Skip to main content

Relationship between genome and epigenome - challenges and requirements for future research

Abstract

Understanding the links between genetic, epigenetic and non-genetic factors throughout the lifespan and across generations and their role in disease susceptibility and disease progression offer entirely new avenues and solutions to major problems in our society. To overcome the numerous challenges, we have come up with nine major conclusions to set the vision for future policies and research agendas at the European level.

The Human Genome Project was completed in 2003 and led to the identification of all human genes. However, the fundamental question that remains unanswered is how do genes function and how are they regulated? Epigenetics may provide many crucial answers. Epigenetics encompasses all processes that lead to heritable changes in gene expression as cells divide, while epigenomics refers to analysis of epigenetic changes across the whole genome in a cell or entire organism [1, 2]. Typically, in a multi-cellular organism, each cell type will be characterised by the same genome, along with as many epigenomes as there are distinct cell types. Epigenetics combined with genetics is a rapidly growing field with promising implications for health and disease because many common diseases result from the interplay between the genetic make-up of individuals and the environmental factors to which they are exposed [3]. Currently, however, there is limited knowledge on the combined role of genetic and non-genetic factors thus hampering personalised medicine. A conceptual goal is to identify a cascade of genetic/epigenetic factors that underlie the development of chronic diseases. For example, a number of candidate genes have been associated with irritable bowel syndrome, but little research has examined the mechanistic impact on epigenetics [4]. Likewise, even though environmental factors such as stress, life-style, nutrition, air pollution and infections lead to allergies, the genetic and epigenetic contributions are not well understood [5, 6].

The reversible nature of epigenetic changes has attracted interest in exploring their potential as targets for the development of novel and more individualised medical treatments.

Europe, with additional effort from Member States, is showing leadership in the field of epigenetics and epigenomics and more than €200 Million were invested in research projects and infrastructure through Framework Programmes 6 and 7 (Table 1). For example, the BLUEPRINT project is focusing on distinct types of haematopoietic cells from healthy individuals and their malignant leukaemic counterparts with the aim of generating at least 100 reference epigenomes and studying them to advance and exploit knowledge of the underlying biological processes and mechanisms in health and disease [7].

Table 1 FP7 Cooperation projects and network of excellence that were represented at the workshop

With this aim, the European Commission's Directorate General for Research and Innovation (DG RTD) and Cooperation in Science and Technology (COST) organised a joint strategic workshop “Relationship between genome and epigenome”. The workshop addressed the links between genetic, epigenetic and non-genetic factors throughout the lifespan and across generations, their role in health and disease including disease susceptibility and progression, and the associated challenges of data handling/storage and interpretation. The outcomes of the workshop will inform future research priorities and are summarised in Figure 1.

Figure 1
figure 1

Understanding the relationship between genome and epigenome and their role in health and disease enables the development of tools for personalized medicine including risk prediction, disease prevention and treatment. The EU funding provides a platform, enables collaborative work and facilitates to achieve the set aims in order to consolidate Europe's leadership position in Epigenetics.

Major issues for future research include the following points:

1) In order to identify good surrogate epigenomic marks that would corroborate the influence of environmental exposure on the epigenome (including periconception environment, lifestyle, reproductive factors, microbiome etc.) and allow for the prediction and prevention of the development of chronic diseases, detailed research in humans and model organisms and careful sample acquisition (more tissue and cell specific epigenomes, time series, epigenomic variation etc.) is required. Parental conditions before, during and after conception (periconception period) may induce epigenetic changes in gametes and embryos [8]. Such changes may adversely affect the offsprings’ future health, development, productivity and fertility [3]. The connection between the perinatal factors and later outcomes in life was illustrated by describing the relationship between birth weight and incidence of diseases in older age such as cardiac disease [9]. Studies of historical famines already yielded key evidence for the association of early life environmental exposure and differences in the adult epigenome [10]. Like the field itself, these studies are in their infancy and ongoing genome-wide studies are expected to result in the identification of epigenetic alterations that are triggered by non-genetic factors leading to particular disease phenotype. The microbiome has strong parallels with the epigenome in that it is complex and may reflect environmental exposure (of the host from which the micobiome was obtained) and might also impact on how non-genetic factors lead to epigenetic changes (i.e. by modulating hormonal levels [11]). Accumulating data demonstrate a crucial impact of the microbiome on health and disease.

2) With the increase of chronological age, an increase of gene promoter methylation paralleled by global hypomethylation across the genome can be observed. This is remarkably similar to the DNA methylation changes seen in cancer [12] suggesting that similar underlying mechanisms may be involved. More age-stratified data are required to understand the relationship between the epigenome, the microbiome and the environment during the course of life and its impact on allergy and chronic diseases.

