• No results found

University of Groningen Opportunities and Challenges of Epigenetic Editing in Human Diseases Goubert, Désirée

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Opportunities and Challenges of Epigenetic Editing in Human Diseases Goubert, Désirée"

Copied!
19
0
0

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

Hele tekst

(1)

Opportunities and Challenges of Epigenetic Editing in Human Diseases

Goubert, Désirée

DOI:

10.33612/diss.173201281

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: 2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Goubert, D. (2021). Opportunities and Challenges of Epigenetic Editing in Human Diseases: Towards the Curable Genome. University of Groningen. https://doi.org/10.33612/diss.173201281

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)

1

147043 Goubert BNW.indd 6

(3)

G

Geenneerraall iinnttrroodduuccttiioonn

General Introduction

(4)

An introduction to Epigenetics

Epigenetics is a layer on top of the DNA that causes heritable, yet reversible changes in gene expression, without affecting the primary DNA sequence (1, 2). Epigenetic gene regulation is very stable but also highly flexible. Epigenetic states can persist over many cell generations, whilst on the other hand epigenetic regulations make sure that genes can dynamically respond to external signals (3). During mammalian development cellular differentiation is regulated by the epigenetics. Even though most cells within complex multicellular organisms contain the same DNA, there are many different cell types, making up the various tissues and organs. Epigenetic modifications underlie cell identity by switching cell-type specific genes on or off (4, 5). Furthermore, this regulation of gene expression patterns is maintained throughout cell divisions (e.g. proliferating liver cells, remain liver cells) and can even be inherited by the offspring (6, 7).

Epigenetic modifications are chemical or physical changes of the DNA and chromatin catalysed by enzymes in a highly dynamic way. Epigenetics represents a molecular explanation as to why healthy living and making responsible choices is so important and has even been defined as “The missing link between nature and nurture” (8, 9). Epigenetic marks instruct gene expression patterns by stimulating or denying transcription of genes through regulating access of transcription factors (TFs), transcriptional initiation complexes, and RNA polymerase to DNA (10). The accessibility of DNA thus causes densely packed genes not to be expressed (or less efficiently), whilst a loosely packed gene will lead to an increased expression. The epigenetic signatures, which are unique for each cell-type, are placed by epigenetic enzymes that write or erase epigenetic marks. These marks include methylation of cytosine (described in detail in Chapter 3) and a wide variety of posttranslational

modifications of histone proteins, where the DNA is wrapped around. Epigenetic marks cause functionally relevant changes to the genome that do not involve changes in the primary DNA sequence (11, 12).

(5)

Epigenetics in human diseases

Disruptions in the epigenetic layer can have severe consequences on gene expression and cellular behaviour and contribute to the development of many different diseases (13-16) as well as sensitivity to treatment and development of resistance (17, 18). In cancer for example, tumour suppressor genes, which normally block cancer growth, are often DNA hypermethylated, which is associated with a silenced gene expression state (19). Unravelling the link between DNA methylation and cancer progression is aiding to the development of clinical applications, for example by using site specific presence or absence of DNA methylation as a diagnostic marker (20, 21). As epigenetic changes are a naturally occurring and reversable process, they present an attractive target for therapeutic intervention. It is therefore not surprising that from 2006 onwards so called epi-drugs have entered the clinical area (22). These drugs target epigenetic enzymes that write or erase epigenetic marks. Such epi-drugs include DNA methyltransferase inhibitors (DNMTIs: e.g. 5-azacytidine and decitabine), histone deacetylase inhibitors (HDACIs: e.g., vorinostat, Zolinza and romidepsin) and histone methyltransferase inhibitors (HMTIs) and they are used in the clinic for the treatment of haematological malignancies, solid tumours and neurological disorders (22-33). Unfortunately, wide-spread clinical application of epi-drugs is hampered by the fact that these drugs act genome-wide rather than in a gene-specific manner, causing unwanted effects, including upregulation of pro-metastatic genes (34, 35) or of genes encoding drug resistance-associated proteins (36).

Epigenetic Editing

Therefore, more targeted epigenetic approaches are desirable and one such approach is Epigenetic Editing. In this approach, which is described in more detail in Chapter 2, epigenetic

marks are placed or removed from specific target genes, without changing the primary DNA sequence (37-43). Artificial Transcription Factors (ATFs), combined with DNA binding platforms, can also be used to modulate gene expression in a targeted way (44-47). Such ATFs represent transient transcriptional repressors (e.g. super KRAB domain (SKD)) (48, 49) or transient transcriptional activators (e.g. the tetramer of herpes simplex virus protein VP16 (VP64)) fused to DNA binding domains. In contrast to ATFs, epigenetic writers or erasers

