SlideShare ist ein Scribd-Unternehmen logo
1 von 14
Downloaden Sie, um offline zu lesen
REVIEWS

                                   DNA METHYLATION AND
                                   HUMAN DISEASE
                                   Keith D. Robertson
                                   Abstract | DNA methylation is a crucial epigenetic modification of the genome that is involved
                                   in regulating many cellular processes. These include embryonic development, transcription,
                                   chromatin structure, X chromosome inactivation, genomic imprinting and chromosome
                                   stability. Consistent with these important roles, a growing number of human diseases have
                                   been found to be associated with aberrant DNA methylation. The study of these diseases has
                                   provided new and fundamental insights into the roles that DNA methylation and other
                                   epigenetic modifications have in development and normal cellular homeostasis.

CPG ISLAND                        With the completion of the Human Genome Project,             transposable elements (LINES), short interspersed
A genomic region of ~1 kb that    we have a nearly complete list of the genes needed to        transposable elements (SINES) and endogenous retro-
has a high G–C content, is rich   produce a human. However, the situation is far more          viruses)6. CPG ISLANDS, particularly those associated with
in CpG dinucleotides and is
                                  complex than a simple catalogue of genes. Of equal           promoters, are generally unmethylated, although an
usually hypomethylated.
                                  importance is a second system that cells use to determine    increasing number of exceptions are being identified7,8.
                                  when and where a particular gene will be expressed dur-      DNA methylation represses transcription directly, by
                                  ing development. This system is overlaid on DNA in the       inhibiting the binding of specific transcription factors,
                                  form of epigenetic marks that are heritable during cell      and indirectly, by recruiting methyl-CpG-binding pro-
                                  division but do not alter the DNA sequence.                  teins and their associated repressive chromatin remod-
                                      The only known epigenetic modification of DNA in         elling activities (see online supplementary information
                                  mammals is methylation of cytosine at position C5 in         S1 (table)). Little is known about how DNA methyla-
                                  CpG dinucleotides1. By contrast, the other main group        tion is targeted to specific regions; however, this prob-
                                  of epigenetic modifications — the post-translational         ably involves interactions between DNMTs and one or
                                  modification of histones — shows a high level of diver-      more chromatin-associated proteins3.
                                  sity and complexity2. The mammalian DNA methyla-                 Properly established and maintained DNA methyla-
                                  tion machinery is composed of two components, the            tion patterns are essential for mammalian development
                                  DNA methyltransferases (DNMTs), which establish and          and for the normal functioning of the adult organism.
                                  maintain DNA methylation patterns, and the methyl-           DNA methylation is a potent mechanism for silencing
                                  CpG binding proteins (MBDs), which are involved in           gene expression and maintaining genome stability in
                                  ‘reading’ methylation marks. An in-depth discussion          the face of a vast quantity of repetitive DNA, which can
                                  of these proteins is not provided here, but their main       otherwise mediate illegitimate recombination events
                                  features are given in online supplementary informa-          and cause transcriptional deregulation of nearby genes.
                                  tion S1 (table) (and were recently reviewed by REFS 3,4).    Embryonic stem cells that are deficient for DNMTs are
Department of Biochemistry
and Molecular Biology,            There is also clear evidence that a DNA demethylase          viable, but die when they are induced to differentiate9.
Shands Cancer Center,             contributes to regulating DNA methylation patterns           Mouse knockout studies have shown that Dnmt1 and
University of Florida,            during embryonic development, although the activity          Dnmt3b are essential for embryonic development
Box 100245, 1600 S.W.             responsible for this has not been identified5.               and that mice that lack Dnmt3a die within a few
Archer Road, Gainesville,
Florida 32610, USA.                   In normal cells, DNA methylation occurs predomi-         weeks of birth10,11. In addition, loss of normal DNA
e-mail: keithr@ufl.edu            nantly in repetitive genomic regions, including satellite    methylation patterns in somatic cells results in loss of
doi:10.1038/nrg1655               DNA and parasitic elements (such as long interspersed        growth control.


NATURE REVIEWS | GENETICS                                                                                              VOLUME 6 | AUGUST 2005 | 597
                                                              © 2005 Nature Publishing Group
REVIEWS


Centromere                                                                                               also contribute to aberrant gene regulation in cancer
                                                                                                         by TRANSCRIPTIONAL INTERFERENCE or the generation of
                                                                                     TSG
                                                                                                         antisense transcripts17. Loss of genomic methylation
                                   Hypermethylated                     CpG island
                                   pericentromeric                     (hypomethylated)
                                                                                                         is a frequent and early event in cancer, and correlates
                                   heterochromatin                                                       with disease severity and metastatic potential in many
                                                                                                         tumour types18.
                                             Hypomethylation                     Hypermethylation
                                                                                                             Gene-specific effects of hypomethylation also occur.
                                                                                                         For example, the melanoma antigen (MAGE) family of
                                   Mitotic recombination,            Transcriptional repression,         cancer–testis genes, which encode tumour antigens of
                                   genomic instability               loss of TSG expression
     DNA repeat                                                                                          unknown function, are frequently demethylated and
                                                                                                         re-expressed in cancer19. Demethylation accompanied
     Methylated                                                                                          by increased expression has been reported for the S100
     Unmethylated                                           Cancer                                       calcium binding protein A4 (S100A4) gene in colon
                                                                                                         cancer20, the serine protease inhibitor gene SERPINB5
Figure 1 | DNA methylation and cancer. The diagram shows a representative region of                      (also known as maspin) in gastric cancer21, and the puta-
genomic DNA in a normal cell. The region shown contains repeat-rich, hypermethylated                     tive oncogene γ-synuclein (SNCG) in breast and ovarian
pericentromeric heterochromatin and an actively transcribed tumour suppressor gene (TSG)                 cancers22. Global demethylation early in tumorigenesis
associated with a hypomethylated CpG island (indicated in red). In tumour cells, repeat-rich             might predispose cells to genomic instability and further
heterochromatin becomes hypomethylated and this contributes to genomic instability, a                    genetic changes, whereas gene-specific demethylation
hallmark of tumour cells, through increased mitotic recombination events. De novo methylation
                                                                                                         could be a later event that allows tumour cells to adapt to
of CpG islands also occurs in cancer cells, and can result in the transcriptional silencing of
growth-regulatory genes. These changes in methylation are early events in tumorigenesis.
                                                                                                         their local environment and promotes metastasis.
                                                                                                             Research on genome-wide demethylation in can-
                                                                                                         cer cells has been largely overshadowed by studies of
                                            The importance of DNA methylation is empha-                  gene-specific hypermethylation events, which occur
                                        sized by the growing number of human diseases that               concomitantly with the hypomethylation events dis-
                                        are known to occur when this epigenetic information              cussed above. Aberrant hypermethylation in cancer
                                        is not properly established and/or maintained, and               usually occurs at CpG islands (FIG. 1), most of which
                                        there is increasing interest in developing ways of phar-         are unmethylated in normal somatic cells8,23, and the
                                        macologically reversing epigenetic abnormalities12.              resulting changes in chromatin structure (such as
                                        Further interest in this area comes from important               histone hypoacetylation) effectively silence transcrip-
                                        new evidence that concerns the regulation of DNA                 tion. Indeed, a distinct subset of many tumour types
                                        methylation, indicating interactions between DNA                 has a CpG-island-methylator phenotype, which has
                                        methylation and the histone modification machin-                 been defined as a 3–5 fold increase in the frequency
                                        ery, and showing a potential role of small RNAs13.               of aberrant hypermethylation events24. Genes involved
                                        Here, I discuss a diverse group of diseases associated           in cell-cycle regulation, tumour cell invasion, DNA
                                        with abnormalities of DNA methylation, studies of                repair, chromatin remodelling, cell signalling, tran-
                                        which have provided insights into how DNA meth-                  scription and apoptosis are known to become aber-
                                        ylation patterns are regulated and the pathological              rantly hypermethylated and silenced in nearly every
                                        consequences of their disruption. I also propose two             tumour type (see online supplementary information
                                        related models for how a single common defect might              S2 and S3 (tables)). This provides tumour cells with
                                        be able to give rise to the diverse array of defective           a growth advantage, increases their genetic instability
                                        DNA methylation patterns in human diseases. It                   (allowing them to acquire further advantageous genetic
                                        should be noted that I focus only on those diseases for          changes), and allows them to metastasize. In tumours
                                        which defective DNA methylation patterns have been               with a well-defined progression, such as colon cancer,
                                        demonstrated. For this reason, I do not discuss Rett             aberrant hypermethylation is detectable in the earliest
TRANSCRIPTIONAL
INTERFERENCE                            syndrome, which involves a defect in the machinery               precursor lesions, indicating that it directly contributes
Repression of one                       that reads methylation marks14.                                  to transformation and is not a late event that arises
transcriptional unit by another                                                                          from genetic alterations25.
such unit that is linked in cis.        DNA methylation and cancer                                           Use of RESTRICTION LANDMARK GENOMIC SCANNING to ana-
RESTRICTION LANDMARK
                                        A link between DNA methylation and cancer was first              lyze the methylation status of 1,184 CpG islands from
GENOMIC SCANNING                        demonstrated in 1983, when it was shown that the                 98 tumour samples showed that de novo methylation of
(RLGS). A genome-wide                   genomes of cancer cells are hypomethylated relative              CpG islands is widespread in tumour cells. In this study,
method for analyzing the DNA            to their normal counterparts15. Hypomethylation in               the extent of methylation varied between individual
methylation status of CpG
                                        tumour cells is primarily due to the loss of methylation         tumours and tumour types, and an average of 608 CpG
islands. Radiolabelled
fragments obtained by digestion         from repetitive regions of the genome6 (FIG. 1), and the         islands were aberrantly hypermethylated26. Because
with NotI (a methylation-               resulting genomic instability is a hallmark of tumour            the hypermethylation of CpG islands is relatively
sensitive restriction enzyme)           cells. Rearrangements that involve the large block of            rare in normal cells, is an early event in transforma-
are separated by two-                   pericentromeric heterochromatin on chromosome 1,                 tion, and robust assays can detect methylated DNA in
dimensional gel electrophoresis,
allowing differentiation
                                        for example, are among the most frequent genomic                 bodily fluids, it represents a good potential biomarker
between methylated and                  instabilities in many tumour types16. Reactivation of            for early cancer detection27. The underlying cause of
unmethylated regions.                   transposon promoters following demethylation might               methylation defects in cancer remains unknown, but


598 | AUGUST 2005        | VOLUME 6                                                                                              www.nature.com/reviews/genetics
                                                                        © 2005 Nature Publishing Group
REVIEWS


