Stable Caenorhabditis Elegans Chromatin Domains Separate Broadly Expressed and Developmentally Regulated Genes

Total Page:16

File Type:pdf, Size:1020Kb

Stable Caenorhabditis Elegans Chromatin Domains Separate Broadly Expressed and Developmentally Regulated Genes Stable Caenorhabditis elegans chromatin domains separate broadly expressed and developmentally regulated genes Kenneth J. Evansa, Ni Huanga, Przemyslaw Stempora, Michael A. Chesneya, Thomas A. Downa, and Julie Ahringera,1 aDepartment of Genetics, The Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, United Kingdom Edited by Paul W. Sternberg, California Institute of Technology, Pasadena, CA, and approved September 20, 2016 (received for review May 24, 2016) Eukaryotic genomes are organized into domains of differing three organisms (10). However, because chromatin differences structure and activity. There is evidence that the domain organiza- also exist, the jointly derived chromatin states are not ideal for tion of the genome regulates its activity, yet our understanding of C. elegans-specific analyses, and no other C. elegans chromatin domain properties and the factors that influence their formation is state maps have been published. poor. Here, we use chromatin state analyses in early embryos and Previous studies have described broad properties of C. elegans third-larval stage (L3) animals to investigate genome domain orga- genome organization. The distal “arms” and central regions of nization and its regulation in Caenorhabditis elegans.Atbothstages the autosomal chromosomes show differences in transcriptional we find that the genome is organized into extended chromatin activity, chromatin composition, and recombination rate (6, 7, domains of high or low gene activity defined by different subsets 11, 13, 14). Central regions have higher average gene expression, of states, and enriched for H3K36me3 or H3K27me3, respectively. moderate enrichment of histone modifications associated with The border regions between domains contain large intergenic re- active transcription, and lower meiotic recombination than distal gions and a high density of transcription factor binding, suggesting arm regions. In contrast, most features associated with hetero- a role for transcription regulation in separating chromatin domains. chromatin, such as H3K9 methylation and nuclear envelope Despite the differences in cell types, overall domain organization is association, are found on the chromosome arms (7, 15). How- remarkably similar in early embryos and L3 larvae, with conserva- ever, the chromosome arms are not purely heterochromatic. tion of 85% of domain border positions. Most genes in high- Actively transcribed genes reside on the chromosome arms and activity domains are expressed in the germ line and broadly across these genes are marked by histone modifications associated with cell types, whereas low-activity domains are enriched for genes gene activity, as in the central regions (7). In addition, the X that are developmentally regulated. We find that domains are chromosome shows extensive chromatin differences compared regulated by the germ-line H3K36 methyltransferase MES-4 with autosomes because of dosage compensation (16). These and that border regions show striking remodeling of H3K27me1, previous studies have provided a large-scale picture of C. elegans supporting roles for H3K36 and H3K27 methylation in regulating chromosome organization. domain structure. Our analyses of C. elegans chromatin domain Here, we investigate C. elegans chromatin and genome organi- structure show that genes are organized by type into domains zation and its regulation through the generation and analyses of that have differing modes of regulation. C. elegans-specific chromatin state maps for early embryos (EE) and third larval (L3) stages. As in other organisms, chromatin chromatin states | chromatin domains | C. elegans | boundary states correlate with many biological features, including enhancers, promoters, transcription elongation, gene ends, repeat regions, he complete genome sequence, which provides the information and inactive genes. Analyzing patterns of states revealed that Tnecessary for constructing an organism, is interpreted in the chromatin domains of differing activity separate germ-line and context of chromatin. Covalent modifications of histone tails and broadly expressed genes from developmentally regulated genes. histone variants can regulate or reflect genome