(6)

change the epigenetic modifications at a target gene, and are therefore more likely to be maintained by cellular processes even after removal of the Epigenetic Editing tool (50-52). In comparison to epigenetic enzymes, ATFs do not possess any enzymatic activity on their own, but they recruit other epigenetic players to have an effect on gene expression. Epigenetic Editing tools consist out of at least two components: a DNA binding component, which functions as a molecular GPS, guiding the tool towards the intended genomic location; and an effector domain (ED), which places or erases epigenetic marks. The two DNA-binding platforms used in this thesis are the CRISPR-dCas9 platform (53) and Zinc Finger (ZF) proteins. CRISPR is an abbreviation of Clustered Regularly Interspaced Short Palindromic Repeats and has been first reported as a bacterial immune response against viruses. Every time a bacterium gets invaded by a virus short DNA copies are made of the viral DNA. The RNA products of these DNA copies, called single guide RNAs (sgRNAs), recognise the viral DNA when it invades a second time and recruit the CRISPR binding partner Cas9. Cas9 then makes a double stranded cut, disabling the DNA and thereby inactivating the virus, rendering it harmless. Since 2012 Jennifer Doudna and Emmanuelle Charpentier demonstrated that it is possible to use this mechanism in human cells for bioengineering by delivering the Cas9 enzyme together with a sgRNA that is designed to target a gene of interest (54). Currently, CRIPSR-Cas9 is used in clinical trials for gene editing purposes, mostly ex vivo (patient cells are taken out of the body, treated with gene editing compounds, and put back into the patient) (55-58), but recently also for the first time in vivo (in this case administrated to the eye of a patient) (59). In gene editing, the Cas9 enzyme is targeted towards a mutated gene, where it makes a cut, rendering the gene inactive and incapable of performing its harmful function (Figure 1). It is also possible to insert a correct gene by providing the cell, after Cas9 DNA cleavage, with a correct piece of DNA. This corrected gene is inserted in the cleaved DNA via its homologues overhangs with the cleaved site and used to repair the cut (60).

The CRISPR-dCas9 platform used for Epigenetic Editing consists of the same DNA-binding component, in the form of sgRNAs, however the Cas9 enzyme has been deactivated. Two mutations introduced into the Cas9 enzyme render it incapable of making a cut, but it still can be used as a molecular shuttle bringing for example an epigenetic ED to a specific location within the genome. Epigenetic EDs can then affect the epigenetic environment of that specific

(7)

gene, and increase or decrease its expression. ZF proteins are small naturally occurring transcription factors, forming the largest group of all transcription factors in the human genome (61), that can also be fused to epigenetic EDs or ATFs and designed to target gene-specific DNA sequences (44, 62, 63). The relatively small size and low immunogenicity of ZF proteins are an advantage compared to other DNA-targeting proteins (39, 63).

Figure 1: Schematic representation of gene editing using CRISPR-Cas9. The Cas9 enzyme inactivates the

mutated target gene by making a cut in the DNA (Left side) or corrects a gene when provided with the correct donor DNA (Right side) sgRNA: Single Guide RNA.

Targeted overwriting of epigenetic marks using these platforms holds promise to permanently reprogram gene expression and has successfully been applied in diverse therapeutic models (64, 65). For example, Thakore et al. successfully silenced the transcription of Pcsk9, a regulator of cholesterol levels, in the liver of adult mice. Systemic administration of inactivated viral particles carrying dCas9-KRAB and sgRNAs targeting of the

Pcsk9 gene led to a moderate immune response and an efficient repression of Pcsk9 (64). Xu et al. showed that in vivo reactivation of several anti-fibrotic genes is also possible using

Epigenetic Editing. Targeted demethylation using TET enzymes led to specific gene reactivation and attenuation of fibrosis (65). Epigenetic Editing thus has the potential to develop into clinically relevant one-and-done approaches, where patients are treated once, after which the modified epigenetic marks are remembered and maintained on the target

(8)

gene, and could lead to breakthroughs in curing diseases that are currently uncurable: “The curable epigenome”.

Epigenetics in the development of endocrine therapy resistance

One of the diseases that this thesis focuses on is breast cancer, more specifically on oestrogen receptor alpha (ERα) positive (ER+) breast cancer (which accounts for 70% of all breast cancer cases). These ER+ tumours depend on the female growth hormone oestrogen for their growth and can be treated with endocrine therapy (66). Endocrine therapy in a premenopausal setting makes use of drugs that selectively modulate oestrogen receptor binding (e.g. Tamoxifen) or downregulate the oestrogen receptor (e.g. Fulvestrant), in a postmenopausal setting drugs are used that inhibit the synthesis of the hormone oestrogen (e.g. Aromatase inhibitors such as Letrazol). Unfortunately, over 40% of patients who receive endocrine therapy eventually develop resistance to the treatment (67), which is accompanied by poor prognosis (17). For therapy resistance to arise, a tumour cell undergoes molecular changes, or has intrinsically present characteristics that allow its continuous cell proliferation under treatment conditions, providing these resistant tumour cells with a selective advantage. Acquired resistance to endocrine therapy is a long-term process in which genetic alterations are expected to act synergistically with epigenetic changes (17). Epigenetic reprogramming has been shown to contribute to the development of endocrine therapy resistance by inducing changes in gene regulatory networks (68-70). For example as shown by Magnani et

al. endocrine therapy resistant cells activate lipid synthesis (endogenous cholesterol) through

alterations in epigenetic histone modifications, both in resistant breast cancer cells and in patients (69). The increased cholesterol synthesis caused an activation of the oestrogen receptor, circumventing the reliance of the cells in the presence of oestrogen.