3) The genome-epigenome interaction is also crucially involved in the biology, character and extent of an established disease and not just in disease development. This is reflected for instance in the role that the chromatin and epigenome plays in DNA damage repair [13]. Epigenetic markers allow for the prediction of the natural behaviour of a disease (prognostic markers) and the likelihood of responding to a specific treatment (predictive markers). Testing and validating these markers in clinical trials and benchmarking against established strategies will be crucial in order to improve disease outcome.

4) Studies of the effects and the mechanistic impact of epidrugs (drugs that can effect epigenomic modifications) and their impact on the genome, development and validation of new epigenetic drug candidates and rational design of combination therapies of genetic and epigenetic drugs should be encouraged to cure diseases or at least improve the efficacy of current treatment modalities as recently demonstrated [14]. Structural and functional information from chromatin and DNA modifying enzymes and the development of small molecules active on specific epi-targets are crucial for the development of new therapeutic approaches. Epigenetic therapy tries to reverse such aberrations following disruption of the epigenetic signal balance through the use of both natural compounds and synthetic molecules [15]. For instance, pharmacological inhibition of EZH2 (enhancer of zeste homolog 2, a Histone-lysine N-methyltransferase) was recently shown as a promising new tool with which to treat cancer [16]. Many clinical trials are already ongoing, and epigenetic therapy (azacytidine) has recently been approved by the United States Food and Drug Administration (US FDA) for use in the treatment of Myelodysplastic Syndrome (MDS) and Primary Cutaneous T-cell Lymphoma (CTCL) [17].

5) Studies to identify functional relationships between epigenetics and genetics require analysis of ex vivo samples of primary cells, and therefore the sampling, sorting and analytical procedures need to be optimised and adapted. Cell heterogeneity (variation among cells) is a challenge in gaining a thorough understanding of genome status, gene expression and the role of underlying epigenetic mechanisms. This is true for many cellular processes, such as genome remodeling during reprogramming or the conversion of somatic cells to pluripotent cells. Therefore collecting the most appropriate samples in order to address a specific set of questions and miniaturization of technologies for the analyses of single cells [18, 19] is crucial.

6) Epigenomic and genomic data sets are complex and multi-dimensional, and their interpretation requires the further development of data analysis tools/software. A large amount of data has already been acquired and is highly multidimensional and multimodal; therefore it is the analysis that remains the challenge. DNA and chromatin exist in a 3D space. Transcriptome data are complex: all transcripts, including non-coding (nc) RNAs, overlap other transcripts and quantification is not trivial. Performing data analysis by integrating data from different repositories (some of which are difficult to find) is problematic because of the different methodologies used to acquire the data sets [20]. There is a need to establish robust benchmarks for data analysis for the comparison of different analytical approaches/software.

7) Integrating the findings from -omics research into clinical practice is one of the major challenges of the future. Systems biology approaches are advantageous in providing predictive models of associations between epigenomic/genomic data and phenotypes offering an entry point for assays into functional relationships. Understanding the functional/mechanistic role of epigenetic marks is highly desirable, but that in many cases it may be difficult to directly obtain such insight. Systems biological approaches could identify predictive models from multi-modal data to support associations that can then be tested in functional models.

8) Improved collaborations should be fostered by the establishment and harmonization of standard operating procedures for sample processing, data acquisition and formatting; and by the development of software that is user-friendly for the non-specialist as well as facilitating an Open Access policy to allow free data sharing and automatic mining of publications. Current European effort should be aligned with those of the International Human Epigenome Consortium (http://www.ihec-epigenomes.org/) coordinating epigenome mapping and characterisation worldwide to avoid redundant research effort, to implement high data quality standards, to coordinate data storage, management and analysis and to provide free access to the epigenomes produced.

9) European Union (EU) consortia and COST Actions have tremendously shaped and consolidated Europe’s leadership position in Epigenetics and can provide indispensable means for young researchers to become principal investigators and future European leaders by integrating them into networks of experienced scientists/clinicians. EC funding schemes should devote further effort to principal investigators career development.

The European Union is currently funding over 300 epigenetics projects (a High Impact Project, Collaborative Projects, Networks of Excellence, ERC (European Research Council) Starting Grants, ERC Advanced Grants, Marie Curie Actions) with a total contribution of more than €200 Million.

