The Epigenome Tools 2: Chip-Seq and Data Analysis

Total Page:16

File Type:pdf, Size:1020Kb

The Epigenome Tools 2: Chip-Seq and Data Analysis The Epigenome Tools 2: ChIP-Seq and Data Analysis Chongzhi Zang [email protected] http://zanglab.com PHS5705: Public Health Genomics March 20, 2017 1 Outline • Epigenome: basics review • ChIP-seq overview • ChIP-seq data analysis 2 Epigenome histone nucleosome The epigenome is a multitude of chemical compounds that can tell the genome what to do. The epigenome is made up of chemical compounds and proteins that can attach to DNA and direct such actions as turning genes on or off, controlling the production of proteins in particular cells. -- from genome.gov Original figure from ENCODE, Darryl Leja (NHGRI), Ian Dunham (EBI) 3 Epigenomic marks • DNA methylation • Histone marks – Covalent modifications – Histone variants • Chromatin regulators – Histone modifying enzymes – Chromatin remodeling complexes • * Transcription factors 4 Histone modifications • Nucleosome Core Particles • Core Histones: H2A, H2B, H3, H4 Notation: H3K4me3 • Covalent modifications on histone tails include: methylation (me), acetylation (ac), phosphorylation … • Histone variants • Histone modifications are implicated in influencing gene expression. Allis C. et al. Epigenetics. 2006 5 LETTERS Figure 3 Patterns of histone modifications at a IL2RA CD28RE b CNS22 IFNG Chr12: 66810000 66830000 66850000 enhancers. (a) The histone modification pattern at Chr10: 6145000 6150000 6155000 6160000 H3K4me3 25 * H3K4me3 184 H3K4me2 11 the CD28RE enhancer (highlighted in red) of the H3K4me2 15 H3K4me1 30 H3K4me1 39 * H3K4ac 4 IL2RA gene. Significant modifications are * 8 H3K4ac * H3K27me3 4 H3K27me3 indicated by asterisks on the left. (b) Histone 4 H3K27me2 5 H3K27me2 5 H3K27me1 6 modification patterns at the IFNG gene and its H3K27me1 10 * H3K27ac 17 * H3K27ac * 23 H3K9me3 5 downstream enhancer, CNS22, are shown. H3K9me3 2 Histone modificationsH3K9me2 associate5 with H3K9me2 3 H3K9me1 12 Significant modifications at CNS22 are indicated H3K9me1 15 * H3K9ac 3 * H3K9ac 13 by asterisks on the left. (c) The fractions of H3K79me3 2 H3K79me3 7 regulation of geneH3K79me2 expression 2 enhancers associated with each of the 38 H3K79me2 6 H3K79me1 3 H3K79me1 5 H3R2me2 4 modifications. (d) Patterns of histone H3R2me2 3 H3R2me1 5 H3R2me1 modifications at 4,179 DNase hypersensitive 5 H3K36me3 11 H3K36me3 4 H3K36me1 5 sites. The y axis indicates the number of patterns, H3K36me1 7 H3K36ac 5 H3K36ac 8 * H4K20me3 4 and x axis indicates the number of hypersensitive H4K20me3 3 H4K20me1 3 H4K20me1 6 sites associated with each pattern. (e) Correlation H2BK5me1 5 H2BK5me1 9 H2BK5ac 6 * H2BK5ac 15 analysis of gene expression with the ten largest * H4R3me2 3 H4R3me2 5 change) - H3K14ac 7 modification patterns by assigning an enhancer to H3K14ac 5 H3K18ac 9 H3K18ac 17 * the TSS of the nearest known gene. All, all * H3K23ac 16 H3K23ac 8 * * H4K5ac 10 H4K5ac 8 * DNase I hypersensitive sites. The number of * H4K8ac 17 H4K8ac 4 * * H4K12ac 4 hypersensitive sites associated with each H4K12ac 7 H4K16ac 6 H4K16ac 6 * H4K91ac 4 pattern is indicated. H4K91ac 10 (fold log2 * * H2BK12ac 7 H2BK12ac 6 * Differentialexpression H2BK20ac 5 H2BK20ac 12 * H2BK120ac 7 H2BK120ac 14 * * H2AK5ac 8 H2AK5ac 12 * H2AK9ac 3 H2AK9ac 3 H2A.Z 23 H2A.Z 16 * * detected patterns, 1,174 are associated with c 0.35 multiple genes and 3,165 with only one gene 0.30 each (Fig. 2a). The 13 most prevalent patterns 0.25 are each associated with more than 62 genes. 