function, and so are The properties of domains and the border regions between them markers of chromatin state and genomic activity (1). For example, H3K4me3 often marks active promoters, H3K36me3 transcription Significance elongation, and H3K27me3 Polycomb-silenced regions. Previous studies have shown that genomic regions of similar activity harbor Genomes are organized into domains of different structure and shared combinations of modifications, termed chromatin states, activity, yet our understanding of their formation and regulation and that subdividing the genome according to these combinations is poor. We show that Caenorhabditis elegans chromatin domain is a powerful method for annotation and uncovering novel func- – organization in early embryos and third-larval stage animals is tional regions (2 5). Here, we apply chromatin state mapping to remarkably similar despite the two developmental stages con- two developmental stages of the model organism Caenorhabditis taining very different cell types. Chromatin domains separate elegans and use the resulting maps to investigate genome domain genes into those with stable versus developmentally regulated organization and its regulation. expression. Analyses of chromatin domain structure suggest C. elegans is highly amenable for global studies of chromatin that transcription regulation and germ-line chromatin regulation structure and function because it has a small, well-annotated ge- × play roles in separating chromatin domains. Our results further nome (30 smaller than human), and work of the modENCODE our understanding of genome domain organization. consortium has provided a large number of datasets mapping the locations of chromatin-associated factors, such as histone modi- Author contributions: K.J.E., T.A.D., and J.A. designed research; K.J.E. and N.H. performed fications and transcription factors (TFs) (6–10). C. elegans chro- research; K.J.E., N.H., P.S., M.A.C., and J.A. analyzed data; and K.J.E. and J.A. wrote matin shows features in common with those of other organisms, the paper. such as the type of marking at regulatory regions and at active or The authors declare no conflict of interest. inactive genes (7, 10–12). Additionally, the derivation of a single This article is a PNAS Direct Submission. set of chromatin states for C. elegans, Drosophila, and human using 1To whom correspondence should be addressed. Email: [email protected]. a single joint genome analysis and data from eight histone modi- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. fications highlighted the common properties of chromatin in the 1073/pnas.1608162113/-/DCSupplemental. E7020–E7029 | PNAS | Published online October 25, 2016 www.pnas.org/cgi/doi/10.1073/pnas.1608162113 Downloaded by guest on September 30, 2021 point to roles for transcription regulatory regions and germ-line for each stage we generated a separate map for autosomes and the PNAS PLUS chromatin marking in domain separation. Our results provide a X chromosome (Fig. 1A, Fig. S1,andDataset S1). The EE and L3 framework for future studies of chromatin structure and function autosomal chromatin states show much greater similarity to each in C. elegans. other than do the EE and L3 chromosome X states (Figs. S1 and S2), consistent with alterations in chromatin structure and marking Results induced by dosage compensation after the EE stage (16). The Twenty State Models of C. elegans Chromatin. To investigate features chromatin states were annotated by analyzing the associations of and domain organization of C. elegans chromatin, we derived 20 states with a range of different genomic features (Fig. 1 and Figs. state EE and L3 chromatin state maps, using hidden Markov S1, S3,andS4). As well as differences in enrichment levels for models (HMM) and ChIP data for 17 histones or modifications histone modifications, the states show differences in median length (Materials and Methods). Patterns and levels of enrichment of many (250–1,250 bp), genomic coverage (2.2–9%), and GC content (25– histone modifications differ on C. elegans autosomes compared with 44%) (Fig. S1). Below, we briefly describe chromatin state anno- the X chromosome, reflecting dosage compensation (7, 16–18), tation and properties using L3 autosomes as an example. We then which caused whole-genome chromatin state maps to subdivide into use the states to investigate autosomal chromatin domain organi- separate autosomal and X-chromosome–specific states. Therefore, zation