                                                                                              imprinted loci, regulates the ability of distant ENHANC
 Box 1 | A comparison of the CTCF and BORIS proteins                                                                  30
                                                                                              ERS to access promoters . In several ICRs, in vitro and
 CTCF (CCCTC-binding factor)                                                                  in vivo studies have shown that CTCF binds only to the
 • Contains an 11 zinc finger (ZF) DNA-binding region30.                                      unmethylated parental allele31,32, providing an elegant
 • Binds diverse DNA sequences, including most ICRs and many CpG islands30,137.               means for ICR-regulated genes to be expressed in
 • Binding is methylation sensitive (if binding sites contain CpGs)31.                        an allele-specific manner. CTCF binding might also
                                                                                              be essential to protect DMRs from de novo methyla-
 • Has insulator function (regulates access of enhancers to promoters)30,70.
                                                                                              tion33.
 • Has boundary element function (blocks the spread of heterochromatin)30,70.                     Loss of imprinting (LOI) is the disruption of
 • Protects regions from DNA methylation138,139.                                              imprinted epigenetic marks through gain or loss
 • Is ubiquitously expressed, except in spermatocytes126.                                     of DNA methylation, or simply the loss of normal
 • Overexpression inhibits cell proliferation140.                                             allele-specific gene expression28. Studies of LOI have
                                                                                              provided important insights into the role of methyla-
 • Gene is located at 16q22, a region of frequent loss of heterozygosity in cancer, and is
                                                                                              tion in imprinting, and have highlighted the impor-
   often mutated in cancer141.
                                                                                              tance of CTCF as a regulator of both imprinted gene
 BORIS                                                                                        expression and DNA methylation patterns. Below, I
 • Contains an 11 ZF DNA-binding region that has high similarity to the CTCF ZF               discuss several human diseases for which the role of
   region30.                                                                                  aberrant genomic imprinting has been particularly
 • Presumed to show significant overlap with CTCF binding sites.                              well established. Features of these diseases are sum-
 • Methylation sensitivity of binding unknown.                                                marized in the online supplementary information
                                                                                              S3 (table). Importantly, imprinted DNA methylation
 • Insulator function unknown.
                                                                                              patterns are frequently disrupted by in vitro manipula-
 • Boundary element function unknown                                                          tion of embryos in both animals and humans34–36, and
 • Methylation protection function unknown                                                    the use of assisted reproductive technologies has been
 • Expression is normally restricted to testis (BORIS is a member of the cancer–testis        associated with an increased risk for several imprinting
   gene family), and is reactivated in cancers126.                                            diseases37 BOX 2.
 • Overexpression promotes cell proliferation30.
                                                                                              LOI in cancer. LOI of a growth-promoting imprinted
 • Gene is located at 20q13, a region that is frequently amplified in cancer.
                                                                                              gene, leading to activation of the normally silent allele,
                                                                                              results in abnormally high expression of the gene prod-
                                 possible mechanisms are discussed at the end of this         uct and gives cells a growth advantage. The imprinted
                                 review. DNA methylation and cancer are also related          IGF2/H19 locus (FIG. 2a) encodes both IGF2, an autocrine
                                 through the loss of imprinted methylation patterns in        growth factor with an important role in many types of
                                 many tumours, as discussed below.                            cancer, and H19, a non-coding RNA of unknown func-
                                                                                              tion with growth suppressive properties38. IGF2 and H19
                                 DNA methylation and imprinting disorders                     are normally expressed from the paternal and maternal
                                 Genomic imprinting — a brief overview. Genomic               alleles, respectively, and relaxed silencing of the mater-
                                 imprinting is defined as an epigenetic modification          nal IGF2 allele results in increased IGF2 expression and
                                 of a specific parental chromosome in the gamete or           reduced H19 expression. LOI of IGF2 is the most com-
                                 zygote that leads to differential expression of the two      mon LOI event across the widest range of tumour types,
                                 alleles of a gene in the somatic cells of the offspring28.   including colon, liver, lung, and ovarian cancer, as well
                                 Differential expression can occur in all cells, or in        as Wilms’ tumour — the embryonic kidney cancer in
                                 specific tissues or developmental stages. About 80           which LOI was first discovered39,40.
                                 genes are known to be imprinted, and this number is              Additional evidence for the role of LOI at IGF2/H19
                                 continually increasing.                                      in cancer came from a recent study that used a mouse
                                     Differential allele-specific DNA methylation is one      model. Animals with LOI showed a twofold increase
                                 of the hallmarks of imprinted regions and is usually         in intestinal tumours, and normal intestinal epithe-
                                 localized to regions termed differentially methylated        lium was shown to be less well differentiated than in
                                 regions (DMRs). DMRs include imprinting con-                 wild-type mice41. LOI defects in IGF2/H19 vary with
                                 trol regions (ICRs), which control gene expression           tumour type. For example, in Wilms’ tumour42 and
                                 within imprinted domains, often over large distances.        colorectal cancer43, the regulatory region ICR1 on the
                                 Whereas DMRs can be reprogrammed during devel-               maternal allele becomes methylated de novo, which
                                 opment, differential methylation in ICRs is generally        inhibits CTCF binding and thereby allows enhancers
                                 established in germ cells and maintained throughout          upstream of H19 to access the IGF2 promoter on both
ENHANCER                         development29.                                               alleles (FIG. 2a). An alternative mechanism has also been
A regulatory DNA element that        CCCTC-binding factor (CTCF), an 11-zinc-finger           proposed, in which DMRs upstream of IGF2 become
usually binds several            protein that binds to highly divergent sequences using       demethylated44.
transcription factors and can    different combinations of zinc fingers, is an important          LOI of growth-inhibitory imprinted genes, through
activate transcription from a
promoter at great distance and
                                 regulator of imprinted gene expression BOX 1). CTCF         silencing of the one normally active allele, might also
in an orientation-independent    is a chromatin insulator that separates the genome           result in deregulated cell growth. An example of this
manner.                          into independent functional domains or, in the case of       involves the maternally expressed cyclin-dependent


NATURE REVIEWS | GENETICS                                                                                             VOLUME 6 | AUGUST 2005 | 599
                                                             © 2005 Nature Publishing Group
REVIEWS


                                 kinase inhibitor 1C (CDKN1C) (also known as p57KIP2)              several imprinted genes (FIG. 2a). This large region
                                 gene, which encodes a cyclin-dependent kinase inhibi-             consists of two independently regulated imprinted
                                 tor that mediates G1/S-phase arrest. In Wilms’ tumour,            domains39,40,48. The more telomeric domain contains
                                 LOI of CDKN1C occurs in ~10% of tumours (LOI of                   IGF2 and H19, followed by an intervening region
                                 IGF2/H19 occurs in 70% of these tumours)28,45. Other              with several genes that do not seem to be imprinted.
                                 imprinted genes that undergo LOI include the RAS-                 The second, more centromeric, domain contains the
                                 related gene DIRAS3 (also known as ARHI) in breast and            maternally expressed gene KCNQ1 (potassium volt-
                                 ovarian cancer46 and the mesoderm specific transcript             age-gated channel, KQT-like subfamily, member 1; also
                                 homologue (MEST) gene in breast, lung, and colon can-             known as KvLQT1), the paternally expressed KCNQ1
                                 cer47. Although the number of genes that show LOI in              antisense transcript KCNQ1OT1 (KCNQ1 overlap-
                                 cancer seems small from this discussion, the frequency            ping transcript 1; also known as LIT1), and several
                                 of such alterations is probably similar in extent to the          other maternally expressed genes, including CDKN1C.
                                 DNA methylation abnormalities of non-imprinted genes              ICR1, which is located upstream of H19, controls
                                 discussed in the previous section. Examples of LOI seem           differential expression of the IGF2/H19 domain,
                                 limited only because relatively few of the estimated total        and there are also several DMRs upstream of IGF2;
                                 number of imprinted genes have been identified and                ICR2 lies at the 5′-end of KCNQ1OT1 REFS 43,48,49.
                                 even fewer have been well characterized.                          In normal cells, the paternal and maternal alleles of
                                                                                                   ICR1 and ICR2, respectively, are methylated, and both
                                 Beckwith–Wiedemann syndrome (BWS). BWS is                         contain binding sites for CTCF31,50.
                                 predominantly a maternally transmitted disorder and                  Several perturbations of this complex genomic
                                 involves fetal and postnatal overgrowth and a predispo-           region are associated with BWS. These include paternal
                                 sition to embryonic tumours, such as Wilms’ tumour                UNIPARENTAL DISOMY (UPD) (resulting in increased IGF2
                                 (see online supplementary information S3 (table)).                and reduced CDKN1C expression), LOI of KCNQ1OT1
                                 The BWS locus, at 11p15.5, spans ~1 Mb and includes               in ~50% of cases that do not involve UPD, and LOI at


                                    Box 2 | DNA-methylation abnormalities and assisted reproductive technology
                                    A link between assisted reproductive technology and birth defects
                                    It has been estimated that one in ten individuals of reproductive age are infertile. The use of assisted reproductive
                                    technologies (ARTs), such as intracytoplasmic sperm injection and in vitro fertilization, has therefore been
                                    increasing, and children conceived by ART now account for 1–3% of all births in some Western countries. Although
                                    the vast majority of these children develop normally, recent studies have raised concerns that these procedures
                                    might lead to epigenetic defects. ART is associated with an increase in multiple births and low birth weight, and this
                                    risk has been known for some time142. Although the numbers are small and larger studies are needed, ARTs have
                                    been linked to a 3–6-fold increase in the occurrence of Beckwith–Wiedemann syndrome (BWS) and Angelman
                                    syndrome (AS)37,143,144 (see online supplementary information S3 (table)).
                                    An epigenetic basis
                                    Germ cell and embryonic development are two periods that are characterized by profound changes in DNA
                                    methylation patterns, also known as epigenetic reprogramming. Primordial germ cells are globally demethylated as
                                    they mature (including at imprinted regions), and then become methylated de novo during gametogenesis, which is
                                    also the time at which most DNA methylation imprints are established. The second period of pronounced change
                                    occurs after fertilization, where the paternal genome is rapidly and actively demethylated, followed by passive
                                    replication-dependent demethylation of the maternal genome (here, imprinting marks seem to resist
                                    demethylation). A wave of de novo methylation once again establishes the somatic-cell pattern of DNA methylation
                                    following implantation145,146.
                                      The increased incidence of BWS and AS associated with ART all arise from epigenetic, rather than genetic, defects.
                                    In most BWS cases studied, loss of imprinting (LOI) at KCNQ1OT1 was observed, and in AS cases the defect was
                                    found to be LOI at SNRPN (loss of maternal methylation, see FIG. 2a,b)37,143. Epigenetic mechanisms and imprinted
                                    genes also seem to be involved in the regulation of birth weight in mouse models; therefore, the disruption of
                                    imprints at currently unknown loci following ART in humans might also contribute to the increased incidence of this
                                    problem147. Low birth weight is a serious issue in its own right as it contributes to long-term adverse health effects
                                    such as cardiovascular disease148.
                                    Causes of epigenetic defects
                                    There has been a trend in fertility clinics towards extended culture time of embryos and transfer of blastocysts, rather
                                    than earlier cleavage-stage embryos. This allows selection of the most robust embryos, proper temporal
                                    synchronization of the embryo and the uterus, and the possibility of maintaining a high pregnancy rate while reducing
                                    the risk of multiple pregnancy (because fewer embryos need to be transferred)35. However, several studies have
                                    demonstrated that in vitro culture conditions have notable effects on imprinting marks, gene expression and
                                    developmental potential in mouse models34,149. Whether the culture conditions used for human embryos provide all of
UNIPARENTAL DISOMY                  the necessary factors for proper epigenetic control is unknown. It is also possible that the epigenetic defects might be
Inheritance of a chromosome or      the cause of the infertility itself and are present in the gametes of the mother and/or father. Therefore, rather than ARTs
chromosome region from a            causing epigenetic defects directly, they might simply be unmasking an existing defect.
single parent.



600 | AUGUST 2005      | VOLUME 6                                                                                           www.nature.com/reviews/genetics
                                                                © 2005 Nature Publishing Group
REVIEWS


a BWS 11p15.5                Cancer LOI         BWS breakpoint region
                             (CDKN1C)                                                                                                ICR1
                                                ICR2

                                                                                              Cancer LOI (IGF2)
Centromere                                                                                                                                                  Telomere
                                     CTCF                                                                                   CTCF
                                                                                                                                                Enhancer


    NAP1L4               CDKN1C CTCF                       KCNQ1                      ASCL2               INS               CTCF            H19
         TSSC3       TSSC5                                                         PHEMX    TH                     IGF2                                   L23
                                                                               CD81
                                              KNCQ1OTI
                                                                           TSSC4                            DMRs 0,1,2
                                                ICR2                                                                                 ICR1

b PWS/AS 15q11–q13                PWS region                                     AS region



Centromere                              PWS ICR                         UBE3A-                                                                              Telomere
                                                1            2      3     AS



                       NDN                      SNURF/SNRPN                UBE3A                                   GABRG3           OCA2            HERC2
     MKRN3                                                                      ATP10C         GABRB3           GABRA5
        MAGE-L2
                                     AS ICR

c AHO/PHP-Ia/PHP-Ib 20q13
                                                                                 LOI in PHP-Ib
Centromere                                                                       patients                                                                   Telomere
                                                                                                                            GNAS
                               NESP55-AS                                                                                    (Gsα)


                      NESP55                                XLα S                      Exon 1A                            Exon 1            Exons 2–13

                                                                                             Paternal expression      Unmethylated           Promoter (not imprinted)
d TNDM 6q24
                                                     CpG island                              Maternal expression      Methylated             ICR or DMR
Telomere
                                                                                                                ZAC-AS                                    Centromere



                      FK506                    ICR         HYMAI                       PLAGL1                                               KIAA0680