Abbreviations

EC-COST:

European Commission's Cooperation in Science and Technology

DG RTD:

European Commission's Directorate General for Research and Innovation

DNA:

Deoxyribonucleic acid

EZH2:

Enhancer of zeste homolog 2

US FDA:

United States Food and Drug Administration

MDS:

Myelodysplastic Syndrome

CTCL:

Primary Cutaneous T-cell Lymphoma

nc:

Non-coding

RNAs:

Ribonucleic acids

EU:

European Union

ERC:

European Research Council.

References

  1. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, Nery JR, Lee L, Ye Z, Ngo QM, Edsall L, Antosiewicz-Bourget J, Stewart R, Ruotti V, Millar AH, Thomson JA, Ren B, Ecker JR: Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009, 462: 315-322. 10.1038/nature08514 [doi]

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  2. Bird A: Perceptions of epigenetics. Nature. 2007, 447: 396-398. 10.1038/nature05913 [doi]

    Article  CAS  PubMed  Google Scholar 

  3. Mill J, Heijmans BT: From promises to practical strategies in epigenetic epidemiology. Nat Rev Genet. 2013, 14: 585-594. 10.1038/nrg3405 [doi]

    Article  CAS  PubMed  Google Scholar 

  4. Kapeller J, Houghton LA, Monnikes H, Walstab J, Moller D, Bonisch H, Burwinkel B, Autschbach F, Funke B, Lasitschka F, Gassler N, Fischer C, Whorwell PJ, Atkinson W, Fell C, Buchner KJ, Schmidtmann M, Van DV I, Wisser AS, Berg T, Rappold G, Niesler B: First evidence for an association of a functional variant in the microRNA-510 target site of the serotonin receptor-type 3E gene with diarrhea predominant irritable bowel syndrome. Hum Mol Genet. 2008, 17: 2967-2977. 10.1093/hmg/ddn195 [doi]

    Article  CAS  PubMed  Google Scholar 

  5. Anto JM, Pinart M, Akdis M, Auffray C, Bachert C, Basagana X, Carlsen KH, Guerra S, Von HL, Illi S, Kauffmann F, Keil T, Kiley JP, Koppelman GH, Lupinek C, Martinez FD, Nawijn MC, Postma DS, Siroux V, Smit HA, Sterk PJ, Sunyer J, Valenta R, Valverde S, Akdis CA, Annesi-Maesano I, Ballester F, Benet M, Cambon-Thomsen A, Chatzi L, et al: Understanding the complexity of IgE-related phenotypes from childhood to young adulthood: a Mechanisms of the Development of Allergy (MeDALL) seminar. J Allergy Clin Immunol. 2012, 129: 943-954. 10.1016/j.jaci.2012.01.047 [doi]

    Article  PubMed  Google Scholar 

  6. Krauss-Etschmann S, Bush A, Bellusci S, Brusselle GG, Dahlen SE, Dehmel S, Eickelberg O, Gibson G, Hylkema MN, Knaus P, Konigshoff M, Lloyd CM, Macciarini P, Mailleux A, Marsland BJ, Postma DS, Roberts G, Samakovlis C, Stocks J, Vandesompele J, Wjst M, Holloway J: Of flies, mice and men: a systematic approach to understanding the early life origins of chronic lung disease. Thorax. 2013, 68: 380-384. 10.1136/thoraxjnl-2012-201902 [doi]

    Article  PubMed  Google Scholar 

  7. Adams D, Altucci L, Antonarakis SE, Ballesteros J, Beck S, Bird A, Bock C, Boehm B, Campo E, Caricasole A, Dahl F, Dermitzakis ET, Enver T, Esteller M, Estivill X, Ferguson-Smith A, Fitzgibbon J, Flicek P, Giehl C, Graf T, Grosveld F, Guigo R, Gut I, Helin K, Jarvius J, Kuppers R, Lehrach H, Lengauer T, Lernmark A, Leslie D, et al: BLUEPRINT to decode the epigenetic signature written in blood. Nat Biotechnol. 2012, 30: 224-226. 10.1038/nbt.2153 [doi]

    Article  CAS  PubMed  Google Scholar 

  8. Habibi E, Brinkman AB, Arand J, Kroeze LI, Kerstens HH, Matarese F, Lepikhov K, Gut M, Brun-Heath I, Hubner NC, Benedetti R, Altucci L, Jansen JH, Walter J, Gut IG, Marks H, Stunnenberg HG: Whole-genome bisulfite sequencing of two distinct interconvertible DNA methylomes of mouse embryonic stem cells. Cell Stem Cell. 2013, 13: 360-369. 10.1016/j.stem.2013.06.002 [doi]

    Article  CAS  PubMed  Google Scholar 

  9. Barker DJ, Winter PD, Osmond C, Margetts B, Simmonds SJ: Weight in infancy and death from ischaemic heart disease. Lancet. 1989, 2: 577-580.

