Bussemaker
- Department of Biological Sciences, Columbia University, New York, New York, USA.,Department of Systems Biology, Columbia University Medical Center, New York, New York, USA
| | - Bas van Steensel
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
350
|
Ehrlich KC, Paterson HL, Lacey M, Ehrlich M. DNA Hypomethylation in Intragenic and Intergenic Enhancer Chromatin of Muscle-Specific Genes Usually Correlates with their Expression. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2016; 89:441-455. [PMID: 28018137 PMCID: PMC5168824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tissue-specific enhancers are critical for gene regulation. In this study, we help elucidate the contribution of muscle-associated differential DNA methylation to the enhancer activity of highly muscle-specific genes. By bioinformatic analysis of 44 muscle-associated genes, we show that preferential gene expression in skeletal muscle (SkM) correlates with SkM-specific intragenic and intergenic enhancer chromatin and overlapping foci of DNA hypomethylation. Some genes, e.g., CASQ1 and FBXO32, displayed broad regions of both SkM- and heart-specific enhancer chromatin but exhibited focal SkM-specific DNA hypomethylation. Half of the genes had SkM-specific super-enhancers. In contrast to simple enhancer/gene-expression correlations, a super-enhancer was associated with the myogenic MYOD1 gene in both SkM and myoblasts even though SkM has < 1 percent as much MYOD1 expression. Local chromatin differences in this super-enhancer probably contribute to the SkM/myoblast differential expression. Transfection assays confirmed the tissue-specificity of the 0.3-kb core enhancer within MYOD1's super-enhancer and demonstrated its repression by methylation of its three CG dinucleotides. Our study suggests that DNA hypomethylation increases enhancer tissue-specificity and that SkM super-enhancers sometimes are poised for physiologically important, rapid up-regulation.
Collapse
Affiliation(s)
- Kenneth C. Ehrlich
- Program in Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA
| | | | - Michelle Lacey
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA,Mathematics Department, Tulane University, New Orleans, LA
| | - Melanie Ehrlich
- Program in Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA,Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA,Hayward Genetics Center, Tulane University Health Sciences Center, New Orleans, LA,To whom all correspondence should be addressed: Melanie Ehrlich, PhD, Hayward Genetics Center, Tulane University Health Sciences Center, 1430 Tulane Ave., New Orleans, LA 70112; Tele: 504-988-2449; Fax: 504-988-1763;
| |
Collapse
|