Also other epigenetic mechanisms including DNA hypermethylation, overexpression of histone deacetylases, enhancer regulation, non‑coding RNAs, chromatin remodelling, post‑translational histone modifications and histone variants have been shown to contribute to the development of endocrine therapy resistance (71). Stone et al. showed that DNA hypermethylation in endocrine resistant cells mainly occurred at oestrogen-responsive enhancers (72). Enhancers are regulatory DNA sequences that can alter the transcription of

(9)

an associated gene when they are activated. DNA methylation of these enhancers led to reduced oestrogen receptor binding and subsequently reduced expression of key regulators of ERα activity. Furthermore, significantly higher DNA methylation levels in these enhancers were also noted in breast cancer patients that relapsed from endocrine treatment (72). Yamamoto et al. investigated the role of the histone H3 lysine 4 (H3K4) demethylase JARID1B in luminal breast tumours (73). They showed that JARID1B is frequently amplified and overexpressed in luminal breast cancer and that a high JARID1B activity is associated with resistance to endocrine therapy and a worse clinical outcome. An additional level of epigenetic complexity is added by histone variants, which can replace original histones and contribute to changes in compaction of the chromatin structure. Both histone variants H2A.Z (an H2A variant) and HIST1H2BE (an H2B variant) have been linked to endocrine resistance (74, 75). H2A.Z overexpression led to an increased proliferation under oestrogen-deprived conditions (74) and HIST1H2BE was overexpressed in endocrine-resistant cell lines, as well as in tumours from patients that relapsed after aromatase inhibitor treatment (75).

These and other studies show the high urge to further identify which epigenetic changes are responsible for the development of endocrine therapy resistance and to develop diagnostic (71) and therapeutic tools (27). To this end, the international research consortium called EpiPredict “Epigenetic regulation of endocrine therapy resistance in breast cancer”, was initiated of which this PhD project is part. This innovative training network (ITN) is funded by EU H2020 MSCA-ITN-2014 and coordinated by Dr PJ Verschure, UvA, NL. In this consortium, 12 PhD students from eight different countries and scientific institutes have studied the role of epigenetic regulation in resistance development for endocrine therapy in ER+ breast cancer. My specific role as ”Early Stage Researcher number 8” (ESR8) from Work Package 5 was to design, construct and test gene-specific epigenetic interference tools to prevent and/or reverse endocrine therapy resistance. Together with the scopes of EpiPredict this is further elaborated on in Chapter 2.

As mentioned earlier, one of the epigenetic marks, and perhaps also the most well-known one, is DNA methylation, and changes in DNA methylation have been reported in multiple tumours, including breast cancer (76, 77). DNA methyltransferases are the enzymes responsible for adding methyl groups to cytosine bases and promoter regions of genes where

(10)

a lot of cytosines (CpG island, CGI) are methylated are associated with transcription inhibition (78). Chapter 3 explores DNA methylation and writing of this specific epigenetic signature.

Available tools for editing DNA methylation are described as well as their applications. In addition, it is explained how to achieve targeted DNA methylation/demethylation and the advantages and disadvantages of this approach are discussed.

Using Epigenetic Editing to achieve functional effects

The ultimate goal of Epigenetic Editing is to achieve changes in cell functioning, for example to render cells less resistant to a therapy. In Chapter 4 the ability of Epigenetic Editing to

accomplish functional changes was explored in different breast cancer cell subtypes, in which the oncogenic potential of the PLAU gene was investigated. Experimental inhibition of the

PLAU gene has previously been shown to reduce tumour growth, aggressiveness and

metastasis. As these experimental methods are not yet clinically possible, other compounds have been investigated to inhibit PLAU. One such promising compound is plant-derived Withaferin A (WA), a natural compound with wide-ranging pharmacological activities including cardio-protective, inflammatory, immuno-modulatory, angiogenic, anti-metastatic and anti-carcinogenic effects (79, 80). WA decreases the expression of PLAU, but also of other genes involved in cell adhesion, inflammation, and metastasis in vitro, as well as

in vivo (81-84). Furthermore, several studies have shown that DNA methylation is essential to

control PLAU expression (85-88). We therefore set out to use Epigenetic Editing as a method to manipulate PLAU expression and investigate the functional impact of PLAU in a targeted approach.

Sustainability of Epigenetic Editing

One of the early-time dogmas about Epigenetic Editing is its ability to lead to long-term, sustained effects, and there is still contradiction in the field whether this is possible. Some reports show sustained epigenetic reprogramming through Epigenetic Editing (52, 89-92), whilst this could not be repeated for other genes in different contexts (93, 94). The current consensus states that for sustained epigenetic reprogramming multiple EDs are required for long-term repressive effects (95-98). For clinical applications however, the requirement of

(11)

multiple components would be a serious limitation. In Chapter 5 we tested whether the

fibrosis- and cancer-associated PLOD2 gene can be repressed by the DNA methyltransferase M.SssI, or by the non-catalytic Krüppel associated box (KRAB) repressor targeted to the PLOD2 promoter via ZF- or CRISPR-dCas9-mediated targeting. KRAB is an ATF, which are generally considered to act transient in somatic cells (43, 44, 52, 95-97, 99), whilst for the induction of stable heterochromatin, direct editing of epigenetic marks, e.g. DNA methylation, is assumed to be more effective (90, 100). Here, we tested these two EDs for their capability to induce direct and indirect epigenetic modifications, (long-term) gene repression and their effect on transcription when having induced a heterochromatic state.