                              Figure 2 | Regions involved in disease-associated genomic imprinting defects. Sense and antisense transcripts are
                              indicated by arrows above and below the genes, respectively. In each panel, the methylation status of the paternal allele is indicated
                              on the top of the chromosome, and that of the maternal allele on the bottom of the chromosome. a | The Beckwith–Wiedemann
                              syndrome (BWS) locus and cancer-associated loss of imprinting (LOI). The region can be divided into two imprinted domains
                              (TSSC3 to KCNQ1, and INS to H19) that are regulated independently. CTCF binds to the unmethylated alleles of both imprinting
                              control regions, ICR1 and ICR2. Cancer-associated LOI commonly occurs within the IGF2/H19 region (resulting in increased IGF2
                              and decreased H19 expression) and less commonly at CDKN1C. BWS arises from several defects in this region, including LOI at
                              ICR1, ICR2 and the differentially methylated regions (DMRs 0, 1 and 2) upstream of IGF2; paternal UPD; and mutations and
                              translocations on the maternal allele. b | The Prader–Willi syndrome (PWS)/Angelman syndrome (AS) locus. The ICR for this locus
                              has a bipartite structure, with the more centromeric component acting as the AS ICR and the more telomeric region, which
                              contains the promoter of the SNURF/SNRPN gene, acting as the PWS ICR. SNURF/SNRPN produces an extremely long and
                              complicated transcript that encodes not only the SNURF/SNRPN (1), and IPW transcripts (2), but also several small nucleolar RNAs
                              (snoRNAs) (3), which actually flank IPW (not shown), that are processed from its introns. This transcript is also thought to inhibit
                              expression of UBE3A and ATP10C on the paternal allele through an antisense mechanism. PWS is a multi-gene disorder that arises
                              from the loss of expression of genes in this region from the paternal allele. AS arises from loss of maternally expressed UBE3A. c |
                              The GNAS locus, which is the site of the molecular defects that underlie Albright hereditary osteodystrophy (AHO), pseudohypopar-
                              athyroidism Ia (PHP-Ia) and PHP-Ib. Four alternative first exons (those of NESP55, XLα s, exon 1A and exon 1), each driven by its
                              own promoter, splice onto a common set of downstream exons (2–13). The promoters of NESP55, XLα s/NESP55-AS and exon
                              1A are differentially methylated and imprinted. Exon 1 (Gsα ) demonstrates preferential maternal expression in only certain tissues
                              (such as the renal proximal tubules); however, it is not differentially methylated. The location of the LOI defect at exon 1A in PHP-Ib
                              patients is shown. AHO and PHP-Ia arise from paternal and maternal transmission of Gsα mutations, respectively, while PHP-Ib is
                              due to LOI at exon 1A and, in some cases, also at the upstream DMRs. d | The transient neonatal diabetes mellitus (TNDM) locus.
                              A differentially methylated CpG island upstream of HYMAI acts as an ICR for this region, which contains three imprinted genes. In
                              TNDM patients, DNA methylation is lost from the maternal allele and expression of PLAGL1 is upregulated. CDKN1C, cyclin-
                              dependent kinase inhibitor 1C; HYMAI, hydatidiform mole associated and imprinted; IGF2, insulin-like growth factor 2; INS, insulin;
                              IPW, imprinted in Prader–Willi syndrome; KCNQ1, potassium voltage-gated channel, KQT-like subfamily, member 1; NESP55,
                              neuroendocrine secretory protein 55; PLAGL1, pleiomorphic adenoma gene-like 1; SNRPN, small nuclear ribonucleoprotein
                              polypeptide N; SNURF, SNRPN upstream reading frame; TSSC3, tumour-suppressing STF cDNA 3 (also known as PHLDA2);
                              UBE3A, ubiquitin protein ligase E3A.



NATURE REVIEWS | GENETICS                                                                                                           VOLUME 6 | AUGUST 2005 | 601
                                                              © 2005 Nature Publishing Group
REVIEWS


                                     IGF2 in ~20% of these cases (both resulting in biallelic        from Snurf/Snrpn to Ube3a60 contribute to aspects
                                     expression of the respective genes). Mutations and              of PWS, but further models are needed to define the
                                     translocations that involve the maternal allele account         contribution of all paternally expressed genes in this
                                     for the remaining cases. LOI at ICR2 involves loss of           region. The finding that a DNA methylation inhibitor
                                     maternal-allele-specific DNA methylation, whereas               restores expression of the silent maternal gene copy of
                                     at ICR1 the defect is usually hypermethylation of the           SNURF/SNRPN is intriguing, and indicates that such
                                     maternal allele, as seen in most Wilms’ tumours49,51.           inhibitors might be useful for PWS treatment61.
                                     Defects in the centromeric imprinted domain (silenc-
                                     ing of maternally expressed genes) are thought to give          Angelman syndrome (AS). AS occurs in ~1 in 15,000
                                     rise predominantly to the BWS phenotype (anatomi-               births and its main characteristics include mental
                                     cal malformation) whereas defects in the telomeric              retardation, speech impairment and behavioural
                                     imprinted domain (activation of maternally repressed            abnormalities (see online supplementary information
                                     IGF2) could be the main driving force for tumorigen-            S3 (table))55,62. The AS defect lies within the imprinted
                                     esis 52. Hypomethylation of the maternal ICR2 and acti-         domain at 15q11–q13 and is due to the loss of mater-
                                     vation of KCNQ1OT1 causes a marked downregulation               nally expressed genes (FIG. 2b). Unlike PWS, however,
                                     of genes that are centromeric to this, such as CDKN1C;          AS arises from the loss of a single gene — maternally
                                     however, the molecular mechanism of this regulation             expressed UBE3A — which is imprinted only in the
                                     is not understood53. One model is that ICR2 functions           brain and encodes an E3 ubiquitin ligase involved in
                                     in a similar way to ICR1 — that is, it might regulate,          the UBIQUITINPROTEASOME DEGRADATION PATHWAY .
                                     through binding of CTCF, the ability of an enhancer                 The most common molecular defects that give rise
                                     to access the CDKN1C and KCNQ1OT1 promoters                     to AS are maternally-derived deletions of 15q11–q13
                                     differentially on the two alleles49.                            (~65–70% of cases), paternal UPD (~5% of cases),
                                                                                                     maternal UBE3A mutations (~10% of cases, and up
                                                                                                     to ~40% in one study), and imprinting defects (~5%
                                     Prader–Willi syndrome (PWS). PWS occurs in ~1                   of cases). About 10% of cases arise from an unknown
                                     in 20,000 births and is characterized by a failure to           defect62. Analysis of samples from AS patients with
                                     thrive during infancy, hyperphagia and obesity during           microdeletions allowed the AS ICR to be mapped to an
                                     early childhood, mental retardation, and behavioural            880 bp region ~35 kb upstream of SNURF/SNRPN63.
                                     problems (see online supplementary information S3               Imprinting defects involved in AS include loss of mater-
                                     (table))54. The molecular defect is complex and involves        nal DNA methylation or maternal ICR deletion, and
                                     a ~2 Mb imprinted domain at 15q11–q13 that con-                 patients with these defects tend to have the mildest AS
                                     tains both paternally and maternally expressed genes            phenotypes62. Important unanswered questions related
                                     (FIG. 2b). One maternally expressed gene, ubiquitin             to AS biology include the nature of the molecular defects
                                     protein ligase E3A (UBE3A), is imprinted only in the            in the ~10% of cases that have no identifiable UBE3A
                                     brain and is discussed in the next section.                     mutation and the role of the nearby gene ATP10C
                                         PWS arises from loss of paternally expressed genes in       (ATPase, class V, type 10A), which is often co-deleted
                                     this region, although no single gene has been shown to          with UBE3A, in modulating the AS phenotype.
                                     cause PWS when its expression is lost55. The analysis of
                                     samples from PWS patients with microdeletions allowed           Albright hereditary osteodystrophy (AHO), pseudo-
                                     the PWS ICR to be mapped to a segment of ~4 kb that             hypoparathyroidism type Ia (PHP-Ia) and PHP-Ib.
                                     spans the first exon and promoter of the small nuclear          These three diseases are related because they all arise
BALANCED TRANSLOCATION               ribonucleoprotein polypeptide N (SNRPN)/SNRPN                   from defects at a complex imprinted locus on chromo-
A condition in which two pieces      upstream reading frame (SNURF) locus56. In normal               some 20q13 called GNAS, which encodes the α subunit
of chromosomal material have         cells, the 5′-end of the ICR, which is needed for mater-        (Gsα) of the heterotrimeric GTP-binding protein Gs
switched places, but the correct
                                     nal gene expression and is involved in Angelman syn-            (FIG. 2c; and see online supplementary information
number of chromosomes has
been maintained.                     drome (discussed below), is methylated on the maternal          S3(table)). AHO is a paternally inherited disorder that
                                     allele55,57. The 3′ end of the ICR is required for expression   occurs within PHP-Ia pedigrees, and involves mental
UBIQUITINPROTEASOME                 of the paternally expressed genes (the PWS region shown         retardation and subcutaneous ossification.
DEGRADATION PATHWAY
                                     in FIG. 2b) and is also the origin of the extremely long           PHP-Ia is maternally transmitted in a dominant
Degradation pathway in which
a protein that has been post-
                                     SNURF/SNRPN transcript. The maternally expressed                manner and patients show all the AHO symptoms,
translationally modified with        genes are not differentially methylated, and their silenc-      plus resistance to the peripheral action of several
several ubiquitin polypeptides       ing on the paternal allele is probably regulated by an          hormones involved in activating pathways coupled to
is targeted for destruction to the   antisense RNA generated from SNURF/SNRPN58.                     Gs. Individuals with PHP-Ib, which is also maternally
proteasome, a large cytosolic
                                         The most common molecular defects in PWS are                inherited, show normal Gsα activity and only renal
protein complex with several
proteolytic activities.              paternal deletions spanning the 15q11–q13 region                parathyroid hormone resistance, with no other endo-
                                     (65–70% of cases) and maternal UPD (~20–30% of                  crine deficiencies64.
CHROMOGRANINS                        cases). Paternally derived microdeletions of the ICR,              The GNAS locus encodes three proteins and an
A group of acidic, soluble,          LOI at the ICR (de novo methylation of the pater-               untranslated RNA from four alternative first exons
secretory proteins that are
produced by neurons and
                                     nal allele), and BALANCED TRANSLOCATIONS that disrupt           that splice onto common exons 2–13 (FIG. 2c). NESP55,
neuroendocrine cells.                SNURF/SNRPN, account for the remainder51. Mouse                 generated from the most centromeric of the first exons,
                                     knockout studies indicate that Ndn59 and the region             encodes a CHROMOGRANIN-like protein, XLαs encodes a


602 | AUGUST 2005         | VOLUME 6                                                                                        www.nature.com/reviews/genetics
                                                                   © 2005 Nature Publishing Group
REVIEWS


