Volume 10, Issue 2, Pages (January 2015)

Slides:



Advertisements
Similar presentations
Volume 4, Issue 5, Pages (September 2013)
Advertisements

circRNA Biogenesis Competes with Pre-mRNA Splicing
Microprocessor Activity Controls Differential miRNA Biogenesis In Vivo
Volume 55, Issue 1, Pages (July 2014)
Volume 9, Issue 5, Pages (December 2014)
Volume 23, Issue 3, Pages (April 2018)
Volume 11, Issue 11, Pages (June 2015)
The INO80 Complex Removes H2A
Global Mapping of Human RNA-RNA Interactions
Steven J. Petesch, John T. Lis  Cell 
RNA Exosome Depletion Reveals Transcription Upstream of Active Human Promoters by Pascal Preker, Jesper Nielsen, Susanne Kammler, Søren Lykke-Andersen,
Transient N-6-Methyladenosine Transcriptome Sequencing Reveals a Regulatory Role of m6A in Splicing Efficiency  Annita Louloupi, Evgenia Ntini, Thomas.
Volume 19, Issue 3, Pages (April 2017)
Volume 15, Issue 9, Pages (May 2016)
Human Senataxin Resolves RNA/DNA Hybrids Formed at Transcriptional Pause Sites to Promote Xrn2-Dependent Termination  Konstantina Skourti-Stathaki, Nicholas J.
circRNA Biogenesis Competes with Pre-mRNA Splicing
MYO5A Gene Is a Target of MITF in Melanocytes
Volume 29, Issue 2, Pages (February 2008)
Exon Circularization Requires Canonical Splice Signals
Anne L. Sapiro, Patricia Deng, Rui Zhang, Jin Billy Li  Cell Reports 
Volume 25, Issue 9, Pages (September 2017)
Volume 7, Issue 1, Pages (April 2014)
Widespread Inhibition of Posttranscriptional Splicing Shapes the Cellular Transcriptome following Heat Shock  Reut Shalgi, Jessica A. Hurt, Susan Lindquist,
Volume 5, Issue 6, Pages (December 2013)
Joseph Rodriguez, Jerome S. Menet, Michael Rosbash  Molecular Cell 
Volume 23, Issue 1, Pages 9-22 (January 2013)
Minchul Kim, Taekhoon Kim, Randy L. Johnson, Dae-Sik Lim  Cell Reports 
Daniel F. Tardiff, Scott A. Lacadie, Michael Rosbash  Molecular Cell 
Volume 16, Issue 4, Pages (November 2004)
Volume 19, Issue 12, Pages (June 2017)
Volume 8, Issue 6, Pages (September 2014)
Volume 10, Issue 7, Pages (February 2015)
Volume 11, Issue 2, Pages (April 2015)
Volume 20, Issue 6, Pages (August 2017)
Long Noncoding RNAs with Enhancer-like Function in Human Cells
Volume 9, Issue 5, Pages (November 2017)
The INO80 Complex Removes H2A
A Genetic Screen Identifies TCF3/E2A and TRIAP1 as Pathway-Specific Regulators of the Cellular Response to p53 Activation  Zdenek Andrysik, Jihye Kim,
Shane M. Harding, Jonathan A. Boiarsky, Roger A. Greenberg 
Baekgyu Kim, Kyowon Jeong, V. Narry Kim  Molecular Cell 
Imprinted Chromatin around DIRAS3 Regulates Alternative Splicing of GNG12-AS1, a Long Noncoding RNA  Malwina Niemczyk, Yoko Ito, Joanna Huddleston, Anna.
Chromosomal Translocation Formation Is Sufficient to Produce Fusion Circular RNAs Specific to Patient Tumor Cells  Loelia Babin, Marion Piganeau, Benjamin.
Volume 20, Issue 9, Pages (August 2017)
ADAR Regulates RNA Editing, Transcript Stability, and Gene Expression
Alterations in mRNA 3′ UTR Isoform Abundance Accompany Gene Expression Changes in Human Huntington’s Disease Brains  Lindsay Romo, Ami Ashar-Patel, Edith.
Volume 23, Issue 7, Pages (May 2018)
Volume 21, Issue 9, Pages (November 2017)
Volume 13, Issue 1, Pages (October 2015)
Volume 11, Issue 11, Pages (June 2015)
Volume 47, Issue 4, Pages (August 2012)
Volume 63, Issue 3, Pages (August 2016)
Human Telomerase RNA Processing and Quality Control
Maternally Inherited Stable Intronic Sequence RNA Triggers a Self-Reinforcing Feedback Loop during Development  Mandy Li-Ian Tay, Jun Wei Pek  Current.
Volume 15, Issue 5, Pages (May 2016)
Assessing the Functional Characteristics of Synonymous and Nonsynonymous Mutation Candidates by Use of Large DNA Constructs  A.M. Eeds, D. Mortlock, R.
Short Telomeres in ESCs Lead to Unstable Differentiation
Volume 17, Issue 3, Pages (September 2009)
Volume 8, Issue 6, Pages (September 2014)
Volume 15, Issue 2, Pages (April 2016)
Volume 5, Issue 4, Pages (November 2013)
Volume 41, Issue 2, Pages (January 2011)
Volume 25, Issue 3, Pages e7 (October 2018)
The Biogenesis of Nascent Circular RNAs
Pervasive Targeting of Nascent Transcripts by Hfq
Influence of RdDM on DCL4 Transcript Isoform Expression.
Volume 2, Issue 4, Pages (October 2012)
Enhancer Control of Transcriptional Bursting
A Splicing-Independent Function of SF2/ASF in MicroRNA Processing
Volume 20, Issue 5, Pages (August 2017)
SUPT4H1 Depletion Leads to a Global Reduction in RNA
Presentation transcript:

Volume 10, Issue 2, Pages 170-177 (January 2015) Analysis of Intron Sequences Reveals Hallmarks of Circular RNA Biogenesis in Animals  Andranik Ivanov, Sebastian Memczak, Emanuel Wyler, Francesca Torti, Hagit T. Porath, Marta R. Orejuela, Michael Piechotta, Erez Y. Levanon, Markus Landthaler, Christoph Dieterich, Nikolaus Rajewsky  Cell Reports  Volume 10, Issue 2, Pages 170-177 (January 2015) DOI: 10.1016/j.celrep.2014.12.019 Copyright © 2015 The Authors Terms and Conditions

Cell Reports 2015 10, 170-177DOI: (10.1016/j.celrep.2014.12.019) Copyright © 2015 The Authors Terms and Conditions

Figure 1 RCMs between or within Introns and circRNA Biogenesis (A) Model for the possible influence of RCMs on circRNA biogenesis. RCMs between different introns (lower panel) in competition with intron internal RCMs (upper panel) may promote hairpin formation and circularization of the embedded exon. (B) Enrichment of RCMs and SMs in introns bracketing C. elegans circRNAs. Left: distribution of RCM and SM counts per intron pair that flanks circRNAs (green) and length-matched controls (gray). Right: ratio of the number of intron pairs bracketing circRNAs that contain only RCMs but no SMs to the number of intron pairs bracketing circRNAs with only SMs but no RCMs (p value: Fisher’s exact test). See also Figures S1, S4, and S5. Cell Reports 2015 10, 170-177DOI: (10.1016/j.celrep.2014.12.019) Copyright © 2015 The Authors Terms and Conditions