Technical improvements

Like with all novel techniques, there are quite some challenges to overcome when optimizing and validating Epigenetic Editing. Therefore, a big part of this PhD project focussed on improving transfection efficiency, delivery of the constructs to the cells and the ease of read-outs of laboratory systems. In Chapter 6, different approaches (e.g. the MS2 system,

fluorescent assisted cell sorting and several methyltransferase variants) are applied to modulate the expression of UCHL1, a gene with relevance in cancer and airway diseases. By overcoming technical hurdles, our understanding of underlying biological phenomena can increase even further, and in this way Epigenetic Editing can rise to its full potential as a flexible one-and-done research tool with various clinical applications.

One way assumed to overcome the problem of delivery in a laboratory setting is to make stable cell lines, a method that has been optimized and described in detail in chapter 7.

Briefly, in this procedure cells are infected with lentiviruses encoding dCas9 fused to an ED. This construct will randomly integrate into the cell’s genome, and will be expressed continuously. The gene-specific sgRNAs can then be transfected transiently to assess long-term effects of modified epigenetic marks on gene expression and cellular behaviour. As the plasmids for sgRNAs are much smaller than the dCas9-ED containing plasmids, transfection efficiency is expected to increase compared to transiently transfecting both the sgRNAs and the dCas9-ED. Furthermore, it might be essential to transfect sgRNAs targeted towards

(12)

multiple genes, which is of importance in diseases like e.g. cancer where multiple genes are involved.

Ways ahead

Even though there are still a lot of challenges to overcome by future researchers within this field, every piece of information is valuable and gets us a bit closer to using Epigenetic Editing in a clinical setting and a step closer towards a curable genome. There are rapid developments in epi-drug discovery (22), mainly focused on targeting the epigenetic enzymes (readers, writers and erasers of epigenetic marks) using small-molecule inhibitors (101-103) and a wide range of epigenetic-based drugs are undergoing clinical trials. Epigenetic drugs that are used in the clinic however, are mostly limited to haematological malignancies (104, 105). Many other diseases could benefit from the potential of epigenetic drugs (106-109), but they would require more targeted approaches. In this respect, Epigenetic Editing provides the possibility to target many different epigenetic enzymes to a specific locus of interest to rewrite epigenetic marks in a sustained manner. Furthermore, ongoing trials using DNA-targeting approaches (Sangamo trials, explained in more detail in the general discussion of this thesis), are giving us valuable information with regard to delivery, efficiency and safety of gene targeting. This knowledge can thus be used to translate Epigenetic Editing into a clinical applicable tool. The future is therefore looking bright in the field of targeted epigenetic approaches.

(13)

References

1. Russo VEA, Martienssen RA, Riggs AD. Epigenetic mechanisms of gene regulation. United States: Cold Spring Haror Laboratory Press; 1996.

2. Wu Ct, Morris JR. Genes, genetics, and epigenetics: a correspondence. Science. 2001;293(5532):1103-5.

3. Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational definition of epigenetics. Genes Dev. 2009;23(7):781-3.

4. Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447(7143):425-32.

5. Berdasco M, Esteller M. Aberrant epigenetic landscape in cancer: how cellular identity goes awry. Dev Cell. 2010;19(5):698-711.

6. Xavier MJ, Roman SD, Aitken RJ, Nixon B. Transgenerational inheritance: how impacts to the epigenetic and genetic information of parents affect offspring health. Hum Reprod Update. 2019;25(5):518-40.

7. Ben Maamar M, King SE, Nilsson E, Beck D, Skinner MK. Epigenetic transgenerational inheritance of parent-of-origin allelic transmission of outcross pathology and sperm epimutations. Dev Biol. 2020;458(1):106-19.

8. Carey N. Masterpieces of Epigenetics: The Missing Link between Nature and Nurture. Conference Auditorium, University of Leeds, Leeds LS2 9JT, UK: Biochemical Society and the University of Leeds; 2012.

9. Tammen SA, Friso S, Choi SW. Epigenetics: the link between nature and nurture. Mol Aspects Med. 2013;34(4):753-64.

10. Talbert PB, Meers MP, Henikoff S. Old cogs, new tricks: the evolution of gene expression in a chromatin context. Nat Rev Genet. 2019;20(5):283-97.

11. Tarakhovsky A. Tools and landscapes of epigenetics. Nat Immunol. 2010;11(7):565-8.

12. Torres IO, Fujimori DG. Functional coupling between writers, erasers and readers of histone and DNA methylation. Curr Opin Struct Biol. 2015;35:68-75.

13. Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429(6990):457-63.

14. Portela A, Esteller M. Epigenetic modifications and human disease. Nat Biotechnol. 2010;28(10):1057-68.

15. Dekker AD, De Deyn PP, Rots MG. Epigenetics: the neglected key to minimize learning and memory deficits in Down syndrome. Neurosci Biobehav Rev. 2014;45:72-84.