0.20 We next examined the expression of genes in 0.15 these patterns, using the mean expression of 0.10 all genes as a reference (Fig. 2b). It seems that Fractions of enhancers 0.05 we can roughly classify these top patterns into 0 three classes (I, II and III in Fig. 2b) accord- ing to expression. Four of six patterns in class H2A.Z H3K4ac H3K9ac H4K5ac H4K8ac H3K14ac H3K18ac H3K23ac H3K27ac H3K36ac H4K12ac H4K16ac H4K91ac H2AK5ac H2AK9ac H2BK5ac H3K4me1 H3K4me2 H3K4me3 H3K9me1 H3K9me2 H3K9me3 H3R2me1 H3R2me2 I contain H3K27me3 and correlate with low H2BK12ac H2BK20ac H3K27me1 H3K27me2 H3K27me3 H3K36me1 H3K36me3 H3K79me1 H3K79me2 H3K79me3 H4K20me1 H4K20me3 H2BK5me1 H2BK120ac 6 Wang, Zang et al. Nat Genet 2008expression. These patterns also contain H2AZ, H3K4me1, H3K4me2, H3K4me3, H3K9me1: 21 deH2AZ, H3K4me1: 36 H3K4me1/2/3, H3K9me1 and H2A.Z but H2AZ, H3K4me2, H3K4me3, H3K9me1: 37 10,000 100 H2AZ, H3K4me3: 20 no acetylations. The patterns containing H2AZ: 67 90 H3K4me1, H4K20me1: 21 H3K4me1: 64 only H3K4me3 or no modification also 80 H3K4me3: 34 1,000 belong to this class. Class II contains 70 H3K27me3: 65 H3K36me3: 41 60 H4K20me1: 42 H3K36me3 or a modification backbone con- H2AK5ac: 63 100 50 No modification: 1,817 sisting of 17 modifications (as discussed All enhancers: 4,179 40 below), or the backbone plus H4K16ac, 30 10 higher expression which correlates with intermediate gene 20 expression. Class III shows the highest expres- Percentage of enhancers with 10 Number of combinatorial patterns sion, and it includes H2BK5me1, H4K16ac, 1 0 1 10 100 1,000 10,000 1 10 100 1,000 10,000 H4K20me1 and H3K79me1/2/3 in addition Number of enhancers associated with one pattern Enhancers-associated gene expression to the modification backbone (Fig. 2b). Our Gene Ontology analysis suggests that genes histone acetyl transferases (HATs) can associate with different involved in cellular physiology and metabolism are enriched in the regions of genes. For example, PCAF associates with the elongation- active class III patterns, consistent with their house-keeping roles (data competent RNA Pol II, whereas p300 interacts with the initiation- not shown). In contrast, many genes involved in development, cell– competent RNA Pol II (ref. 19). Additionally, depletion of GCN5 cell signaling and synaptic transmission are enriched in the inactive or PCAF, but not CBP or p300, affects H4K8ac and H3K14ac20. class I patterns, consistent with their not being required for mature The distribution patterns of these histone acetylations and histone T-cell function. methylations are exemplified by the genomic locus for ZMYND8 (also To correlate each modification with gene expression, we compared known as PRKCBP1), which is expressed in CD4+ T cells (Fig. 1g). The the average gene expression with or without each modification promoter region (highlighted in red), which was defined as a 2-kb (Fig. 2c). H3K27me3 was among a group of repressive marks also region surrounding the TSS, is associated with 25 modifications including H3K27me2, H3K9me2, H3K9me3 and H4K20me3, whereas 7 (P o 10À ). most other modifications correlated with activation. Although the To identify the patterns of histone modifications in an