Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Available online at www.sciencedirect.

com Current Opinion in

ScienceDirect Toxicology

Progress in identifying epigenetic mechanisms of


xenobiotic-induced non-genotoxic carcinogenesis
Rémi Terranovaa,e, Antonio Vitobelloa,e,
Alberto Del Rio Espinolaa, C. Roland Wolfb,e,
Michael Schwarzc,e, John Thomsond,e, Richard Meehand,e and
Jonathan Moggsa,e

Abstract
Determining the human relevance of structurally and func-
Current Opinion in Toxicology 2017, 3:62–70
tionally distinct non-genotoxic carcinogenic compounds that
This review comes from a themed issue on Risk Assessment in
induce a diverse range of tissue-, gender-, strain- and species-
Toxicology – Experimental Toxicology
specific tumours in animals remains a major challenge for
Available online 8 June 2017
toxicologists. Nevertheless, elucidating mechanisms of
xenobiotic-induced tumours in animals can provide industry, For a complete overview see the Issue and the Editorial
environmental and regulatory scientists with valuable tools for http://dx.doi.org/10.1016/j.cotox.2017.06.005
cancer hazard identification and risk assessment. The dis- 2468-2020/© 2017 Elsevier B.V. All rights reserved.
covery that aberrant epigenetic events frequently accompany
genetic mutations in human cancers has stimulated efforts to
Keywords
deploy integrated epigenomic and transcriptomic profiling of Non-genotoxic carcinogenesis, Cancer risk assessment, Epigenetics,
xenobiotic-induced non-genotoxic carcinogenesis (NGC) in Genetics, Epigenome.
animal models, enabling enhanced mechanistic interpretation
and novel early biomarker discovery. Recent advances in the
1. Introduction
mapping and functional characterization of mammalian tissue-
Concerns regarding the appropriateness of extrapolating
specific epigenomes also provides new opportunities to char-
lifetime rodent carcinogenicity study findings to humans
acterize the cross-strain/-species chromatin architecture of
have been extensively reviewed [1e3]. If xenobiotic
non-genotoxic carcinogen effector genes and to predict their
exposure in animals is found to be associated with either
potential for modulation by xenobiotics in human tissue. Since
tumour induction or early indicators of neoplastic
xenobiotic-induced perturbations of gene regulation are inti-
hazard, then a weight of evidence-based cancer risk
mately associated with the underlying DNA sequence, there is
assessment is generally recommended. A key contrib-
a need to integrate the impact of genotype on susceptibility to
uting factor to the weight of evidence approach for
NGC. Furthermore, the potential association of xenobiotic
xenobiotic-induced non-genotoxic carcinogenesis
target modulation with tumorigenic phenotypes can be
(NGC) is the determination of a mechanism or mode of
assessed using genetic models and cancer genome re-
action since this provides an entry point for subsequent
sources. Finally, we discuss how epigenomic profiling may be
assessments of potential human relevance [4e6]. A
used to critically assess the comparability and validity of
molecular basis for species-specific non-genotoxic
cellular NGC models versus in vivo-derived tissue samples
carcinogenesis has been proposed for a number of
and some of key challenges associated with incorporating
compounds [4e10]. However, the diverse range of
epigenetic mechanisms and biomarkers into cancer risk
xenobiotic-induced tumour types that are typically
assessment.
observed in animal carcinogenicity studies, often
Addresses exhibiting tissue-, gender-, strain- and/or species-
a
Preclinical Safety, Translational Medicine, Novartis Institutes for
specificities, make the determination of mechanism
BioMedical Research, Basel, CH-4057, Switzerland
b
Division of Cancer Research, University of Dundee, United Kingdom and assessment of potential relevance to humans very
c
Department of Toxicology, University of Tübingen, Germany challenging. Furthermore, there is very little data on
d
MRC Human Genetics Unit, Institute of Genetics & Molecular Medi- potential association of xenobiotic exposure with non-
cine, University of Edinburgh, United Kingdom genotoxic carcinogenesis in humans due to the likely
e
Member of IMI MARCAR Consortium, United Kingdom
latency, very low incidence, and difficulty of deconvo-
Corresponding author: Moggs, Jonathan, Preclinical Safety, luting environmental versus intrinsic factors for malig-
Translational Medicine, Novartis Institutes for BioMedical Research, nancy development. Some insight may be gained from
Basel, CH-4057, Switzerland. (jonathan.moggs@novartis.com) somatic mutational signatures of human tumours that