16. Keating ST, El-Osta A. Epigenetics and metabolism. Circ Res. 2015;116(4):715-36.

17. Badia E, Oliva J, Balaguer P, Cavaillès V. Tamoxifen resistance and epigenetic modifications in breast cancer cell lines. Curr Med Chem. 2007;14(28):3035-45.

18. Patten DK, Corleone G, Győrffy B, Perone Y, Slaven N, Barozzi I, et al. Enhancer mapping uncovers phenotypic heterogeneity and evolution in patients with luminal breast cancer. Nat Med. 2018;24(9):1469-80.

19. Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358(11):1148-59.

20. Moran S, Martínez-Cardús A, Sayols S, Musulén E, Balañá C, Estival-Gonzalez A, et al. Epigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysis. Lancet Oncol. 2016;17(10):1386-95.

(14)

21. Dumitrescu RG. Early Epigenetic Markers for Precision Medicine. Methods Mol Biol. 2018;1856:3-17.

22. Ganesan A, Arimondo PB, Rots MG, Jeronimo C, Berdasco M. The timeline of epigenetic drug discovery: from reality to dreams. Clin Epigenetics. 2019;11(1):174.

23. Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol. 2007;25(1):84-90.

24. Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nat Biotechnol. 2010;28(10):1069-78.

25. Howell PM, Liu Z, Khong HT. Demethylating Agents in the Treatment of Cancer. Pharmaceuticals (Basel). 2010;3(7):2022-44.

26. Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1(7):598-607.

27. Falahi F, van Kruchten M, Martinet N, Hospers GA, Rots MG. Current and upcoming approaches to exploit the reversibility of epigenetic mutations in breast cancer. Breast Cancer Res. 2014;16(4):412.

28. Prebet T, Sun Z, Figueroa ME, Ketterling R, Melnick A, Greenberg PL, et al. Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: results of the US Leukemia Intergroup trial E1905. J Clin Oncol. 2014;32(12):1242-8.

29. Diesch J, Zwick A, Garz AK, Palau A, Buschbeck M, Götze KS. A clinical-molecular update on azanucleoside-based therapy for the treatment of hematologic cancers. Clin Epigenetics. 2016;8:71.

30. Ganesan A. Multitarget Drugs: an Epigenetic Epiphany. ChemMedChem. 2016;11(12):1227-41. 31. Connolly RM, Li H, Jankowitz RC, Zhang Z, Rudek MA, Jeter SC, et al. Combination Epigenetic Therapy in Advanced Breast Cancer with 5-Azacitidine and Entinostat: A Phase II National Cancer Institute/Stand Up to Cancer Study. Clin Cancer Res. 2017;23(11):2691-701.

32. Kantarjian HM, Roboz GJ, Kropf PL, Yee KWL, O'Connell CL, Tibes R, et al. Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial. Lancet Oncol. 2017;18(10):1317-26.

33. Delgado-Morales R, Agís-Balboa RC, Esteller M, Berdasco M. Epigenetic mechanisms during ageing and neurogenesis as novel therapeutic avenues in human brain disorders. Clin Epigenetics. 2017;9:67.

34. Yu Y, Zeng P, Xiong J, Liu Z, Berger SL, Merlino G. Epigenetic drugs can stimulate metastasis through enhanced expression of the pro-metastatic Ezrin gene. PLoS One. 2010;5(9):e12710. 35. Duijkers FA, de Menezes RX, Goossens-Beumer IJ, Stumpel DJ, Admiraal P, Pieters R, et al.

Epigenetic drug combination induces genome-wide demethylation and altered gene expression in neuro-ectodermal tumor-derived cell lines. Cell Oncol (Dordr). 2013;36(5):351-62.

36. Hauswald S, Duque-Afonso J, Wagner MM, Schertl FM, Lübbert M, Peschel C, et al. Histone deacetylase inhibitors induce a very broad, pleiotropic anticancer drug resistance phenotype in acute myeloid leukemia cells by modulation of multiple ABC transporter genes. Clin Cancer Res. 2009;15(11):3705-15.

37. Jurkowski TP, Ravichandran M, Stepper P. Synthetic epigenetics-towards intelligent control of epigenetic states and cell identity. Clin Epigenetics. 2015;7(1):18.

(15)

39. Falahi F, Sgro A, Blancafort P. Epigenome engineering in cancer: fairytale or a realistic path to the clinic? Front Oncol. 2015;5:22.

40. Köeferle A, Stricker SH, Beck S. Brave new epigenomes: the dawn of epigenetic engineering. Genome Med. 2015;7(1):59.

41. Zentner GE, Henikoff S. Epigenome editing made easy. Nat Biotechnol. 2015;33(6):606-7. 42. Kungulovski G, Jeltsch A. Epigenome Editing: State of the Art, Concepts, and Perspectives.

Trends Genet. 2016;32(2):101-13.