unbiased modification patterns do not uniquely determine the extent of way, we examined each of the 12,541 gene promoters for association expression, the H3K79me3 and H2BK5ac modifications showed with each of the 18 acetylations, 19 methylations and H2A.Z. Of the weak correlation with expression within a modification pattern possible patterns, only a small fraction exists at promoters. Of 4,339 (Supplementary Fig. 5 online). NATURE GENETICS VOLUME 40 [ NUMBER 7 [ JULY 2008 899 “Functions” of histone marks Table 3. Distinctive Chromatin Features of Genomic Elements Functional Annotation Histone Marks References Promoters H3K4me3 Bernstein et al., 2005; Kim et al., 2005; Pokholok et al., 2005 Bivalent/Poised Promoter H3K4me3/H3K27me3 Bernstein et al., 2006 Transcribed Gene Body H3K36me3 Barski et al., 2007 Enhancer (both active and poised) H3K4me1 Heintzman et al., 2007 Poised Developmental Enhancer H3K4me1/H3K27me3 Creyghton et al., 2010; Rada-Iglesias et al., 2011 Active Enhancer H3K4me1/H3K27ac Creyghton et al., 2010; Heintzman et al., 2009; Rada-Iglesias et al., 2011 Polycomb Repressed Regions H3K27me3 Bernstein et al., 2006; Lee et al., 2006 Heterochromatin H3K9me3 Mikkelsen et al., 2007 survey DNA enrichment at 500 representative loci using the steps in a row, or Sequential-ChIP-seq, can uncover histone nCounter probe system. High-quality antibodies are distinctRivera &PTMs Ren Cell on2013 the7 same molecule or chromatin-associated proteins from IgG patterns and the occupancy distribution correlates in the same complex. Several groups combined bisulfite with a logical set of chromatin states. Validating antibody re- sequencing with ChIP giving rise to BisChIP-seq and ChIP-BS- agents for enrichment specificity and robustness ensures good seq (Brinkman et al., 2012; Statham et al., 2012). Long-distance quality ChIP-seq data sets with high signal to noise ratios. DNA interactions mediated by a specific protein can be profiled Given that ChIP-seq is a mature technology, the technical using chromatin interaction analysis by paired-end-tag restrictions of the technique are well defined by its users. These sequencing, or ChIA-PET (Fullwood et al., 2009). We anticipate restrictions include the need for large amounts of starting mate- other inventive uses of ChIP technology to continue to uncover rial, limited resolution, and the dependence on antibodies. undiscovered roles of histone modifications and histone variants Improvements to ChIP-seq have been developed to address in transcriptional regulation. these limitations and expand the possibilities of its use. Collect- ing enough starting material for ChIP-seq can be challenging Mapping of Chromatin Structures because experiments typically require 1 million (histone modifi- Nucleosome Positioning cations) to 5 million (TFs and chromatin modifiers) cells. Although Moving up the hierarchy of genomic organization, we now look this is feasible when studying fast dividing cell lines, the chal-
Recommended publications
  • Dedifferentiation by Adenovirus E1A Due to Inactivation of Hippo Pathway Effectors YAP and TAZ
