• No results found

University of Groningen Epigenetic editing Cano Rodriguez, David

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Epigenetic editing Cano Rodriguez, David"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Epigenetic editing

Cano Rodriguez, David

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2017

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Cano Rodriguez, D. (2017). Epigenetic editing: Towards sustained gene expression reprogramming in

diseases. University of Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)
(3)
(4)

12

General Introduction

The field of epigenetics studies changes in gene expression that are independent of the underlying DNA sequence. Although all cells within an organism contain the same DNA, there are many different cell types, tissues and organs present. Different subsets of genes that are activated depending on their regulation lead to different properties and therefore to different cell types and tissues1,2. The organization of DNA and

histones into chromatin is an important aspect in gene regulation, through which the access of trans-cription complexes to the DNA can be regulated. Chromatin serves as a macromolecular scaffold in nuclei of eukaryotic cells that consists of high order structures called nucleosomes. The nucleosomal architecture is built up of octameric units of dimers of the four core histones H2A, H2B, H3 and H4 wra-pped around by 147 base pairs of DNA3. Higher-order folding of the nucleosomes can result in either

less condensed, active euchromatin or highly condensed, silent heterochromatin. Epigenetic gene regulation is mediated by several mechanisms including DNA methylation and the post-translational modifications (PTM) of the histone tails, both of which may activate or repress transcription according to the specific context4. These modifications can directly or indirectly influence chromatin structure

by modulating DNA-histone interactions and form docking sites to facilitate recruitment of proteins to the chromatin5,6. This form of epigenetic regulation is important for the maintenance of cell identity

and therefore it is implicated in processes such as proliferation, development and differentiation7,8.

DNA methylation occurs predominantly at CpG dinucleotides and is mediated by DNA methyltransferases (DNMTs). DNMTs are enzymes that catalyze the transfer of methyl groups from S-adenosyl-L-methionine to the 5’ position of cytosine, resulting in 5-methylcytosine (5mC)9,10. The DNMT family in humans

consists of DNMT3A and 3B, which catalyze de novo DNA methylation during embryonic development, and DNMT1, which is responsible for the maintenance of established DNA methylation patterns during replication (i.e. with participation of DNMT3A and 3B). Another family member, DNMT3L (DNMT3-like), resembles DNMT3 enzymes while itself lacking catalytic activity11-13.

Methylated CpG dinucleotides are not distributed evenly across the genome. There are re-petitive and foreign elements that are mainly hypermethylated, and focused locations within gene promoters that are hypomethylated. These focused locations are short (approx. 1 kilobase (kb)) CpG-rich regions called CpG islands (CGIs)14-17. The presence of 5mC within the germline is thought to lead to the loss of CpGs, because 5mC can be deaminated spontaneously or enzymatically, leading to the conversion of 5mC to thymine. CGIs are consequently thought to exist because they are never or only transiently methylated in the germline and therefore escape the pressure to be converted 18,19. Such CGIs are found in approx. 40% of promoters in the mammalian genome. Their methyla-tion has been associated with long-term transcripmethyla-tional silencing and a closed chromatin state, which is known to occur e.g. at imprinted genes, genes on the inactive X chromosome and genes that are exclusively expressed in germ cells14,18,20. Interestingly, differences in CpG-density within promoter regions have been associated with specific methylation and expression patterns. This was shown by use of methylated DNA immunoprecipitation and microarray analysis in human primary fibroblasts19. It appears that low-CpG (i.e. non-CGI) promoters are often methylated, even in the active state.

