Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

C h a p t e r

8
The Human Epigenome:
Implications for the
Understanding of Human
Disease
Maria Berdasco n Manel Esteller

INTRODUCTION directed toward analyzing the evolution of the


genome, differences between individuals (polymorph-
Epigenetic processes, defined as the heritable patterns of isms), and environmental effects. The near-complete
gene expression that do not involve changes in the sequence has provided a rich source of scientific
sequence of the genome, and their effects on gene knowledge and has initiated new fields of genomic
repression are increasingly understood to be such a way applications. However, the sequence itself does not
of modulating phenotype transmission and develop- predict how the genome is packaged in chromatin to
ment. The patterns of DNA methylation, histone modi- ensure the differential expression of genes, which is
fications, microRNAs, and several chromatin-related essential for development and differentiation [1]. Epi-
proteins of sick cells usually differ from those of healthy genetic processes, defined as the heritable patterns of
cells, highlighting the importance of epigenetic regula- gene expression that do not involve changes in the
tion in most human pathologies. The aim of the present sequence of the genome, and their effects on gene
review is to provide an overview of how epigenetic factors activation and inactivation are increasingly understood
contribute to the development of human diseases such to be such a way of modulating phenotype transmis-
as abnormal imprinting-causative pathologies, cancer sion and development. Epigenetics links the fields of
malignancies, as well as autoimmune, cardiovascular, genetics and developmental biology and can explain,
and neurological disorders. These studies have provided at least in part, the biological process by which identi-
extensive information about the mechanisms that con- cal genotypes establish patterns of differential gene
tribute to the phenotype of human diseases, but also expression that are stable through cell division.
provided opportunities for therapy.

The Human Epigenome Project


EPIGENETIC REGULATION To date, small-scale studies of specific epigenetic
OF THE GENOME marks have provided limited information about the
regulation of genes from different pathways, for exam-
The primary goal of the Human Genome Project was ple, the hypermethylation-dependent silencing of
to determine the sequence of the three billion base tumor suppressor genes in cancer or the mutational
pairs that make up the human genome and to identify inactivation of MeCP2 in Rett patients. However, we
its approximately 25,000 genes from physical and func- need to develop an understanding of these processes
tional standpoints. Most of the human genome (95%) that is based on a broader perspective. A range of mat-
had been sequenced by the end of 2003; of the ters remains to be resolved, such as the relationships
remainder it is likely that the centromeres and telo- between the epigenetic players (the epigenetic code)
meres will remain unsequenced until suitable new and how the environment and/or aging modulate
technology is developed. Current research efforts are the epigenetic marks. Much of this could be achieved

Molecular Pathology # 2009, Elsevier, Inc. All Rights Reserved. 151


Part II Concepts in Molecular Biology and Genetics

by analyzing the epigenetic patterns on a genome-wide phases of development. Nucleus transplantation experi-
scale, an approach that has at last become possible ments in mouse zygotes carrying reciprocal transloca-
thanks to recent technological advances. For example, tions carried out in the early 1980s suggested that
comprehensive DNA-methylation maps (called the imprinting may be fundamental to mammalian devel-
methylome) could be assessed by combining the opment [9]. Assays confirmed that normal gene
methyl-DIP strategy with tilling or promoter microarray expression and development in mice require the con-
analyses [2–4]. In a similar manner, the use of the tribution of both maternal and paternal alleles. How-
chromatin immunoprecipitation technique followed ever, it was not until 1991 that the first imprinted
by genomic microarray hybridization (ChIP-on-chip) genes, insulin-like growth factor 2 (IGF2) and its
has begun to provide extensive maps of histone modifi- receptor (IGF2R), were identified [10]. Since then,
cations [5–7]. Although the groundbreaking discov- 83 imprinted genes have been identified in mice and
eries in the field of human disease were initially humans, about one-third of which are imprinted in
performed in cancer cells, the characterization of the both species [11]. It has recently been predicted that
error-bearing epigenomes underlying other disorders, 600 genes have a high probability of being imprinted
such as neurological, cardiovascular, and immunologi- in the mouse genome, and a similar genome-wide
cal pathologies, has only just begun [8]. The fact that analysis predicts humans to have about half as many
epigenetic aberrations control the function of the imprinted genes [12]. The molecular mechanisms
human genome and contribute to normal and patho- underlying genomic imprinting are poorly under-
logical states justifies carrying out a comprehensive stood. As imprinting is a dynamic process and the pro-
human epigenome project. The goal of the Human file of imprinted genes varies during development,
Epigenome Project is “to identify all the chemical regulation must be epigenetic. DNA methylation has
changes and relationships among chromatin constitu- been widely described as the major mechanism
ents that provide function to the DNA code.” This “will involved in the control of genes subjected to imprint-
allow a fuller understanding of normal development, ing. One model for this regulation is based on the
aging, abnormal gene control in cancer, and other dis- cluster organization of imprinted genes. This structure
eases as well as the role of the environment in human within clusters allows them to share common regu-
health” [1]. It is important to bear in mind that there latory elements, such as noncoding RNAs and differ-
is no single epigenome, but rather many different ones entially methylated regions (DMRs). DMRs are up to
that are characteristic of normal and diverse human dis- several kilobases in size, rich in CpG dinucleotides
orders, so it is essential to define the chosen starting (such as CpG islands), and may contain repetitive
material [1]. The information extracted from the sequences. DNA methylation of DMRs is thought
whole-genome assays will help us understand the role to interact with histone modifications and other chro-
of the epigenetic marks, and could have translational matin proteins to regulate parental allele-specific
research benefits for diagnosis, prognostic, and thera- expression of imprinted genes. Furthermore, the
peutic treatment [8]. Defining human epigenomes aforementioned regulatory elements usually control
associated with human disorders will help select patients the imprinting of more than one gene, giving rise to
who are likely to benefit from epigenomic therapies or imprinting control regions (ICRs). This cluster organi-
prevention strategies, determine their efficacy and spec- zation, observed in 80% of imprinted genes, and the
ificity, and lead to the identification of surrogate mar- specific DNA methylation patterns associated with
kers and end-points of its effects. The aim of the DMRs are two of the main characteristics of imprinted
present review is to provide an overview of how epige- genes. Deletions or aberrations in DNA methylation of
netic factors contribute to the development of human ICRs lead to loss of imprinting (LOI) and inappropri-
diseases such as abnormal imprinting-causative patholo- ate parental gene expression [13]. Imprinted genes
gies, cancer malignancies, as well as autoimmune, car- have diverse roles in growth and cellular proliferation,
diovascular, and neurological disorders with aberrant and specific patterns of genomic imprinting are estab-
epigenetic profiles. lished in somatic and germline cells [12,14]. Imprint-
ing is erased in germline cells, and reprogramming
involving a de novo methyltransferase is necessary to
GENOMIC IMPRINTING ensure sex-specific gene expression in the individual.
Methyl groups are incorporated into most ICRs in
Epigenetic Regulation of Imprinted Genes oocytes, although only some ICRs are methylated dur-
Genomic imprinting is a genetic phenomenon by ing spermatogenesis. After fertilization, the specific
which epigenetic chromosomal modifications drive methylation profiles of ICRs must be maintained dur-
differential gene expression according to the parent- ing development in order to mediate the allelic expres-
of-origin. Expression is exclusively due either to the sion of imprinted genes. In the primordial germline
allele inherited from the mother (such as the H19 cells of the developed individual, these imprinting
and CDKN1C genes) or to that inherited from the marks must be freshly erased by DNA demethylation
father (such as IGF2). It is an inheritance process that to allow the subsequent establishment of new oocyte-
is independent of the classical Mendelian model. Most specific and sperm-specific imprints. As a consequence,
imprinted genes, which have been identified in mammalian imprinting can be described as a develop-
insects, mammals, and flowering plants, are involved ment-dependent cycle based on germline establish-
in the establishment and maintenance of particular ment, somatic maintenance, and erasure.