                                  Golgi-specific Gsα isoform, and exon 1 encodes Gsα.               Three mechanisms give rise to TNDM: paternal UPD
                                  Transcripts derived from exon 1A do not seem to be            of chromosome 6, paternal duplications of 6q24, and
                                  translated. Each alternative first exon is driven from its    LOI at the TNDM CpG-island-associated ICR, which
                                  own promoter, and the first three promoters are dif-          leads to increased PLAGL1 expression67. TNDM patients
                                  ferentially methylated. NESP55 is maternally expressed        who lack chromosomal abnormalities demonstrate LOI
                                  and its promoter region is methylated on the paternal         of the ICR, specifically loss of DNA methylation from
                                  allele. Gsα (exon 1) is preferentially expressed from the     the maternal allele68. Because the methylated ICR acts as
                                  maternal allele, although its promoter region seems           a repressor of PLAGL1 transcription, LOI has the same
                                  to be unmethylated on both alleles. Gsα is imprinted          consequences as paternal UPD and 6q24 duplication.
                                  only in certain hormone-responsive tissues, such as the           The TNDM locus has a much simpler organiza-
                                  renal proximal tubules, which is the probable cause of        tion than those that are involved in BWS, PWS and
                                  the tissue-specific effects of these disorders. By con-       AHO/PHP. However, this further emphasizes that most
                                  trast, XLαs, a NESP55 antisense transcript, and exon          imprinted genes are found in clusters under the control
                                  1A are all paternally expressed, and their promoters          of regulatory regions that are often distant from the
                                  are methylated on the maternal allele64.                      genes themselves, and that this control is dependent
                                      The molecular defects that underlie AHO, PHP-Ia,          on allele-specific DNA methylation.
                                  and PHP-Ib, are well characterized. AHO and PHP-Ia
                                  arise from the paternal and maternal transmission of          A wider role of imprinting in disease. These examples
                                  Gsα mutations, respectively64. By contrast, PHP-Ib            of diseases associated with aberrant imprinting are
                                  involves LOI at exon 1A (loss of maternal-specific            probably only the tip of the iceberg. Unlike the global
                                  methylation), which occurs in all cases. Interestingly,       genomic hypomethylation and widespread CpG island
                                  it was recently demonstrated that sporadic PHP-Ib             hypermethylation seen in cancer and cancer-associated
                                  (the most common form) also involves imprinting               LOI, which probably affects many imprinted regions,
                                  defects at the upstream DMRs, in the form of biallelic        the DNA methylation defects in imprinting disorders
                                  methylation at NESP55 and hypomethylation at the              are subtle. Numerous other pathologies demonstrate
                                  NESP antisense (NESPAS)/XLαs promoter region. By              parent-of-origin effects or result from UPD, indicating
                                  contrast, familial PHP-Ib is associated with maternally       that imprinted genes might be involved69. Interestingly,
                                  transmitted deletions within the nearby syntaxin 16           accumulating evidence from the diseases described
                                  (STX16) gene and LOI at the exon 1A DMR only65.               above indicates that loss of normal CTCF function has
                                  NESP55, XLαs and exon 1A are probably regulated by            an important role in imprinting disorders. Studies of
                                  antisense transcription and/or differential promoter          the mechanism by which CTCF regulates DNA meth-
                                  methylation. However it is still unclear how G s α            ylation patterns in imprinted regions also indicate that
                                  exon 1 is regulated, as it is not differentially methylated   it could have a much broader role in regulating DNA
                                  and no antisense transcript has been identified. The          methylation patterns throughout the genome than is
                                  deleted region in STX16 might contain a cis-element           currently appreciated70.
                                  that acts over a large distance to establish imprinting
                                  at the exon 1A DMR65. LOI at exon 1A (and biallelic           Methylation and repeat-instability diseases
                                  Gsα expression) also occurs in a subset of growth-            DNA methylation defects have been linked to members
                                  hormone-secreting pituitary adenomas, supporting              of a large group of human diseases that are associated
                                  the idea that imprinting of each GNAS promoter is             with repeat instability. Expansion of trinucleotide
                                  independently regulated66.                                    repeats (TNRs) during gametogenesis leads to muta-
                                                                                                tion or silencing of associated genes, with pathological
                                  Transient neonatal diabetes mellitus (TNDM). TNDM             consequences71. The causes of TNR instability remain
                                  is a rare form of diabetes (occuring in ~1 in 400,000         largely unknown, although DNA polymerase slippage,
                                  births) that presents during the first few weeks after        DNA repair mechanisms, transcription, nucleosome
                                  birth. Remission usually occurs after 3–6 months, but         positioning and aberrant OKAZAKIFRAGMENT process-
                                  a significant proportion of patients develop diabetes         ing within the repeat have been proposed72. Some of
                                  later in life67. The TNDM locus on chromosome 6q24            these diseases involve expansions of non-methylatable
                                  spans ~400 kb and contains three imprinted genes              repeats, such as CAG in Huntington’s disease (HD) and
                                  — the paternally expressed genes hydatidiform mole            several forms of spinocerebellar ataxia (SCA1, 2, 3, 6,
                                  associated and imprinted (HYMAI) and pleiomorphic             7, and 17), CTG in myotonic dystrophy type 1 (DM1)
                                  adenoma gene-like 1 (PLAGL1; also known as ZAC or             and SCA8, and GAA in Friedreich’s ataxia (FRDA)71.
                                  LOT1), and the maternally expressed PLAGL1 anti-              Expansion of methylatable CGG repeats occurs at
                                  sense transcript (ZAC-AS) (FIG. 2d). HYMAI gives rise to      several sites that are known or likely to be involved
                                  an apparently untranslated RNA of unknown function,           in human disease, and increased DNA methylation
                                  whereas PLAGL1 encodes a zinc finger protein that is a        is thought to have a role in pathogenesis. In contrast
                                  putative tumour suppressor and transcriptional regula-        to the TNR expansion diseases, facioscapulohumeral
                                  tor involved in modulating the cell cycle and apoptosis.      muscular dystrophy (FSHD) is due to the contraction
OKAZAKI FRAGMENTS
Short pieces of DNA that are
                                  A putative ICR within the CpG island at the 5′-end of         of a much larger repeat accompanied by hypomethyla-
synthesized on the lagging        HYMAI is methylated on the maternal allele and acts as        tion. These diseases offer insights into possible mecha-
strand at the replication fork.   a transcriptional repressor when methylated68.                nisms for the targeting of de novo DNA methylation.


NATURE REVIEWS | GENETICS                                                                                               VOLUME 6 | AUGUST 2005 | 603
                                                               © 2005 Nature Publishing Group
REVIEWS


a FRAXA Xq27.3                                   CGG repeat                                                       The molecular basis of FRAXA lies in the highly
                                                                     ATG                                      polymorphic CGG repeat within the 5′-untranslated
5′                                                                                                  3′        region of FMR1 (FIG. 3a). Normally, 6–52 copies of the
                                                                                                              repeat are present, which increases to 52–200 copies
                              TATA                 5′ UTR           FMR1 gene                                 in the premutation state (now recognized as a separate
                                                                                                              disease called fragile X tremor/ataxia syndrome76),
                                                       Repeat expansion                                       and to more than 200 repeats in individuals with
                                                                                                              FRAXA73,77. Affected individuals show de novo methy-
                                             CGG repeat                                                       lation of the expanded CGG repeat and silencing of
                                                                     ATG
                                                                                                              FMR1 transcription77. DNA methylation is clearly an
5′                                                                                                  3′        important mediator of silencing as DNMT inhibitors
Targeting to DNA hairpins?                                                                                    reactivate transcription of the full-mutation allele,
                                                                             FMR1 hairpin RNA                 although FMRP is not produced78.
                     DNMT1
                                    Methylation?
                                                                                                              Other folate-sensitive fragile sites. The FRAXA locus
                                                              RNAi                    Dicer
                                                              machinery                                       is one of more than 20 folate-sensitive fragile sites in
                                                                                                              the genome. CGG repeat expansion and hypermeth-
                                                                                                              ylation have been documented for several other such
b FSHD 4q35                                                                                                   sites associated with genes (FRAXE, FRAXF, FRA10A,
                                                                          Repressor                           FRA11B and FRA16A) although only FRAXE has been
                        ?              ?                ?                 complex
                                                                                                              firmly linked to human disease (nonspecific X-linked
                                                                                                              mental retardation)74,79. It is reasonable to assume
5′                                                                                                  3′        that all folate-sensitive fragile sites similarly occur at
        ANT1             FRG1               FRG2                 D4Z4 (>11)                                   CGG repeat expansions that are accompanied by DNA
                                                                                                              methylation. All fragile sites characterized so far that
                                                   Repeat contraction                                         have a genomic structure similar to the FMR1 gene (a
                                                   and hypomethylation                                        CGG repeat in the untranslated region and a nearby
               ?                ?                  ?
                                                                                                              CpG island) show hypermethylation of both regions
                                                                                                              in the repeat-expanded state, and transcription of the
5′                                                                                                  3′        associated gene is silenced74.
        ANT1             FRG1               FRG2            D4Z4 (1–10)
                                                                                                              Facioscapulohumeral muscular dystrophy. FSHD
Figure 3 | DNA methylation and diseases associated with repeat instability.
                                                                                                              is an autosomal dominant disease that affects ~1 in
a | Trinucleotide repeat instability associated with aberrant DNA methylation in FRAXA.
A schematic representation of the 5′-end of the FMR1 gene is shown. The CGG repeat in
                                                                                                              20,000 individuals (see online supplementary infor-
FMR1 lies in the 5′ untranslated region (UTR), which is embedded in a hypomethylated CpG                      mation S3 (table)). The FSHD locus maps to 4q35 and
island in the normal state. In fully affected individuals with large repeat expansions, the repeat            the underlying molecular defect is well established,
and CpG island become de novo methylated, resulting in gene silencing. Models for how the                     involving contraction of the polymorphic 3.3 kb D4Z4
repeat is targeted for methylation include the formation of unusual DNA structures that attract               repeat. Unaffected individuals have 11–150 copies of
DNA methyltransferase 1 (DNMT1), and the formation of hairpin RNA from the transcribed                        this repeat, whereas in most FSHD cases the repeat is
repeat, which might be cleaved by Dicer and subsequently recruit the RNAi silencing
machinery, including DNMTs. b | Facioscapulohumeral muscular dystrophy (FSHD) is caused
                                                                                                              reduced to 1–10 copies, although some patients (~5%)
by contraction of the D4Z4 repeat on chromosome 4q35 and loss of DNA methylation.                             do not show repeat contraction. The abnormality that is
Whereas repeat contraction is not always present in FSHD, repeat hypomethylation is an                        common to all FSHD patients is loss of DNA methyla-
invariant feature. D4Z4 repeat contraction and hypomethylation are believed to cause the                      tion from D4Z4, indicating that aberrant hypomethy-
abnormal expression of adjacent genes (FRG2 is considered a good candidate) or genes in                       lation is the main causal factor80. However, the genes
other regions of the genome. One model has proposed that a repressor complex binds to the                     affected by this hypomethylation are unknown.
repeats and regulates the expression of nearby genes, although the role of DNA methylation
                                                                                                                  Most studies have focused on the idea that con-
was not addressed in this study. ANT1, solute carrier family 25, also known as SLC25A4;
FRG2, FSHD region gene 2 protein.                                                                             traction and hypomethylation of D4Z4 alters the
                                                                                                              transcription of one or more genes that flank the
                                                                                                              repeat. Genes in the vicinity of D4Z4 (FIG. 3b) include
                                           Fragile X syndrome (FRAXA). FRAXA, an X-linked                     the adenine nucleotide translocator SLC25A4 (also
                                           disorder, is a common cause of inherited mental                    known as ANT1), the overexpression of which induces
                                           retardation and occurs in ~1 in 4000 males (see online             apoptosis (characteristic of dystrophic muscle); FSHD
                                           supplementary information S3 (table))73,74. The disease            region gene 1 (FRG1) which is ubiquitously expressed
                                           maps to the fragile X mental retardation 1 (FMR1)                  and might be involved in RNA processing; and FRG2
FOLATESENSITIVE                           gene at Xq27.3, which is also a FOLATESENSITIVE FRAGILE           (which is closest to D4Z4), the function of which is
FRAGILE SITE                               SITE. The FMR1 protein (FMRP) is an RNA-binding                    unknown but which has been shown to induce mor-
A region of chromatin that fails           protein expressed in many fetal and adult tissues, with            phological changes in transfected myoblasts81. Results
to compact normally during                 particularly high levels in brain and testis. It seems to          from different laboratories have yielded conflicting
mitosis and that can be
observed after culturing cells in
                                           be involved in regulating translation and mRNA trans-              data about the degree of overexpression of these genes
media that is deficient in folic           port, and is believed to regulate synaptic plasticity by           in FSHD muscle, although FRG2 does seem to be a
acid and thymidine.                        controlling translation at synapses75.                             promising candidate for the disease gene81–83.