43. Thakore PI, Black JB, Hilton IB, Gersbach CA. Editing the epigenome: technologies for programmable transcription and epigenetic modulation. Nat Methods. 2016;13(2):127-37. 44. de Groote ML, Verschure PJ, Rots MG. Epigenetic Editing: targeted rewriting of epigenetic marks

to modulate expression of selected target genes. Nucleic Acids Res. 2012;40(21):10596-613. 45. van Tol N, van der Zaal BJ. Artificial transcription factor-mediated regulation of gene expression.

Plant Sci. 2014;225:58-67.

46. Huisman C, Wisman GB, Kazemier HG, van Vugt MA, van der Zee AG, Schuuring E, et al. Functional validation of putative tumor suppressor gene C13ORF18 in cervical cancer by Artificial Transcription Factors. Mol Oncol. 2013;7(3):669-79.

47. Huisman C, van der Wijst MG, Falahi F, Overkamp J, Karsten G, Terpstra MM, et al. Prolonged re-expression of the hypermethylated gene EPB41L3 using artificial transcription factors and epigenetic drugs. Epigenetics. 2015;10(5):384-96.

48. Huntley S, Baggott DM, Hamilton AT, Tran-Gyamfi M, Yang S, Kim J, et al. A comprehensive catalog of human KRAB-associated zinc finger genes: insights into the evolutionary history of a large family of transcriptional repressors. Genome Res. 2006;16(5):669-77.

49. Groner AC, Meylan S, Ciuffi A, Zangger N, Ambrosini G, Dénervaud N, et al. KRAB-zinc finger proteins and KAP1 can mediate long-range transcriptional repression through heterochromatin spreading. PLoS Genet. 2010;6(3):e1000869.

50. Falahi F, Huisman C, Kazemier HG, van der Vlies P, Kok K, Hospers GA, et al. Towards sustained silencing of HER2/neu in cancer by epigenetic editing. Mol Cancer Res. 2013;11(9):1029-39. 51. Stolzenburg S, Rots MG, Beltran AS, Rivenbark AG, Yuan X, Qian H, et al. Targeted silencing of

the oncogenic transcription factor SOX2 in breast cancer. Nucleic Acids Res. 2012;40(14):6725-40.

52. Stolzenburg S, Beltran AS, Swift-Scanlan T, Rivenbark AG, Rashwan R, Blancafort P. Stable oncogenic silencing in vivo by programmable and targeted de novo DNA methylation in breast cancer. Oncogene. 2015;34(43):5427-35.

53. Rusk N. CRISPRs and epigenome editing. Nat Methods. 2014;11(1):28.

54. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346(6213):1258096.

55. Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020;367(6481).

56. Therapeutics C. Immuno-Oncology 2020 [

57. Offord C. US Companies Launch CRISPR Clinical Trial. The Scientist; 2018.

58. Curran K. Gene editing enters into human clinical trials. San Diego, CA: Rising Tide Biology; 2020. 59. Terry M. Allergan and Editas Dose First Patient in Historic CRISPR Trial for Inherited Blindness.

BioSpace; 2020.

(16)

60. Rodríguez-Rodríguez DR, Ramírez-Solís R, Garza-Elizondo MA, Garza-Rodríguez ML, Barrera-Saldaña HA. Genome editing: A perspective on the application of CRISPR/Cas9 to study human diseases (Review). Int J Mol Med. 2019;43(4):1559-74.

61. Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM. A census of human transcription factors: function, expression and evolution. Nat Rev Genet. 2009;10(4):252-63.

62. Vandevenne M, Jacques DA, Artuz C, Nguyen CD, Kwan AH, Segal DJ, et al. New insights into DNA recognition by zinc fingers revealed by structural analysis of the oncoprotein ZNF217. J Biol Chem. 2013;288(15):10616-27.

63. Mussolino C, Morbitzer R, Lütge F, Dannemann N, Lahaye T, Cathomen T. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 2011;39(21):9283-93.

64. Thakore PI, Kwon JB, Nelson CE, Rouse DC, Gemberling MP, Oliver ML, et al. RNA-guided transcriptional silencing in vivo with S. aureus CRISPR-Cas9 repressors. Nat Commun. 2018;9(1):1674.

65. Xu X, Tan X, Tampe B, Wilhelmi T, Hulshoff MS, Saito S, et al. High-fidelity CRISPR/Cas9- based gene-specific hydroxymethylation rescues gene expression and attenuates renal fibrosis. Nat Commun. 2018;9(1):3509.

66. Clarke R, Tyson JJ, Dixon JM. Endocrine resistance in breast cancer--An overview and update. Mol Cell Endocrinol. 2015;418 Pt 3:220-34.

67. Normanno N, Di Maio M, De Maio E, De Luca A, de Matteis A, Giordano A, et al. Mechanisms of endocrine resistance and novel therapeutic strategies in breast cancer. Endocr Relat Cancer. 2005;12(4):721-47.

68. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27-36. 69. Magnani L, Stoeck A, Zhang X, Lánczky A, Mirabella AC, Wang TL, et al. Genome-wide

reprogramming of the chromatin landscape underlies endocrine therapy resistance in breast cancer. Proc Natl Acad Sci U S A. 2013;110(16):E1490-9.