152
Chapter 8 The Human Epigenome: Implications for the Understanding of Human Disease

Imprinted Genes and Human gene expression depends on parental origin. The clinical
Genetic Diseases features of both syndromes are quite similar; they are neu-
rological disorders with mental retardation and develop-
Since expression of imprinted genes is monoallelic, and mental aberrations [21]. PWS is characterized by
thereby functionally haploid, there is no protection from diminished fetal activity, feeding difficulties, obesity, mus-
recessive mutations that the normal diploid genetic com- cular hypotonia, mental retardation, poor physical coordi-
plement would provide [12]. For this reason, genetic and nation, short stature, hypogonadism, and small hands and
epigenetic aberrations in imprinted genes are linked to a feet, amongst other traits. AS is characterized by mental
wide range of diseases [15]. Modulation of perinatal retardation, movement or balance disorder, characteristic
growth and human pregnancy has played a central role abnormal behaviors, increased sensitivity to heat, absent
in the evolution of imprinting, and many of the diseases or little speech, and epilepsy. The prevalence of the two
associated with imprinted genes involve some disorders syndromes is not accurately known, but is estimated to
of embryogenesis. This is the case of the hydatidiform be between 1 in 12,000 and 1 in 15,000 live births, respec-
mole disorder, where all nuclear genes are inherited tively. Most cases of PWS are caused by the deficiency of
from the father. In most cases, this androgenesis arises the paternal copies of the imprinted genes on chromo-
when an anuclear egg is fertilized by a single sperm, after some 15 located in the 15q11–q13 region, while AS affects
which all the chromosomes and genes are duplicated. maternally imprinted genes in the same region. This defi-
However, fertilization by two haploid sperm (diandric ciency could be due to the deletion of the 15q11–q13
diploidy) may occasionally occur. Most cases are sporadic region (3–4 Mb), parental uniparental disomy of chromo-
and androgenetic, but recurrent hydatidiform mole has some 15, or imprinting defects [22]. The imprinted
biparental inheritance with disrupted DNA methylation domain on human chromosome 15q11–q13 is regulated
of DMRs at imprinted loci [16]. In contrast, the disorder by an ICR that is responsible for establishing the imprint-
of ovarian dermoid cysts arises from the spontaneous acti- ing in the gametes and for maintaining the patterns dur-
vation of an ovarian oocyte that leads to the duplication ing the embryonic phases. ICR regulates differential
of the maternal genome [17]. These abnormalities sug- DNA methylation and chromatin structure, and in conse-
gest that normal human development is possible only quence, differential gene expression affecting the two
when the paternal and maternal genomes are correctly parental alleles. The ICR in 15q11–q13 appears to have a
transmitted. Parent-of-origin effects involved in behav- bipartite structure; one part seems to be responsible for
ioral and brain disorders have been widely reported at the control of paternal expression and the other for
the prenatal and postnatal stages of development [18]. maternal gene expression [23]. PWS is in effect a contigu-
A postnatal growth retardation syndrome associated with ous gene syndrome resulting from deficiency of the pater-
the MEST gene expression is an illustrative example. The nal copies of the imprinted SNRF/SNRPN gene, the
effect of introducing a targeted deletion into the coding necdin gene, and possibly other genes [24,25]. It has been
sequence of the mouse MEST gene strongly depends on estimated that the region could contain more than 30
the paternal allele [19]. When the deletion is paternally genes, so PWS probably results from a stochastic partial
derived, Mestþ/– mice are viable and fertile, but mutant inactivation of important genes [26]. While PWS appears
mice show growth retardation and high mortality. to be more closely related to deficiencies caused by chro-
Mest–/þ animals, with a maternally derived deletion, matin aberrations affecting several genes, AS is associated
show none of these effects. This suggests that the pheno- with mutations in single genes. For instance, the most
typic consequences of this mutation are detected only common genetic defect leading to AS is a 4 Mb maternal
through paternal inheritance and are the result of deletion in chromosomal region 15q11–13, which causes
imprinting. There is also evidence that some imprinting an absence of UBE3A expression in the maternally
effects are associated with increased susceptibility to can- imprinted brain regions. Mutations in the gene encoding
cer. Absence of expression of a tumor suppressor gene the ubiquitin-protein ligase E3A (UBE3A) have been iden-
could be the result of LOI or uniparental disomy tified in 25% of AS patients [27]. The UBE3A gene is pres-
(UPD) in imprinted genes. Conversely, LOI or UPD of ent on both the maternal and paternal chromosomes,
an imprinted gene that promotes cell proliferation (an but differs in its pattern of methylation [28]. Paternal
oncogene) may allow gene expression to be inappropri- silencing of the UBE3A gene occurs in a brain-region-spe-
ately increased. These aberrations could have a wide- cific manner, with the maternal allele being active almost
spread effect if the aberrant imprinting occurs in an exclusively in the Purkinje cells, hippocampus, and cere-
ICR, resulting in the epigenetic dysregulation of multiple bellum [29]. Another maternally expressed gene,
imprinted oncogenes and/or tumor suppressor genes ATP10C (aminophospholipid-transporting ATPase), is
[20]. The most common genetic disorders associated also located in this region and has been implicated in
with imprinting aberrations are described next. AS [30]. Like UBE3A, it exhibits imprinted, preferential
maternal expression in human brain [31].

Prader-Willi Syndrome and


Angelman Syndrome Beckwith-Wiedemann Syndrome
Prader-Willi syndrome (PWS; OMIM #176270) and Angel- Beckwith-Wiedemann syndrome (BWS; OMIM #130650)
man syndrome (AS; OMIM #105830) are very rare genetic is a well-characterized human disease involving imprin-
disorders with autosomal dominant inheritance in which ted genes that are epigenetically regulated, and studies

153
Part II Concepts in Molecular Biology and Genetics

of BWS patients have contributed much to the under- cell differentiation, and other homeostatic functions.
standing of normal imprinting. BWS is a rare genetic However, recent research suggests that these altera-
or epigenetic overgrowth syndrome with an estimated tions could also be due to epigenetic disruption. The
prevalence of about 1 in 15,000 births and a high mor- study of epigenetic mechanisms in cancer, such as
tality rate in the newborn (about 20%). Neonatal DNA methylation, histone modification, nucleosome
patients are mainly characterized by exomphalos, positioning and microRNA expression, has provided
macroglossia, and gigantism, but other symptoms may extensive information about the mechanisms that con-
occur, such as organomegaly, adrenocortical cytome- tribute to the neoplastic phenotype through the regu-
galy, hemihypertrophy, and neonatal hypoglycemia lation of expression of genes critical to transformation
[32]. There is also an increased risk of developing spe- pathways. These alterations and their involvement in
cific tumors, such as Wilms’ tumor and hepatoblastoma tumor development are briefly reviewed in the follow-
[33]. The imprinted domain BWS-related genes are ing sections.
located on 11p15 and are regulated by a bipartite
ICR. Two clusters of imprinting genes have been
described [34]: (i) H19/IGF2 (imprinted, maternally
DNA Hypomethylation in Cancer Cells
expressed, untranslated mRNA/insulin-like growth fac- The low level of DNA methylation in tumors compared
tor 2); and (ii) p57KIP2 (a cyclin-dependent kinase with that in their normal-tissue counterparts was one
inhibitor), TSSC3 (a pleckstrin homology-like domain), of the first epigenetic alterations to be found in
SLC22A1 (an organic cation transporter), KvLQT1 (a human cancer [44]. It has been estimated that 3–6%
voltage-gated potassium channel), and LIT1 (KCNQ1 of all cytosines are methylated in normal human
overlapping transcript 1). Both clusters are regulated DNA, although cancer cell genomes are usually hypo-
by two DMRs differentially methylated regions: DMR1, methylated with malignant cells, featuring 20–60% less
which is responsible for H19/IGF2 control and is genomic 5-methylcytosine than their normal counter-
methylated on the paternal but not the maternal allele, parts [45]. Global hypomethylation in cancer cells
and DMR2, which is located upstream of LIT1 and is generally due to decreased methylation in CpGs
is normally methylated on the maternal but not the dispersed throughout repetitive sequences, which
paternal allele. BWS can appear as a consequence of account for 20–30% of the human genome, as well as
two separate mechanisms [35]. First, some patients in the coding regions and introns of genes [45]. A
are characterized by UPD, which consists of the com- chromosome-wide and large-promoter-specific study
plete genetic replacement of the maternal allele region of DNA methylation in a colorectal cancer cell line
with a second paternal copy, and/or LOI affecting using the methyl-DIP approach has revealed extensive
the IGF2-containing region [36] and/or the LIT1 gene hypomethylated genomic regions located in gene-poor
[37], which causes a switch in the epigenotype of areas [2]. Importantly, the degree of hypomethylation
the H19/IGF2 subdomain or the p57/ KvLQT1/LIT1 of genomic DNA increases as the lesion progresses
subdomain, respectively. When UPD affects IGF2, it from a benign cellular proliferation to an invasive
yields a double dose of this autocrine factor, resulting cancer [46]. From a functional point of view, hypo-
in tissue overgrowth and increased cancer risk [38]. methylation in cancer cells is associated with a number
The LOI mechanism involves aberrant methylation of of adverse outcomes, including chromosome instab-
the maternal H19 DMR [39]. Second, maternal repla- ility, activation of transposable elements, and LOI
cements of the allele and localized abnormalities of (Figure 8.1). Decreased methylation of repetitive
allele-specific chromatin modification on p57KIP2 sequences in the satellite DNA of the pericentric
(also known as CDKN1C) [40] or LIT1 could also con- region of chromosomes is associated with increased
tribute to the BWS phenotype [41]. In conclusion, chromosomal rearrangements, mitotic recombination,
BWS is a model for the hierarchical organization of epi- and aneuploidy [47,48]. Intragenomic endoparasitic
genetic regulation in progressively larger domains DNA, such as L1 (long interspersed nuclear elements)
[42]. Additionally, mutations in NSD1 (nuclear receptor- [49] and Alu (recombinogenic sequence) repeats, are
binding SET domain-containing protein 1), the major silenced in somatic cells and become reactivated in
cause of the Sotos overgrowth syndrome, have been human cancer. Deregulated transposons could cause
described in BWS patients, demonstrating the role this transcriptional deregulation, insertional mutations,
gene plays in imprinting the chromosome 11p15 DNA breaks, and an increased frequency of recombi-
region [43]. nation, contributing to genome disorganization,
expression changes, and chromosomal instability
[49]. DNA methylation underlies the control of several
imprinted genes, so the effect on the loss of imprint-
CANCER EPIGENETICS ing must also be considered. Wilms’ tumor, a nephro-
Cancer encompasses a fundamentally heterogeneous blastoma that typically occurs in children, is the best-
group of disorders affecting different biological pro- characterized imprinting effect associated with
cesses, and is caused by abnormal gene/pathway func- increased susceptibility to cancer [50]. Other changes
tion arising from specific alterations in the genome. in the expression of imprinted genes caused by
Initially, cancer was thought to be solely a consequence changes in methylation have been demonstrated in
of genetic changes in key tumor suppressor genes and malignancies such as osteosarcoma [51], hepatocellu-
oncogenes that regulate cell proliferation, DNA repair, lar carcinoma [52], and bladder cancer [53]. Finally,