Epigenetic

gene regulation

DNA

(5)

13

Histone

modifications

The N-terminal tails of the histone proteins may be chemically modified at lysine, arginine or serine residues by for instance, acetylation, me-thylation, ubiquitination and phosphorylation5,8. The pattern of PTM’s that

occur on one or more histone tails form a so-called histone code that can be deciphered by other proteins These proteins can alter the struc-ture of higher-order chromatin and recruit effector molecules22. Histone PTM’s are reversible and

can be associated with either euchromatin or heterochromatin and therefore can control gene ex-pression. The transcriptionally inert heterochromatin state is negatively regulated by hypoacetyla-tion of H3 and H4 and tri-methylahypoacetyla-tion of lysine residues 9 and 27 on histone H3 (H3K9me3, H3K-27me3) and tri-methylation of lysine residue 20 on histone H4 (H4K20me3). In contrast, acetylation of histones H3 and H4 and tri-methylation of lysine residue 4 and 79 on histone H3 (H3K4me3, H3K79me3) situated at transcription start sites (TSS) are associated with transcriptionally active euchromatin23-28. Through these modifications, histones can regulate gene transcription by

chan-ging chromatin structure, altering electrostatic charge or by providing protein recognition sites7. The machinery that regulates histone methylation generally consists of different classes of histone methyltransferases (HMTs) and histone demethylases (HDMs)29. Histone 3 lysine 4 di- and

trimethylation (H3K4me2/3) is mainly associated with transcriptionally active genes, although in mam-mals much lower levels of this mark have also been observed at silent genes. Nevertheless, the mark is thought to positively regulate transcription by recruitment of nucleosome remodeling enzymes and histone acetylases30. The establishment of the histone mark is catalyzed by the trithorax group (trxG)

of proteins through activity of the SET1 or MLL family members, which reside within multimeric protein complexes. The first of these complexes containing SET1 was identified in yeast and was named COMPASS, for complex of proteins associated with SET131. HDMs known to abolish the H3K4me2/3 mark are KDM1/LSD1 family members (remove H3K4me1/2) and members of the JmjC-domain con-taining family (remove H3K4me3)18. H3K4me3 is one of the most studied active histone marks, and

one of the most known models for histone PTMs crosstalk. The tight regulation between monoubi-quitination of H2B and the trimethylation of H3K4 and H3K79 is a hallmark of active promoters32-36.

Histone repressive marks have been well studied, such as methylation of H3K9 and H3K27. In mammalian cells, H3K9me3 is a hallmark of heterochromatin and is also required for transcriptional silencing of genes and retroviral elements37,38. Transcriptional repression

invol-ves heterochromatin protein 1 (HP1), which specifically binds to methylated H3K9, which would, in turn, recruit DNA methyltransferases28,39-41. Between the enzymes identified to be required for

H3K9 methylation are SETDB1, SUV39H1 and G9a. Additionally, H3K27me3 marks are associa-ted with transcriptional repression, which is thought to be mediaassocia-ted by the promotion of a com-pact chromatin structure42. These marks are induced by Polycomb-group (PcG) proteins residing

within the polycomb repressive complex 2 (PRC2). PRC2 consists of Suz12, EED and EZH2, the latter being a SET1 domain-containing HMT43. The repressive H3K27me3 mark can

furthermo-re be furthermo-recognized by PRC1, which mediates the ubiquitylation of H2AK119, a mark that is thought to further maintain silencing18,43. Binding of PRC1 is thought to block the recruitment of

transcrip-tional activation factors and to prevent initiation of transcription by RNA polymerase II44. The

de-methylation of H3K27 is catalyzed by the KDM6 family, better known as UTX and JmjD318.

Different histone modifications may influence each other (e.g. H3K36 methyla-tion inhibits H3K27 methylamethyla-tion) or may influence other epigenetic modifiers or epigene-tic marks (e.g. H3K27me3 may recruit PRC1, and H3K4me3 may inhibit DNA methylation). Methylation of H3K4, H3K9 and H3K27 has been shown to mediate mitotic inheritance of linea-ge-specific gene expression patterns and accordingly have key developmental functions42. For

this reason, these epigenetic marks are important targets for epigenetic editing, in order to achie-ve sustained gene expression reprogramming. Moreoachie-ver, as cancer cells frequently remain in an undifferentiated state45, these histone marks are of particular interest in cancer research.