    Downloaded from genesdev.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press Dedifferentiation by adenovirus E1A due to inactivation of Hippo pathway effectors YAP and TAZ Nathan R. Zemke, Dawei Gou, and Arnold J. Berk Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA Adenovirus transformed cells have a dedifferentiated phenotype. Eliminating E1A in transformed human embryonic kidney cells derepressed ∼2600 genes, generating a gene expression profile closely resembling mesenchymal stem cells (MSCs). This was associated with a dramatic change in cell morphology from one with scant cytoplasm and a globular nucleus to one with increased cytoplasm, extensive actin stress fibers, and actomyosin-dependent flat- tening against the substratum. E1A-induced hypoacetylation at histone H3 Lys27 and Lys18 (H3K27/18) was reversed. Most of the increase in H3K27/18ac was in enhancers near TEAD transcription factors bound by Hippo signaling-regulated coactivators YAP and TAZ. E1A causes YAP/TAZ cytoplasmic sequestration. After eliminating E1A, YAP/TAZ were transported into nuclei, where they associated with poised enhancers with DNA-bound TEAD4 and H3K4me1. This activation of YAP/TAZ required RHO family GTPase signaling and caused histone acetylation by p300/CBP, chromatin remodeling, and cohesin loading to establish MSC-associated enhancers and then superenhancers. Consistent results were also observed in primary rat embryo kidney cells, human fibroblasts, and human respiratory tract epithelial cells. These results together with earlier studies suggest that YAP/TAZ function in a developmental checkpoint controlled by signaling from the actin cytoskeleton that prevents differ- entiation of a progenitor cell until it is in the correct cellular and tissue environment.
    [Show full text]
  • Functional Roles of Bromodomain Proteins in Cancer
    cancers Review Functional Roles of Bromodomain Proteins in Cancer Samuel P. Boyson 1,2, Cong Gao 3, Kathleen Quinn 2,3, Joseph Boyd 3, Hana Paculova 3 , Seth Frietze 3,4,* and Karen C. Glass 1,2,4,* 1 Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Colchester, VT 05446, USA; [email protected] 2 Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; [email protected] 3 Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA; [email protected] (C.G.); [email protected] (J.B.); [email protected] (H.P.) 4 University of Vermont Cancer Center, Burlington, VT 05405, USA * Correspondence: [email protected] (S.F.); [email protected] (K.C.G.) Simple Summary: This review provides an in depth analysis of the role of bromodomain-containing proteins in cancer development. As readers of acetylated lysine on nucleosomal histones, bromod- omain proteins are poised to activate gene expression, and often promote cancer progression. We examined changes in gene expression patterns that are observed in bromodomain-containing proteins and associated with specific cancer types. We also mapped the protein–protein interaction network for the human bromodomain-containing proteins, discuss the cellular roles of these epigenetic regu- lators as part of nine different functional groups, and identify bromodomain-specific mechanisms in cancer development. Lastly, we summarize emerging strategies to target bromodomain proteins in cancer therapy, including those that may be essential for overcoming resistance. Overall, this review provides a timely discussion of the different mechanisms of bromodomain-containing pro- Citation: Boyson, S.P.; Gao, C.; teins in cancer, and an updated assessment of their utility as a therapeutic target for a variety of Quinn, K.; Boyd, J.; Paculova, H.; cancer subtypes.