154
Chapter 8 The Human Epigenome: Implications for the Understanding of Human Disease

Promoter of TSGs Repetitive sequence


RNA HDAC
Pol MBD
TF
HMT
Me Me Rb
Ac Ac Ac Me
MeMe Ac
MeMe MeMe MeMe

K16H4 K8H4 K4H3 K16H4 K20H4 K9H3 K27H3


K5H4 K12H4

Normal Cell

Normal Cell
TRANSCRIPTION REPRESSION

Unmethylated CpG
Methylated CpG HDAC
MBD
HMT Me
Ac Ac Ac
Me MeMe MeMe
MeMe MeMe
K16H4 K8H4 K4H3 K20H4
K9H3 K27H3 K5H4 K12H4
Cancer Cell

Cancer Cell
A SILENCING B ACTIVATION

Figure 8.1 A model for the disruption of histone modifications and DNA methylation patterns in cancer cells.
Nucleosome arrays are located in the context of genomic regions that include (A) promoters of tumor suppressor genes
(TSGs), (B) repetitive sequences in heterochromatin regions. Nucleosomes consisting of two copies of histones H2A, H2B,
H3, and H4 are represented as gray cylinders. DNA (black lines) is wrapped around each nucleosome. In normal cells,
TSG promoter regions are unmethylated and enriched in histone modification marks associated with active transcription,
such as acetylation of histone H4 (at lysine K5, K8, K12, and K16), or trimethylation of histone H3 (at lysine K4).
Transcription machinery recognizes these active marks and transcription of TSGs is allowed. In the same cells, the
repetitive genomic DNA is silenced due to the high degree of DNA methylation and histone-repressive marks: histone H4 is
densely trimethylated at lysine 20, and histone H3 is dimethylated at lysine 9 and trimethylated at lysine k27. This
epigenetic profile is disrupted in transformed cells. TSG promoters are silenced by the loss of the histone-active marks and
gain of promoter hypermethylation. Repetitive sequences are activated by replacement of the repressive marks, leading to
activation of endoparasitic sequences, genomic instability, or loss of imprinting.

DNA methylation acts a mechanism for controlling a hypomethylation-dependent manner. Activation of


cellular differentiation, allowing the expression only PAX2, a gene that encodes a transcription factor
of tissue-specific and housekeeping genes in somatic involved in proliferation and other important cell
differentiated cells. It is possible that some tissue- activities, and let-7a-3, an miRNA gene, have been
specific genes became reactivated in cancer [54] in implicated in endometrial and colon cancer [55,56].

155
Part II Concepts in Molecular Biology and Genetics

Hypermethylation of Tumor sites after genistein induction (an isoflavone found in


Suppressor Genes the soybean with tumor suppressor properties), in the
absence of p21 promoter methylation, have been
Aberrations in DNA methylation patterns of the CpG reported [64]. The association between DNA methyla-
islands in the promoter regions of tumor suppressor tion and histone modification aberrations in cancer also
genes are accepted as being a common feature of occurs at the global level. In human and mouse tumors,
human cancer (Figure 8.1). The initial discovery of histone H4 undergoes a loss of monoacetylated and tri-
silencing was performed in the promoter of the retino- methylated lysines 16 and 20, respectively, especially in
blastoma (Rb) tumor suppressor gene [57], but hyper- the repetitive DNA sequences [46]. Subsequent studies
methylation of genes like VHL (associated with von showed that loss of trimethylation at H4K20 is involved
Hippel-Lindau disease), p16INK4a, 8-11 hMLH1 (a in disrupting heterochromatic domains and may reduce
homologue of Escherichia coli MutL), and BRCA1 the response to DNA damage of cancer cells [65]. Immu-
(breast-cancer susceptibility gene 1) has also been nohistochemical staining of primary prostatectomy tis-
described [58]. The presence of CpG island promoter sue samples revealed that patterns of H3 and H4 were
hypermethylation affects genes from a wide range of predictors of clinical outcome independently of tumor
cellular pathways, such as cell cycle, DNA repair, toxic stage, preoperative prostate-specific antigen levels, and
catabolism, cell adherence, apoptosis, and angiogene- capsule invasion in prostate cancer [66].
sis, among others [45], and may occur at various stages
in the development of cancer. In recent years, a CpG-
island hypermethylation profile of human primary Epigenetic Regulation of microRNAs
tumors has emerged, which shows that the CpG island in Cancer
hypermethylation profiles of tumor suppressor genes
are specific to the cancer type [59,60]. Each tumor miRNA expression patterns must be tightly regulated
type can be assigned a specific, defining DNA “hyper- during development in a tissue-specific manner, and
methylome,” rather like a physiological or cytogenetic play important roles in cell proliferation, apoptosis,
marker. These marks of epigenetic inactivation occur and differentiation [67]. miRNA expression profiles dif-
not only in sporadic tumors but also in inherited cancer fer between normal and tumor tissues, and also among
syndromes, in which hypermethylation may be the sec- tumor types [68], whereby some microRNAs are down-
ond lesion in Knudson’s two-hit model of cancer devel- regulated in cancer (like tumor suppressor genes). This
opment [61]. It is expected that improvements in comparison suggests that miRNAs could be silenced by
genome-wide epigenomic studies will increase the num- epigenetic mechanisms [69]. DNA methylation has
ber of hypermethylated tumor suppressor genes in a been shown to be the regulatory mechanism in at least
broad spectrum of tumors. However, to date 100–400 two microRNAs, miR-127 and miR-124a [69]. miR-127,
instances of gene-specific methylation have been noted which negatively regulates the protooncogene BCL6
in a given tumor. Sometimes the epigenetic alteration (B-Cell Lymphoma 6), is usually expressed in normal
of a tumor suppressor gene has genetic consequences, cells but is silenced by DNA methylation in cancer cells
for example, when a DNA repair gene (hMLH1, BRCA1, [70]. Similarly, the hypermethylation-dependent silenc-
MGMT, or Werner’s syndrome gene) is silenced by pro- ing of miR-124a results in an increase in expression of
moter methylation and functionally blocked [58,62]. cyclin D-kinase 6 oncogene (CDK6), and is recognized
as a common feature of a wide range of tumors [71].

Histone Modifications of Cancer Cells Aberrations in Histone-Modifier Enzymes


The histone modification network is very complex. Aberrations in the epigenetic profiles, with respect to
Histone modifications can occur in various histone pro- DNA methylation and histone modifications, could also
teins (including H2B, H3, H4) and variants (such as be a consequence of genetic disruption of the epige-
H3.3) and affect different histone residues (including netic machinery. A preliminary set of genes involved
lysine, arginine, serine). Several chemical groups in epigenetic modifications with mutations in cancer
(methyl, acetyl, phosphate) may be added in different cells but not in the normal counterparts has been
degrees (for example, monomethylation, dimethylation, found[45]. A list of genes involved in epigenetic modi-
or trimethylation). Furthermore, the significance of fications that are disrupted in human cancer is pre-
each modification depends on the organism, the sented in Table 8.1. Although alterations in the levels
biological process, and the chromatin-genomic region. of DNMTs and MBD-containing proteins are com-
Due to this diversity of permutations and combinations, monly observed in human tumors [72,73], no genetic
little is known about the patterns of histone modification lesion has been described in the DNA-methylation
disruption in human tumors. Recent results have shown machinery in cancer cells. The picture is different for
that the CpG promoter-hypermethylation event in histone modifier enzymes. In leukemia and sarcoma,
tumor suppressor genes in cancer cells is associated with chromosomal translocations that involve histone-modi-
a particular combination of histone markers: deacetyla- fier genes, such as histone acetyltransferases (HATs)
tion of histones H3 and H4, loss of H3K4 trimethylation, [74] [such as cyclic AMP response-element-binding
and gain of H3K9 methylation and H3K27 trimethyla- protein (CREB)-binding protein-monocytic leukemia
tion (Figure 8.1) [63]. Increased acetylated histones H3 zinc finger (CBP-MOZ)] and histone methyltransferases
and H4, and H3/K4 at the p21 and p16 transcription start (HMTs) [such as mixed-lineage leukemia 1 (MLL1)]
156
Chapter 8 The Human Epigenome: Implications for the Understanding of Human Disease

Table 8.1 Disruption of Genes Involved in DNA Methylation and Histone Modifications in Cancer

Gene Alteration Tumor type

Alterations affecting DNA methylation enzymes (DNMTs)


DNMT1 Overexpression Various
DNMT3b Overexpression Various
Alterations involving Methyl-CpG-binding proteins (MBPs)
MeCP2 Overexpression, rare mutations Various
MBD1 Overexpression, rare mutations Various
MBD2 Overexpression, rare mutations Various
MBD3 Overexpression, rare mutations Various
MBD4 Mutations in microsatellite instable tumors Colon, stomach, endometrium
Alterations disrupting histone acetyltransferases (HATs)
p300 Mutations in microsatellite instable tumors Colon, stomach, endometrium
CBP Mutations, translocations, deletions Colon, stomach, endometrium, lung, leukemia
pCAF Rare mutations Colon
MOZ Translocations Hematological malignancies
MORF Translocations Hematological malignancies, leiomyomata
Alterations disrupting histone deacetylases (HDACs)
HDAC1 Aberrant expression Various
HDAC2 Aberrant expression, mutations in microsatellite instable Various
tumors
Alterations affecting histone methyltransferases (HMTs)
MLL1 Translocation Hematological malignancies
MLL2 Gene amplification Glioma, pancreas
MLL3 Deletion Leukemia
NSD1 Translocation Leukemia
EZH2 Gene amplification, overexpression Various
RIZ1 Promoter CpG-island hypermethylation Various
Alterations affecting histone demethylases
GASC1 Gene amplification Squamous cell carcinoma

Adapted from [45].