This implies that low concentrations of 5mC do not foreclose gene activity19,21. High-CpG (i.e CGI) promoters on the other hand are mostly unmethylated, even when inactive. This implies that the-se regions are somehow protected from methylation, not only in the germline, but also in somatic cells19. The fact that these CGI promoter genes can still be inactivated, suggests an additional le-vel of epigenetic control. Indeed, such a regulatory lele-vel is formed by the modification of histones.

(6)

14

Reading and writing

the epigenetic code

At certain stages of the cell cycle or in response to stimuli, the chromatin folding must decrease to facilitate access of appropriate factors or it must condense to render the DNA in-accessible to those factors. This level of regulation is achie-ved through the abundance and impact of specific PTM’s. This balance is regulated by the balance of enzymes that can ‘write’ or ‘erase’ a specific PTM22,46.

Writers such as acetylases, methylases and phosphorylases can introduce distinct PTM’s on his-tones. Erasers such as deacetylases, demethylases and phosphatases antagonize the function of writers by removing those histone modifications47.

Readers are proteins that contain conserved domains that can recognize histone modifications48.

These readers are for example bromodomains, chromodomains, PHD fingers or WD-40 repeats that can interpret PTMS’s to evoke specific functional outcomes such as remodeling of the chromatin, stabilization of the higher order-chromatin, further posttranslational modification (through writer or eraser) or other gene regulatory effects such as direct recruitment of RNA polymerase II machinery46.

Cancer

Epigenetics

Failure to correctly regulate chromatin condensation or decondensation can lead to an increase in DNA damage and abnormal gene expression, which on their turn can lead to genome instability. Genomic instability is implicated in many pathogenic events such as human syndromes and cancers49-51. Indeed it

is shown that there is a strong relationship between aberrant post-translational histone modifications and tumorigenesis. The gene expression abnormalities observed in cancer are frequently associated with aberrant epigenetic profiles52. For example, tumor suppressor genes can

frequently become silenced during tumor development. The promoter regions of these genes can be subject to hypermethylation, which is associated with gene silencing. In addition, a global DNA hypo-methylation is frequently involved in genomic instability and transformation into malignant cells. Mo-reover altered histone modification patterns also play a critical role in tumorigenesis of tumor suppres-sor genes and oncogenes, including an enrichment or loss of active or repressive histone marks53.

The fact that the various epigenetic marks are reversible makes it an ideal target for therapeutic intervention. Currently, US Food and drug administration (FDA)-approved epigenetic drugs, e.g. histo-ne deacetylase inhibitors and DNA demethylating agents have been demonstrated to have potent an-ti-cancer effects in clinics54-56. However, it has been demonstrated that the chemotherapeutic agents can

induce undesired off-target effects throughout the genome, e.g. induction of expression of the pro-me-tastatic gene Ezrin57. Therefore it is essential for future therapeutic intervention to target cancer in a

ge-ne-specific way. The interactions between proteins that can read and write or erase the code provide re-gulatory opportunities for the transmission of epigenetic marks along the genome, and the use of current targeting platforms has open new avenues to study the effect of epigenetic marks in health and disease.

Epigenetic Editing

An elegant method to modulate gene expression at will is depic-ted by artificial transcription factors (ATFs). ATFs that are engi-neered to bind to a promoter region of a gene of interest are able

Figure 1. Epigenetic editing. By coupling or fusing an

epigenetic effector domain to a DNA binding domain, any region in the human genome can be targeted and gene expression can be modulated

1

to specifically affect gene expression. Previous-ly, the potentials of ATF treatment have been de-monstrated for several genes involved in cancer, including SOX2, Maspin, C13ORF18 and EP-B41L358-61. In general, ATFs consist of a

DNA-bin-ding domain (DBD) and a transcriptional activator/ repressor to positively or negatively regulate trans-cription. Yet, a major disadvantage of ATF treat-ment is the transient effect on gene expression modulation. Hence, a sustained gene expression modulation can only be achieved by reprogram-ming of the cellular epigenetic context in a mitoti-cally stable manner. The most promising approach to meet this requirement is illustrated by epige-netic editing62 (Figure 1) . The goal of epigenetic

editing is to rewrite epigenetic marks at any locus at will to permanently modulate the expression of endogenous genes. Since the dynamic remodeling of the chromatin landscape (and the stable CpG dinucleotide methylation) is tightly regulated by a conglomerate of enzymes and other macro-molecules, there is a huge array of epigenetic effector domains for gene expression modulation.