    Article  CAS  PubMed  Google Scholar 

  10. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, Slagboom PE, Lumey LH: Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A. 2008, 105: 17046-17049. 10.1073/pnas.0806560105 [doi]

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, Von BM, McCoy KD, Macpherson AJ, Danska JS: Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013, 339: 1084-1088. 10.1126/science.1233521 [doi]

    Article  CAS  PubMed  Google Scholar 

  12. Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, Campan M, Noushmehr H, Bell CG, Maxwell AP, Savage DA, Mueller-Holzner E, Marth C, Kocjan G, Gayther SA, Jones A, Beck S, Wagner W, Laird PW, Jacobs IJ, Widschwendter M: Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010, 20: 440-446. 10.1101/gr.103606.109 [doi]

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Soria G, Polo SE, Almouzni G: Prime, repair, restore: the active role of chromatin in the DNA damage response. Mol Cell. 2012, 46: 722-734. 10.1016/j.molcel.2012.06.002 [doi]

    Article  CAS  PubMed  Google Scholar 

  14. Schenk T, Chen WC, Gollner S, Howell L, Jin L, Hebestreit K, Klein HU, Popescu AC, Burnett A, Mills K, Casero RA, Marton L, Woster P, Minden MD, Dugas M, Wang JC, Dick JE, Muller-Tidow C, Petrie K, Zelent A: Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med. 2012, 18: 605-611. 10.1038/nm.2661 [doi]

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Dell'Aversana C, Lepore I, Altucci L: HDAC modulation and cell death in the clinic. Exp Cell Res. 2012, 318: 1229-1244. 10.1016/j.yexcr.2012.01.025 [doi]

    Article  PubMed  Google Scholar 

  16. McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, Liu Y, Graves AP, Della PA, Diaz E, LaFrance LV, Mellinger M, Duquenne C, Tian X, Kruger RG, McHugh CF, Brandt M, Miller WH, Dhanak D, Verma SK, Tummino PJ, Creasy CL: EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012, 492: 108-112. 10.1038/nature11606 [doi]

    Article  CAS  PubMed  Google Scholar 

  17. Gronbaek K, Hother C, Jones PA: Epigenetic changes in cancer. APMIS. 2007, 115: 1039-1059.

    Article  PubMed  Google Scholar 

  18. Bock C, Tomazou EM, Brinkman AB, Muller F, Simmer F, Gu H, Jager N, Gnirke A, Stunnenberg HG, Meissner A: Quantitative comparison of genome-wide DNA methylation mapping technologies. Nat Biotechnol. 2010, 28: 1106-1114. 10.1038/nbt.1681 [doi]

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Stunnenberg HG, Hubner NC: Genomics meets proteomics: identifying the culprits in disease. Hum Genet. 2013, 10: 1007/s00439-013-1376-2 [doi]

    Google Scholar 

  20. Pettifer S, Ison J, Kalas M, Thorne D, McDermott P, Jonassen I, Liaquat A, Fernandez JM, Rodriguez JM, Pisano DG, Blanchet C, Uludag M, Rice P, Bartaseviciute E, Rapacki K, Hekkelman M, Sand O, Stockinger H, Clegg AB, Bongcam-Rudloff E, Salzemann J, Breton V, Attwood TK, Cameron G, Vriend G: The EMBRACE web service collection. Nucleic Acids Res. 2010, 38: W683-W688. 10.1093/nar/gkq297 [doi]

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Martin Widschwendter.

Additional information

Competing interests

The authors declare that they have no competing interest.

Authors’ contributions

All authors, GA, LA, BA, NA, DB, NB, CB, EB-R, JB, SB, BB-deP, MB, LC, AC, XE, AF, NG, IG, BTH, SH, JH-D, II, JI, RK, SK-E, PL, SL, AL, GM, EM, GM, NM, EM, CM, KM, MM-V, GM, DN, BN, MN, JN, SO,MP, RP, UP, MR, JR, MR, MDR, BR, SS, DS, ES, TS, HGS, ET, M-ET-P, RT, AVS, MV and MW, contributed to this report equally as members of the workshop. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Almouzni, G., Altucci, L., Amati, B. et al. Relationship between genome and epigenome - challenges and requirements for future research. BMC Genomics 15, 487 (2014). https://doi.org/10.1186/1471-2164-15-487

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2164-15-487

Keywords