70. Jansen MP, Knijnenburg T, Reijm EA, Simon I, Kerkhoven R, Droog M, et al. Hallmarks of aromatase inhibitor drug resistance revealed by epigenetic profiling in breast cancer. Cancer Res. 2013;73(22):6632-41.

71. Fontes-Sousa M, Amorim M, Salta S, Palma De Sousa S, Henrique R, Jerónimo C. Predicting resistance to endocrine therapy in breast cancer: It's time for epigenetic biomarkers (Review). Oncol Rep. 2019;41(3):1431-8.

72. Stone A, Zotenko E, Locke WJ, Korbie D, Millar EK, Pidsley R, et al. DNA methylation of oestrogen-regulated enhancers defines endocrine sensitivity in breast cancer. Nat Commun. 2015;6:7758.

73. Yamamoto S, Wu Z, Russnes HG, Takagi S, Peluffo G, Vaske C, et al. JARID1B is a luminal lineage-driving oncogene in breast cancer. Cancer Cell. 2014;25(6):762-77.

74. Svotelis A, Gévry N, Grondin G, Gaudreau L. H2A.Z overexpression promotes cellular proliferation of breast cancer cells. Cell Cycle. 2010;9(2):364-70.

75. Nayak SR, Harrington E, Boone D, Hartmaier R, Chen J, Pathiraja TN, et al. A Role for Histone H2B Variants in Endocrine-Resistant Breast Cancer. Horm Cancer. 2015;6(5-6):214-24. 76. Heyn H, Esteller M. DNA methylation profiling in the clinic: applications and challenges. Nat Rev

Genet. 2012;13(10):679-92.

77. Amorim M, Salta S, Henrique R, Jerónimo C. Decoding the usefulness of non-coding RNAs as breast cancer markers. J Transl Med. 2016;14:265.

(17)

78. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484-92.

79. Kupchan SM, Anderson WK, Bollinger P, Doskotch RW, Smith RM, Renauld JA, et al. Tumor inhibitors. XXXIX. Active principles of Acnistus arborescens. Isolation and structural and spectral studies of withaferin A and withacnistin. J Org Chem. 1969;34(12):3858-66.

80. Mohan R, Hammers HJ, Bargagna-Mohan P, Zhan XH, Herbstritt CJ, Ruiz A, et al. Withaferin A is a potent inhibitor of angiogenesis. Angiogenesis. 2004;7(2):115-22.

81. Vanden Berghe W, Sabbe L, Kaileh M, Haegeman G, Heyninck K. Molecular insight in the multifunctional activities of Withaferin A. Biochem Pharmacol. 2012;84(10):1282-91.

82. Szarc vel Szic K, Op de Beeck K, Ratman D, Wouters A, Beck IM, Declerck K, et al. Pharmacological levels of Withaferin A (Withania somnifera) trigger clinically relevant anticancer effects specific to triple negative breast cancer cells. PLoS One. 2014;9(2):e87850.

83. Szarc Vel Szic K, Declerck K, Crans RAJ, Diddens J, Scherf DB, Gerhäuser C, et al. Epigenetic silencing of triple negative breast cancer hallmarks by Withaferin A. Oncotarget. 2017;8(25):40434-53.

84. Hassannia B, Logie E, Vandenabeele P, Vanden Berghe T, Vanden Berghe W. Withaferin A: From ayurvedic folk medicine to preclinical anti-cancer drug. Biochem Pharmacol. 2020;173:113602. 85. Zambelli F, Prazzoli GM, Pesole G, Pavesi G. Cscan: finding common regulators of a set of genes by using a collection of genome-wide ChIP-seq datasets. Nucleic Acids Res. 2012;40(Web Server issue):W510-5.

86. Pakneshan P, Têtu B, Rabbani SA. Demethylation of urokinase promoter as a prognostic marker in patients with breast carcinoma. Clin Cancer Res. 2004;10(9):3035-41.

87. Guo Y, Pakneshan P, Gladu J, Slack A, Szyf M, Rabbani SA. Regulation of DNA methylation in human breast cancer. Effect on the urokinase-type plasminogen activator gene production and tumor invasion. J Biol Chem. 2002;277(44):41571-9.

88. Chik F, Machnes Z, Szyf M. Synergistic anti-breast cancer effect of a combined treatment with the methyl donor S-adenosyl methionine and the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Carcinogenesis. 2014;35(1):138-44.

89. Cano-Rodriguez D, Gjaltema RA, Jilderda LJ, Jellema P, Dokter-Fokkens J, Ruiters MH, et al. Writing of H3K4Me3 overcomes epigenetic silencing in a sustained but context-dependent manner. Nat Commun. 2016;7:12284.

90. Rivenbark AG, Stolzenburg S, Beltran AS, Yuan X, Rots MG, Strahl BD, et al. Epigenetic reprogramming of cancer cells via targeted DNA methylation. Epigenetics. 2012;7(4):350-60. 91. Hathaway NA, Bell O, Hodges C, Miller EL, Neel DS, Crabtree GR. Dynamics and memory of

heterochromatin in living cells. Cell. 2012;149(7):1447-60.