(7)

15 In order to rewrite the epigenetic landscape, the catalytic domain of an epigenetic writer or an eraser is fused to a gene-specific DBD to enforce the presence of this effector domain at a particular DNA sequence. Induced epigenetic changes can be assessed by chromatin immuno-precipitation (ChIP) or bisulfite sequencing and the effect of targeting epigenetic enzymes on gene expression can be assessed by measuring gene expression levels of genes that are in close proximity of the DBD recognition site. The most used targeting DBDs used are Zinc Finger Proteins (ZFPs)63, Transcription Activator Like Effectors

(TALEs)64 and the clustered, regularly interspaced, short palindromic repeat-CRISPR associated

pro-tein (CRISPR-Cas) RNA-guided endonuclease system that can be targeted to any loci of interest 65,66.

After the introduction of the concept by us, the field of epigenetic editing has been in increa-sed development67-74. Especially the introduction of a new, cheap and flexible DNA binding platform

(CRISPR-Cas) has sparked the interest in this approach.

Several genes and epigenetic enzymes have been used to repress or reactivate gene expression at will. Most of these studies, however, have only addressed short term epigenetic reprogram-ming and are lacking sustained gene expression modulation. For this reason we have deve-loped several platforms to address stable epigenetic reprogramming after epigenetic editing.

Aims and scope

of the thesis

The aim of this thesis was to induce stable activation or repression of endogenous target genes by epigenetic editing. We develop no-vel modalities of gene expression modulation that exploits epige-netics to induce inheritable and robust states of transcriptional re-programming at desired loci. To this end, we took advantage of our expertise in developing programmable DNA Binding Domain (such as ZFPs and CRISPR/dCas9) to target the promoter of endogenous genes, and couple these to effector domains from natura-lly occurring epigenetic enzymes (e.g. histone modifying enzyme and DNA methytransferases). Previous studies showed that ATFs can be used to modulate gene transcription. However, their tran-sient activity was associated with reacquisition of the previous transcriptional state. This outcome likely reflects the inability of the chosen transcriptional regulators to recreate self-propagating epi-genetic changes at the target loci. To overcome this limitation, we hypothesized that targeting of multiple epigenetic regulators might mimic natural conditions to allow the formation of transcriptional complexes capable of creating self-sustaining epigenetic reprogramming. The hit-and-run approach of our epigenetic platform should overcome the limitations associated with the current technologies, paving the way for its therapeutic application. Finally, this epigenetic editing platform can be rapidly adopted in basic biology to interrogate the function of mammalian regulatory elements and to un-derstand the mechanistic relationship among chromatin states, gene regulation and cell phenotype. The first three chapters deal with the overall development of the field of epigenetic editing. In Chap-ter 2, we set out to present the latest efforts and achievements in the field of epigenetic editing. In this review we aimed to show the current techniques and targeting platforms used, as well as their advantages and disadvantages. We also present the most used epigenetic enzymes targe-ted at different regulatory regions, the clinical application and the future perspectives. Chapter 3 aims to provide a clear protocol for the field covering the methodology to create Zinc Finger Pro-teins and to target the promoter of a hypermethylated gene (ICAM-1) using epigenetic editing to de-methylate DNA using the catalytic domain of Tet2. In Chapter 4, we aimed to increase effective-ness of epigenetic editing by identifying main barriers in targeting genes and subsequent epigenetic reprogramming. As DNA methylation at CpG islands has been shown to be a hurdle when targe-ting loci, addressing this issue will help in achieving the optimal possibilities for genome targetarge-ting. The next chapters serve as an example for the potential use of epigenetic editing in clinical research and basic fundamental biology research. Chapter 5 explores the possibility of targeting two different promoters of the RASSF1 gene, with the aim to demonstrate that two transcripts from the same gene, but regulated from different promoters, have opposing effects in cancer. It has already been proven by exogenous overexpression or silencing that the two transcript act differently in cells, and we aimed to show that endogenous regulation of the two promoters by epigenetic editing has the same results.