Current Opinion in Toxicology 2017, 3:62–70 www.sciencedirect.com


Epigenetic mechanisms of carcinogenesis Terranova et al. 63

are associated with known mutagenic exposures [11] but 2. Leveraging recent advances in cancer
there will inevitably be overlap between intrinsic and epigenetics to enhance mechanistic
extrinsic mechanisms [12]. Despite these challenges, interpretation and biomarkers of non-
there is a need for rigorous cancer risk assessment of genotoxic carcinogenesis
xenobiotics to which humans are exposed including Epigenetics describes mechanisms that operate in con-
treatment with novel therapeutics [13] and occupa- cert with the underlying DNA sequence to regulate
tional or environmental exposure to chemicals [3]. gene expression and determine the overall phenotype of
cell. Epigenetic marks include the methylation of DNA
Characterizing the molecular mechanisms underlying cytosine bases, post-translational modifications of his-
xenobiotic-induced non-genotoxic carcinogenesis has tone proteins, nucleosome remodelling, and non-coding
great potential for providing industry, environmental RNAs. These epigenetic marks are dynamically regu-
and regulatory scientists with valuable tools for cancer lated by numerous enzymes and binding proteins that
hazard identification and risk assessment. This is enable cells to read, write or erase chromatin modifica-
exemplified by phenobarbital-induced hepatocarcino- tions. Epigenetic formatting of the genome contributes
genesis where Constitutive Androstane Receptor to spatio-temporal patterns of gene expression and
(CAR)-mediated stimulation of mouse hepatocyte pro- controls lineage choice, differentiation and cellular
liferation represents a mode of action that has not been functions [25]. Epigenetic variation, together with ge-
reproduced in human hepatocytes in vitro [14e16]. netic variation and meta-genomic variation, thus repre-
Since there is no clear evidence for phenobarbital- sents one of the key drivers of phenotypic variation in
associated liver cancer risk in humans (based on epide- health and disease. The acquisition of cancer hallmarks
miological data from a large number of clinical studies such as sustained proliferative signalling and resistance
including long-term therapeutic treatment of epileptics; to cell death [26] is facilitated by a combination of
[17]), CAR-mediated liver non-genotoxic carcinogen- genome instability, mutation and epigenomic disruption
esis is not generally considered to be human-relevant [27]. Epigenetic perturbations associated with cancer
[4,10]. Humanized rodent models in which mouse aetiology and progression include widespread mutations
livers have been engineered to express human CAR in epigenetic regulatory proteins and aberrant expres-
supported proliferative responses and tumour promo- sion of stem cell reprogramming genes [28]. Epigenetic
tion following exposure to PB [18e20]. In contrast, mechanisms of carcinogenesis that are well character-
human hepatocytes did not support hyperplastic re- ized in humans include oestrogen exposure and breast
sponses to the phenobarbital in chimeric mice with cancer [29]. Importantly, from a toxicologic perspective,
humanized liver [21]. It is noteworthy that the observed inflammatory responses to tissue injury and chronic
plasma phenobarbital exposures in these humanized exposure to environmental factors have been proposed
models were comparable to those obtained in human as mechanisms for inducing cancer-predisposing epige-
subjects receiving therapeutic doses of this drug. Thus, netic changes in vulnerable populations of somatic stem
understanding the opposing outcomes of these models cells and progenitor compartments [30]. It is note-
will require further characterization of: i) quantitative worthy that environmental influences on intermediary
exposureeresponse relationships; ii) the influence of metabolism such as nutrient availability and utilisation
human nuclear receptor-mouse gene regulatory protein can also result in tumour growth-promoting modifica-
interactions; iii) the influence of mouse host cellular tions of the epigenetic landscape by altering substrates
environment on grafted human hepatocytes; and iv) and inhibitors of chromatin-modifying enzymes [30e
comparability at the molecular, biochemical and cellular 34]. Epigenetic signatures of environmental exposure
levels of engineered or grafted hepatocytes to human (e.g. to cigarette smoke) have already been identified in
donor-derived liver tissue. Importantly, phenobarbital humans and are likely to exist for many other types of
induces extensive changes in chromatin modification xenobiotic exposure in both humans and animals [35].
patterns across the regulatory regions of CAR target
genes in mouse liver [22e24] and it is thus plausible Together, these observations are stimulating efforts to
that differences in the genetic and epigenetic archi- investigate whether xenobiotic-induced perturbations
tecture of phenobarbital effector genes play a role in of DNA methylation, chromatin modification, non-
determining species-specific susceptibility to CAR- coding RNAs, and/or transcription factor accessibility
mediated hepatocarcinogenesis. contribute to non-genotoxic carcinogenesis [23,24,36e
42].
Here we describe how recent advances in epigenetic
regulation of the genome can be leveraged to provide
3. Elucidating early epigenetic molecular
new insights into molecular mechanisms of xenobiotic-
indicators of non-genotoxic carcinogenesis
induced non-genotoxic carcinogenesis, to identify early
Integrated epigenomic and transcriptomic profiling of
biomarkers and susceptibility factors, and to enable new
target tissues for xenobiotic-induced tumours repre-
approaches for assessing potential human relevance.
sents a powerful approach for elucidating early

www.sciencedirect.com Current Opinion in Toxicology 2017, 3:62–70


64 Risk Assessment in Toxicology – Experimental Toxicology

Fig. 1

Current Opinion in Toxicology 2017, 3:62–70 www.sciencedirect.com


Epigenetic mechanisms of carcinogenesis Terranova et al. 65

molecular indicators of non-genotoxic carcinogenesis. phenobarbital-induced loss of 5hmC in a specific subset


This is exemplified by a series of mechanistic in vivo of gene loci predicts subsequent aberrant promoter 5mC
rodent studies conducted within the EU IMI MARCAR hypermethylation in resultant phenobarbital-promoted
consortium (www.imi-marcar.eu) in which tumour pro- mouse liver tumours [41]. Recent evidence suggests
moting doses of phenobarbital were used as a reference that genomic loci exhibiting elevated levels of 5hmC
rodent liver tissue-specific non-genotoxic carcinogen. In represent dynamic chromatin states in contrast to loci
addition to the anticipated activation of CAR/b-catenin marked by 5mC which are typically regarded to indicate
signalling pathways and induction of proliferation inactive chromatin states. Consistent with this notion,
markers, novel early biomarkers include the aberrant up- the majority of enriched 5hmC is found to be associated
regulation of pluripotency-associated non-coding RNAs, with gene bodies, a large number of enhancer elements,
dynamic changes in locus-specific chromatin modifica- and a small subset in gene promoters, in a transcription-
tions and altered transcription factor activity. dependent manner. Thus, the profiling of xenobiotic-
induced changes in tissue-specific 5hmC levels has
Phenobarbital progressively upregulated long non- significant potential for classifying the mode of action of
coding RNAs encoded by an epigenetically imprinted potentially carcinogenic agents [48].
Dlk1-Dio3 gene cluster in mouse perivenous hepatocytes
in a CAR and b-catenin dependent manner [39]. Significant progress has been made in identifying target
Importantly, perturbation of this gene locus has been genes for a diverse range of NGC modes of action via
associated with mouse stem cell pluripotency [43,44] transcriptomic profiling of rodent tissues [49,50]. Prox-
and also with a stem-cell like phenotype in a subset of imal regulatory transcription factors and upstream
human hepatocellular carcinomas [45]. Furthermore, signalling pathways can often be inferred for well char-
hepatocytes in the same perivenous region of mouse acterized target genes and have been extensively vali-
liver were recently associated with Wnt-signalling dated for the NGC-associated nuclear receptors CAR
dependent stem cell-like properties [46]. Together and Peroxisome Proliferator Activated Receptor alpha
these observations raise the intriguing possibility that (PPARa) using biochemical, cellular and rodent genetic
PB-induces dedifferentiation/reprogramming of adult models. A complementary and powerful approach for
hepatocytes to a stem cell-like state during mouse identifying gene regulatory interactions underlying
hepatocarcinogenesis. The notion that functional and NGC involves computational modelling of predicted
molecular hallmarks of pluripotent stem cells [47] transcription factor binding sites within NGC-
might represent a valuable source of early NGC bio- responsive genes and has been used to identify novel
markers warrants further research. roles for E2F and ZFP161 transcription factors in regu-
lating PB-mediated hepatocyte proliferation and sub-
It is widely accepted that deregulation of normal DNA sequent tumour promotion [40].
methylation and gene expression patterns can aid cancer
cells to evolve more rapidly and thus contribute to 4. Towards functional and cross-species
increased invasiveness, metastatic potential and poten- epigenomic characterization of xenobiotic-
tially drug resistance [48]. Phenobarbital induced both induced non-genotoxic carcinogenesis
acute and long-lasting changes in the mouse liver DNA Importantly, multiple layers of epigenetic marks and
methylome with dynamic and reciprocal changes in 5- gene regulatory factors control genome structure and
methylcytosine (5mC) and 5-hydroxymethylcytosine function. Recent advances in the development of deep
(5hmC) levels within regulatory regions of CAR and b- sequencing based chromatin modifications and accessi-
catenin gene targets [24]. Furthermore, early bility assays enable genome-wide functional