[75], nuclear-receptor binding SET domain protein 1 cells, but nowadays there is increasing interest in under-
(NSD1) [76], and nuclear-receptor binding SET- standing the role of epigenetic modifications in the etiol-
domain protein 3 (NSD3) [77], create aberrant fusion ogy of human disease. This section summarizes the
proteins [8]. In solid tumors, both HMT genes such as involvement of specific epigenetic aberrations in immu-
EZH2 [78], mixed-lineage leukemia 2 (MLL2) [79], or nological, cardiovascular, and neurological disorders.
NSD3 [80], and a demethylase [Jumonji domain-con- DNA methylation is the best-characterized epigenetic
taining protein 2C (JMJD2C/GASC1)] are known to be modification in many pathways of immunology, and is
amplified [81]. Genetic aberrations also disrupt expres- the source of much of our knowledge about the molecu-
sion of histone deacetylases, such as histone deacetylase lar network of the immune system. Classical autoimmune
2 (HDAC2), which could be affected by mutational disorders, such as systemic lupus erythematosus (SLE),
frameshift inactivation in colon cancer [82], and chro- an autoimmune disease characterized by the production
matin remodeling proteins, such as HLTF (helicase-like of a variety of antibodies against nuclear components and
transcription factor) [83], BRG1 (Brahma-related gene which causes inflammation and injury of multiple
1) [84], and other components of the SWI/SNF family organs, and rheumatoid arthritis, a chronic systemic
of proteins. autoimmune disorder which primarily causes inflamma-
tion and destruction of the joints, are characterized
by massive genomic hypomethylation [85,86]. This
HUMAN DISORDERS ASSOCIATED decrease in DNA methylation levels is highly reminiscent
WITH EPIGENETICS of the global demethylation observed in the DNA of
tumor cells compared with their normal tissue counter-
Aberrant Epigenetic Profiles Underlying parts [8]. How this hypomethylation in T-cells induces
Immunological, Cardiovascular, SLE is not well understood. It has been proposed that
demethylation induces overexpression of integrin adhe-
Neurological, and Metabolic Disorders sive receptors and leads to an autoreactive response.
The patterns of DNA methylation, histone modifications, Identification of the full set of genes deregulated by
microRNAs, and several chromatin-related proteins of DNA hypomethylation could help to explain these
sick cells usually differ from those of healthy cells, high- immunological disorders and will also enable the devel-
lighting the importance of epigenetic regulation in most opment of effective therapies to cure SLE [85]. More
human pathologies. Most of our knowledge regarding pathologies with epigenetic regulation of immunology
human epigenetic diseases was first obtained from cancer have been reported, including the epigenetic silencing
157
Part II Concepts in Molecular Biology and Genetics

of the ABO histo-blood group genes [87], the silencing of Syndromes of Disordered Chromatin Remodeling
human leukocyte antigen (HLA) class I antigens [88],
and the melanoma antigen-encoding gene (MAGE) fam- Alpha-Thalassemia X-Linked Mental Retardation
ily. It is known that MAGE gene expression is epigeneti- Syndrome Alpha-thalassemia X-linked mental retar-
cally repressed by promoter CpG methylation in most dation (ATRX; OMIM #301040) syndrome is charac-
cells, but MAGE genes may be expressed in various tumor terized by distinctive craniofacial features, genital
types via CpG demethylation and can act as antigens that anomalies, and severe developmental delays with hypo-
are recognized by cytolytic T-lymphocytes [89]. Altera- tonia and mental retardation. Some individuals with
tions of specific genomic DNA methylation levels have ATRX syndrome have an unusual mild form of hemo-
been described not only in the fields of oncology and globin H disease, but alpha-thalassemia is not a constant
immunology but also in a wide range of biomedical and characteristic of the syndrome [94]. ATRX is the gene
scientific fields. In neurology, for example, mutations in associated with ATRX syndrome and codes for a cen-
MeCP2, which encodes methyl CpG binding protein 2, tromeric heterochromatin-binding protein from the
cause most cases of Rett syndrome (RTT) and autism SNF2 family of helicase/ATPases. Centromeric ATRX
[90]. Future research needs to determine whether aber- is required for maintaining a bipolar metaphase II spin-
rant DNA methylation is important in the complex etiol- dle and for establishing correct chromosome alignment
ogy of other frequent neurological pathologies, such as during meiosis. Its activity has been associated with
schizophrenia and Alzheimer’s disease. Beyond this, epigenetic modifications, such as histone deacetylation
DNA methylation changes are also known to be involved and DNA methylation [95].
in cardiovascular disease, the biggest killer in Western
countries [8]. For example, aberrant CpG island hyper- CHARGE Syndrome The acronym CHARGE (OMIN
methylation has been described in atherosclerotic lesions #214800) stands for Coloboma of the eye, Heart
[91]. Since DNA methylation and histone modifications anomaly, choanal Atresia, Retardation of mental and
are mechanistically linked, it is likely that different somatic development, Genital and/or urinary abno-
changes in DNA methylation are associated with changes rmalities, and Ear abnormalities and/or deafness
in histone modifications in human diseases. To date, we [96]. The estimated incidence of CHARGE syndrome
have been largely ignorant of how these histone modifi- is 1 in 8,500–12,000 births. It is an autosomal domi-
cation markers are disrupted in human diseases. A pre- nant condition with genotypic heterogeneity. Most
view of the patterns of histone modifications and their cases are due to the mutation or deletion of a mem-
cellular location has only been described for cancer ber of the chromodomain helicase DNA-binding
malignancies [45]. For other human pathologies, our domain (CHD7) family of ATP-dependent chromatin-
knowledge of the alterations in histone modification remodeling enzymes [97], and in consequence, a pro-
patterns comes from the use of epigenetic drugs. Thera- tein involved in chromatin structure and gene
peutic assays have demonstrated that histone deacetylase expression.
(HDAC) inhibitors can improve deficits in synaptic plas-
ticity, cognition, and stress-related behaviors in a wide Cockayne Syndrome Patients with Cockayne syndrome
range of neurological and psychiatric disorders, includ- type B (CSB; OMIM #133540) present with failure to
ing Huntington’s and Parkinson’s diseases, anxiety and thrive, short stature, premature aging, neurological
mood disorders, and Rubinstein-Taybi and Rett syn- alterations, photosensitivity, delayed eruption of the
dromes [92]. Abnormal histone modification patterns primary teeth, congenital absence of some permanent
associated with specific gene expression have also been teeth, partial macrodontia, atrophy of the alveolar
described in lupus CD4þ T-cells [93], and HDAC inhibi- process, and caries [98]. CSB is a genetic disorder with
tors are able to reverse gene expression significantly [85]. autosomal recessive inheritance that is caused by muta-
tion of the excision-repair cross-complementing group
6 (ERCC6) gene. The protein, with ATP-stimulated
Genetic Aberrations Involving Epigenetic ATPase activity, is part of the nucleotide excision repair
Genes (NER) pathway, a complex system that eliminates a
Genetic alterations of genes coding for enzymes that broad spectrum of structural DNA lesions, including
mediate chromatin structure could result in a loss of ade- ultraviolet (UV)-induced dimers and DNA cross-links
quate regulation of chromatin compaction, and finally, [99].
the deregulation of gene transcription and inappropriate
protein expression. Although the consequences in cancer Schimke Immunoosseous Dysplasia Schimke immu-
malignancies have been widely described, in this section noosseous dysplasia (SIOD; OMIM #242900) is an
we extend the review to include other genetic diseases autosomal recessive disorder with the diagnostic
involving the function of several enzymes of the epigenetic features of growth retardation, renal failure, recur-
machinery. The phenotype of these diseases also helps to rent infections, cerebral infarcts, slowly progressive
clarify the role of various chromatin proteins in cell prolif- immune defects, such as T-cell immunodeficiency
eration and differentiation. These include disorders aris- and skin pigmentation, that begin in childhood
ing from alterations in chromatin remodeling factors, [100]. Mutations in a component of the SWI/SNF
alterations of the components of the DNA methylation complex, the SMARCAL1 gene, are responsible for
machinery, and aberrations disturbing histone modifiers. this condition [101].