Figure 2 Prediction of circRNAs Based on Sequence Analysis of Introns (A) C. elegans. The matrix represents the average circularization scores for intron pairs of circRNA producing genes. (0,0) coordinates correspond to circRNA flanking introns. Upstream and downstream introns are enumerated with decreasing and increasing numbers, respectively (with 0 being directly adjacent, −1/+1 being the preceding/next intron, etc.; see Supplemental Experimental Procedures). (B) C. elegans. Different prediction methods are used. The x axis is the fraction of all introns pairs per gene sorted by H scores. Small (high) x values denote intron pairs with high (low) scores for bracketing a circRNA. The y axis is the fraction of 1,111 exonic circRNAs recovered by H score. (C and D) Analyses for human circRNAs (Memczak et al., 2013), analogous to (A) and (B). See also Figures S4 and S5, and Tables S1, S2, and S3. Cell Reports 2015 10, 170-177DOI: (10.1016/j.celrep.2014.12.019) Copyright © 2015 The Authors Terms and Conditions

Figure 3 Experimental Validation of Human circRNAs Predicted by RCM Analyses (A) CircRNA candidates from high-, medium-, and low-expression sets were assayed by qPCR with divergent primers and RNase R treatment. Linear control: VCL, GAPDH, TFRC; positive control: a known circRNA (Memczak et al., 2013). Sanger sequencing of amplicons confirmed in all tested cases the predicted head-to-tail junctions (candidates 9 and 11 contained an additional exon: marked with ∗). CircRNA candidates that were >10-fold resistant to RNase R treatment compared with Vinculin were counted as positive (+). As negative controls, we selected exons from highly expressed genes with H score = 0. Error bars, SEM; n = 4. (B) The chromatogram of a Sanger sequencing experiment confirms the presence of the predicted head-to-tail junction of the candidate circRNA from the PTGES3 gene locus. See also Figure S5 and Tables S1, S2, and S3. Cell Reports 2015 10, 170-177DOI: (10.1016/j.celrep.2014.12.019) Copyright © 2015 The Authors Terms and Conditions

Figure 4 ADAR Antagonizes circRNA Expression (A) Normalized distribution of A-to-I conversions upstream/downstream of the head/tail splice sites of circRNAs (Memczak et al., 2013). As controls, we selected length-matched introns and introns from the same genes that produce circRNAs. Introns smaller than 1.5 kb were removed from the analysis. The curves were smoothed within ±10 bp at each position and normalized to the total number of analyzed introns. (B) Position of the nearest RCMs (or SMs) around circRNA splice sites. For each intron pair, we selected the top-scoring RCMs within the 1.5 kb region around the splice site. The curves were smoothed within a ±20 bp window and normalized to the total number of analyzed intron pairs. (C) Comparison of the differential expression (ADAR1 and ADAR2 codepletion against untreated control) of linear RNA and circRNA in two independent replicates (merged). The y axis is the log2 fold change of the reads (+5) supporting the head-to-tail splice sites. The x axis is log2 fold change of the reads supporting linear splice sites of circRNA host genes. For the analysis, we selected only circRNAs with at least ten reads in ADAR1/2 KD or control samples. The numbers tally the count of circRNAs/corresponding linear hosts in each segment of the graph. (D) Depletion of ADAR1 by RNAi. ADAR1 was depleted from HEK293 cells for 96 hr using two different siRNAs and compared with untreated and ADAR2-depleted cells. Upper panel: qRT-PCR of ADAR1 and ADAR2 transcripts, as well as GAPDH and SNCA as controls. Lower panel: western blot using ADAR1- and ADAR2-specific antibodies. Error bars, SD; n = 3. (E) Quantification of circRNAs and host transcript levels. Using circRNA-specific primers (upper panel) or exon-spanning primers that were not part of the circRNA (lower panel), we quantified RNA abundances relative to untreated cells by qRT-PCR (Delta-Delta Ct values were normalized to C. elegans spike-in). Error bars, SD; n = 3. See also Figures S2, S3, and S5, and Tables S1, S2, and S3. Cell Reports 2015 10, 170-177DOI: (10.1016/j.celrep.2014.12.019) Copyright © 2015 The Authors Terms and Conditions