Xenobiotic signalling to chromatin and potential for species differences in regulation of NGC effector genes. A) Potential for species differences in NGC
signalling mechanisms have been extensively explored at the level of ligand–receptor interactions. For example, phenobarbital activation of the CAR
nuclear receptor in hepatocytes and subsequent transcriptional up-regulation of xenobiotic response genes is highly conserved between rodents and
humans. In contrast, human hepatocytes appear to be refractory to the proliferative effects of phenobarbital that are seen in rodents [14–16] and this is
not fully accounted for by species differences in CAR-ligand interactions alone. We hypothesise that, although the molecular mechanisms responsible for
the acute xenobiotic responses appear to be highly conserved across species (Target Gene a/A), secondary effectors implicated in the transduction of the
extracellular signals to the nucleus and the chromatin landscape configuration of NGC effector genes are likely to contribute to strain and species dif-
ferences in susceptibility to non-genotoxic carcinogens (Target Gene b/B). Such mechanistic differences in NGC molecular signalling pathways need to
be integrated with potential quantitative differences in the potency/level of exposure of xenobiotics and intrinsic tissue mutation rates. B) Illustration of
conserved (blue) and species-specific (red) differences in chromatin accessibility of liver tissue gene loci based on visualization of ENCODE DNase I
mouse and human datasets via the UCSC browser (https://genome.ucsc.edu/; [51,77]; GEO dataset accession numbers GSE90306 and GSM1014195,
John Stamatoyannopoulos, UW). Underlying DNA sequence similarity is shown as dotplots based on local genomic alignment between orthologous
mouse (y axis) and human (x axis) regions (images obtained using YASS tool; [78]). Housekeeping gene (GAPDH), liver tissue-specific gene (ALB),
xenobiotic response gene (CYP2B6; orthologous to mouse Cyp2b10), candidate non-coding RNA biomarker for CAR-mediated liver tumour promotion
response gene (MEG3).

www.sciencedirect.com Current Opinion in Toxicology 2017, 3:62–70


66 Risk Assessment in Toxicology – Experimental Toxicology

characterization of mammalian tissue-specific epige- that the potential human relevance of well-defined
nomes. This is exemplified by the recent completion of NGC mechanisms in animals could be explored by
genome-wide epigenome profiles for >100 human tis- evaluating the degree of conservation and dynamic range
sues and cell types [51,52] (http://epigenomesportal.ca/ of chromatin modifications/accessibility at regulatory
ihec/index.html) that is dramatically enhancing our regions of orthologous non-genotoxic carcinogen
understanding of genome function and regulation. effector genes in human tissue samples (Fig. 1). It is
noteworthy that comparison of mode of action-related
Genome-wide chromatin profiling methods including target gene architecture in human versus animal tissue
DNase I hypersensitivity and Assay for Transposase may circumvent some of the challenges associated with
Accessible Chromatin (ATAC) assays have been inte- the comparability of cultured human cells or humanized
grated with histone post-translational modification rodent models to human tissue. Importantly, any pro-
mapping to identify the open cis-regulatory DNA re- posed species-differences in NGC molecular signalling
gions (also known as the cistrome) that are recognised pathways needs to be integrated with quantitative dif-
by trans-acting regulatory factors. Cistrome elements ferences in the potency and level/exposure of xenobi-
include gene enhancers, promoters, insulators, silencers otics and intrinsic tissue mutation rates.
and locus control regions. Cistrome profiling has iden-
tified hundreds of thousands of enhancer regions in the 5. Influence of genotype on epigenetic
human and mouse genomes, encompassing 1e3% of the mechanisms of xenobiotic-induced non-
genome, and vastly outnumbering the number protein genotoxic carcinogenesis
coding genes, overall highlighting the importance of the Since xenobiotic-induced perturbations of gene regula-
regulatory fraction of the genome [51,53,54]. Enhancers tion are intimately associated with the underlying DNA
are bound by regulatory transcription factors and inte- sequence, there is a need to integrate the impact of
grate extracellular signalling pathways with intracellular genotype on susceptibility to non-genotoxic
cell fate information to elicit cell type-specific tran- carcinogenesis.
scriptional responses [55] The functional binding of
regulatory transcription factors to these cis-acting Distinct strains and species of preclinical animal models
accessible sites orchestrates long-range regulatory in- have been shown to widely vary in their susceptibility to
teractions in the 3D space of the nucleus, enabling cell- xenobiotic-induced carcinogenicity. For example, mouse
type specific, spatiotemporal, control of gene expression stocks and inbred strains can significantly differ in their
patterns which drive cell identity and function [56e58]. susceptibility to treatment-induced liver neoplasia, with
Furthermore, enhancers represent regulatory modules C3H males being highly sensitive compared to highly
associated with the most conserved phase of vertebrate resistant C57BL/6 males, although this is likely to be
embryogenesis and are subject to conserved develop- based on both genetic and non-genetic factors [65].
mental regulation by epigenetic modifiers [59]. Impor- Such strain differences in xenobiotic-induced liver tu-
tantly, the role of enhancer dysfunction in cancer has mours primarily affect tumour progression (or tumour
been extensively characterized [60,61]. Single en- size) and to a much lower part tumour number. Whilst
hancers and dense clusters of enhancers (known as specific liver tumour susceptibility genes have not yet
“Super”-enhancers in the literature) appear to control been identified, several hepatocarcinogenesis suscepti-
the expression of core transcription factors (TFs) un- bility (Hcs) loci have been identified using mouse
derlying cell identity [61e63] and are often deregulated backcrosses and linkage analysis [66,67]. Intriguingly,
in cancer leading to aberrant activation of growth related hsc3 maps within 6 megabases of the PB-responsive
genes and deregulation of important lineage specific Dlk1-Dio3 imprinted gene cluster on chromosome 12
differentiation drivers [61,64]. [39,68]. Thus, transcriptional responses from this
epigenetically imprinted gene cluster may be influenced
Together these observations suggest that integrated by strain-specific genetic factors that could be further
cistrome, chromatin modification and transcriptome characterized via deep sequencing of appropriate inbred
profiling of NGC target tissues will provide novel mouse strains.
mechanistic insights and may provide unique opportu-
nity to characterize the cross-strain and cross-species The recent integration of human genome regulatory
chromatin architecture and transcription factor accessi- DNA and disease- and trait-associated genetic variants
bility of non-genotoxic carcinogen effector genes. reveals a disproportionate (>80%) enrichment of
disease-associated genetic variants in non-coding
Although access to blood or tissue samples from humans enhancer regions, potentially disrupting important
exposed to putative NGCs is very rare, a variety of transcription factor-based regulatory interactions in a
human tissue-specific epigenomes representing both cell-type specific manner [56,62,69,70]. These cis-
healthy and disease states (http://ihec-epigenomes.org/) acting non-coding regulatory variants range from rare
can in principle be leveraged as comparators to NGC to common and are associated with a broad range of
target tissue epigenomes in animal models. We envisage
Current Opinion in Toxicology 2017, 3:62–70 www.sciencedirect.com
Epigenetic mechanisms of carcinogenesis Terranova et al. 67