158
Chapter 8 The Human Epigenome: Implications for the Understanding of Human Disease

DNA Methylation-Associated Diseases with putative broad-ranging effects on expression of


genes that otherwise are not mutated [111]. It has been
Immunodeficiency-Centromeric Instability-Facial proposed that MeCP2 disruption in RTT patients causes
Anomalies Syndrome aberrant upregulation of the genes that should nor-
mally be silenced in the nervous system, like the BDNF
Immunodeficiency-centromeric instability-facial ano-
gene or imprinted genes [112]. The discovery that
malies syndrome (ICF syndrome; OMIM #242860) is
MeCP2 was the gene responsible for RTT syndrome
characterized by immunodeficiency in association with
allows it to be used for early diagnosis and prenatal
centromere instability of chromosomes 1, 9, and 16,
detection. In addition, the finding that epigenetic regu-
and facial anomalies. ICF syndrome patients exhibit
lation has a role in the pathogenesis of RTT has sug-
facial anomalies, such as hypertelorism, low-set ears,
gested opportunities for therapy.
epicanthal folds, and macroglossia. It also features
variable reductions in serum immunoglobulin levels
that cause most ICF syndrome patients to succumb to
infectious diseases before adulthood [102]. It is a rare Human Malignancies Associated with
autosomal recessive disorder that is notable for the Alterations in Histone Modifications
variable severity of chromosomal abnormalities. Essen-
tially, these involve juxtacentromeric heterochromatin Coffin-Lowry Syndrome
formation of chromosomes 1, 9, and 16; an increased The symptoms of Coffin-Lowry syndrome (CLS; OMIM
frequency of somatic recombination between the arms #303600) are usually more severe in males than in
of these chromosomes; and a marked tendency to form females since it is an X-linked condition. Males with
multibranched configurations [103]. The observation CLS syndrome usually exhibit severe-to-profound
that the cytogenetic abnormalities in the phenotype of mental retardation and delayed development, but in
ICF syndrome could be compared with the decondensa- female patients this may be entirely absent, or mild-to-
tion effect found in a normal pro-B lymphoblastoid profound [113]. Other features, such as skeletal malfor-
cell line treated with the DNA methylation inhibitors mations, distinctive facial features, microcephaly, and
5-azacytidine or 5-azadeoxycytidine suggested the involve- soft hands with short-tapered fingers, may be observed.
ment of genomic DNA methylation in this disorder. CLS has an estimated incidence of 1 in 40,000–50,000
In fact, 40% of ICF syndrome patients owe their condi- births. It is caused by mutations in the RSK2 (ribosomal
tion to mutations in the DNMT3B gene, which encodes protein S6 kinase) gene [114]. RSKs are involved in
a de novo DNA-methyltransferase that acts on CpG-rich activating the mitogen-activated kinase cascade and in
satellite DNAs [104]. In this way, ICF syndrome can be stimulating cell proliferation (transition between the
biochemically characterized by hypomethylation of G0 and G1 phases of the cell cycle), and differentiation.
CpG sites in some heterochromatic regions. RSK2 is responsible for the phosphorylation of the tran-
scription factor CREB (cyclic AMP-responsive-binding
protein) and histone H3 as a mechanism for respond-
Rett Syndrome ing to mitogenic stimulation by epidermal growth
Rett syndrome (RTT; OMIM #312750) is a severe neuro- factor (EGF) [115]. Analysis of mRNA and protein
developmental disorder that primarily occurs in females expression of RSK2 on lymphocytes extracted directly
at an estimated frequency of 1 in 15,000 births. It is char- from blood of patients revealed a mutation at a putative
acterized by arrested development between 6 and 18 phosphorylation site that would be critical for RSK2
months of age, regression of acquired skills, loss of activity. Both assays are rapid and practical techniques
speech, stereotypical movements (classically of the for the diagnosis of CLS [116].
hands), microcephaly, seizures, and mental retardation.
Rarely, RTT has also been described in male patients
with somatic mosaicism or an extra X chromosome
Rubinstein-Taybi Syndrome
[105]. Mutations in the coding regions of the MeCP2 Rubinstein-Taybi syndrome (RSTS; OMIM #180849) is
(methyl CpG-binding protein 2) gene are found in an autosomal dominant disorder characterized by
approximately 70–80% of classic RTT patients [106], mental retardation, broad thumbs and toes, distinctive
while the remaining cases result from large deletions facial abnormalities, and short stature. RSTS patients
of the MeCP2 gene [107], mutations in noncoding have an increased risk of developing tumors, especially
regions of the MeCP2 gene, and mutations in other congenital glaucoma. The incidence has been esti-
genes [108]. An atypical Rett syndrome with infantile mated to be in 1 in 100,000 births [117]. In almost
spasms that may be related to mutations in the cyclin- all cases, the syndrome is caused by mutation of the
dependent kinase-like 5 (CDKL5) gene has also been gene encoding the transcriptional coactivator CREB-
described [109]. The MeCP2 gene encodes a protein binding protein (CREBBP) [118]. Mutations in the
that preferentially binds methylated CpG dinucleotides EP300 gene are responsible for a small percentage of
and, in turn, mediates transcriptional repression RSTS cases [119]. Both CREBBP and EP300 are his-
through the recruitment of histone deacetylases, his- tone acetyltransferases. A direct connection between
tone methyltransferases, and polycomb proteins, loss of acetyltransferase activity and RSTS has been
among others [110]. In addition, cells of RTT patients described [120], which indicates that the disorder is
may have an abnormal secondary chromatin structure, caused by aberrant chromatin regulation.

159
Part II Concepts in Molecular Biology and Genetics

Sotos Syndrome megaloblastic anemia, hemolytic uremic syndrome, and


pulmonary hypertension, among others [129]. Exposure
Sotos syndrome (OMIM #117550) is a well-known over- to metals, such as arsenic [130], cadmium [131], lead
growth syndrome with autosomal dominant inheritance [132], nickel [133], and chromium [134], is linked to
characterized by excessive growth during childhood, changes in the expression of epigenetically controlled
advanced bone age, macrocephaly, characteristic facial genes via interactions with DNA-methylation-associated
gestalt, and various degrees of developmental delay enzymes, histone acetyltransferase, and histone deacety-
[121]. It may also be associated with variable minor lase enzymes. Endocrine-active chemicals, like estrogenic
features, including cardiac and renal anomalies, and anti-androgenic toxins, decrease male fertility by
seizures, and scoliosis. The risk of neoplasm formation manipulating DNA methylation levels [135]. Among
in Sotos patients has been estimated to be about 2–3% the behavioral factors, maternal grooming is an influen-
[122], which is greater than in the general population. tial environmental factor affecting the methylation
The most frequent tumor types are leukemia and lym- state of the glucocortocoid receptor in the hippocampus
phoma, but others, such as neural crest tumors, small- of rat pups [136]. Reproductive factors like assisted
cell lung cancer, and sacrococcygeal teratomas have reproductive technology act as an environmental modu-
been reported [123]. Sotos syndrome is caused by hap- lator of the epigenome, and are associated with the
loinsufficiency of the nuclear receptor-binding SET hypomethylation noted in Beckwith-Wiedemann and
domain-containing protein 1 gene (NSD1). Several Angelman syndromes [137]. Studies performed with
mutations cause the loss of NSD1 function in 90% of genetically identical individuals are an excellent method
patients with Sotos syndrome [121], such as truncating for understanding the environment-epigenome interac-
mutations, missense mutations, 5q35 macrodeletions tion [138,139]. In this way, the epigenome of human
and microdeletions encompassing the NSD1 gene or monozygotic twins could also vary because of differences
the t(5;11)(q35;p15.5) translocation, which fuses NSD1 in environmental factors. Recently, it was noted that
to nucleoporin-98 (NUP98 ). NSD1 contains a SET although twins are epigenetically indistinguishable
domain that confers intrinsic histone methyltransferase during the early years of life, older monozygous twins
activity that is specific to lysine 36 of histone H3 exhibit marked differences in their overall content and
(H3K36) and lysine 20 of histone H4 (H4K20), epige- genomic distribution of 5-methylcytosine DNA and
netic marks with ascribed roles in chromatin structure histone acetylation, which affects their gene-expression
and gene function. portrait [139].