and its regulation. A Annotation 1 2 3 4 Txn elongation II: exon and intron 5 Txn elongtion III: exon and gene end 6 Txn elongation IV low expression and repeats 7 Txn elongation V: introns and repeats 8 9 Enhancer II: intergenic GENETICS 10 Enhancer III: weak 11 12 13 Retrotransposons, pseudogenes, H3K9me3, H3K27me3 14 15 Repeats, RNA pseudogenes, H3K9me2 16 17 Pc/H3K27me3 I:low expr/silent and pseudogenes 18 Pc/H3K27me3 II: low expr/silent 19 20 H3 HTZ1 H4K8ac H3K27ac H3K4me2 H3K4me1 H3K9me3 H3K9me2 H3K4me3 H4K20me1 H3K36me2 H3K79me3 H3K36me3 H3K36me1 H3K79me2 H3K27me3 H3K27me1 -2 -1 0 1 2 B Chr I: 7,996,451- 8,337,598 genes EE domains L3 domains EE states 1-5 EE states 6-15 EE states 16-20 L3 states 1-5 L3 states 6-15 L3 states 16-20 EE H3K36me3 EE H3K27me3 L3 H3K36me3 L3 H3K27me3 Fig. 1. Chromatin states
Recommended publications
  • Active Motif Technical Data Sheet (TDS)
    Histone H4K8ac antibody (pAb) Catalog Nos: 61103, 61104 Quantities: 100 µg, 10 µg RRID: AB_2793506 Purification: Protein A Chromatography Isotype: IgG Host: Rabbit Application(s): ChIP, ChIP-Seq, DB, ICC, IF, WB Concentration: 1 µg/µl Reactivity: Human, Mouse, Wide Range Predicted Molecular Weight: 8 kDa Background: Histone H4 is one of the core components of the nucleosome. The nucleosome is the smallest subunit of chromatin and consists of 147 base pairs of DNA wrapped around an octamer of core histone proteins (two each of Histone H2A, Histone H2B, Histone H3 and Histone H4). Histone H1 is a linker histone, present at the interface between the nucleosome core and DNA entry/exit points; it is responsible for establishing higher-order chromatin structure. Chromatin is subject to a variety of chemical modifications, including post-translational modifications of the histone proteins and the methylation of cytosine residues in the DNA. Reported histone modifications include acetylation, methylation, phosphorylation, ubiquitylation, glycosylation, ADP-ribosylation, carbonylation and SUMOylation; they play a major role in regulating gene expression. Lysine N-ε-acetylation is a dynamic, reversible and tightly regulated protein and histone modification that plays a major role in chromatin remodeling and in the regulation of gene expression in various cellular functions. The chromatin-remodeling complex SWI/SNF is recruited to promoters through the interaction of the bromodomain of the protein BRG1, belonging to the SWI/SNF complex, and CBP-acetylated histone H4 Lysine 8, leading to a chromatin remodeling. Immunogen: This Histone H4 acetyl Lys8 antibody was raised against a peptide containing acetyl Lys8 of human Histone H4.
    [Show full text]
  • Active Motif Technical Data Sheet (TDS)
    Histone H4K8ac antibody (mAb) Catalog No: 61525 Quantity: 100 µg RRID: AB_2793669 Purification: Protein G Chromatography Clone: MABI 0408 Host: Mouse Isotype: IgG1 Concentration: 0.66 µg/µl Application(s): ChIP, DB, WB Molecular Weight: 8 kDa Reactivity: Human, Wide Range Predicted Background: Histone H4 is one of the core components of the nucleosome. The nucleosome is the smallest subunit of chromatin and consists of 147 base pairs of DNA wrapped around an octamer of core histone proteins (two each of Histone H2A, Histone H2B, Histone H3 and Histone H4). Histone H1 is a linker histone, present at the interface between the nucleosome core and DNA entry/exit points; it is responsible for establishing higher-order chromatin structure. Chromatin is subject to a variety of chemical modifications, including post-translational modifications of the histone proteins and the methylation of cytosine residues in the DNA. Reported histone modifications include acetylation, methylation, phosphorylation, ubiquitylation, glycosylation, ADP-ribosylation, carbonylation and SUMOylation; they play a major role in regulating gene expression. Lysine N-ε-acetylation is a dynamic, reversible and tightly regulated protein and histone modification that plays a major role in chromatin remodeling and in the regulation of gene expression in various cellular functions. The chromatin-remodeling complex SWI/SNF is recruited to promoters through the interaction of the bromodomain of the protein BRG1, belonging to the SWI/SNF complex, and CBP-acetylated histone H4 Lysine 8, leading to a chromatin remodeling. Immunogen: This antibody was raised against a synthetic peptide containing acetyl-lysine 8 of human Histone H4. Buffer: Purified IgG in PBS with 30% glycerol and 0.035% sodium azide.