phenotypic effects driven by sometimes subtle effects human tissue responses to xenobiotic exposure. Given
on target gene expression [71]. The role of non-coding the likely importance of epigenetic mechanisms during
sequence variants in cancer is currently being explored NGC it is particularly noteworthy that the DNA
[72] but it seems likely that genetic variants in non- methylation profiles of mammalian cells differ from
coding enhancer regions will contribute to tissue-, those of the primary tissues from which they were
strain- and species-specific responses to NGC. derived due to rapid reprogramming of epigenetic and
transcriptional profiles following adaptation of primary
It is also noteworthy that most rodent carcinogenicity cells to culture [74]. The recent refinement of cistrome,
studies are currently performed in genetically undefined epigenome and transcriptome profiling tools for assess-
outbred stocks (e.g. CD1 mouse and/or Wistar rat) that ing cross-species conservation of tissue-specific genome
represent a very narrow range of genetic diversity functions provides a valuable opportunity to critically
compared to humans. Whilst using more diverse panels of assess the comparability and validity of cellular NGC
rodent strains might conceivably enable improved pre- models versus in vivo-derived tissue samples. Further-
dictions of inter-individual human variability [73], the more, 5-hydroxymethylcytosine profiling is proving to be
feasibility of testing xenobiotics for carcinogenicity in a powerful tool for cell lineage tracing [48] and thus may
multiple rodent strains is limited by ethical and economic help deconvolute the cell type specificity of xenobiotic
factors. Nevertheless, the genetic diversity in mice pro- responses within complex tissues.
vides a powerful approach for establishing mechanism.
Finally, there are several challenges associated with
In the context of pharmaceutical and agrochemical incorporating epigenetic mechanisms and biomarkers
product development, the ability to deploy early into cancer risk assessment. These include the need to
mechanism-based approaches for cancer hazard identi- define the normal inter-individual dynamics of healthy
fication prior to entering more resource intensive and versus disease-state epigenomes across tissues and
costly late phase preclinical and/or clinical studies would species, and also to determine what constitutes adverse
be highly advantageous. Molecular characterization of versus adaptive epigenetic changes in response to
the potential association of drug target modulation with xenobiotic exposure [75,76]. However, efforts are
tumorigenic phenotypes via genetic models, germline already underway to characterize tissue-specific epige-
mutation databases and cancer genome resources has nomes from rodent stocks and strains that are regularly
recently been proposed as an innovative strategy for used for carcinogenicity testing (http://ceficlri.org/
enhanced cancer hazard identification [13]. Where the projects/c3-ed-a-comprehensive-epigenomic-profile-of-
molecular target of non-therapeutic xenobiotic NGCs liver-tissue from-rat and mouse) and phenotypic
can be defined through mechanistic studies, similar anchoring of mechanism-based epigenetic NGC bio-
genetic approaches could be leveraged to help derisk markers provides a way forward towards defining
the potential association of xenobiotic target modulation adversity at the molecular pathway level.
with tumorigenic phenotypes. For example, is there a
cancer phenotype associated with genetic variants or References
genetic modifications of xenobiotic target genes? Are Papers of particular interest, published within the period of review,
have been highlighted as:
tumour suppressor-like somatic mutation spectrums
associated with genes encoding xenobiotic targets in  of special interest
 of outstanding interest
human cancers? Emerging cancer epigenome mapping
resources should also be integrated to strengthen these 1. Downes N, Foster J: Regulatory forum opinion piece: carcin-
xenobiotic target gene assessments. ogen risk assessment: the move from screens to science.
Toxicol Pathol 2015, 43:1064–1073.
2. Friedrich A, Olejniczak K: Evaluation of carcinogenicity studies
6. Conclusion and perspectives of medicinal products for human use authorised via the Eu-
Whilst substantial progress has been made in identifying ropean centralized procedure (1995-2009). Regul Toxicol
Pharmacol 2011, 60:225.
epigenetic mechanisms and biomarkers of xenobiotic-
induced non-genotoxic carcinogenesis in animal 3. Hsu C-H, Stedeford T. Cancer risk assessment: chemical carci-
nogenesis, hazard evaluation, and risk quantification. Wiley;
models, determining the human relevance of structur- 2010. ISBN: 978-0-470-23822-6.
ally and functionally distinct non-genotoxic carcinogenic
4. Holsapple MP, Pitot HC, Cohen SM, Boobis AR, Klaunig JE,
compounds that induce a diverse range of tissue-, Pastoor T, Dellarco VL, Dragan YP: Mode of action in relevance
gender-, strain- and species-specific tumours in animals of rodent liver tumors to human cancer risk. Toxicol Sci 2006,
89:51–56.
still remains a major challenge for toxicologists, although
several rodent NGC modes of action are generally 5. van der Laan JW, Kasper P, Silva Lima B, Jones DR, Pasanen M:
Critical analysis of carcinogenicity study outcomes. Rela-
accepted as not being relevant for humans [4e10]. A tionship with pharmacological properties. Crit Rev Toxicol
number of human cellular models and humanised rodent 2016, 46:587–614.
genetic models have been deployed for NGC hazard 6. Meek ME, Boobis A, Cote I, Dellarco V, Fotakis G, Munn S,
identification but these are unlikely to fully recapitulate Seed J, Vickers C: New developments in the evolution and