604 | AUGUST 2005           | VOLUME 6                                                                                                www.nature.com/reviews/genetics
                                                                             © 2005 Nature Publishing Group
REVIEWS


                                      There are also diverse models for how repeat con-            number in FSHD could reduce the efficiency of RNAi
                                   traction and hypomethylation alter gene expression and          targeting, eventually resulting in hypomethylation.
                                   give rise to FSHD. One possible mechanism involves              The molecular defects that underlie these diseases
                                   the binding of a repressor complex to the D4Z4 repeat,          are therefore consistent with the growing body of
                                   which is reduced as the repeat size decreases, causing          evidence that the insulator factor CTCF and/or
                                   elevated expression of nearby genes (although this study        RNAi are involved in regulating DNA methylation
                                   did not address the role of methylation)82. A looping           patterns.
                                   model has also been proposed, whereby the D4Z4
                                   repeat binds transcriptional regulatory factors and             Specific defects of the methylation machinery
                                   loops over to distant genes, which might be well outside        Human diseases that arise from mutations in the DNA
                                   the immediate vicinity of D4Z4. Repeat contraction and          methylation machinery have provided important
                                   hypomethylation might disrupt the binding of crucial            insights into the functional specialization of DNMTs.
                                   regulatory proteins or negatively affect the ability of the     They have also shown that improper regulation of
                                   repeat to interact with other regulatory regions83.             DNA methylation can occur in specific cell types, with
                                                                                                   pathological consequences.
                                   Is there a wider role for DNA methylation in diseases
                                   of repeat instability? Studies of repeat-instability dis-       Systemic lupus erythematosus (SLE). SLE is an autoim-
                                   eases demonstrate that repetitive DNA, in the form of           mune disease, with diverse clinical manifestations, that
                                   both naturally occurring repeats and pathologically             is 8–10 times more frequent in females than in males
                                   expanded TNRs, is efficiently targeted by the methyla-          (~1 in 2000 in the overall population) (see online sup-
                                   tion machinery. Why only some expanded TNRs are                 plementary information S3 (table)). Common features
                                   targeted for de novo methylation remains unknown. It            in all cases are autoantibody production and the pres-
                                   might simply be the availability of a methylatable site         ence of antibodies against nuclear components, such as
                                   within or near the repeat expansion that makes CGG              DNA, chromatin factors and small nuclear ribonuclear
                                   rather than CTG or CAG TNRs attractive to the meth-             protein particles92. There is also evidence for defective
                                   ylation machinery. However, several lines of evidence           T-cell function, which provides the main connection
                                   indicate that the silencing machinery might be recruited        with DNA methylation93.
                                   to expanded TNRs, regardless of their sequence. For                 T cells are believed to drive the autoantibody
RNAI                               example, a CpG island adjacent to the CTG repeat                response in SLE, and this has been proposed to result
The process whereby double-        involved in DM1 (which also contains binding sites for          from loss of DNA methylation in these cells. The
stranded RNAs are cleaved into     CTCF) becomes hypermethylated in the severe con-                genomes of SLE T cells are globally hypomethylated
21-23 nucleotide duplexes
termed small interfering RNAs,
                                   genital form of the disease84,85. In both DM1 and FRDA,         (15–20% reduction) and DNMT1 levels are reduced94.
leading to inhibition of           the associated genes (DMPK and FRDA, respectively)              Treatment of normal CD4 T CELLS with the DNA methy-
                                                                                                                            +


expression of genes that contain   are downregulated, and the expanded repeat adopts a             lation inhibitor 5-aza-2′-deoxycytidine renders them
a complementary sequence.          repressive chromatin structure86. This indicates that           autoreactive, and ADOPTIVE TRANSFER of these cells
                                   TNR expansions might commonly be targeted for                   causes an SLE-like disease in mice95. Hypomethylated
DICER
A ribonuclease that processes      silencing at the chromatin level, and that additional           T cells are also able to promiscuously kill SYNGENEIC
dsRNAs to ~21 nucleotide           factors might influence whether DNA methylation                 MACROPHAGES; this increased apoptosis, combined with
siRNAs (for RNAi) or excises       also occurs. In addition, DNA methylation stabilizes            reduced clearance of apoptotic material, is likely to
microRNAs from their hairpin       expanded repeats in model systems regardless of                 have a role in inducing the anti-DNA antibodies that
precursors.
                                   whether they contain a CpG site87,88.                           characterise SLE. In addition, several genes relevant to
CD4+ T CELL                            Other aspects that might influence methylation              the SLE phenotype are methylated in normal T cells
Also known as a helper T cell.     targeting include whether the repeats involved are              but are demethylated by both 5-aza-2′-deoxycytidine
Initiates both antibody            transcribed, as they are in FRAXA and DM1, or                   and in SLE patients96,97. An alternative hypothesis
production by B cells and
                                   transcribed and translated, as in HD, where there is            for the role of DNA methylation in SLE involves the
stimulates the activation of
other immune cells, such as        no evidence of disease-associated hypermethylation89.           hypomethylation of endogenous retroviruses (HERV)
macrophages, after recognizing     Another factor might be the propensity of a repeat to           and the re-expression of viral proteins. Antibodies to
a portion of a protein antigen     form DNA hairpins, which have been shown to be pre-             HERV-encoded proteins have been detected in SLE
on the surface of an antigen       ferred substrates for DNMT190. The finding that FMR1            patients, and peptides derived from these proteins
presenting cell.
                                   mRNA also forms hairpin structures, and the discovery           cause aberrant CD4+ T-cell responses98.
ADOPTIVE TRANSFER                  that they are substrates for the RNAI -associated DICER ribo-       Exposure to several DNA methylation inhibitors
The process of conferring          nuclease provides a possible alternative explanation.           also induces a lupus-like disease in humans, provid-
immunity to an individual by       Components of the histone-modification machinery                ing further support for a role of aberrant DNA methy-
transferring cells or serum from
                                   that cause transcriptional repression are recruited to          lation in SLE93,99. DNMT1 is upregulated in normal
another individual that has
been immunized with a specific     repeats by the RNAi pathway, which probably also                T cells following stimulation by signals transmitted
antigen.                           recruits the DNA methylation machinery, although                through the extracellular signal-regulated kinase
                                   this has not been demonstrated directly (see later for          (ERK) pathway, which is impaired in lupus T cells.
SYNGENEIC MACROPHAGES
                                   a further discussion)13,91. In support of this model,           The drug hydralazine is thought to cause hypomethy-
macrophages (immune cells
that engulf foreign particles)
                                   the affected repeats and/or adjacent sequences adopt            lation in T cells and an SLE-like disease in humans
that are transferred between       tightly packed chromatin structures in FRAXA, DM1               by inhibiting this pathway. Impaired ERK signalling
genetically identical mice.        and FRDA78,86. Conversely, reduction in D4Z4 repeat             probably contributes to reduced DNMT1 levels and


NATURE REVIEWS | GENETICS                                                                                                  VOLUME 6 | AUGUST 2005 | 605
                                                                 © 2005 Nature Publishing Group
REVIEWS


                                   hypomethylation of SLE T cells 100 . SLE therefore           toid cell lines, indicating a defect in differentiation
                                   demonstrates not only that DNA methylation defects           or activation. However, these experiments did not
                                   can be restricted to only one or a few cell types, but       provide obvious candidate genes to explain the devel-
                                   also that DNA methylation is regulated differently,          opmental and neurological aspects of ICF syndrome.
                                   or responds to different stimuli, depending on cell          Interestingly, genes with altered expression in ICF
                                   type.                                                        cells are not hypomethylated. This implies either that
                                                                                                DNMT3B regulates gene expression independently of
                                   Immunodeficiency, centromeric instability and                its ability to methylate DNA, or that the affected genes
                                   facial anomalies (ICF) syndrome. ICF syndrome is             show altered expression owing to downstream effects
                                   an extremely rare autosomal recessive disease that           of other genes, such as transcription factors, which
                                   is characterized by profound immunodeficiency, and           might be activated by hypomethylation113. The former
                                   which results from an absence or significant reduction       idea is supported by recent findings that DNMT3B
                                   of at least two immunoglobulin isotypes. Other more          interacts with histone deacetylases (HDACs), histone
                                   variable features include impaired cellular immunity         methylases and chromatin-remodelling enzymes114,115.
                                   and unusual facial features101,102. All ICF patients show    In addition, DNMT3B can profoundly influence neu-
                                   marked hypomethylation and decondensation of peri-           ronal differentiation of multipotent cells in a manner
                                   centromeric heterochromatin on chromosomes 1 and             that depends on its ability to interact with HDACs, but
                                   16, and, to a lesser extent, chromosome 9 (see online        not on its ability to methylate DNA116. Both methyla-
                                   supplementary information S3 (table)). This abnor-           tion-related and unrelated defects might therefore be
                                   mality is observed almost exclusively in ICF B cells or      important in ICF syndrome.
                                   in LYMPHOBLASTOID CELL LINES that have been stimulated
                                   with mitogen101,103,104.                                     ATRX: a connection with chromatin remodelling
                                       The molecular defect in 60–70% of ICF patients is        Alpha-thalassemia/mental retardation syndrome,
                                   mutation of the DNMT3B gene at 20q11.2, leading to           X-linked (ATRX) is a very rare X-linked disease that
                                   defective methylation11,105,106. Patients without detect-    occurs in <1 in 100,000 males. It is characterized by
                                   able DNMT3B mutations might have abnormalities in            severe mental retardation and genital abnormalities,
                                   other regions of the gene that have not been exam-           with alpha-thalassemia in many, but not all, patients.
                                   ined (such as the promoter and 5′ and 3′ UTRs), or           The affected gene, ATRX, is located at Xq13 (see online
                                   might arise from mutations in a gene that regulates          supplementary information S3 (table)) and encodes
                                   DNMT3B. Mutations in DNMT3B are usually het-                 an ATP-dependent chromatin-remodelling protein of
                                   erozygous and target the catalytic domain. Complete          the chromodomain and helicase-like domains (CHD)
                                   loss-of-function of the encoded enzyme is probably           subclass of SNF2LIKE PROTEINS117.
                                   embryonic lethal in humans, as it is in mice11, and sev-         Functional domains of ATRX include a PHD zinc
                                   eral of the catalytic domain mutations examined so far       finger-like motif at its N-terminus (homologous to
                                   do not result in complete loss of enzyme activity105,107.    the PHD motifs in DNMT3A and DNMT3B), and a
                                   The clinical variability of the disease might therefore      helicase domain at its C-terminus. Mutations in ATRX
                                   be related to the level of residual DNMT3B activity.         are clustered in these two domains and are thought to
                                   It was recently suggested that different methylation         impair its nuclear localization, protein–protein inter-
                                   defects are present in ICF patients with and without         actions, or chromatin-remodelling functions118–120.
                                   DNMT3B mutations, but the molecular basis behind             ATRX localizes to pericentromeric heterochromatin
                                   these differences is not known108.                           and PROMYELOCYTIC LEUKEMIA PML NUCLEAR BODIES in a
                                       The instability of repeat-rich pericentromeric           cell-cycle-dependent manner, and exists in a complex
                                   heterochromatin in this disease is probably due to           with the transcriptional regulatory death-associated
                                   its hypomethylation in ICF cells. Loss of DNA meth-          protein 6 (DAXX)120–122. It is therefore thought to
LYMPHOBLASTOID CELL LINE
                                   ylation in ICF syndrome is region specific and affects       regulate transcription, although its target genes and
Immortalized B cell line created
by infecting primary B cells       mainly the pericentromeric satellite 2 and 3 repeats         the nature of the regulation (positive or negative) are
with Epstein-Barr virus.           and non-pericentromeric regions that include the             unknown.
                                   inactive X chromosome, cancer–testis genes, and                  Interestingly, ATRX patients also have DNA-
SNF2LIKE PROTEINS
                                   other sporadic repeats (such as NBL2 and the tar-            methylation defects. These are diverse, and include
ATP-dependent chromatin
remodelling enzymes that
                                   get in FSHD, D4Z4)109–111. This indicates that these         both aberrant hypermethylation (at the DYZ2
contain a region homologous to     regions are bona-fide targets for DNMT3B. Satellite          Y-chromosome repeat) and hypomethylation events
an extended family of proteins     2 and 3 hypomethylation is common in cancer cells,           (at ribosomal DNA repeats), but no overall change in
that include known RNA and         and although no elevated cancer incidence has been           5-methylcytosine levels123. It remains unclear, how-
DNA helicases.
                                   reported in ICF patients, the small number of these          ever, if methylation abnormalities directly contribute
PML BODIES
                                   patients and their short lifespan make it possible that a    to the ATRX phenotype. There have been important
Dot-like structures in the         slightly increased incidence has gone undetected112.         advances in recent years in our understanding of the
nucleus of most mammalian              The role of DNMT3B mutations in the invariant            direct and indirect connections between the DNA
cells. These were originally       immune defects in ICF syndrome is far less clear.            methylation and chromatin-remodelling machin-
defined by the localization of
the PML protein, which is
                                   Microarray analyses have revealed that many genes            eries, and the identification of DNA methylation
involved in transcriptional        involved in immune function and B-cell immunoglob-           defects in ATRX patients has reinforced this link in
regulation.                        ulin production are deregulated in ICF lymphoblas-           human cells124.


606 | AUGUST 2005        | VOLUME 6                                                                                    www.nature.com/reviews/genetics
                                                               © 2005 Nature Publishing Group
002 & 003   Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14

Weitere ähnliche Inhalte

Was ist angesagt?