    [Show full text]
  • Glycolytic Metabolism Influences Global Chromatin Structure
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No.6 Glycolytic metabolism influences global chromatin structure Xue-Song Liu, John B. Little, Zhi-Min Yuan Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA 02115, USA Correspondence to: Zhi-Min Yuan, e-mail: [email protected] Keywords: glycolysis, acetylation, chromatin structure, chemosensitivity Received: September 29, 2014 Accepted: December 15, 2014 Published: January 13, 2015 ABSTRACT Metabolic rewiring, specifically elevated glycolytic metabolism is a hallmark of cancer. Global chromatin structure regulates gene expression, DNA repair, and also affects cancer progression. But the interrelationship between tumor metabolism and chromatin architecture remain unclear. Here we show that increased glycolysis in cancer cells promotes an open chromatin configuration. Using complementary methods including Micrococcal nuclease (MNase) digestion assay, electron microscope and immunofluorescence staining, we demonstrate that glycolysis inhibition by pharmacological and genetic approaches was associated with induction of compacted chromatin structure. This condensed chromatin status appeared to result chiefly from histone hypoacetylation as restoration of histone acetylation with an HDAC inhibitor reversed the compacted chromatin state. Interestingly, glycolysis inhibition-induced chromatin condensation impeded DNA repair efficiency leading to increased sensitivity of cancer cells to DNA damage drugs, which may represent a novel molecular mechanism that can be exploited for cancer therapy. INTRODUCTION Tumor cells metabolize most glucose into lactate and thus generate abundant glycolytic intermediates as Pathologist have observed for long time that the precursors for macromolecular biosynthesis, which nucleus of cancer cells show distinct morphological enables tumor cells to meet their increased anabolic and alterations compared to the nucleus of normal cells, and energetic demands due to rapid tumor growth [8].
    [Show full text]
  • H4k8ac Antibody
    08/14 For research use only BioVision H4K8ac Antibody ALTERNATE NAMES: Histone H4 CATALOG #: 6807-50 AMOUNT: 50 µl HOST/ISOTYPE: Rabbit To determine the titer, an ELISA was performed using a IMMUNOGEN: KLH-conjugated synthetic peptide of Histone H4 containing the HeLa cells (15 µg) were analysed by serial dilution of the antibody. The antigen used was a acetylated lysine 8 WB blot using the H4K8ac antibody. peptide containing the histone modification of interest. By plotting the absorbance against the antibody dilution the FORM: Liquid titer of the antibody was estimated to be 1:17,500. FORMULATION: In PBS with 0.05% (W/V) sodium azide. PURIFICATION: Whole antiserum from rabbit SPECIES REACTIVITY: Human. STORAGE CONDITIONS: Store at -20°C; for long storage, store at -80°C. Avoid multiple freeze-thaw cycles. DESCRIPTION: Histones are the main constituents of the protein part of chromosomes of eukaryotic cells. They are rich in the amino acids arginine and lysine and have been greatly conserved during evolution. Histones pack the DNA into tight masses of chromatin. Histone tails undergo numerous post-translational modifications, which either directly or indirectly alter chromatin structure to facilitate transcriptional activation or repression or other nuclear processes. In addition to the genetic code, combinations of the different histone modifications ChIP assays were performed using human reveal the so-called “histone code”. Histone methylation and demethylation is dynamically osteosarcoma (U2OS) cells and the antibody and regulated by respectively histone methyl transferases and histone demethylases. Acetylation optimized PCR primer sets for qPCR. A titration of the A Dot Blot analysis was performed to test the of histone H4 is associated with active gene transcription.
    [Show full text]
  • RNF8 Regulates Active Epigenetic Modifications and Escape Gene Activation from Inactive Sex Chromosomes in Post-Meiotic Spermatids
    Downloaded from genesdev.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press RNF8 regulates active epigenetic modifications and escape gene activation from inactive sex chromosomes in post-meiotic spermatids Ho-Su Sin,1,2,3 Artem Barski,3,4,5 Fan Zhang,3,6 Andrey V. Kartashov,3,4,5 Andre Nussenzweig,7 Junjie Chen,8 Paul R. Andreassen,4,5,6 and Satoshi H. Namekawa1,2,3,9 1Division of Reproductive Sciences, 2Division of Developmental Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229, USA; 3Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 49229, USA; 4Division of Allergy and Immunology, 5Division of Human Genetics, 6Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 45229, USA; 7Laboratory of Genome Integrity, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA; 8Department of Experimental Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA Sex chromosomes are uniquely subject to chromosome-wide silencing during male meiosis, and silencing persists into post-meiotic spermatids. Against this background, a select set of sex chromosome-linked genes escapes silencing and is activated in post-meiotic spermatids. Here, we identify a novel mechanism that regulates escape gene activation in an environment of chromosome-wide silencing in murine germ cells. We show that RNF8- dependent ubiquitination of histone H2A during meiosis establishes active epigenetic modifications, including dimethylation of H3K4 on the sex chromosomes. RNF8-dependent active epigenetic memory, defined by dimethylation of H3K4, persists throughout meiotic division.