www.sciencedirect.com Current Opinion in Toxicology 2017, 3:62–70


68 Risk Assessment in Toxicology – Experimental Toxicology

application of the WHO/IPCS framework on mode of action/ Terranova R, Moggs JG: Phenobarbital induces cell cycle
species concordance analysis. J Appl Toxicol 2014, 34:1–18. transcriptional responses in mouse liver humanized for
constitutive androstane and pregnane x receptors. Toxicol Sci
7. Corton JC, Cunningham ML, Hummer BT, Lau C, Meek B, 2014, 139:501–511.
Peters JM, Popp JA, Rhomberg L, Seed J, Klaunig JE: Mode of
action framework analysis for receptor-mediated toxicity: the 21. Yamada T, Okuda Y, Kushida M, Sumida K, Takeuchi H,
peroxisome proliferator-activated receptor alpha (PPARa) as Nagahori H, Fukuda T, Lake BG, Cohen SM, Kawamura S:
a case study. Crit Rev Toxicol 2014, 44:1–49. Human hepatocytes support the hypertrophic but not the
hyperplastic response to the murine nongenotoxic hepato-
8. Cohen SM: Evaluation of possible carcinogenic risk to carcinogen sodium phenobarbital in an in vivo study using a
humans based on liver tumors in rodent assays: the two-year chimeric mouse with humanized liver. Toxicol Sci 2014, 142:
bioassay is no longer necessary. Toxicol Pathol 2010, 38: 137–157.
487–501.
22. Lempiäinen H, Müller A, Brasa S, Teo SS, Roloff TC, Morawiec L,
9. Cohen SM, Arnold LL: Critical role of toxicologic pathology in a Zamurovic N, Vicart A, Funhoff E, Couttet P, Schübeler D,
short-term screen for carcinogenicity. J Toxicol Pathol 2016, Grenet O, Marlowe J, Moggs J, Terranova R: Phenobarbital
29:215–227. mediates an epigenetic switch at the constitutive androstane
receptor (CAR) target gene Cyp2b10 in the liver of B6C3F1
10. Elcombe CR, Peffer RC, Wolf DC, Bailey J, Bars R, Bell D,
mice. PLoS One 2011, 6:e18216.
Cattley RC, Ferguson SS, Geter D, Goetz A, Goodman JI,
Hester S, Jacobs A, Omiecinski CJ, Schoeny R, Xie W, Lake BG: 23. Phillips JM, Goodman JI: Multiple genes exhibit phenobarbital-
Mode of action and human relevance analysis for nuclear induced constitutive active/androstane receptor-mediated
receptor-mediated liver toxicity: a case study with pheno- DNA methylation changes during liver tumorigenesis and in
barbital as a model constitutive androstane receptor (CAR) liver tumors. Toxicol Sci 2009, 108:273–289.
activator. Crit Rev Toxicol 2014, 44:64–82.
24. Thomson JP, Hunter JM, Lempiäinen H, Müller A, Terranova R,
11. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Moggs JG, Meehan RR: Dynamic changes in 5-
Biankin AV, Bignell GR, Bolli N, Borg A, Børresen-Dale AL, hydroxymethylation signatures underpin early and late
Boyault S, Burkhardt B, Butler AP, Caldas C, Davies HR, events in drug exposed liver. Nucleic Acids Res 2013, 41:
Desmedt C, Eils R, Eyfjörd JE, Foekens JA, Greaves M, 5639–5654.
Hosoda F, Hutter B, Ilicic T, Imbeaud S, Imielinski M, Jäger N,
Jones DT, Jones D, Knappskog S, Kool M, Lakhani SR, López- 25. Allis CD, Jenuwein T: The molecular hallmarks of epigenetic
Otín C, Martin S, Munshi NC, Nakamura H, Northcott PA, Pajic M,  control. Nat Rev Genet 2016, 17:487–500.
Papaemmanuil E, Paradiso A, Pearson JV, Puente XS, Raine K,
Ramakrishna M, Richardson AL, Richter J, Rosenstiel P, 26. Hanahan D, Weinberg RA: Hallmarks of cancer: the next
Schlesner M, Schumacher TN, Span PN, Teague JW, Totoki Y, generation. Cell 2011, 144:646–674.
Tutt AN, Valdés-Mas R, van Buuren MM, van ’t Veer L, Vincent-
27. Shen H, Laird PW: Interplay between the cancer genome and
Salomon A, Waddell N, Yates LR, Australian Pancreatic Cancer
epigenome. Cell 2013, 153:38–55.
Genome Initiative, ICGC Breast Cancer Consortium, ICGC
MMML-Seq Consortium, ICGC PedBrain, Zucman-Rossi J, 28. Feinberg AP, Koldobskiy MA, Göndör A: Epigenetic modulators,