    [Show full text]
  • Four Enzymes Cooperate to Displace Histone H1 During the First Minute of Hormonal Gene Activation
    Downloaded from genesdev.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Four enzymes cooperate to displace histone H1 during the first minute of hormonal gene activation Guillermo Pablo Vicent,1,2 A. Silvina Nacht,1 Jofre Font-Mateu, Giancarlo Castellano, Laura Gaveglia, Cecilia Ballare´, and Miguel Beato Centre de Regulacio´ Geno` mica (CRG), Universitat Pompeu Fabra (UPF), E-08003 Barcelona, Spain Gene regulation by external signals requires access of transcription factors to DNA sequences of target genes, which is limited by the compaction of DNA in chromatin. Although we have gained insight into how core histones and their modifications influence this process, the role of linker histones remains unclear. Here we show that, within the first minute of progesterone action, a complex cooperation between different enzymes acting on chromatin mediates histone H1 displacement as a requisite for gene induction and cell proliferation. First, activated progesterone receptor (PR) recruits the chromatin remodeling complexes NURF and ASCOM (ASC-2 [activating signal cointegrator-2] complex) to hormone target genes. The trimethylation of histone H3 at Lys 4 by the MLL2/MLL3 subunits of ASCOM, enhanced by the hormone-induced displacement of the H3K4 demethylase KDM5B, stabilizes NURF binding. NURF facilitates the PR-mediated recruitment of Cdk2/CyclinA, which is required for histone H1 displacement. Cooperation of ATP-dependent remodeling, histone methylation, and kinase activation, followed by H1 displacement, is a prerequisite for the subsequent displacement of histone H2A/H2B catalyzed by PCAF and BAF. Chromatin immunoprecipitation (ChIP) and sequencing (ChIP-seq) and expression arrays show that H1 displacement is required for hormone induction of most hormone target genes, some of which are involved in cell proliferation.
    [Show full text]
  • DNA Methylation Regulates Discrimination of Enhancers From
    Sharifi-Zarchi et al. BMC Genomics (2017) 18:964 DOI 10.1186/s12864-017-4353-7 RESEARCHARTICLE Open Access DNA methylation regulates discrimination of enhancers from promoters through a H3K4me1-H3K4me3 seesaw mechanism Ali Sharifi-Zarchi1,2,3,4†, Daniela Gerovska5†, Kenjiro Adachi6, Mehdi Totonchi3, Hamid Pezeshk7,8, Ryan J. Taft9, Hans R. Schöler6,10, Hamidreza Chitsaz2, Mehdi Sadeghi8,11, Hossein Baharvand3,12* and Marcos J. Araúzo-Bravo5,13,14* Abstract Background: DNA methylation at promoters is largely correlated with inhibition of gene expression. However, the role of DNA methylation at enhancers is not fully understood, although a crosstalk with chromatin marks is expected. Actually, there exist contradictory reports about positive and negative correlations between DNA methylation and H3K4me1, a chromatin hallmark of enhancers. Results: We investigated the relationship between DNA methylation and active chromatin marks through genome- wide correlations, and found anti-correlation between H3K4me1 and H3K4me3 enrichment at low and intermediate DNA methylation loci. We hypothesized “seesaw” dynamics between H3K4me1 and H3K4me3 in the low and intermediate DNA methylation range, in which DNA methylation discriminates between enhancers and promoters, marked by H3K4me1 and H3K4me3, respectively. Low methylated regions are H3K4me3 enriched, while those with intermediate DNA methylation levels are progressively H3K4me1 enriched. Additionally, the enrichment of H3K27ac, distinguishing active from primed enhancers, follows a plateau in the lower range of the intermediate DNA methylation level, corresponding to active enhancers, and decreases linearly in the higher range of the intermediate DNA methylation. Thus, the decrease of the DNA methylation switches smoothly the state of the enhancers from a primed to an active state.