Dna methylation pattern during development
Dna methylation pattern during developmentDna methylation pattern during development
Dna methylation pattern during development
Bhupendra Bhuppe
 

Was ist angesagt? (20)

Dna methylation
Dna methylationDna methylation
Dna methylation
 
Inheritence of dna methylation
Inheritence of dna methylationInheritence of dna methylation
Inheritence of dna methylation
 
Epigenetic role in plant
Epigenetic role in plant Epigenetic role in plant
Epigenetic role in plant
 
The Role of DNA Methylation in Coronary Artery Disease
 The Role of DNA Methylation in Coronary Artery Disease The Role of DNA Methylation in Coronary Artery Disease
The Role of DNA Methylation in Coronary Artery Disease
 
Epigenetics
Epigenetics Epigenetics
Epigenetics
 
Dna methyltransferase screening
Dna methyltransferase screeningDna methyltransferase screening
Dna methyltransferase screening
 
DNA Methylation
DNA MethylationDNA Methylation
DNA Methylation
 
Dna methylation pattern during development
Dna methylation pattern during developmentDna methylation pattern during development
Dna methylation pattern during development
 
Ibica2014 p(8) visualizing and identifying the dna methylation
Ibica2014 p(8) visualizing and identifying the dna methylationIbica2014 p(8) visualizing and identifying the dna methylation
Ibica2014 p(8) visualizing and identifying the dna methylation
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Epigenetics final
Epigenetics finalEpigenetics final
Epigenetics final
 
dna Imprinting
 dna Imprinting dna Imprinting
dna Imprinting
 
Epigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plantsEpigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plants
 
Dna methylation field guide 20130806
Dna methylation field guide 20130806Dna methylation field guide 20130806
Dna methylation field guide 20130806
 
Epigenetic
EpigeneticEpigenetic
Epigenetic
 
Epigenetic
EpigeneticEpigenetic
Epigenetic
 
Epigenetics role in diseases
Epigenetics role in diseasesEpigenetics role in diseases
Epigenetics role in diseases
 
Definition of epigenetics
Definition of epigeneticsDefinition of epigenetics
Definition of epigenetics
 
The role of DNA methylation in complex diseases
The role of DNA methylation in complex diseasesThe role of DNA methylation in complex diseases
The role of DNA methylation in complex diseases
 

Ähnlich wie 002 & 003 Dna Methyl And Human Disease 14

Genomic imprinting: recognition and marking of imprinted loci
Genomic imprinting: recognition and marking of imprinted lociGenomic imprinting: recognition and marking of imprinted loci
Genomic imprinting: recognition and marking of imprinted loci
Bárbara Pérez
 
Gene regulation eukaryote spptx
Gene regulation eukaryote spptxGene regulation eukaryote spptx
Gene regulation eukaryote spptx
aljeirou
 
Integrative Analysis of Epigenomics and miRNA data in Immune System Models
Integrative Analysis of Epigenomics and miRNA data in Immune System ModelsIntegrative Analysis of Epigenomics and miRNA data in Immune System Models
Integrative Analysis of Epigenomics and miRNA data in Immune System Models
COST action BM1006
 

Ähnlich wie 002 & 003 Dna Methyl And Human Disease 14 (20)

Dna methylation
Dna methylationDna methylation
Dna methylation
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
 
Genomic imprinting: recognition and marking of imprinted loci
Genomic imprinting: recognition and marking of imprinted lociGenomic imprinting: recognition and marking of imprinted loci
Genomic imprinting: recognition and marking of imprinted loci
 
Epigenetics : overview and concepts
Epigenetics : overview and conceptsEpigenetics : overview and concepts
Epigenetics : overview and concepts
 
Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2
 
Cancer epigenetics
Cancer epigenetics Cancer epigenetics
Cancer epigenetics
 
Genetic instability
Genetic instabilityGenetic instability
Genetic instability
 
Cancer Epigenetics: Concepts, Challenges and Promises
Cancer Epigenetics: Concepts, Challenges and PromisesCancer Epigenetics: Concepts, Challenges and Promises
Cancer Epigenetics: Concepts, Challenges and Promises
 
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
 
cancer nice paper.pdf
cancer nice paper.pdfcancer nice paper.pdf
cancer nice paper.pdf
 
DNA methylation_2023.pptx
DNA methylation_2023.pptxDNA methylation_2023.pptx
DNA methylation_2023.pptx
 
Epigenetics 12
Epigenetics 12Epigenetics 12
Epigenetics 12
 
Cancer
CancerCancer
Cancer
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Biologia plegable
Biologia plegableBiologia plegable
Biologia plegable
 
DNA methylation
DNA methylationDNA methylation
DNA methylation
 
Gene regulation eukaryote spptx
Gene regulation eukaryote spptxGene regulation eukaryote spptx
Gene regulation eukaryote spptx
 
Integrative Analysis of Epigenomics and miRNA data in Immune System Models
Integrative Analysis of Epigenomics and miRNA data in Immune System ModelsIntegrative Analysis of Epigenomics and miRNA data in Immune System Models
Integrative Analysis of Epigenomics and miRNA data in Immune System Models
 
DNA Methylation & C Value.pdf
DNA Methylation & C Value.pdfDNA Methylation & C Value.pdf
DNA Methylation & C Value.pdf
 

Kürzlich hochgeladen

1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
QucHHunhnh
 
Gardella_PRCampaignConclusion Pitch Letter
Gardella_PRCampaignConclusion Pitch LetterGardella_PRCampaignConclusion Pitch Letter
Gardella_PRCampaignConclusion Pitch Letter
MateoGardella
 

Kürzlich hochgeladen (20)

Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 
Mattingly "AI & Prompt Design: The Basics of Prompt Design"
Mattingly "AI & Prompt Design: The Basics of Prompt Design"Mattingly "AI & Prompt Design: The Basics of Prompt Design"
Mattingly "AI & Prompt Design: The Basics of Prompt Design"
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
 
Unit-IV- Pharma. Marketing Channels.pptx
Unit-IV- Pharma. Marketing Channels.pptxUnit-IV- Pharma. Marketing Channels.pptx
Unit-IV- Pharma. Marketing Channels.pptx
 
APM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAPM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across Sectors
 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptx
 
Mehran University Newsletter Vol-X, Issue-I, 2024
Mehran University Newsletter Vol-X, Issue-I, 2024Mehran University Newsletter Vol-X, Issue-I, 2024
Mehran University Newsletter Vol-X, Issue-I, 2024
 
Web & Social Media Analytics Previous Year Question Paper.pdf
Web & Social Media Analytics Previous Year Question Paper.pdfWeb & Social Media Analytics Previous Year Question Paper.pdf
Web & Social Media Analytics Previous Year Question Paper.pdf
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy Consulting
 
Mixin Classes in Odoo 17 How to Extend Models Using Mixin Classes
Mixin Classes in Odoo 17  How to Extend Models Using Mixin ClassesMixin Classes in Odoo 17  How to Extend Models Using Mixin Classes
Mixin Classes in Odoo 17 How to Extend Models Using Mixin Classes
 
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptxINDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
 
Measures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDMeasures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SD
 
Holdier Curriculum Vitae (April 2024).pdf
Holdier Curriculum Vitae (April 2024).pdfHoldier Curriculum Vitae (April 2024).pdf
Holdier Curriculum Vitae (April 2024).pdf
 
Nutritional Needs Presentation - HLTH 104
Nutritional Needs Presentation - HLTH 104Nutritional Needs Presentation - HLTH 104
Nutritional Needs Presentation - HLTH 104
 
This PowerPoint helps students to consider the concept of infinity.
This PowerPoint helps students to consider the concept of infinity.This PowerPoint helps students to consider the concept of infinity.
This PowerPoint helps students to consider the concept of infinity.
 
Sports & Fitness Value Added Course FY..
Sports & Fitness Value Added Course FY..Sports & Fitness Value Added Course FY..
Sports & Fitness Value Added Course FY..
 
Gardella_PRCampaignConclusion Pitch Letter
Gardella_PRCampaignConclusion Pitch LetterGardella_PRCampaignConclusion Pitch Letter
Gardella_PRCampaignConclusion Pitch Letter
 
Class 11th Physics NEET formula sheet pdf
Class 11th Physics NEET formula sheet pdfClass 11th Physics NEET formula sheet pdf
Class 11th Physics NEET formula sheet pdf
 