Futreal PA, McDermott U, Lichter P, Meyerson M, Grimmond SM,  modifiers and mediators in cancer aetiology and progres-
Siebert R, Campo E, Shibata T, Pfister SM, Campbell PJ, sion. Nat Rev Genet 2016, 17:284–299.
Stratton MR: Signatures of mutational processes in human
cancer. Nature 2013, 500:415–421. 29. Karsli-Ceppioglu S, Dagdemir A, Judes G, Ngollo M, Penault-
Llorca F, Pajon A, Bignon YJ, Bernard-Gallon D: Epigenetic
12. Thomas F, Roche B, Ujvari B: Intrinsic versus extrinsic cancer mechanisms of breast cancer: an update of the current
risks: the debate continues. Trends Cancer 2016, 2:68–69. knowledge. Epigenomics 2014, 6:651–664.
13. Moggs JG, MacLachlan T, Martus H-J, Bentley P: Derisking 30. Gut P, Verdin E: The nexus of chromatin regulation and
 drug-induced carcinogenicity for novel therapeutics. Trends intermediary metabolism. Nature 2013, 502:489–498.
Cancer 2016, 2:398–408.
31. Berger SL, Sassone-Corsi P: Metabolic signaling to chromatin.
14. Hasmall SC, Roberts RA: The perturbation of apoptosis and  Cold Spring Harb Perspect Biol 2016, 8. pii: a019463.
mitosis by drugs and xenobiotics. Pharmacol Ther 1999, 82:
63–70. 32. Sciacovelli M, Gonçalves E, Johnson TI, Zecchini VR, da
Costa AS, Gaude E, Drubbel AV, Theobald SJ, Abbo SR,
15. Hirose Y, Nagahori N, Yamada T, et al.: Comparison of the ef- Tran MG, Rajeeve V, Cardaci S, Foster S, Yun H, Cutillas P,
fects of the synthetic pyrethroid metofluthrin and phenobar- Warren A, Gnanapragasam V, Gottlieb E, Franze K, Huntly B,
bital on CYP2B form induction and replicative DNA synthesis Maher ER, Maxwell PH, Saez-Rodriguez J, Frezza C: Fumarate
in cultured rat and human hepatocytes. Toxicology 2009, 258: is an epigenetic modifier that elicits epithelial-to-
64–69. mesenchymal transition. Nature 2016, 537:544–547.
16. Parzefall W, Erber E, Sedivy R, Schulte-Hermann R: Testing for 33. Kottakis F, Nicolay BN, Roumane A, Karnik R, Gu H, Nagle JM,
induction of DNA synthesis in human hepatocyte primary Boukhali M, Hayward MC, Li YY, Chen T, Liesa M,
cultures by rat liver tumor promoters. Cancer Res 1991, 51: Hammerman PS, Wong KK, Hayes DN, Shirihai OS, Dyson NJ,
1143–1147. Haas W, Meissner A, Bardeesy N: LKB1 loss links serine
metabolism to DNA methylation and tumorigenesis. Nature
17. La Vecchia C, Negri E: A review of epidemiological data on
2016, 539:390–395.
epilepsy, phenobarbital, and risk of liver cancer. Eur J Cancer
Prev 2014, 23:1–7. 34. Højfeldt JW, Helin K: Regional tumour glutamine supply af-
fects chromatin and cell identity. Nat Cell Biol 2016, 18:
18. Huang W, Zhang J, Washington M, Liu J, Parant JM, Lozano G,
1027–1029.
Moore DD: Xenobiotic stress induces hepatomegaly and liver
tumors via the nuclear receptor constitutive androstane re- 35. Marczylo EL, Jacobs MN, Gant TW: Environmentally induced
ceptor. Mol Endocrinol 2005, 19:1646–1653. epigenetic toxicity: potential public health concerns. Crit Rev
Toxicol 2016, 46:676–700.
19. Braeuning A, Gavrilov, Brown S, Wolf CR, Henderson CJ,
Schwarz M: Phenobarbital-mediated tumor promotion in 36. Watson RE, Goodman JI: Effects of phenobarbital on DNA
transgenic mice with humanized CAR and PXR. Toxicol Sci methylation in GC-rich regions of hepatic DNA from mice that
2014, 140:259–270. exhibit different levels of susceptibility to liver tumorigen-
esis. Toxicol Sci 2002, 68:51–58.
20. Luisier R, Lempiäinen H, Scherbichler N, Braeuning A,
Geissler M, Dubost V, Müller A, Scheer N, Chibout SD, Hara H, 37. Phillips JM, Yamamoto Y, Negishi M, Maronpot RR, Goodman JI:
Picard F, Theil D, Couttet P, Vitobello A, Grenet O, Grasl- Orphan nuclear receptor constitutive active/androstane
Kraupp B, Ellinger-Ziegelbauer H, Thomson JP, Meehan RR, receptor-mediated alterations in DNA methylation during
Elcombe CR, Henderson CJ, Wolf CR, Schwarz M, Moulin P,