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • Quantitative Proteomics Methods for the Analysis of Histone Post-Translational Modifications
    Université de Montréal Quantitative Proteomics Methods for the Analysis of Histone Post-translational Modifications par Nebiyu Ali Abshiru Département de Chimie Faculté des arts et des sciences Thèse présentée à la Faculté des études supérieures et postdoctorales en vue de l’obtention du grade de philosophiae doctor (Ph.D.) en chimie Septembre 2015 ©Nebiyu Ali Abshiru, 2015 i Résumé Les histones sont des protéines nucléaires hautement conservées chez les cellules des eucaryotes. Elles permettent d’organiser et de compacter l’ADN sous la forme de nucléosomes, ceux-ci representant les sous unités de base de la chromatine. Les histones peuvent être modifiées par de nombreuses modifications post-traductionnelles (PTMs) telles que l’acétylation, la méthylation et la phosphorylation. Ces modifications jouent un rôle essentiel dans la réplication de l’ADN, la transcription et l’assemblage de la chromatine. L’abondance de ces modifications peut varier de facon significative lors du developpement des maladies incluant plusieurs types de cancer. Par exemple, la perte totale de la triméthylation sur H4K20 ainsi que l’acétylation sur H4K16 sont des marqueurs tumoraux spécifiques a certains types de cancer chez l’humain. Par conséquent, l’étude de ces modifications et des événements determinant la dynamique des leurs changements d’abondance sont des atouts importants pour mieux comprendre les fonctions cellulaires et moléculaires lors du développement de la maladie. De manière générale, les modifications des histones sont étudiées par des approches biochimiques telles que les immuno-buvardage de type Western ou les méthodes d’immunoprécipitation de la chromatine (ChIP). Cependant, ces approches présentent plusieurs inconvénients telles que le manque de spécificité ou la disponibilité des anticorps, leur coût ou encore la difficulté de les produire et de les valider.
    [Show full text]
  • Inferring Causal Relationships Among Different Histone Modifications and Gene Expression
    Downloaded from genome.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Inferring Causal Relationships among Different Histone Modifications and Gene Expression Hong Yu*, Shanshan Zhu*, Bing Zhou*, Huiling Xue and Jing-Dong J. Han Chinese Academy of Sciences Key Laboratory of Molecular Developmental Biology, Center for Molecular Systems Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Datun Road, Beijing, 100101, China * These authors contributed equally to this work. Correspondence should be addressed to J.-D.J.H ([email protected]). Running title: histone modification and gene expression network Keywords: histone modification, gene expression, Bayesian network, causal relationship, epigenetics 1 Downloaded from genome.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract Histone modifications are major epigenetic factors regulating gene expression. They play important roles in maintaining stem cell pluripotency and in cancer pathogenesis. Different modifications may combine to form complex ‘histone codes’. Recent high throughput technologies, such as ‘ChIP-chip’ and ‘ChIP-seq’, have generated high resolution maps for many histone modifications on the human genome. Here we use these maps to build a Bayesian network to infer causal and combinatorial relationships among histone modifications and gene expression. A pilot network derived by the same method among polycomb group (PcG) genes and H3K27 trimethylation is accurately supported by current literature. Our unbiased network model among histone modifications is also well supported by cross validation results. It not only confirmed already known relationships, such as those of H3K27me3 to gene silencing, H3K4me3 to gene activation, and the effect of bivalent modification of both H3K4me3 and H3K27me3, but also identified many other relationships that may predict new epigenetic interactions important in epigenetic gene regulation.
    [Show full text]
  • Anti-H3r2me2(Asym) Antibody
    FOR RESEARCH USE ONLY! 01/20 Anti-H3R2me2(asym) Antibody CATALOG NO.: A2025-100 (100 µl) BACKGROUND DESCRIPTION: Histones are basic nuclear proteins that are responsible for the nucleosome structure of the chromosomal fiber in eukaryotes. Nucleosomes consist of approximately 146 bp of DNA wrapped around a histone octamer composed of pairs of each of the four core histones (H2A, H2B, H3, and H4). The chromatin fiber is further compacted through the interaction of a linker histone, H1, with the DNA between the nucleosomes to form higher order chromatin structures. This gene is intronless and encodes a replication-dependent histone that is a member of the histone H3 family. Transcripts from this gene lack poly A tails; instead, they contain a palindromic termination element. This gene is located separately from the other H3 genes that are in the histone gene cluster on chromosome 6p22-p21.3. H3.4; H3/g; H3FT; H3t; HIST3H3; Histone H3; HIST1H3A ALTERNATE NAMES: ANTIBODY TYPE: Polyclonal HOST/ISOTYPE: Rabbit / IgG IMMUNOGEN: A synthetic methylated peptide targeting residues around Arginine 2 of human Histone H3 MOLECULAR WEIGHT: 17 kDa PURIFICATION: Affinity purified FORM: Liquid FORMULATION: In PBS with 0.02% sodium azide, 50% glycerol, pH 7.3 SPECIES REACTIVITY: Human, Mouse, Rat STORAGE CONDITIONS: Store at -20ºC. Avoid freeze / thaw cycles APPLICATIONS AND USAGE: WB 1:500 - 1:2000, IHC 1:50 - 1:200, IF 1:50 - 1:200 Note: This information is only intended as a guide. The optimal dilutions must be determined by the user Western blot analysis of H3R2me2(asym) expression in Dot-blot analysis of methylation peptides using Anti- HeLa cells and H3 protein.