ENVIRONMENT AND THE EPIGENOME REFERENCES


Gene expression strongly depends on environmental 1. Jones PA, Martienssen R. A blueprint for a Human Epigenome
signals—the external and endogenous stimuli—that Project: the AACR Human Epigenome Workshop. Cancer Res.
make up the cellular context. Since epigenetic states 200; 6:11241–11246.
can potentially be reverted, these gene-environment 2. Weber M, Davies JJ, Wittig D, et al. Chromosome-wide and pro-
moter-specific analyses identify sites of differential DNA methyla-
interactions could be integrated by epigenetic modifica- tion in normal and transformed human cells. Nat Genet.
tions of the genome [124]. Disease susceptibility also 2005;37:853–862.
could be modified by environmental factors that alter 3. Zhang X, Yazaki J, Sundaresan A, et al. Genome-wide high-reso-
the profile of DNA methylation and histone modifica- lution mapping and functional analysis of DNA methylation in
arabidopsis. Cell. 2006;126:1189–1201.
tions in normal cells during prenatal and postnatal life. 4. Jacinto FV, Ballestar E, Esteller M. Methyl-DNA immunoprecipi-
Alterations in the epigenome, and subsequent differen- tation (MeDIP): Hunting down the DNA methylome. Biotechni-
tial gene expression, could be directly affected by diet, ques. 2008;44:35, 37, 39 passim.
xenobiotic chemicals, behavior, and exogenous stimuli, 5. Kurdistani SK, Tavazoie S, Grunstein M. Mapping global histone
such as doses of radiation [12]. Aberrant epigenetic pat- acetylation patterns to gene expression. Cell. 2004;117:721–733.
6. Bernstein BE, Kamal M, Lindblad-Toh K, et al. Genomic maps
terns could be a consequence of changes in the metabo- and comparative analysis of histone modifications in human
lism of the groups of donor chemicals, such as methyl or and mouse. Cell. 2005;120:169–181.
acetyl groups. Diets that are deficient in folate and 7. Azuara V, Perry P, Sauer S, et al. Chromatin signatures of plurip-
methionine, which are necessary for normal biosynthe- otent cell lines. Nat Cell Biol. 2006;8:532–538.
8. Esteller M. The necessity of a human epigenome project. Carci-
sis of S-adenosylmethionine (SAM), the methyl donor nogenesis. 2006;27:1121–1125.
for methylcytosine, lead to DNA hypomethylation and 9. Surani MA, Barton SC, Norris ML. Development of reconstituted
aberrant imprinting of insulin-like growth factor mouse eggs suggests imprinting of the genome during gameto-
2 (IGF2) [125]. Mutations in methylenetetrahydrofolate genesis. Nature. 1984;308:548–550.
reductase (MTHFR-677T allele), which is critical for 10. DeChiara TM, Robertson EJ, Efstratiadis A. Parental imprinting of
the mouse insulin-like growth factor II gene. Cell. 1991;64:849–859.
directing the folate pool to remethylate homocysteine 11. Morison IM, Ramsay JP, Spencer HG. A census of mammalian
to methionine, reduce the levels of DNA methylation imprinting. Trends Genet. 2005;21:457–465.
responsible for the pathogenesis of diseases related to 12. Dolinoy DC, Weidman JR, Jirtle RL. Epigenetic gene regulation:
neural tube defects [126], spina bifida [127], and colo- Linking early developmental environment to adult disease.
Reprod Toxicol. 2007;23:297–307.
rectal cancer [128]. Associations are also known 13. Murrell A, Ito Y, Verde G, et al. Distinct methylation changes at
between deficiency of methionine synthase, a folate- the IGF2-H19 locus in congenital growth disorders and cancer.
dependent and vitamin B(12)-dependent enzyme, and PLoS ONE. 2008;3:e1849.

160
Chapter 8 The Human Epigenome: Implications for the Understanding of Human Disease

14. Feil R, Berger F. Convergent evolution of genomic imprinting in 39. Cerrato F, Sparago A, Verde G, et al. Different mechanisms
plants and mammals. Trends Genet. 2007;23:192–199. cause imprinting defects at the IGF2/H19 locus in Beckwith-
15. Ubeda F, Wilkins JF. Imprinted genes and human disease: An Wiedemann syndrome and Wilms’ tumour. Hum Mol Genet.
evolutionary perspective. Adv Exp Med Biol. 2008;626:101–115. 2008;17:1427–1435.
16. Kou YC, Shao L, Peng HH, et al. A recurrent intragenic genomic 40. Diaz-Meyer N, Yang Y, Sait SN, et al. Alternative mechanisms
duplication, other novel mutations in NLRP7 and imprinting associated with silencing of CDKN1C in Beckwith-Wiedemann
defects in recurrent biparental hydatidiform moles. Mol Hum syndrome. J Med Genet. 2005;42:648–655.
Reprod. 2008;14:33–40. 41. Enklaar T, Zabel BU, Prawitt D. Beckwith-Wiedemann syndrome:
17. Oosterhuis JW, Looijenga LH. Testicular germ-cell tumours in a Multiple molecular mechanisms. Expert Rev Mol Med. 2006;8:1–19.
broader perspective. Nat Rev Cancer. 2005;5:210–222. 42. Feinberg AP. Phenotypic plasticity and the epigenetics of human
18. Morison IM, Reeve AE. A catalogue of imprinted genes and par- disease. Nature. 2007;447:433–440.
ent-of-origin effects in humans and animals. Hum Mol Genet. 43. Baujat G, Rio M, Rossignol S, et al. Paradoxical NSD1 mutations
1998;7:1599–1609. in Beckwith-Wiedemann syndrome and 11p15 anomalies in
19. Lefebvre L, Viville S, Barton SC, et al. Abnormal maternal beha- Sotos syndrome. Am J Hum Genet. 2004;74:715–720.
viour and growth retardation associated with loss of the imprinted 44. Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes
gene Mest. Nat Genet. 1998;20:163–169. of some human cancers from their normal counterparts. Nature.
20. Falls JG, Pulford DJ, Wylie AA, et al. Genomic imprinting: Impli- 1983;301:89–92.
cations for human disease. Am J Pathol. 1999;154:635–647. 45. Esteller M. Cancer epigenomics: DNA methylomes and histone-
21. Chen C, Visootsak J, Dills S, et al. Prader-Willi syndrome: An modification maps. Nat Rev Genet. 2007;8:286–298.
update and review for the primary pediatrician. Clin Pediatr 46. Fraga MF, Ballestar E, Villar-Garea A, et al. Loss of acetylation at
(Phila). 2007;46:580–591. Lys16 and trimethylation at Lys20 of histone H4 is a common
22. Camprubi C, Coll MD, Villatoro S, et al. Imprinting center anal- hallmark of human cancer. Nat Genet. 2005;37:391–400.
ysis in Prader-Willi and Angelman syndrome patients with typical 47. Eden A, Gaudet F, Waghmare A, et al. Chromosomal instability
and atypical phenotypes. Eur J Med Genet. 2007;50:11–20. and tumors promoted by DNA hypomethylation. Science. 2003;
23. Kantor B, Shemer R, Razin A. The Prader-Willi/Angelman 300:455.
imprinted domain and its control center. Cytogenet Genome Res. 48. Karpf AR, Matsui S. Genetic disruption of cytosine DNA methyl-
2006;113:300–305. transferase enzymes induces chromosomal instability in human
24. Maina EN, Webb T, Soni S, et al. Analysis of candidate imprinted cancer cells. Cancer Res. 2005;65:8635–8639.
genes in PWS subjects with atypical genetics: A possible inactivat- 49. Schulz WA. L1 retrotransposons in human cancers. J Biomed Bio-
ing mutation in the SNURF/SNRPN minimal promoter. J Hum technol. 2006;2006:83672.
Genet. 2007;52:297–307. 50. Bjornsson HT, Brown LJ, Fallin MD, et al. Epigenetic specificity
25. Watrin F, Le Meur E, Roeckel N, et al. The Prader-Willi syn- of loss of imprinting of the IGF2 gene in Wilms tumors. J Natl
drome murine imprinting center is not involved in the spatio- Cancer Inst. 2007;99:1270–1273.
temporal transcriptional regulation of the Necdin gene. BMC 51. Li Y, Meng G, Guo QN. Changes in genomic imprinting and
Genet. 2005;6:1. gene expression associated with transformation in a model of
26. Yang T, Adamson TE, Resnick JL, et al. A mouse model for human osteosarcoma. Exp Mol Pathol. 2008;84:234–239.
Prader-Willi syndrome imprinting-centre mutations. Nat Genet. 52. Wu J, Qin Y, Li B, et al. Hypomethylated and hypermethylated
1998;19:25–31. profiles of H19DMR are associated with the aberrant imprinting
27. Kishino T, Lalande M, Wagstaff J. UBE3A/E6-AP mutations of IGF2 and H19 in human hepatocellular carcinoma. Genomics.
cause Angelman syndrome. Nat Genet. 1997;15:70–73. 2008; 91:443–450.
28. Lalande M, Calciano MA. Molecular epigenetics of Angelman 53. Byun HM, Wong HL, Birnstein EA, et al. Examination of IGF2
syndrome. Cell Mol Life Sci. 2007;64:947–960. and H19 loss of imprinting in bladder cancer. Cancer Res.
29. Fang P, Lev-Lehman E, Tsai TF, et al. The spectrum of mutations 2007;67:10753–10758.
in UBE3A causing Angelman syndrome. Hum Mol Genet. 54. Fan T, Schmidtmann A, Xi S, et al. DNA hypomethylation
1999;8:129–135. caused by Lsh deletion promotes erythroleukemia development.
30. Meguro M, Kashiwagi A, Mitsuya K, et al. A novel maternally Epigenetics. 2008;3:134–142.
expressed gene, ATP10C, encodes a putative aminophospholi- 55. Wu H, Chen Y, Liang J, et al. Hypomethylation-linked activation
pid translocase associated with Angelman syndrome. Nat Genet. of PAX2 mediates tamoxifen-stimulated endometrial carcino-
2001;28:19–20. genesis. Nature. 2005;438:981–987.
31. Herzing LB, Kim SJ, Cook EH Jr, et al. The human aminopho- 56. Brueckner B, Stresemann C, Kuner R, et al. The human let-7a-3
spholipid-transporting ATPase gene ATP10C maps adjacent to locus contains an epigenetically regulated microRNA gene with
UBE3A and exhibits similar imprinted expression. Am J Hum oncogenic function. Cancer Res. 2007;67:1419–1423.
Genet. 2001;68:1501–1505. 57. Sakai T, Toguchida J, Ohtani N, et al. Allele-specific hypermethy-
32. Weksberg R, Shuman C, Smith AC. Beckwith-Wiedemann syn- lation of the retinoblastoma tumor-suppressor gene. Am J Hum
drome. Am J Med Genet C Semin Med Genet. 2005;137C:12–23. Genet. 1991;48:880–888.
33. Scott RH, Stiller CA, Walker L, et al. Syndromes and constitu- 58. Herman JG, Baylin SB. Gene silencing in cancer in association
tional chromosomal abnormalities associated with Wilms with promoter hypermethylation. N Engl J Med. 2003;349:
tumour. J Med Genet. 2006;43:705–715. 2042–2054.
34. Cohen Jr MM. Beckwith-Wiedemann syndrome: Historical, clini- 59. Esteller M, Corn PG, Baylin SB, et al. A gene hypermethylation
copathological, and etiopathogenetic perspectives. Pediatr Dev profile of human cancer. Cancer Res. 2001;61:3225–3229.
Pathol. 2005;8:287–304. 60. Paz MF, Fraga MF, Avila S, et al. A systematic profile of DNA
35. Cooper WN, Luharia A, Evans GA, et al. Molecular subtypes and methylation in human cancer cell lines. Cancer Res. 2003;63:
phenotypic expression of Beckwith-Wiedemann syndrome. Eur J 1114–1121.
Hum Genet. 2005;13:1025–1032. 61. Esteller M, Fraga MF, Guo M, et al. DNA methylation patterns in
36. Joyce JA, Lam WK, Catchpoole DJ, et al. Imprinting of IGF2 and hereditary human cancers mimic sporadic tumorigenesis. Hum
H19: Lack of reciprocity in sporadic Beckwith-Wiedemann syn- Mol Genet. 2001;10:3001–3007.
drome. Hum Mol Genet. 1997;6:1543–1548. 62. Agrelo R, Cheng WH, Setien F, et al. Epigenetic inactivation of
37. Higashimoto K, Soejima H, Saito T, et al. Imprinting disruption the premature aging Werner syndrome gene in human cancer.
of the CDKN1C/KCNQ1OT1 domain: The molecular mechan- Proc Natl Acad Sci USA. 2006;103:8822–8827.
isms causing Beckwith-Wiedemann syndrome and cancer. Cyto- 63. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:
genet Genome Res. 2006;113:306–312. 683–692.
38. DeBaun MR, Niemitz EL, McNeil DE, et al. Epigenetic altera- 64. Majid S, Kikuno N, Nelles J, et al. Genistein induces the
tions of H19 and LIT1 distinguish patients with Beckwith-Wiede- p21WAF1/CIP1 and p16INK4a tumor suppressor genes in pros-
mann syndrome with cancer and birth defects. Am J Hum Genet. tate cancer cells by epigenetic mechanisms involving active chro-
2002;70:604–611. matin modification. Cancer Res. 2008;68:2736–2744.