Current Opinion in Toxicology 2017, 3:62–70 www.sciencedirect.com


Epigenetic mechanisms of carcinogenesis Terranova et al. 69

phenobarbital promotion of liver tumorigenesis. Toxicol Sci 52. Roadmap Epigenomics Consortium: Integrative analysis of 111
2007, 96:72–82.  reference human epigenomes. Nature 2015, 518:317–330.
38. Pogribny IP, Rusyn I: Role of epigenetic aberrations in the 53. Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT,
development and progression of human hepatocellular car- Haugen E, Sheffield NC, Stergachis AB, Wang H, Vernot B,
cinoma. Cancer Lett 2014, 342:223–230. Garg K, John S, Sandstrom R, Bates D, Boatman L, Canfield TK,
Diegel M, Dunn D, Ebersol AK, Frum T, Giste E, Johnson AK,
39. Lempiäinen H, Couttet P, Bolognani F, Müller A, Dubost V, Johnson EM, Kutyavin T, Lajoie B, Lee BK, Lee K, London D,
 Luisier R, Del Rio Espinola A, Vitry V, Unterberger EB, Lotakis D, Neph S, Neri F, Nguyen ED, Qu H, Reynolds AP,
Thomson JP, Treindl F, Metzger U, Wrzodek C, Hahne F, Roach V, Safi A, Sanchez ME, Sanyal A, Shafer A, Simon JM,
Zollinger T, Brasa S, Kalteis M, Marcellin M, Giudicelli F, Song L, Vong S, Weaver M, Yan Y, Zhang Z, Zhang Z,
Braeuning A, Morawiec L, Zamurovic N, Längle U, Scheer N, Lenhard B, Tewari M, Dorschner MO, Hansen RS, Navas PA,
Schübeler D, Goodman J, Chibout SD, Marlowe J, Theil D, Stamatoyannopoulos G, Iyer VR, Lieb JD, Sunyaev SR,
Heard DJ, Grenet O, Zell A, Templin MF, Meehan RR, Wolf RC, Akey JM, Sabo PJ, Kaul R, Furey TS, Dekker J, Crawford GE,
Elcombe CR, Schwarz M, Moulin P, Terranova R, Moggs JG: Stamatoyannopoulos JA: The accessible chromatin land-
Identification of Dlk1-Dio3 imprinted gene cluster noncoding scape of the human genome. Nature 2012 Sep 6, 489(7414):
RNAs as novel candidate biomarkers for liver tumor promo- 75– 82.
tion. Toxicol Sci 2013, 131:375–386.
54. Shen Y, Yue F, McCleary DF, Ye Z, Edsall L, Kuan S, Wagner U,
40. Luisier R, Unterberger EB, Goodman JI, Schwarz M, Moggs J, Dixon J, Lee L, Lobanenkov VV, Ren B: A map of the cis-
Terranova R, van Nimwegen E: Computational modeling regulatory sequences in the mouse genome. Nature 2012,
identifies key gene regulatory interactions underlying 488:116–120.
phenobarbital-mediated tumor promotion. Nucleic Acids Res
2014, 42:4180–4195. 55. Jin F, Li Y, Dixon JR, Selvaraj S, Ye Z, Lee AY, Yen CA,
Schmitt AD, Espinoza CA, Ren B: A high-resolution map of the
41. Thomson JP, Ottaviano R, Unterberger EB, Lempiäinen H, three-dimensional chromatin interactome in human cells.
 Muller A, Terranova R, Illingworth RS, Webb S, Kerr AR, Lyall MJ, Nature 2013, 503:290–294.
Drake AJ, Wolf CR, Moggs JG, Schwarz M, Meehan RR: Loss of
tet1-associated 5-hydroxymethylcytosine is concomitant 56. Pennacchio LA, Bickmore W, Dean A, Nobrega MA, Bejerano G:
with aberrant promoter hypermethylation in liver cancer. Enhancers: five essential questions. Nat Rev Genet 2013, 14:
Cancer Res 2016, 76:3097–3108. 288–295.
42. Shpyleva S, Dreval K, de Conti A, Kindrat I, Melnyk S, Yan J, 57. Sanyal A, Lajoie BR, Jain G, Dekker J: The long-range inter-
Chen T, Beland FA, Pogribny IP: Organ-specific epigenetic action landscape of gene promoters. Nature 2012, 489:
changes induced by the non-genotoxic liver carcinogen 109–113.
methapyrilene in Fischer 344 rats. Toxicol Sci 2016 Dec 24. pii:
kfw242. [Epub ahead of print], http://dx.doi.org/10.1093/toxsci/ 58. Zhang Y, Wong CH, Birnbaum RY, Li G, Favaro R, Ngan CY,
kfw242; 2016 Dec 24. Lim J, Tai E, Poh HM, Wong E, Mulawadi FH, Sung WK,
Nicolis S, Ahituv N, Ruan Y, Wei CL: Chromatin connectivity
43. Liu L, Luo GZ, Yang W, Zhao X, Zheng Q, Lv Z, Li W, Wu HJ, maps reveal dynamic promoter-enhancer long-range asso-
Wang L, Wang XJ, Zhou Q: Activation of the imprinted Dlk1- ciations. Nature 2013:306–310.
Dio3 region correlates with pluripotency levels of mouse
stem cells. J Biol Chem 2010, 285:19483–19490. 59. Bogdanovic O, Smits AH, de la Calle Mustienes E, Tena JJ,
Ford E, Williams R, Senanayake U, Schultz MD, Hontelez S, van
44. Stadtfeld M, Apostolou E, Akutsu H, ukuda A, Follett P, Natesan S, Kruijsbergen I, Rayon T, Gnerlich F, Carell T, Veenstra GJ,
Kono T, Shioda T, Hochedlinger K: Aberrant silencing of Manzanares M, Sauka-Spengler T, Ecker JR, Vermeulen M,
imprinted genes on chromosome 12qF1 in mouse induced Gómez-Skarmeta JL, Lister R: Active DNA demethylation at
pluripotent stem cells. Nature 2010, 465:175–181. enhancers during the vertebrate phylotypic period. Nat Genet
2016, 48:417–426.
45. Luk JM, Burchard J, Zhang C, Liu AM, Wong KF, Shek FH,
Lee NP, Fan ST, Poon RT, Ivanovska I, Philippar U, Cleary MA, 60. Davie K, Jacobs J, Atkins M, Potier D, Christiaens C, Halder G,
Buser CA, Shaw PM, Lee CN, Tenen DG, Dai H, Mao M: DLK1- Aerts S: Discovery of transcription factors and regulatory
DIO3 genomic imprinted microRNA cluster at 14q32.2 de- regions driving in vivo tumor development by ATAC-seq and
fines a stemlike subtype of hepatocellular carcinoma asso- FAIRE-seq open chromatin profiling. PLoS Genet 2015, 11:
ciated with poor survival. J Biol Chem 2011, 286: e1004994.
30706–30713.
61. Sur I, Taipale J: The role of enhancers in cancer. Nat Rev
46. Wang B, Zhao L, Fish M, Logan CY, Nusse R: Self-renewing  Cancer 2016, 16:483–493.
diploid Axin2(+) cells fuel homeostatic renewal of the liver.
Nature 2015, 524:180–185. 62. Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-André V, Sigova AA,
Hoke HA, Young RA: Super-enhancers in the control of cell
47. De Los Angeles A, Ferrari F, Xi R, Fujiwara Y, Benvenisty N, identity and disease. Cell 2013, 155:934–947.
 Deng H, Hochedlinger K, Jaenisch R, Lee S, Leitch HG,
Lensch MW, Lujan E, Pei D, Rossant J, Wernig M, Park PJ, 63. Saint-André V, Federation AJ, Lin CY, Abraham BJ, Reddy J, Lee TI,
Daley GQ: Hallmarks of pluripotency. Nature 2015, 525: Bradner JE, Young RA: Models of human core transcriptional
469–478. regulatory circuitries. Genome Res 2016, 26:385–396.