    [Show full text]
  • SETD2 Mutations in Primary Central Nervous System Tumors Angela N
    Viaene et al. Acta Neuropathologica Communications (2018) 6:123 https://doi.org/10.1186/s40478-018-0623-0 RESEARCH Open Access SETD2 mutations in primary central nervous system tumors Angela N. Viaene1, Mariarita Santi1, Jason Rosenbaum2, Marilyn M. Li1, Lea F. Surrey1 and MacLean P. Nasrallah2,3* Abstract Mutations in SETD2 are found in many tumors, including central nervous system (CNS) tumors. Previous work has shown these mutations occur specifically in high grade gliomas of the cerebral hemispheres in pediatric and young adult patients. We investigated SETD2 mutations in a cohort of approximately 640 CNS tumors via next generation sequencing; 23 mutations were detected across 19 primary CNS tumors. Mutations were found in a wide variety of tumors and locations at a broad range of allele frequencies. SETD2 mutations were seen in both low and high grade gliomas as well as non-glial tumors, and occurred in patients greater than 55 years of age, in addition to pediatric and young adult patients. High grade gliomas at first occurrence demonstrated either frameshift/truncating mutations or point mutations at high allele frequencies, whereas recurrent high grade gliomas frequently harbored subclones with point mutations in SETD2 at lower allele frequencies in the setting of higher mutational burdens. Comparison with the TCGA dataset demonstrated consistent findings. Finally, immunohistochemistry showed decreased staining for H3K36me3 in our cohort of SETD2 mutant tumors compared to wildtype controls. Our data further describe the spectrum of tumors in which SETD2 mutations are found and provide a context for interpretation of these mutations in the clinical setting. Keywords: SETD2, Histone, Brain tumor, Glioma, Epigenetics, H3K36me3 Introduction (H3K36me3) in humans.
    [Show full text]
  • Enhancer Priming by H3K4 Methylation Safeguards Germline Competence
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.07.192427; this version posted July 7, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Title: Enhancer priming by H3K4 methylation safeguards germline competence 1* 1 1,2 Authors: Tore Bleckwehl ​ ,​ Kaitlin Schaaf ,​ Giuliano Crispatzu ,​ Patricia ​ ​ ​ ​ 1,3 1,4 5 5 Respuela ,​ Michaela Bartusel ,​ Laura Benson ,​ Stephen J. Clark ,​ Kristel M. ​ ​ ​ ​ 6 7 1,8 7,9 Dorighi ,​ Antonio Barral ,​ Magdalena Laugsch ,​ Miguel Manzanares ,​ Joanna ​ ​ ​ ​ 6,10,11 5,12,13 1,3,14* Wysocka ,​ Wolf Reik ,​ Álvaro Rada-Iglesias ​ ​ ​ ​ ​ ​ Affiliations: 1 Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany. 2 Department of Internal Medicine 2, University Hospital Cologne. 3 Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), CSIC/University of Cantabria, Spain. 4 Department of Biology, Massachusetts Institute of Technology, USA. 5 Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK. 6 Department of Chemical and Systems Biology, Stanford University School of Medicine, USA. 7 Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain. 8 Institute of Human Genetics, Heidelberg University Hospital, Germany. 9 Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC-UAM, Spain. 10 Department of Developmental Biology, Stanford University School of Medicine, USA. 11 Howard Hughes Medical Institute, Stanford University School of Medicine, USA. 12 Centre for Trophoblast Research, University of Cambridge, CB2 3EG, UK 13 Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK.