    [Show full text]
  • New Developments in Epigenetics and Potential Clinical Applications
    NEW DEVELOPMENTS IN EPIGENETICS AND POTENTIAL CLINICAL APPLICATIONS Susan E. Bates, M.D. Columbia University Medical Center, New York, NY and J.J.P Bronx VA Medical Center, Bronx, NY TARGETING THE EPIGENOME What do we mean? “Targeting the Epigenome” - Meaning that we identify proteins that impact transcriptional controls that are important in cancer. “Epigenetics” – Meaning the right genes expressed at the right time, in the right place and in the right quantities. This process is ensured by an expanding list of genes that themselves must be expressed at the right time and right place. Think of it as a coordinated chromatin dance involving DNA, histone proteins, transcription factors, and over 700 proteins that modify them. Transcriptional Control: DNA Histone Tail Modification Nucleosomal Remodeling Non-Coding RNA HISTONE PROTEIN FAMILY 146 bp DNA wrap around an octomer of histone proteins H2A, H2B, H3, H4 are core histone families H1/H5 are linker histones >50 variants of the core histones Some with unique functions Post translational modification of “histone tails” key to gene expression Post-translational modifications include: – Acetylation – Methylation – Ubiquitination – Phosphorylation – Citrullation – SUMOylation – ADP-ribosylation THE HISTONE CODE Specific histone modifications determine function H3K9Ac, H3K27Ac, H3K36Ac H3K4Me3 – Gene activation H3K36Me2 – Inappropriate gene activation H3K27Me3 – Gene repression; Inappropriate gene repression H2AX S139Phosphorylation – associated with DNA double strand break, repair http://www.slideshare.net/jhowlin/eukaryotic-gene-regulation-part-ii-2013 KEY MODIFICATION - SPECIFIC FUNCTIONS H3K36Me: ac Active transcription, me H3K27Me: RNA elongation me Key Silencing ac ac me me Residue me me H3K4Me: H3K4 ac me Key Activating me me me Residue me H3K9 me H3K36 H3K9Ac:: H3K27 Activating Residue H3K18 H4K20 Activation me me me ac Modified from Mosammaparast N, et al.
    [Show full text]
  • Transcription Shapes Genome-Wide Histone Acetylation Patterns
    ARTICLE https://doi.org/10.1038/s41467-020-20543-z OPEN Transcription shapes genome-wide histone acetylation patterns Benjamin J. E. Martin 1, Julie Brind’Amour 2, Anastasia Kuzmin1, Kristoffer N. Jensen2, Zhen Cheng Liu1, ✉ Matthew Lorincz 2 & LeAnn J. Howe 1 Histone acetylation is a ubiquitous hallmark of transcription, but whether the link between histone acetylation and transcription is causal or consequential has not been addressed. 1234567890():,; Using immunoblot and chromatin immunoprecipitation-sequencing in S. cerevisiae, here we show that the majority of histone acetylation is dependent on transcription. This dependency is partially explained by the requirement of RNA polymerase II (RNAPII) for the interaction of H4 histone acetyltransferases (HATs) with gene bodies. Our data also confirms the targeting of HATs by transcription activators, but interestingly, promoter-bound HATs are unable to acetylate histones in the absence of transcription. Indeed, HAT occupancy alone poorly predicts histone acetylation genome-wide, suggesting that HAT activity is regulated post- recruitment. Consistent with this, we show that histone acetylation increases at nucleosomes predicted to stall RNAPII, supporting the hypothesis that this modification is dependent on nucleosome disruption during transcription. Collectively, these data show that histone acetylation is a consequence of RNAPII promoting both the recruitment and activity of histone acetyltransferases. 1 Department of Biochemistry and Molecular Biology, Life Sciences Institute, Molecular
    [Show full text]
  • H4k8ac Antibody
    09/14 For research use only BioVision H4K8ac Antibody ALTERNATE NAMES: Histone H4 CATALOG #: 6878-25 AMOUNT: 25 µg HOST/ISOTYPE: Rabbit IMMUNOGEN: Polyclonal antibody raised in rabbit against the region of histone H4 containing the acetylated lysine 20 (H4K8ac), using a KLH-conjugated synthetic peptide. HeLa cell s histone extracts (15 µg) were ChIP assays were performed using HeLa cells and the FORM: Liquid analysed by Western blot using the antibody and optimized PCR primer sets for qPCR. A antibody. The position of the protein of titration of the antibody consisting of 1, 2, 5, and 10 µl FORMULATION: In PBS with 0.05% (W/V) sodium azide and 0.05% ProClin 300. interest is indicated on the right; the per ChIP experiment was analysed. IgG (1 µg/IP) was marker (in kDa) is shown on the left. used as negative control. The Fig shows the recovery, PURIFICATION: Affinity purified expressed as a % of input (the relative amount of IP DNA compared