(8)

16

While the first promoter generates a transcript that has tumor suppressive activity, the shortest iso-form, transcribed from an inner promoter is thought to be an oncogene. In Chapter 6, we use epi-genetic editing to show the potential function of the NCO2 gene, which is overexpressed in cancer. The aim of this chapter was to identify the function of this gene by using epigenetic editing to repress its expression in cancer cells and evaluate their behavior. With these two chapters we aimed to show that some genes could serve not only as biomarkers but can also provide therapeutic targets. The last chapters depict the possibility to achieve sustained gene expression reprogramming with epigenetic editing. In Chapter 7 we aim to accomplish sustained transcriptional repression of a can-didate gene dysregulated in Chronic Obstructive Pulmonary Disorder (COPD) by means of compa-rison between the transcriptional repressor Super Krab Domain (SKD) and epigenetic enzymes. In Chapter 8, we investigate the capability of H3K4me3 to induce gene expression, and whether there is dependency on the chromatin microenvironment targeted. We also set out to unravel requirements that need to be achieved in order to stably activate gene expression from hypermethylated genes. In Chapter 9, the most important findings of this thesis are summarized and discussed in the context of existing literature. We also show the potential for future research from the findings of this thesis.

(9)

17

1 Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57-74, doi:10.1038/nature11247 (2012).

2 Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317-330, doi:10.1038/ nature14248 (2015).

3 Felsenfeld, G. & Groudine, M. Controlling the double helix. Nature 421, 448-453, doi:10.1038/nature01411 (2003).

4 Kouzarides, T. Chromatin modifications and their function. Cell 128, 693-705, doi:10.1016/j.cell.2007.02.005 (2007).

5 Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41-45, doi:10.1038/47412 (2000).

6 Turner, B. M. The adjustable nucleosome: an epigenetic signaling module. Trends Genet 28, 436-444, doi:10.1016/j.tig.2012.04.003 (2012).

7 Reik, W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447, 425-432, doi:10.1038/nature05918 (2007).

8 Turner, B. M. Cellular memory and the histone code. Cell 111, 285-291 (2002).

9 Smith, Z. D. & Meissner, A. DNA methylation: roles in mammalian development. Nat Rev Genet 14, 204-220, doi:10.1038/nrg3354 (2013).

10 Goll, M. G. & Bestor, T. H. Eukaryotic cytosine methyltransferases. Annu Rev Biochem 74, 481-514, doi:10.1146/ annurev.biochem.74.010904.153721 (2005).

11 Zhang, Y. et al. Chromatin methylation activity of Dnmt3a and Dnmt3a/3L is guided by interaction of the ADD domain with the histone H3 tail. Nucleic Acids Res 38, 4246-4253, doi:10.1093/nar/gkq147 (2010).

12 Ooi, S. K. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature

448, 714-717, doi:10.1038/nature05987 (2007).

13 Hashimoto, H., Vertino, P. M. & Cheng, X. Molecular coupling of DNA methylation and histone methylation.

Epigenomics 2, 657-669, doi:10.2217/epi.10.44 (2010).

14 Jones, P. A. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13, 484-492, doi:10.1038/nrg3230 (2012).

15 Bird, A. P. CpG-rich islands and the function of DNA methylation. Nature 321, 209-213, doi:10.1038/321209a0 (1986).

16 Thomson, J. P. et al. CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature 464, 1082-1086, doi:10.1038/nature08924 (2010).

17 Wrzodek, C. et al. Linking the epigenome to the genome: correlation of different features to DNA methylation of CpG islands. PLoS One 7, e35327, doi:10.1371/journal.pone.0035327 (2012).

18 van Vlerken, L. E., Hurt, E. M. & Hollingsworth, R. E. The role of epigenetic regulation in stem cell and cancer biology. J Mol Med (Berl) 90, 791-801, doi:10.1007/s00109-012-0917-9 (2012).