    [Show full text]
  • Deciphering the Histone Code to Build the Genome Structure
    bioRxiv preprint doi: https://doi.org/10.1101/217190; this version posted November 20, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Deciphering the histone code to build the genome structure Kirti Prakasha,b,c,* and David Fournierd,* aPhysico-Chimie Curie, Institut Curie, CNRS UMR 168, 75005 Paris, France; bOxford Nanoimaging Ltd, OX1 1JD, Oxford, UK; cMicron Advanced Bioimaging Unit, Department of Biochemistry, University of Oxford, Oxford, UK; dFaculty of Biology and Center for Computational Sciences, Johannes Gutenberg University Mainz, 55128 Mainz, Germany; *Correspondence: [email protected], [email protected] Histones are punctuated with small chemical modifications that alter their interaction with DNA. One attractive hypothesis stipulates that certain combinations of these histone modifications may function, alone or together, as a part of a predictive histone code to provide ground rules for chromatin folding. We consider four features that relate histone modifications to chromatin folding: charge neutrali- sation, molecular specificity, robustness and evolvability. Next, we present evidence for the association among different histone modi- fications at various levels of chromatin organisation and show how these relationships relate to function such as transcription, replica- tion and cell division. Finally, we propose a model where the histone code can set critical checkpoints for chromatin to fold reversibly be- tween different orders of the organisation in response to a biological stimulus. DNA | nucleosomes | histone modifications | chromatin domains | chro- mosomes | histone code | chromatin folding | genome structure Introduction The genetic information within chromosomes of eukaryotes is packaged into chromatin, a long and folded polymer of double-stranded DNA, histones and other structural and non- structural proteins.
    [Show full text]
  • Symmetric-Di-Methyl-Histone H3 (Arg8) Rabbit
    Symmetric -Di-methyl-Histone H3 (Arg8) rabbit pAb ( H3R8me2(sy) pAb) www.ptm-biolab.com.cn Cat#: PTM-672 [email protected] Species: Rabbit 0571-2883 3567 Size: 100 μl Form: supplied in liquid form Recommend Species Swissprot Molecular Application Immunogen dilution Reactivity ID Weight H3R8me2(sy) WB, Dot, ELISA 1:2000 for WB H, M, R, Mk P68431 17 KDa -KLH **Species reactivity is determined by WB. Kept at -20oC after reconstituted. *** Anti-rabbit secondary antibodies must be used to detect this antibody. Source/Purification: This product is produced by immunizing rabbits with a synthetic di-methyl(sy) peptide corresponding to residues surrounding Arg8 of human histone H3. Antibodies are purified by protein A-conjugated agarose followed by di-methylated(sy) histone H3 (Arg8) peptide affinity chromatography. Recommended Applications: ELISA, Dot blot, Western blot. Recommended antibody dilution: WB: 1:2000 NOTE: For WB, incubate membrane with diluted antibody in 5% nonfat milk, 1 x TBS, 0.1% Tween- 20 for two hours at room temperature with gentle shaking. Prepare working dilution immediately before use. Use at an assay dependent concentration. Optimal dilutions/concentrations should be determined by the end user. Not yet tested in other applications Scientific Description: Proteins post-translational Modification (PTM) is an important reversible event controlling protein activity. The amino-terminal tails of core histones undergo multiple modifications in multiple sites, termed as “histone code” or “epigenetic code”. Lysine methylation or arginine methylation in core histones is a major determinant for the formation of active and inactive regions of the genome and therefore plays vital roles in multiple cellular events.
    [Show full text]