92. Saunderson EA, Stepper P, Gomm JJ, Hoa L, Morgan A, Allen MD, et al. Hit-and-run epigenetic editing prevents senescence entry in primary breast cells from healthy donors. Nat Commun. 2017;8(1):1450.

93. Kungulovski G, Nunna S, Thomas M, Zanger UM, Reinhardt R, Jeltsch A. Targeted epigenome editing of an endogenous locus with chromatin modifiers is not stably maintained. Epigenetics Chromatin. 2015;8:12.

94. McDonald JI, Celik H, Rois LE, Fishberger G, Fowler T, Rees R, et al. Reprogrammable CRISPR/Cas9-based system for inducing site-specific DNA methylation. Biol Open. 2016;5(6):866-74.

(18)

95. Amabile A, Migliara A, Capasso P, Biffi M, Cittaro D, Naldini L, et al. Inheritable Silencing of Endogenous Genes by Hit-and-Run Targeted Epigenetic Editing. Cell. 2016;167(1):219-32.e14. 96. O'Geen H, Ren C, Nicolet CM, Perez AA, Halmai J, Le VM, et al. dCas9-based epigenome editing

suggests acquisition of histone methylation is not sufficient for target gene repression. Nucleic Acids Res. 2017;45(17):9901-16.

97. Mlambo T, Nitsch S, Hildenbeutel M, Romito M, Müller M, Bossen C, et al. Designer epigenome modifiers enable robust and sustained gene silencing in clinically relevant human cells. Nucleic Acids Res. 2018;46(9):4456-68.

98. O'Geen H, Bates SL, Carter SS, Nisson KA, Halmai J, Fink KD, et al. Ezh2-dCas9 and KRAB-dCas9 enable engineering of epigenetic memory in a context-dependent manner. Epigenetics Chromatin. 2019;12(1):26.

99. Groner AC, Tschopp P, Challet L, Dietrich JE, Verp S, Offner S, et al. The Krüppel-associated box repressor domain can induce reversible heterochromatization of a mouse locus in vivo. J Biol Chem. 2012;287(30):25361-9.

100. Siddique AN, Nunna S, Rajavelu A, Zhang Y, Jurkowska RZ, Reinhardt R, et al. Targeted methylation and gene silencing of VEGF-A in human cells by using a designed Dnmt3a-Dnmt3L single-chain fusion protein with increased DNA methylation activity. J Mol Biol. 2013;425(3):479-91.

101. Morera L, Lübbert M, Jung M. Targeting histone methyltransferases and demethylases in clinical trials for cancer therapy. Clin Epigenetics. 2016;8:57.

102. Esteller M. Epigenetic drugs: More than meets the eye. Epigenetics. 2017;12(5):307.

103. Velasco G, Francastel C. Genetics meets DNA methylation in rare diseases. Clin Genet. 2019;95(2):210-20.

104. Juo YY, Gong XJ, Mishra A, Cui X, Baylin SB, Azad NS, et al. Epigenetic therapy for solid tumors: from bench science to clinical trials. Epigenomics. 2015;7(2):215-35.

105. Wee S, Dhanak D, Li H, Armstrong SA, Copeland RA, Sims R, et al. Targeting epigenetic regulators for cancer therapy. Ann N Y Acad Sci. 2014;1309:30-6.

106. Berdasco M, Esteller M. Clinical epigenetics: seizing opportunities for translation. Nat Rev Genet. 2019;20(2):109-27.

107. Jones PA, Issa JP, Baylin S. Targeting the cancer epigenome for therapy. Nat Rev Genet. 2016;17(10):630-41.

108. Graça I, Pereira-Silva E, Henrique R, Packham G, Crabb SJ, Jerónimo C. Epigenetic modulators as therapeutic targets in prostate cancer. Clin Epigenetics. 2016;8:98.

109. Dirks RA, Stunnenberg HG, Marks H. Genome-wide epigenomic profiling for biomarker discovery. Clin Epigenetics. 2016;8:122.

(19)

Referenties

GERELATEERDE DOCUMENTEN

Opportunities and Challenges of Epigenetic Editing in Human Diseases Goubert,

We will determine cellular heterogeneity and sub-cell type epigenetic state switching [29] and use CRISPR-dCas9-based Epigenetic Editing [30] to locally overwrite epigenetic

In this study, we were able to silence SPDEF expression in the human alveolar epithelial cell line A549, using a novel strategy: engineered SPDEF targeting ZF proteins

(c) Relati- ve mRNA expression of ICAM1, RASSF1a, EpCAM in HEK293T and A549 cells and (d) PLOD2 in C33a cells, by the indicated dCas9 fusion protein co-transfected with a

However, modulation of the target gene by induction of histone marks in the chromatin context was demonstrated first in 2002, by inducing H3K9 methylation and causing gene

First, we reviewed in chapter 2 the most recent advances in targeting epigenetic effector domains to different regions in the genome, in order to alter gene expression..

In 2012, after finishing his master, he got the great opportunity to obtain a PhD position at the University Medical Center Groningen, from the Rijksuniversiteit Gro- ningen,

unravel the mechanisms underlying stable gene reprogramming, thus different outcomes for gene expression modulation are dependent on the local chromatin