161
Part II Concepts in Molecular Biology and Genetics

65. Fraga MF, Esteller M. Towards the human cancer epigenome: A 90. Nagarajan RP, Hogart AR, Gwye Y, et al. Reduced MeCP2
first draft of histone modifications. Cell Cycle. 2005;4:1377–1381. expression is frequent in autism frontal cortex and correlates
66. Seligson DB, Horvath S, Shi T, et al. Global histone modification with aberrant MECP2 promoter methylation. Epigenetics. 2006;
patterns predict risk of prostate cancer recurrence. Nature. 1:e1–e11.
2005;435:1262–1266. 91. Lund G, Andersson L, Lauria M, et al. DNA methylation poly-
67. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in morphisms precede any histological sign of atherosclerosis
gene regulation. Nat Rev Genet. 2004;5:522–531. in mice lacking apolipoprotein E. J Biol Chem. 2004;279:
68. Calin GA, Croce CM. MicroRNA signatures in human cancers. 29147–29154.
Nat Rev Cancer. 2006;6:857–866. 92. Abel T, Zukin RS. Epigenetic targets of HDAC inhibition in
69. Lujambio A, Esteller M. CpG island hypermethylation of tumor neurodegenerative and psychiatric disorders. Curr Opin Pharma-
suppressor microRNAs in human cancer. Cell Cycle. 2007;6: col. 2008;8:57–64.
1455–1459. 93. Hu N, Qiu X, Luo Y, et al. Abnormal histone modification
70. Saito Y, Liang G, Egger G, et al. Specific activation of microRNA- patterns in lupus CD4þ T cells. J Rheumatol. 2008;35:804–810.
127 with downregulation of the proto-oncogene BCL6 by 94. Gibbons R. Alpha thalassaemia-mental retardation, X linked.
chromatin-modifying drugs in human cancer cells. Cancer Cell. Orphanet J Rare Dis. 2006;1:15.
2006;9:435–443. 95. De La Fuente R, Viveiros MM, Wigglesworth K, et al. ATRX, a
71. Lujambio A, Ropero S, Ballestar E, et al. Genetic unmasking of member of the SNF2 family of helicase/ATPases, is required
an epigenetically silenced microRNA in human cancer cells. for chromosome alignment and meiotic spindle organization
Cancer Res. 2007;67:1424–1429. in metaphase II stage mouse oocytes. Dev Biol. 2004;272:1–14.
72. Feinberg AP, Tycko B. The history of cancer epigenetics. Nat Rev 96. Sanlaville D, Verloes A. CHARGE syndrome: An update. Eur J
Cancer. 2004;4:143–153. Hum Genet. 2007;15:389–399.
73. Lopez-Serra L, Ballestar E, Fraga MF, et al. A profile of methyl-CpG 97. Jongmans MC, Hoefsloot LH, van der Donk KP, et al. Familial
binding domain protein occupancy of hypermethylated promoter CHARGE syndrome and the CHD7 gene: A recurrent missense
CpG islands of tumor suppressor genes in human cancer. Cancer mutation, intrafamilial recurrence and variability. Am J Med
Res. 2006;66:8342–8346. Genet A. 2008;146A:43–50.
74. Yang XJ. The diverse superfamily of lysine acetyltransferases and 98. Spivak G. The many faces of Cockayne syndrome. Proc Natl Acad
their roles in leukemia and other diseases. Nucleic Acids Res. Sci USA. 2004;101:15273–15274.
2004;32:959–976. 99. Fousteri M, Vermeulen W, van Zeeland AA, et al. Cockayne syn-
75. Intini D, Fabris S, Storlazzi T, et al. Identification of a novel IGH- drome A and B proteins differentially regulate recruitment of
MMSET fusion transcript in a human myeloma cell line with the chromatin remodeling and repair factors to stalled RNA poly-
t(4;14)(p16.3;q32) chromosomal translocation. Br J Haematol. merase II in vivo. Mol Cell. 2006;23:471–482.
2004;126:437–439. 100. Boerkoel CF, O’Neill S, Andre JL, et al. Manifestations and
76. Jaju RJ, Fidler C, Haas OA, et al. A novel gene, NSD1, is fused to treatment of Schimke immuno-osseous dysplasia: 14 new cases
NUP98 in the t(5;11)(q35;p15.5) in de novo childhood acute and a review of the literature. Eur J Pediatr. 2000;159:1–7.
myeloid leukemia. Blood. 2001;98:1264–1267. 101. Boerkoel CF, Takashima H, John J, et al. Mutant chromatin
77. Rosati R, La Starza R, Veronese A, et al. NUP98 is fused to the remodeling protein SMARCAL1 causes Schimke immuno-osse-
NSD3 gene in acute myeloid leukemia associated with t(8;11) ous dysplasia. Nat Genet. 2002;30:215–220.
(p11.2;p15). Blood. 2002;99:3857–3860. 102. Brown DC, Grace E, Sumner AT, et al. ICF syndrome (immuno-
78. Bracken AP, Pasini D, Capra M, et al. EZH2 is downstream of the deficiency, centromeric instability and facial anomalies): Inves-
pRB-E2F pathway, essential for proliferation and amplified in tigation of heterochromatin abnormalities and review of
cancer. Embo J. 2003;22:5323–5335. clinical outcome. Hum Genet. 1995;96: 411–416.
79. Huntsman DG, Chin SF, Muleris M, et al. MLL2, the second 103. Ehrlich M. The ICF syndrome, a DNA methyltransferase 3B
human homolog of the Drosophila trithorax gene, maps to deficiency and immunodeficiency disease. Clin Immunol.
19q13.1 and is amplified in solid tumor cell lines. Oncogene. 2003;109:17–28.
1999;18:7975–7984. 104. Xu GL, Bestor TH, Bourc’his D, et al. Chromosome instability
80. Angrand PO, Apiou F, Stewart AF, et al. NSD3, a new SET and immunodeficiency syndrome caused by mutations in a
domain-containing gene, maps to 8p12 and is amplified in DNA methyltransferase gene. Nature. 1999;402:187–191.
human breast cancer cell lines. Genomics. 2001;74:79–88. 105. Moog U, Smeets EE, van Roozendaal KE, et al. Neurodevelop-
81. Cloos PA, Christensen J, Agger K, et al. The putative oncogene mental disorders in males related to the gene causing Rett syn-
GASC1 demethylates tri- and dimethylated lysine 9 on histone drome in females (MECP2). Eur J Paediatr Neurol. 2003;7:5–12.
H3. Nature. 2006;442:307–311. 106. Van den Veyver IB, Zoghbi HY. Methyl-CpG-binding protein
82. Ropero S, Fraga MF, Ballestar E, et al. A truncating mutation of 2 mutations in Rett syndrome. Curr Opin Genet Dev. 2000;
HDAC2 in human cancers confers resistance to histone deacety- 10:275–279.
lase inhibition. Nat Genet. 2006;38:566–569. 107. Laccone F, Junemann I, Whatley S, et al. Large deletions of the
83. Moinova HR, Chen WD, Shen L, et al. HLTF gene silencing MECP2 gene detected by gene dosage analysis in patients with
in human colon cancer. Proc Natl Acad Sci USA. 2002;99: Rett syndrome. Hum Mutat. 2004;23:234–244.
4562–4567. 108. Ballestar E, Ropero S, Alaminos M, et al. The impact of MECP2
84. Gunduz E, Gunduz M, Ouchida M, et al. Genetic and epigenetic mutations in the expression patterns of Rett syndrome patients.
alterations of BRG1 promote oral cancer development. Int J Hum Genet. 2005;116:91–104.
Oncol. 2005;26:201–210. 109. Li MR, Pan H, Bao XH, et al. MECP2 and CDKL5 gene muta-
85. Ballestar E, Esteller M, Richardson BC. The epigenetic face tion analysis in Chinese patients with Rett syndrome. J Hum
of systemic lupus erythematosus. J Immunol. 2006;176:7143–7147. Genet. 2007;52:38–47.
86. Corvetta A, Della Bitta R, Luchetti MM, et al. 5-Methylcytosine 110. Jones PL, Veenstra GJ, Wade PA, et al. Methylated DNA and
content of DNA in blood, synovial mononuclear cells and syno- MeCP2 recruit histone deacetylase to repress transcription.
vial tissue from patients affected by autoimmune rheumatic dis- Nat Genet. 1998;19:187–191.
eases. J Chromatogr. 1991;566:481–491. 111. Esteller M. Rett syndrome: The first forty years: 1966–2006.
87. Kominato Y, Hata Y, Takizawa H, et al. Expression of human Epigenetics. 2007;2:1.
histo-blood group ABO genes is dependent upon DNA methyla- 112. Abuhatzira L, Makedonski K, Kaufman Y, et al. MeCP2 defi-
tion of the promoter region. J Biol Chem. 1999;274:37240–37250. ciency in the brain decreases BDNF levels by REST/CoREST-
88. Menendez L, Walker LD, Matyunina LV, et al. Epigenetic changes mediated repression and increases TRKB production. Epige-
within the promoter region of the HLA-G gene in ovarian tumors. netics. 2007;2:214–222.
Mol Cancer. 2008;7:43. 113. Kesler SR, Simensen RJ, Voeller K, et al. Altered neurodeve-
89. Honda T, Tamura G, Waki T, et al. Demethylation of MAGE pro- lopment associated with mutations of RSK2: A morphometric
moters during gastric cancer progression. Br J Cancer. MRI study of Coffin-Lowry syndrome. Neurogenetics. 2007;
2004;90:838–843. 8:143–147.