48. Thomson JP, Meehan RR: The application of genome-wide 5- 64. Stergachis AB, Neph S, Reynolds A, Humbert R, Miller B,
hydroxymethylcytosine studies in cancer research. Epige- Paige SL, Vernot B, Cheng JB, Thurman RE, Sandstrom R,
nomics 2017:77–91. Haugen E, Heimfeld S, Murry CE, Akey JM,
Stamatoyannopoulos JA: Developmental fate and cellular
49. Ellinger-Ziegelbauer H, Aubrecht J, Kleinjans JC, Ahr HJ: Appli- maturity encoded in human regulatory DNA landscapes. Cell
cation of toxicogenomics to study mechanism of genotox- 2013, 154:888–903.
icity and carcinogenicity. Toxicol Lett 2009, 186:36–44.
65. Maronpot RR: Biological basis of differential susceptibility to
50. Fielden MR, Adai A, Dunn 2nd RR, Olaharski A, Searfoss G, hepatocarcinogenesis among mouse strains. J Toxicol Pathol
Sina J, Aubrecht J, Boitier E, Nioi P, Auerbach S, Jacobson- 2009, 22:11–33.
Kram D, Raghavan N, Yang Y, Kincai A, Sherlock J, Chen SJ,
Car B, Predictive Safety Testing Consortium, Carcinogenicity 66. Dragani TA, Manenti G, Della Porta G: Quantitative analysis of
Working Group: Development and evaluation of a genomic genetic susceptibility to liver and lung carcinogenesis in
signature for the prediction and mechanistic assessment of mice. Cancer Res 1991, 51:6299–6303.
nongenotoxic hepatocarcinogens in the rat. Toxicol Sci 2011,
67. Gariboldi M, Manenti G, Canzian F, Falvella FS, Pierotti MA, Della
12:54–74.
Porta G, Binelli G, Dragani TA: Chromosome mapping of
51. ENCODE Project Consortium: An integrated encyclopedia of murine susceptibility loci to liver carcinogenesis. Cancer Res
DNA elements in the human genome. Nature 2012, 489:57–74. 1993, 53:209–211.

www.sciencedirect.com Current Opinion in Toxicology 2017, 3:62–70


70 Risk Assessment in Toxicology – Experimental Toxicology

68. Drinkwater NR, Ginsler JJ: Genetic control of hepatocarcino- 73. Rusyn I, Gatti DM, Wiltshire T, Kleeberger SR, Threadgill DW:
genesis in C57BL/6J and C3H/HeJ inbred mice. Carcinogen- Toxicogenetics: population-based testing of drug and
esis 1986, 7:1701–1707. chemical safety in mouse models. Pharmacogenomics 2010,
11:1127–1136.
69. Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E,
Wang H, Reynolds AP, Sandstrom R, Qu H, Brody J, Shafer A, 74. Nestor CE, Ottaviano R, Reinhardt D, Cruickshanks HA,
Neri F, Lee K, Kutyavin T, Stehling-Sun S, Johnson AK,  Mjoseng HK, McPherson RC, Lentini A, Thomson JP,
Canfield TK, Giste E, Diegel M, Bates D, Hansen RS, Neph S, Dunican DS, Pennings S, Anderton SM, Benson M, Meehan RR:
Sabo PJ, Heimfeld S, Raubitschek A, Ziegler S, Cotsapas C, Rapid reprogramming of epigenetic and transcriptional pro-
Sotoodehnia N, Glass I, Sunyaev SR, Kaul R, files in mammalian culture systems. Genome Biol 2015, 16:11.
Stamatoyannopoulos JA: Systematic localization of common
disease-associated variation in regulatory DNA. Science 75. Herceg Z, Lambert MP, van Veldhoven K, Demetriou C, Vineis P,
2012, 337:1190–1195. Smith MT, Straif K, Wild CP: Towards incorporating epigenetic
mechanisms into carcinogen identification and evaluation.
70. Mifsud B, Tavares-Cadete F, Young AN, Sugar R, Carcinogenesis 2013, 34:1955–1967.
Schoenfelder S, Ferreira L, Wingett SW, Andrews S, Grey W,
Ewels PA, Herman B, Happe A, Higgs S, LeProust E, Follows GA, 76. Goodman JI, Augustine KA, Cunnningham ML, Dixon D,
Fraser P, Luscombe NM, Osborne CS: Mapping long-range Dragan YP, Falls JG, Rasoulpour RJ, Sills RC, Storer RD,
promoter contacts in human cells with high-resolution cap- Wolf DC, Pettit SD: What do we need to know prior to thinking
ture Hi-C. Nat Genet 2015, 47:598–606. about incorporating an epigenetic evaluation into safety as-
sessments? Toxicol Sci 2010, 116:375–381.
71. Corradin O: Combinatorial effects of multiple enhancer vari-
ants in linkage disequilibrium dictate levels of gene expres- 77. Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH,
sion to confer susceptibility to common traits. Genome Res Zahler AM, Haussler D: The human genome browser at UCSC.
2014, 24:1–13. http://dx.doi.org/10.1101/gr.164079.113. Genome Res 2002, 12:996–1006.

72. Khurana E, Fu Y, Chakravarty D, Demichelis F, Rubin MA, 78. Noe L, Kucherov G: YASS: enhancing the sensitivity of DNA
Gerstein M: Role of non-coding sequence variants in cancer. similarity search. Nucleic Acids Res 2005, 33:W540–W543.
Nat Rev Genet 2016, 17:93–108.

Current Opinion in Toxicology 2017, 3:62–70 www.sciencedirect.com

You might also like