to input DNA after qPCR analysis). SPECIES REACTIVITY: Human, mouse. STORAGE CONDITIONS: Store at -20°C; for long storage, store at -80°C. Avoid multiple freeze-thaw cycles. DESCRIPTION: Histones are the main constituents of the protein part of chromosomes of eukaryotic cells. They are rich in the amino acids arginine and lysine and have been greatly conserved during evolution. Histones pack the DNA into tight masses of chromatin. Two core histones of each class H2A, H2B, H3 and H4 assemble and are wrapped by 146 base pairs of DNA to form one octameric nucleosome. Histone tails undergo numerous post-translational modifications, which either directly or indirectly alter chromatin structure to facilitate transcriptional activation or repression or other nuclear processes.
    [Show full text]
  • 1 Establishment of Regions of Genomic Activity During the Drosophila Maternal to 2 Zygotic Transition 3 4 Xiao-Yong Li1, Melissa M
    1 Establishment of regions of genomic activity during the Drosophila maternal to 2 zygotic transition 3 4 Xiao-Yong Li1, Melissa M. Harrison2, Jacqueline E. Villata1, Tommy Kaplan3*, Michael B. Eisen1,4,5,6* 5 6 1. Howard Hughes Medical Institute, University of California, Berkeley, CA 7 2. Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 8 3. School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel 9 4. Department of Molecular and Cell Biology, University of California, Berkeley, CA 10 5. Department of Integrative Biology, University of California, Berkeley, CA 11 6. QB3 Institute, University of California, Berkeley, CA 12 * co-senior authors 13 14 corresponding authors: MBE([email protected]), TK([email protected]) 15 16 1 17 Abstract 18 19 We describe the genome-wide distributions and temporal dynamics of nucleosomes and 20 post-translational histone modifications throughout the maternal-to-zygotic transition in 21 embryos of Drosophila melanogaster. At mitotic cycle 8, when few zygotic genes are being 22 transcribed, embryonic chromatin is in a relatively simple state: there are few nucleosome 23 free regions, undetectable levels of the histone methylation marks characteristic of mature 24 chromatin, and low levels of histone acetylation at a relatively small number of loci. Histone 25 acetylation increases by cycle 12, but it is not until cycle 14 that nucleosome free regions 26 and domains of histone methylation become widespread. Early histone acetylation is 27 strongly associated with regions that we have previously shown to be bound in early 28 embryos by the maternally deposited transcription factor Zelda, suggesting that Zelda 29 triggers a cascade of events, including the accumulation of specific histone modifications, 30 that plays a role in the subsequent activation of these sequences.
    [Show full text]
  • Histone Onco-Modifications
    Oncogene (2011) 30, 3391–3403 & 2011 Macmillan Publishers Limited All rights reserved 0950-9232/11 www.nature.com/onc REVIEW Histone onco-modifications JFu¨llgrabe, E Kavanagh and B Joseph Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden Post-translational modification of histones provides an (Luger et al., 1997). In eukaryotes, an octamer of important regulatory platform for processes such as gene histones-2 copies of each of the four core histone expression, DNA replication and repair, chromosome proteins histone 2A (H2A), histone 2B (H2B), histone condensation and segregation and apoptosis. Disruption of 3 (H3) and H4—is wrapped by 147 bp of DNA to form these processes has been linked to the multistep process of a nucleosome, the fundamental unit of chromatin carcinogenesis. We review the aberrant covalent histone (Kornberg and Lorch, 1999). Nucleosomal arrays were modifications observed in cancer, and discuss how these observed with electron microscopy as a series of ‘beads epigenetic changes, caused by alterations in histone- on a string’, the ‘beads’ being the individual nucleo- modifying enzymes, can contribute to the development of somes and the ‘string’ being the linker DNA. Linker a variety of human cancers. As a conclusion, a new histones, such as histone H1, and other non-histone terminology ‘histone onco-modifications’ is proposed to proteins can interact with the nucleosomal arrays to describe post-translational modifications of histones, further package the nucleosomes to form higher-order which have been linked to cancer. This new term would chromatin structures (Figure 1a). take into account the active contribution and importance Histones are no longer considered to be simple ‘DNA- of these histone modifications in the development and packaging’ proteins; they are recognized as being progression of cancer.