162
Chapter 8 The Human Epigenome: Implications for the Understanding of Human Disease

114. Field M, Tarpey P, Boyle J, et al. Mutations in the RSK2 A1298C in Brazilian population and its correlation with plasma
(RPS6KA3) gene cause Coffin-Lowry syndrome and nonsyn- homocysteine levels in spina bifida. Am J Med Genet A.
dromic X-linked mental retardation. Clin Genet. 2006;70: 2003;119A:20–25.
509–515. 128. Giovannucci E. Alcohol, one-carbon metabolism, and colorec-
115. Kang S, Dong S, Guo A, et al. Epidermal growth factor stimu- tal cancer: Recent insights from molecular studies. J Nutr.
lates RSK2 activation through activation of the MEK/ERK path- 2004;134:2475S–2481S.
way and src-dependent tyrosine phosphorylation of RSK2 at 129. Labrune P, Zittoun J, Duvaltier I, et al. Haemolytic uraemic syn-
Tyr-529. J Biol Chem. 2008;283:4652–4657. drome and pulmonary hypertension in a patient with methio-
116. Merienne K, Jacquot S, Trivier E, et al. Rapid immunoblot and nine synthase deficiency. Eur J Pediatr. 1999;158:734–739.
kinase assay tests for a syndromal form of X linked mental 130. Benbrahim-Tallaa L, Waterland RA, Styblo M, et al. Molecular
retardation: Coffin-Lowry syndrome. J Med Genet. 1998;35: events associated with arsenic-induced malignant transforma-
890–894. tion of human prostatic epithelial cells: Aberrant genomic
117. Roelfsema JH, Peters DJ. Rubinstein-Taybi syndrome: Clinical DNA methylation and K-ras oncogene activation. Toxicol Appl
and molecular overview. Expert Rev Mol Med. 2007;9:1–16. Pharmacol. 2005;206: 288–298.
118. Bartsch O, Schmidt S, Richter M, et al. DNA sequencing of 131. Poirier LA, Vlasova TI. The prospective role of abnormal
CREBBP demonstrates mutations in 56% of patients with methyl metabolism in cadmium toxicity. Environ Health Perspect.
Rubinstein-Taybi syndrome (RSTS) and in another patient with 2002;110(Suppl 5):793–795.
incomplete RSTS. Hum Genet. 2005;117:485–493. 132. Silbergeld EK, Waalkes M, Rice JM. Lead as a carcinogen:
119. Zimmermann N, Acosta AM, Kohlhase J, et al. Confirmation of Experimental evidence and mechanisms of action. Am J Ind
EP300 gene mutations as a rare cause of Rubinstein-Taybi syn- Med. 2000;38:316–323.
drome. Eur J Hum Genet. 2007;15:837–842. 133. Salnikow K, Costa M. Epigenetic mechanisms of nickel carcino-
120. Roelfsema JH, White SJ, Ariyurek Y, et al. Genetic heterogeneity genesis. J Environ Pathol Toxicol Oncol. 2000;19:307–318.
in Rubinstein-Taybi syndrome: Mutations in both the CBP and 134. Wei YD, Tepperman K, Huang MY, et al. Chromium inhibits
EP300 genes cause disease. Am J Hum Genet. 2005;76:572–580. transcription from polycyclic aromatic hydrocarbon-inducible
121. Tatton-Brown K, Rahman N. Sotos syndrome. Eur J Hum Genet. promoters by blocking the release of histone deacetylase and
2007;15:264–271. preventing the binding of p300 to chromatin. J Biol Chem.
122. Rahman N. Mechanisms predisposing to childhood overgrowth 2004;279:4110–4119.
and cancer. Curr Opin Genet Dev. 2005;15:227–233. 135. Anway MD, Cupp AS, Uzumcu M, et al. Epigenetic transgenera-
123. Martinez-Glez V, Lapunzina P. Sotos syndrome is associated with tional actions of endocrine disruptors and male fertility. Science.
leukemia/lymphoma. Am J Med Genet A. 2007;143A:1244–1245. 2005;308:1466–1469.
124. Jaenisch R, Bird A. Epigenetic regulation of gene expression: 136. Weaver IC, Cervoni N, Champagne FA, et al. Epigenetic pro-
How the genome integrates intrinsic and environmental sig- gramming by maternal behavior. Nat Neurosci. 2004;7:847–854.
nals. Nat Genet. 2003;33(Suppl):245–254. 137. Niemitz EL, Feinberg AP. Epigenetics and assisted reproductive
125. Waterland RA, Lin JR, Smith CA, et al. Post-weaning diet affects technology: A call for investigation. Am J Hum Genet. 2004;74:
genomic imprinting at the insulin-like growth factor 2 (IGF2) 599–609.
locus. Hum Mol Genet. 2006;15:705–716. 138. Rideout WM, 3rd, Eggan K, Jaenisch R. Nuclear cloning and
126. Kirke PN, Mills JL, Molloy AM, et al. Impact of the MTHFR epigenetic reprogramming of the genome. Science. 2001;293:
C677T polymorphism on risk of neural tube defects: Case- 1093–1098.
control study. BMJ. 2004;328:1535–1536. 139. Fraga MF, Ballestar E, Paz MF, et al. Epigenetic differences arise
127. Perez AB, D’Almeida V, Vergani N, et al. Methylenetetrahydro- during the lifetime of monozygotic twins. Proc Natl Acad Sci
folate reductase (MTHFR): Incidence of mutations C677T and USA. 2005;102:10604–10609.

163

You might also like