19 Weber, M. et al. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet 39, 457-466, doi:10.1038/ng1990 (2007).

(10)

18

20 Cedar, H. & Bergman, Y. Programming of DNA methylation patterns. Annu Rev Biochem 81, 97-117, doi:10.1146/ annurev-biochem-052610-091920 (2012).

21 Boyes, J. & Bird, A. Repression of genes by DNA methylation depends on CpG density and promoter strength: evidence for involvement of a methyl-CpG binding protein. EMBO J 11, 327-333 (1992).

22 Torres, I. O. & Fujimori, D. G. Functional coupling between writers, erasers and readers of histone and DNA methylation. Curr Opin Struct Biol 35, 68-75, doi:10.1016/j.sbi.2015.09.007 (2015).

23 Wang, Z. et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138, 1019-1031, doi:10.1016/j.cell.2009.06.049 (2009).

24 Martin, C. & Zhang, Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol 6, 838-849, doi:10.1038/nrm1761 (2005).

25 Lee, K. K. & Workman, J. L. Histone acetyltransferase complexes: one size doesn’t fit all. Nat Rev Mol Cell Biol

8, 284-295, doi:10.1038/nrm2145 (2007).

26 Rice, J. C. & Allis, C. D. Histone methylation versus histone acetylation: New insights into epigenetic regulation.

Current Opinion in Cell Biology 13, 263-273, doi:10.1016/S0955-0674(00)00208-8 (2001).

27 Bannister, A. J., Schneider, R. & Kouzarides, T. Histone methylation: dynamic or static? Cell 109, 801-806 (2002).

28 Kouzarides, T. Histone methylation in transcriptional control. Current opinion in genetics & development 12, 198-209, doi:10.1016/S0959-437X(02)00287-3 (2002).

29 Fischle, W., Wang, Y. & Allis, C. D. Binary switches and modification cassettes in histone biology and beyond.

Nature 425, 475-479, doi:10.1038/nature02017 (2003).

30 Shilatifard, A. Molecular implementation and physiological roles for histone H3 lysine 4 (H3K4) methylation.

Current Opinion in Cell Biology 20, 341-348, doi:10.1016/j.ceb.2008.03.019 (2008).

31 Shilatifard, A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu Rev Biochem 81, 65-95, doi:10.1146/annurev-biochem-051710-134100 (2012).

32 Lee, J.-S. et al. Histone Crosstalk between H2B Monoubiquitination and H3 Methylation Mediated by COM-PASS. Cell 131, 1084-1096, doi:10.1016/j.cell.2007.09.046 (2007).

33 van Ingen, H. et al. Structural insight into the recognition of the H3K4me3 mark by the TFIID subunit TAF3.

Structure (London, England : 1993) 16, 1245-1256, doi:10.1016/j.str.2008.04.015 (2008).

34 Cedar, H. & Bergman, Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev

Genet 10, 295-304, doi:10.1038/nrg2540 (2009).

35 Kim, J. et al. The n-SET Domain of Set1 Regulates H2B Ubiquitylation-Dependent H3K4 Methylation. Molecular

Cell 49, 1121-1133, doi:10.1016/j.molcel.2013.01.034 (2013).

36 Soares, L. M. & Buratowski, S. Histone Crosstalk: H2Bub and H3K4 Methylation. Molecular Cell 49, 1019-1020, doi:10.1016/j.molcel.2013.03.012 (2013).

37 Peters, A. H. et al. Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin. Nat

Genet 30, 77-80, doi:10.1038/ng789 (2002).

38 Nielsen, S. J. et al. Rb targets histone H3 methylation and HP1 to promoters. Nature 412, 561-565, doi:10.1038/35087620 (2001).

39 Lachner, M., O’Carroll, D., Rea, S., Mechtler, K. & Jenuwein, T. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116-120, doi:10.1038/35065132 (2001).