002 & 003 Dna Methyl And Human Disease 14

  • 1. REVIEWS DNA METHYLATION AND HUMAN DISEASE Keith D. Robertson Abstract | DNA methylation is a crucial epigenetic modification of the genome that is involved in regulating many cellular processes. These include embryonic development, transcription, chromatin structure, X chromosome inactivation, genomic imprinting and chromosome stability. Consistent with these important roles, a growing number of human diseases have been found to be associated with aberrant DNA methylation. The study of these diseases has provided new and fundamental insights into the roles that DNA methylation and other epigenetic modifications have in development and normal cellular homeostasis. CPG ISLAND With the completion of the Human Genome Project, transposable elements (LINES), short interspersed A genomic region of ~1 kb that we have a nearly complete list of the genes needed to transposable elements (SINES) and endogenous retro- has a high G–C content, is rich produce a human. However, the situation is far more viruses)6. CPG ISLANDS, particularly those associated with in CpG dinucleotides and is complex than a simple catalogue of genes. Of equal promoters, are generally unmethylated, although an usually hypomethylated. importance is a second system that cells use to determine increasing number of exceptions are being identified7,8. when and where a particular gene will be expressed dur- DNA methylation represses transcription directly, by ing development. This system is overlaid on DNA in the inhibiting the binding of specific transcription factors, form of epigenetic marks that are heritable during cell and indirectly, by recruiting methyl-CpG-binding pro- division but do not alter the DNA sequence. teins and their associated repressive chromatin remod- The only known epigenetic modification of DNA in elling activities (see online supplementary information mammals is methylation of cytosine at position C5 in S1 (table)). Little is known about how DNA methyla- CpG dinucleotides1. By contrast, the other main group tion is targeted to specific regions; however, this prob- of epigenetic modifications — the post-translational ably involves interactions between DNMTs and one or modification of histones — shows a high level of diver- more chromatin-associated proteins3. sity and complexity2. The mammalian DNA methyla- Properly established and maintained DNA methyla- tion machinery is composed of two components, the tion patterns are essential for mammalian development DNA methyltransferases (DNMTs), which establish and and for the normal functioning of the adult organism. maintain DNA methylation patterns, and the methyl- DNA methylation is a potent mechanism for silencing CpG binding proteins (MBDs), which are involved in gene expression and maintaining genome stability in ‘reading’ methylation marks. An in-depth discussion the face of a vast quantity of repetitive DNA, which can of these proteins is not provided here, but their main otherwise mediate illegitimate recombination events features are given in online supplementary informa- and cause transcriptional deregulation of nearby genes. tion S1 (table) (and were recently reviewed by REFS 3,4). Embryonic stem cells that are deficient for DNMTs are Department of Biochemistry and Molecular Biology, There is also clear evidence that a DNA demethylase viable, but die when they are induced to differentiate9. Shands Cancer Center, contributes to regulating DNA methylation patterns Mouse knockout studies have shown that Dnmt1 and University of Florida, during embryonic development, although the activity Dnmt3b are essential for embryonic development Box 100245, 1600 S.W. responsible for this has not been identified5. and that mice that lack Dnmt3a die within a few Archer Road, Gainesville, Florida 32610, USA. In normal cells, DNA methylation occurs predomi- weeks of birth10,11. In addition, loss of normal DNA e-mail: keithr@ufl.edu nantly in repetitive genomic regions, including satellite methylation patterns in somatic cells results in loss of doi:10.1038/nrg1655 DNA and parasitic elements (such as long interspersed growth control. NATURE REVIEWS | GENETICS VOLUME 6 | AUGUST 2005 | 597 © 2005 Nature Publishing Group
  • 2. REVIEWS Centromere also contribute to aberrant gene regulation in cancer by TRANSCRIPTIONAL INTERFERENCE or the generation of TSG antisense transcripts17. Loss of genomic methylation Hypermethylated CpG island pericentromeric (hypomethylated) is a frequent and early event in cancer, and correlates heterochromatin with disease severity and metastatic potential in many tumour types18. Hypomethylation Hypermethylation Gene-specific effects of hypomethylation also occur. For example, the melanoma antigen (MAGE) family of Mitotic recombination, Transcriptional repression, cancer–testis genes, which encode tumour antigens of genomic instability loss of TSG expression DNA repeat unknown function, are frequently demethylated and re-expressed in cancer19. Demethylation accompanied Methylated by increased expression has been reported for the S100 Unmethylated Cancer calcium binding protein A4 (S100A4) gene in colon cancer20, the serine protease inhibitor gene SERPINB5 Figure 1 | DNA methylation and cancer. The diagram shows a representative region of (also known as maspin) in gastric cancer21, and the puta- genomic DNA in a normal cell. The region shown contains repeat-rich, hypermethylated tive oncogene γ-synuclein (SNCG) in breast and ovarian pericentromeric heterochromatin and an actively transcribed tumour suppressor gene (TSG) cancers22. Global demethylation early in tumorigenesis associated with a hypomethylated CpG island (indicated in red). In tumour cells, repeat-rich might predispose cells to genomic instability and further heterochromatin becomes hypomethylated and this contributes to genomic instability, a genetic changes, whereas gene-specific demethylation hallmark of tumour cells, through increased mitotic recombination events. De novo methylation could be a later event that allows tumour cells to adapt to of CpG islands also occurs in cancer cells, and can result in the transcriptional silencing of growth-regulatory genes. These changes in methylation are early events in tumorigenesis. their local environment and promotes metastasis. Research on genome-wide demethylation in can- cer cells has been largely overshadowed by studies of The importance of DNA methylation is empha- gene-specific hypermethylation events, which occur sized by the growing number of human diseases that concomitantly with the hypomethylation events dis- are known to occur when this epigenetic information cussed above. Aberrant hypermethylation in cancer is not properly established and/or maintained, and usually occurs at CpG islands (FIG. 1), most of which there is increasing interest in developing ways of phar- are unmethylated in normal somatic cells8,23, and the macologically reversing epigenetic abnormalities12. resulting changes in chromatin structure (such as Further interest in this area comes from important histone hypoacetylation) effectively silence transcrip- new evidence that concerns the regulation of DNA tion. Indeed, a distinct subset of many tumour types methylation, indicating interactions between DNA has a CpG-island-methylator phenotype, which has methylation and the histone modification machin- been defined as a 3–5 fold increase in the frequency ery, and showing a potential role of small RNAs13. of aberrant hypermethylation events24. Genes involved Here, I discuss a diverse group of diseases associated in cell-cycle regulation, tumour cell invasion, DNA with abnormalities of DNA methylation, studies of repair, chromatin remodelling, cell signalling, tran- which have provided insights into how DNA meth- scription and apoptosis are known to become aber- ylation patterns are regulated and the pathological rantly hypermethylated and silenced in nearly every consequences of their disruption. I also propose two tumour type (see online supplementary information related models for how a single common defect might S2 and S3 (tables)). This provides tumour cells with be able to give rise to the diverse array of defective a growth advantage, increases their genetic instability DNA methylation patterns in human diseases. It (allowing them to acquire further advantageous genetic should be noted that I focus only on those diseases for changes), and allows them to metastasize. In tumours which defective DNA methylation patterns have been with a well-defined progression, such as colon cancer, demonstrated. For this reason, I do not discuss Rett aberrant hypermethylation is detectable in the earliest TRANSCRIPTIONAL INTERFERENCE syndrome, which involves a defect in the machinery precursor lesions, indicating that it directly contributes Repression of one that reads methylation marks14. to transformation and is not a late event that arises transcriptional unit by another from genetic alterations25. such unit that is linked in cis. DNA methylation and cancer Use of RESTRICTION LANDMARK GENOMIC SCANNING to ana- RESTRICTION LANDMARK A link between DNA methylation and cancer was first lyze the methylation status of 1,184 CpG islands from GENOMIC SCANNING demonstrated in 1983, when it was shown that the 98 tumour samples showed that de novo methylation of (RLGS). A genome-wide genomes of cancer cells are hypomethylated relative CpG islands is widespread in tumour cells. In this study, method for analyzing the DNA to their normal counterparts15. Hypomethylation in the extent of methylation varied between individual methylation status of CpG tumour cells is primarily due to the loss of methylation tumours and tumour types, and an average of 608 CpG islands. Radiolabelled fragments obtained by digestion from repetitive regions of the genome6 (FIG. 1), and the islands were aberrantly hypermethylated26. Because with NotI (a methylation- resulting genomic instability is a hallmark of tumour the hypermethylation of CpG islands is relatively sensitive restriction enzyme) cells. Rearrangements that involve the large block of rare in normal cells, is an early event in transforma- are separated by two- pericentromeric heterochromatin on chromosome 1, tion, and robust assays can detect methylated DNA in dimensional gel electrophoresis, allowing differentiation for example, are among the most frequent genomic bodily fluids, it represents a good potential biomarker between methylated and instabilities in many tumour types16. Reactivation of for early cancer detection27. The underlying cause of unmethylated regions. transposon promoters following demethylation might methylation defects in cancer remains unknown, but 598 | AUGUST 2005 | VOLUME 6 www.nature.com/reviews/genetics © 2005 Nature Publishing Group
  • 3. REVIEWS imprinted loci, regulates the ability of distant ENHANC Box 1 | A comparison of the CTCF and BORIS proteins 30 ERS to access promoters . In several ICRs, in vitro and CTCF (CCCTC-binding factor) in vivo studies have shown that CTCF binds only to the • Contains an 11 zinc finger (ZF) DNA-binding region30. unmethylated parental allele31,32, providing an elegant • Binds diverse DNA sequences, including most ICRs and many CpG islands30,137. means for ICR-regulated genes to be expressed in • Binding is methylation sensitive (if binding sites contain CpGs)31. an allele-specific manner. CTCF binding might also be essential to protect DMRs from de novo methyla- • Has insulator function (regulates access of enhancers to promoters)30,70. tion33. • Has boundary element function (blocks the spread of heterochromatin)30,70. Loss of imprinting (LOI) is the disruption of • Protects regions from DNA methylation138,139. imprinted epigenetic marks through gain or loss • Is ubiquitously expressed, except in spermatocytes126. of DNA methylation, or simply the loss of normal • Overexpression inhibits cell proliferation140. allele-specific gene expression28. Studies of LOI have provided important insights into the role of methyla- • Gene is located at 16q22, a region of frequent loss of heterozygosity in cancer, and is tion in imprinting, and have highlighted the impor- often mutated in cancer141. tance of CTCF as a regulator of both imprinted gene BORIS expression and DNA methylation patterns. Below, I • Contains an 11 ZF DNA-binding region that has high similarity to the CTCF ZF discuss several human diseases for which the role of region30. aberrant genomic imprinting has been particularly • Presumed to show significant overlap with CTCF binding sites. well established. Features of these diseases are sum- • Methylation sensitivity of binding unknown. marized in the online supplementary information S3 (table). Importantly, imprinted DNA methylation • Insulator function unknown. patterns are frequently disrupted by in vitro manipula- • Boundary element function unknown tion of embryos in both animals and humans34–36, and • Methylation protection function unknown the use of assisted reproductive technologies has been • Expression is normally restricted to testis (BORIS is a member of the cancer–testis associated with an increased risk for several imprinting gene family), and is reactivated in cancers126. diseases37 BOX 2. • Overexpression promotes cell proliferation30. LOI in cancer. LOI of a growth-promoting imprinted • Gene is located at 20q13, a region that is frequently amplified in cancer. gene, leading to activation of the normally silent allele, results in abnormally high expression of the gene prod- possible mechanisms are discussed at the end of this uct and gives cells a growth advantage. The imprinted review. DNA methylation and cancer are also related IGF2/H19 locus (FIG. 2a) encodes both IGF2, an autocrine through the loss of imprinted methylation patterns in growth factor with an important role in many types of many tumours, as discussed below. cancer, and H19, a non-coding RNA of unknown func- tion with growth suppressive properties38. IGF2 and H19 DNA methylation and imprinting disorders are normally expressed from the paternal and maternal Genomic imprinting — a brief overview. Genomic alleles, respectively, and relaxed silencing of the mater- imprinting is defined as an epigenetic modification nal IGF2 allele results in increased IGF2 expression and of a specific parental chromosome in the gamete or reduced H19 expression. LOI of IGF2 is the most com- zygote that leads to differential expression of the two mon LOI event across the widest range of tumour types, alleles of a gene in the somatic cells of the offspring28. including colon, liver, lung, and ovarian cancer, as well Differential expression can occur in all cells, or in as Wilms’ tumour — the embryonic kidney cancer in specific tissues or developmental stages. About 80 which LOI was first discovered39,40. genes are known to be imprinted, and this number is Additional evidence for the role of LOI at IGF2/H19 continually increasing. in cancer came from a recent study that used a mouse Differential allele-specific DNA methylation is one model. Animals with LOI showed a twofold increase of the hallmarks of imprinted regions and is usually in intestinal tumours, and normal intestinal epithe- localized to regions termed differentially methylated lium was shown to be less well differentiated than in regions (DMRs). DMRs include imprinting con- wild-type mice41. LOI defects in IGF2/H19 vary with trol regions (ICRs), which control gene expression tumour type. For example, in Wilms’ tumour42 and within imprinted domains, often over large distances. colorectal cancer43, the regulatory region ICR1 on the Whereas DMRs can be reprogrammed during devel- maternal allele becomes methylated de novo, which opment, differential methylation in ICRs is generally inhibits CTCF binding and thereby allows enhancers established in germ cells and maintained throughout upstream of H19 to access the IGF2 promoter on both ENHANCER development29. alleles (FIG. 2a). An alternative mechanism has also been A regulatory DNA element that CCCTC-binding factor (CTCF), an 11-zinc-finger proposed, in which DMRs upstream of IGF2 become usually binds several protein that binds to highly divergent sequences using demethylated44. transcription factors and can different combinations of zinc fingers, is an important LOI of growth-inhibitory imprinted genes, through activate transcription from a promoter at great distance and regulator of imprinted gene expression BOX 1). CTCF silencing of the one normally active allele, might also in an orientation-independent is a chromatin insulator that separates the genome result in deregulated cell growth. An example of this manner. into independent functional domains or, in the case of involves the maternally expressed cyclin-dependent NATURE REVIEWS | GENETICS VOLUME 6 | AUGUST 2005 | 599 © 2005 Nature Publishing Group