    [Show full text]
  • Ab115118 – Histone H4 (Acetyl K8) Quantification Kit (Colorimetric)
    ab115118 – Histone H4 (acetyl K8) Quantification Kit (Colorimetric) Instructions for Use For the measurement of global acetylation of Histone H4K8 using a variety of mammalian cells including fresh and frozen tissues, and cultured adherent and suspension cells This product is for research use only and is not intended for diagnostic use. Version 1 Last Updated 2 September 2014 Table of Contents INTRODUCTION 1. BACKGROUND 2 2. ASSAY SUMMARY 3 GENERAL INFORMATION 3. PRECAUTIONS 4 4. STORAGE AND STABILITY 4 5. MATERIALS SUPPLIED 5 6. MATERIALS REQUIRED, NOT SUPPLIED 5 7. LIMITATIONS 6 8. TECHNICAL HINTS 6 ASSAY PREPARATION 9. REAGENT PREPARATION 7 10. SAMPLE PREPARATION 7 11. PLATE PREPARATION 8 ASSAY PROCEDURE 12. ASSAY PROCEDURE 9 DATA ANALYSIS 13. ANALYSIS 10 RESOURCES 14. TROUBLESHOOTING 11 15. NOTES 12 Discover more at www.abcam.com 1 INTRODUCTION 1. BACKGROUND Acetylation of histones including histone H3 and H4 has been involved in the regulation of chromatin structure and recruitment of transcription factors to gene promoters. Histone acetyltransferases (HAT) and histone deacetylases (HDACs) play a critical role in the control of histone H4 acetylation at multiple sites. Acetylation of histone H4 at lysine 8 (H4K8) reflects the hyperacetylated state in histone H4 and is strongly correlated with active states of genes. H4K8 acetylation is related to DNA repair and increased H4K8 acetylation is also observed in cancer and inflammatory diseases. In the absence of this modification, cellular survival following DNA damage is impaired. Histone H4K8 acetylation may be increased by inhibition of HDACs and decreased by HAT inhibition. Thus, quantitative detection of global acetyl histone H4K8 would provide useful information for better understanding epigenetic regulation of gene activation and for developing HAT or HDAC-targeted drugs.
    [Show full text]
  • The Majority of Histone Acetylation Is a Consequence of Transcription
    bioRxiv preprint doi: https://doi.org/10.1101/785998; this version posted September 30, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. The majority of histone acetylation is a consequence of transcription Benjamin J.E. Martin§, Julie Brind’Amour†, Kristoffer N. Jensen†, Anastasia Kuzmin§, Zhen Cheng Liu§, Matthew Lorincz† and LeAnn J. Howe§* §Department of Biochemistry and Molecular Biology and †Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, B.C., Canada, V6T 1Z3 bioRxiv preprint doi: https://doi.org/10.1101/785998; this version posted September 30, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract Histone acetylation is a ubiquitous hallmark of transcriptional activity, but whether the link is of a causal or consequential nature is still a matter of debate. In this study we resolve this question. Using both immunoblot analysis and chromatin immunoprecipitation-sequencing (ChIP-seq) in S. cerevisiae, we show that the majority of histone acetylation is dependent on transcription. Loss of histone H4 acetylation upon transcription inhibition is partially explained by depletion of histone acetyltransferases (HATs) from gene bodies, implicating transcription in HAT targeting. Despite this, HAT occupancy alone poorly predicts histone acetylation, suggesting that HAT activity is regulated at a step post- recruitment.
    [Show full text]