40 Bannister, A. J. et al. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain.

(11)

19

41 Hathaway, Nathaniel A. et al. Dynamics and Memory of Heterochromatin in Living Cells. Cell 149, 1447-1460, doi:10.1016/j.cell.2012.03.052 (2012).

42 Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells.

Cell 125, 315-326, doi:10.1016/j.cell.2006.02.041 (2006).

43 Bracken, A. P. & Helin, K. Polycomb group proteins: navigators of lineage pathways led astray in cancer. Nat

Rev Cancer 9, 773-784, doi:10.1038/nrc2736 (2009).

44 Kondo, Y. et al. Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet 40, 741-750, doi:10.1038/ng.159 (2008).

45 Teschendorff, A. E. et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hall-mark of cancer. Genome Res 20, 440-446, doi:10.1101/gr.103606.109 (2010).

46 Thompson, L. L., Guppy, B. J., Sawchuk, L., Davie, J. R. & McManus, K. J. Regulation of chromatin structure via histone post-translational modification and the link to carcinogenesis. Cancer Metastasis Rev 32, 363-376, doi:10.1007/s10555-013-9434-8 (2013).

47 Tarakhovsky, A. Tools and landscapes of epigenetics. Nat Immunol 11, 565-568, doi:10.1038/ni0710-565 (2010).

48 Taverna, S. D., Li, H., Ruthenburg, A. J., Allis, C. D. & Patel, D. J. How chromatin-binding modules interpret his-tone modifications: lessons from professional pocket pickers. Nat Struct Mol Biol 14, 1025-1040, doi:10.1038/ nsmb1338 (2007).

49 Iacobuzio-Donahue, C. A. Epigenetic changes in cancer. Annu Rev Pathol 4, 229-249, doi:10.1146/annurev. pathol.3.121806.151442 (2009).

50 You, J. S. & Jones, P. A. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell 22, 9-20, doi:10.1016/j.ccr.2012.06.008 (2012).

51 Dekker, A. D., De Deyn, P. P. & Rots, M. G. Epigenetics: the neglected key to minimize learning and memory deficits in Down syndrome. Neurosci Biobehav Rev 45, 72-84, doi:10.1016/j.neubiorev.2014.05.004 (2014). 52 Feinberg, A. P. & Tycko, B. The history of cancer epigenetics. Nat Rev Cancer 4, 143-153, doi:10.1038/nrc1279

(2004).

53 Dawson, M. A. & Kouzarides, T. Cancer epigenetics: from mechanism to therapy. Cell 150, 12-27, doi:10.1016/j. cell.2012.06.013 (2012).

54 Kelly, T. K., De Carvalho, D. D. & Jones, P. A. Epigenetic modifications as therapeutic targets. Nat Biotechnol

28, 1069-1078, doi:10.1038/nbt.1678 (2010).

55 Altucci, L. & Rots, M. G. Epigenetic drugs: from chemistry via biology to medicine and back. Clin Epigenetics 8, 56, doi:10.1186/s13148-016-0222-5 (2016).

56 Heerboth, S. et al. Use of epigenetic drugs in disease: an overview. Genet Epigenet 6, 9-19, doi:10.4137/GEG. S12270 (2014).

57 Yu, Y. et al. Epigenetic drugs can stimulate metastasis through enhanced expression of the pro-metastatic Ezrin gene. PLoS One 5, e12710, doi:10.1371/journal.pone.0012710 (2010).

58 Rivenbark, A. G. et al. Epigenetic reprogramming of cancer cells via targeted DNA methylation. Epigenetics 7, 350-360, doi:10.4161/epi.19507 (2012).

59 Stolzenburg, S. et al. Targeted silencing of the oncogenic transcription factor SOX2 in breast cancer. Nucleic

Acids Research 40, 6725-6740, doi:10.1093/nar/gks360 (2012).

60 Huisman, C. et al. Functional validation of putative tumor suppressor gene C13ORF18 in cervical cancer by Artificial Transcription Factors. Mol Oncol 7, 669-679, doi:10.1016/j.molonc.2013.02.017 (2013).

61 Huisman, C. et al. Prolonged re-expression of the hypermethylated gene EPB41L3 using artificial transcription factors and epigenetic drugs. Epigenetics 10, 384-396, doi:10.1080/15592294.2015.1034415 (2015).

(12)

20

62 de Groote, M. L., Verschure, P. J. & Rots, M. G. Epigenetic Editing: targeted rewriting of epigenetic marks to modulate expression of selected target genes. Nucleic Acids Research 40, 10596-10613, doi:10.1093/nar/ gks863 (2012).

63 Gersbach, C. A., Gaj, T. & Barbas, C. F. Synthetic zinc finger proteins: the advent of targeted gene regulation and genome modification technologies. Acc Chem Res 47, 2309-2318, doi:10.1021/ar500039w (2014). 64 Sun, N. & Zhao, H. Transcription activator-like effector nucleases (TALENs): a highly efficient and versatile tool

for genome editing. Biotechnol Bioeng 110, 1811-1821, doi:10.1002/bit.24890 (2013).

65 Hsu, P. D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 31, 827-832, doi:10.1038/nbt.2647 (2013).

66 Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering.

Cell 157, 1262-1278, doi:10.1016/j.cell.2014.05.010 (2014).

67 Voigt, P. & Reinberg, D. Epigenome editing. Nat Biotechnol 31, 1097-1099, doi:10.1038/nbt.2756 (2013). 68 Rusk, N. CRISPRs and epigenome editing. Nat Methods 11, 28 (2014).

69 Falahi, F., Sgro, A. & Blancafort, P. Epigenome Engineering in Cancer: Fairytale or a Realistic Path to the Clinic?

Frontiers in Oncology 5, 1-11, doi:10.3389/fonc.2015.00022 (2015).

70 Jurkowski, T. P., Ravichandran, M. & Stepper, P. Synthetic epigenetics-towards intelligent control of epigenetic states and cell identity. Clin Epigenetics 7, 18, doi:10.1186/s13148-015-0044-x (2015).

71 Köeferle, A., Stricker, S. H. & Beck, S. Brave new epigenomes: the dawn of epigenetic engineering. Genome

Med 7, 59, doi:10.1186/s13073-015-0185-8 (2015).

72 Zentner, G. E. & Henikoff, S. Epigenome editing made easy. Nat Biotechnol 33, 606-607, doi:10.1038/nbt.3248 (2015).

73 Kungulovski, G. & Jeltsch, A. Epigenome Editing: State of the Art, Concepts, and Perspectives. Trends Genet

32, 101-113, doi:10.1016/j.tig.2015.12.001 (2016).

74 Thakore, P. I., Black, J. B., Hilton, I. B. & Gersbach, C. A. Editing the epigenome: technologies for programma-ble transcription and epigenetic modulation. Nat Methods 13, 127-137, doi:10.1038/nmeth.3733 (2016).

(13)

Referenties

GERELATEERDE DOCUMENTEN

Known and unknown functions of TET dioxygenases: the potential of inducing DNA modifications in Epigenetic Editing..

Epigenetic mechanisms, including DNA methylation and post-translational histone modifications, regulate gene expression and mounting evidence indicates epigenetics to play an

Epigenetic editing: Towards sustained gene expression reprogramming in diseases.. University

Below we discuss the most used epigenetic effector domains in epigenetic editing (Table 1)... Epigenetic editing tools available. a) Zinc finger proteins can recognize

Specifically in cancer, transcriptional silencing by hypermethylation has been reported for key regu- latory genes related with cell cycle or apoptosis; in this kind of

faster to design and produce a short sgRNA for any given new target sequence compared to ZFPs and TALEs. However, among these technologies, Cas9 is the biggest, around 160

genous gene-targeting with zinc finger proteins in four cell lines. a) Activation of endogenous RASS- F1A in four cancer cell lines (two hypermethylated cell lines

During the screening of the en- tire antibody library on tissue microarrays (TMAs) carrying cancerous and normal formalin-fixed para- ffin-embedded (FFPE) samples (5 samples /tumor