  • 4. REVIEWS kinase inhibitor 1C (CDKN1C) (also known as p57KIP2) several imprinted genes (FIG. 2a). This large region gene, which encodes a cyclin-dependent kinase inhibi- consists of two independently regulated imprinted tor that mediates G1/S-phase arrest. In Wilms’ tumour, domains39,40,48. The more telomeric domain contains LOI of CDKN1C occurs in ~10% of tumours (LOI of IGF2 and H19, followed by an intervening region IGF2/H19 occurs in 70% of these tumours)28,45. Other with several genes that do not seem to be imprinted. imprinted genes that undergo LOI include the RAS- The second, more centromeric, domain contains the related gene DIRAS3 (also known as ARHI) in breast and maternally expressed gene KCNQ1 (potassium volt- ovarian cancer46 and the mesoderm specific transcript age-gated channel, KQT-like subfamily, member 1; also homologue (MEST) gene in breast, lung, and colon can- known as KvLQT1), the paternally expressed KCNQ1 cer47. Although the number of genes that show LOI in antisense transcript KCNQ1OT1 (KCNQ1 overlap- cancer seems small from this discussion, the frequency ping transcript 1; also known as LIT1), and several of such alterations is probably similar in extent to the other maternally expressed genes, including CDKN1C. DNA methylation abnormalities of non-imprinted genes ICR1, which is located upstream of H19, controls discussed in the previous section. Examples of LOI seem differential expression of the IGF2/H19 domain, limited only because relatively few of the estimated total and there are also several DMRs upstream of IGF2; number of imprinted genes have been identified and ICR2 lies at the 5′-end of KCNQ1OT1 REFS 43,48,49. even fewer have been well characterized. In normal cells, the paternal and maternal alleles of ICR1 and ICR2, respectively, are methylated, and both Beckwith–Wiedemann syndrome (BWS). BWS is contain binding sites for CTCF31,50. predominantly a maternally transmitted disorder and Several perturbations of this complex genomic involves fetal and postnatal overgrowth and a predispo- region are associated with BWS. These include paternal sition to embryonic tumours, such as Wilms’ tumour UNIPARENTAL DISOMY (UPD) (resulting in increased IGF2 (see online supplementary information S3 (table)). and reduced CDKN1C expression), LOI of KCNQ1OT1 The BWS locus, at 11p15.5, spans ~1 Mb and includes in ~50% of cases that do not involve UPD, and LOI at Box 2 | DNA-methylation abnormalities and assisted reproductive technology A link between assisted reproductive technology and birth defects It has been estimated that one in ten individuals of reproductive age are infertile. The use of assisted reproductive technologies (ARTs), such as intracytoplasmic sperm injection and in vitro fertilization, has therefore been increasing, and children conceived by ART now account for 1–3% of all births in some Western countries. Although the vast majority of these children develop normally, recent studies have raised concerns that these procedures might lead to epigenetic defects. ART is associated with an increase in multiple births and low birth weight, and this risk has been known for some time142. Although the numbers are small and larger studies are needed, ARTs have been linked to a 3–6-fold increase in the occurrence of Beckwith–Wiedemann syndrome (BWS) and Angelman syndrome (AS)37,143,144 (see online supplementary information S3 (table)). An epigenetic basis Germ cell and embryonic development are two periods that are characterized by profound changes in DNA methylation patterns, also known as epigenetic reprogramming. Primordial germ cells are globally demethylated as they mature (including at imprinted regions), and then become methylated de novo during gametogenesis, which is also the time at which most DNA methylation imprints are established. The second period of pronounced change occurs after fertilization, where the paternal genome is rapidly and actively demethylated, followed by passive replication-dependent demethylation of the maternal genome (here, imprinting marks seem to resist demethylation). A wave of de novo methylation once again establishes the somatic-cell pattern of DNA methylation following implantation145,146. The increased incidence of BWS and AS associated with ART all arise from epigenetic, rather than genetic, defects. In most BWS cases studied, loss of imprinting (LOI) at KCNQ1OT1 was observed, and in AS cases the defect was found to be LOI at SNRPN (loss of maternal methylation, see FIG. 2a,b)37,143. Epigenetic mechanisms and imprinted genes also seem to be involved in the regulation of birth weight in mouse models; therefore, the disruption of imprints at currently unknown loci following ART in humans might also contribute to the increased incidence of this problem147. Low birth weight is a serious issue in its own right as it contributes to long-term adverse health effects such as cardiovascular disease148. Causes of epigenetic defects There has been a trend in fertility clinics towards extended culture time of embryos and transfer of blastocysts, rather than earlier cleavage-stage embryos. This allows selection of the most robust embryos, proper temporal synchronization of the embryo and the uterus, and the possibility of maintaining a high pregnancy rate while reducing the risk of multiple pregnancy (because fewer embryos need to be transferred)35. However, several studies have demonstrated that in vitro culture conditions have notable effects on imprinting marks, gene expression and developmental potential in mouse models34,149. Whether the culture conditions used for human embryos provide all of UNIPARENTAL DISOMY the necessary factors for proper epigenetic control is unknown. It is also possible that the epigenetic defects might be Inheritance of a chromosome or the cause of the infertility itself and are present in the gametes of the mother and/or father. Therefore, rather than ARTs chromosome region from a causing epigenetic defects directly, they might simply be unmasking an existing defect. single parent. 600 | AUGUST 2005 | VOLUME 6 www.nature.com/reviews/genetics © 2005 Nature Publishing Group
  • 5. REVIEWS a BWS 11p15.5 Cancer LOI BWS breakpoint region (CDKN1C) ICR1 ICR2 Cancer LOI (IGF2) Centromere Telomere CTCF CTCF Enhancer NAP1L4 CDKN1C CTCF KCNQ1 ASCL2 INS CTCF H19 TSSC3 TSSC5 PHEMX TH IGF2 L23 CD81 KNCQ1OTI TSSC4 DMRs 0,1,2 ICR2 ICR1 b PWS/AS 15q11–q13 PWS region AS region Centromere PWS ICR UBE3A- Telomere 1 2 3 AS NDN SNURF/SNRPN UBE3A GABRG3 OCA2 HERC2 MKRN3 ATP10C GABRB3 GABRA5 MAGE-L2 AS ICR c AHO/PHP-Ia/PHP-Ib 20q13 LOI in PHP-Ib Centromere patients Telomere GNAS NESP55-AS (Gsα) NESP55 XLα S Exon 1A Exon 1 Exons 2–13 Paternal expression Unmethylated Promoter (not imprinted) d TNDM 6q24 CpG island Maternal expression Methylated ICR or DMR Telomere ZAC-AS Centromere FK506 ICR HYMAI PLAGL1 KIAA0680 Figure 2 | Regions involved in disease-associated genomic imprinting defects. Sense and antisense transcripts are indicated by arrows above and below the genes, respectively. In each panel, the methylation status of the paternal allele is indicated on the top of the chromosome, and that of the maternal allele on the bottom of the chromosome. a | The Beckwith–Wiedemann syndrome (BWS) locus and cancer-associated loss of imprinting (LOI). The region can be divided into two imprinted domains (TSSC3 to KCNQ1, and INS to H19) that are regulated independently. CTCF binds to the unmethylated alleles of both imprinting control regions, ICR1 and ICR2. Cancer-associated LOI commonly occurs within the IGF2/H19 region (resulting in increased IGF2 and decreased H19 expression) and less commonly at CDKN1C. BWS arises from several defects in this region, including LOI at ICR1, ICR2 and the differentially methylated regions (DMRs 0, 1 and 2) upstream of IGF2; paternal UPD; and mutations and translocations on the maternal allele. b | The Prader–Willi syndrome (PWS)/Angelman syndrome (AS) locus. The ICR for this locus has a bipartite structure, with the more centromeric component acting as the AS ICR and the more telomeric region, which contains the promoter of the SNURF/SNRPN gene, acting as the PWS ICR. SNURF/SNRPN produces an extremely long and complicated transcript that encodes not only the SNURF/SNRPN (1), and IPW transcripts (2), but also several small nucleolar RNAs (snoRNAs) (3), which actually flank IPW (not shown), that are processed from its introns. This transcript is also thought to inhibit expression of UBE3A and ATP10C on the paternal allele through an antisense mechanism. PWS is a multi-gene disorder that arises from the loss of expression of genes in this region from the paternal allele. AS arises from loss of maternally expressed UBE3A. c | The GNAS locus, which is the site of the molecular defects that underlie Albright hereditary osteodystrophy (AHO), pseudohypopar- athyroidism Ia (PHP-Ia) and PHP-Ib. Four alternative first exons (those of NESP55, XLα s, exon 1A and exon 1), each driven by its own promoter, splice onto a common set of downstream exons (2–13). The promoters of NESP55, XLα s/NESP55-AS and exon 1A are differentially methylated and imprinted. Exon 1 (Gsα ) demonstrates preferential maternal expression in only certain tissues (such as the renal proximal tubules); however, it is not differentially methylated. The location of the LOI defect at exon 1A in PHP-Ib patients is shown. AHO and PHP-Ia arise from paternal and maternal transmission of Gsα mutations, respectively, while PHP-Ib is due to LOI at exon 1A and, in some cases, also at the upstream DMRs. d | The transient neonatal diabetes mellitus (TNDM) locus. A differentially methylated CpG island upstream of HYMAI acts as an ICR for this region, which contains three imprinted genes. In TNDM patients, DNA methylation is lost from the maternal allele and expression of PLAGL1 is upregulated. CDKN1C, cyclin- dependent kinase inhibitor 1C; HYMAI, hydatidiform mole associated and imprinted; IGF2, insulin-like growth factor 2; INS, insulin; IPW, imprinted in Prader–Willi syndrome; KCNQ1, potassium voltage-gated channel, KQT-like subfamily, member 1; NESP55, neuroendocrine secretory protein 55; PLAGL1, pleiomorphic adenoma gene-like 1; SNRPN, small nuclear ribonucleoprotein polypeptide N; SNURF, SNRPN upstream reading frame; TSSC3, tumour-suppressing STF cDNA 3 (also known as PHLDA2); UBE3A, ubiquitin protein ligase E3A. NATURE REVIEWS | GENETICS VOLUME 6 | AUGUST 2005 | 601 © 2005 Nature Publishing Group
  • 6. REVIEWS IGF2 in ~20% of these cases (both resulting in biallelic from Snurf/Snrpn to Ube3a60 contribute to aspects expression of the respective genes). Mutations and of PWS, but further models are needed to define the translocations that involve the maternal allele account contribution of all paternally expressed genes in this for the remaining cases. LOI at ICR2 involves loss of region. The finding that a DNA methylation inhibitor maternal-allele-specific DNA methylation, whereas restores expression of the silent maternal gene copy of at ICR1 the defect is usually hypermethylation of the SNURF/SNRPN is intriguing, and indicates that such maternal allele, as seen in most Wilms’ tumours49,51. inhibitors might be useful for PWS treatment61. Defects in the centromeric imprinted domain (silenc- ing of maternally expressed genes) are thought to give Angelman syndrome (AS). AS occurs in ~1 in 15,000 rise predominantly to the BWS phenotype (anatomi- births and its main characteristics include mental cal malformation) whereas defects in the telomeric retardation, speech impairment and behavioural imprinted domain (activation of maternally repressed abnormalities (see online supplementary information IGF2) could be the main driving force for tumorigen- S3 (table))55,62. The AS defect lies within the imprinted esis 52. Hypomethylation of the maternal ICR2 and acti- domain at 15q11–q13 and is due to the loss of mater- vation of KCNQ1OT1 causes a marked downregulation nally expressed genes (FIG. 2b). Unlike PWS, however, of genes that are centromeric to this, such as CDKN1C; AS arises from the loss of a single gene — maternally however, the molecular mechanism of this regulation expressed UBE3A — which is imprinted only in the is not understood53. One model is that ICR2 functions brain and encodes an E3 ubiquitin ligase involved in in a similar way to ICR1 — that is, it might regulate, the UBIQUITINPROTEASOME DEGRADATION PATHWAY . through binding of CTCF, the ability of an enhancer The most common molecular defects that give rise to access the CDKN1C and KCNQ1OT1 promoters to AS are maternally-derived deletions of 15q11–q13 differentially on the two alleles49. (~65–70% of cases), paternal UPD (~5% of cases), maternal UBE3A mutations (~10% of cases, and up to ~40% in one study), and imprinting defects (~5% Prader–Willi syndrome (PWS). PWS occurs in ~1 of cases). About 10% of cases arise from an unknown in 20,000 births and is characterized by a failure to defect62. Analysis of samples from AS patients with thrive during infancy, hyperphagia and obesity during microdeletions allowed the AS ICR to be mapped to an early childhood, mental retardation, and behavioural 880 bp region ~35 kb upstream of SNURF/SNRPN63. problems (see online supplementary information S3 Imprinting defects involved in AS include loss of mater- (table))54. The molecular defect is complex and involves nal DNA methylation or maternal ICR deletion, and a ~2 Mb imprinted domain at 15q11–q13 that con- patients with these defects tend to have the mildest AS tains both paternally and maternally expressed genes phenotypes62. Important unanswered questions related (FIG. 2b). One maternally expressed gene, ubiquitin to AS biology include the nature of the molecular defects protein ligase E3A (UBE3A), is imprinted only in the in the ~10% of cases that have no identifiable UBE3A brain and is discussed in the next section. mutation and the role of the nearby gene ATP10C PWS arises from loss of paternally expressed genes in (ATPase, class V, type 10A), which is often co-deleted this region, although no single gene has been shown to with UBE3A, in modulating the AS phenotype. cause PWS when its expression is lost55. The analysis of samples from PWS patients with microdeletions allowed Albright hereditary osteodystrophy (AHO), pseudo- the PWS ICR to be mapped to a segment of ~4 kb that hypoparathyroidism type Ia (PHP-Ia) and PHP-Ib. spans the first exon and promoter of the small nuclear These three diseases are related because they all arise BALANCED TRANSLOCATION ribonucleoprotein polypeptide N (SNRPN)/SNRPN from defects at a complex imprinted locus on chromo- A condition in which two pieces upstream reading frame (SNURF) locus56. In normal some 20q13 called GNAS, which encodes the α subunit of chromosomal material have cells, the 5′-end of the ICR, which is needed for mater- (Gsα) of the heterotrimeric GTP-binding protein Gs switched places, but the correct nal gene expression and is involved in Angelman syn- (FIG. 2c; and see online supplementary information number of chromosomes has been maintained. drome (discussed below), is methylated on the maternal S3(table)). AHO is a paternally inherited disorder that allele55,57. The 3′ end of the ICR is required for expression occurs within PHP-Ia pedigrees, and involves mental UBIQUITINPROTEASOME of the paternally expressed genes (the PWS region shown retardation and subcutaneous ossification. DEGRADATION PATHWAY in FIG. 2b) and is also the origin of the extremely long PHP-Ia is maternally transmitted in a dominant Degradation pathway in which a protein that has been post- SNURF/SNRPN transcript. The maternally expressed manner and patients show all the AHO symptoms, translationally modified with genes are not differentially methylated, and their silenc- plus resistance to the peripheral action of several several ubiquitin polypeptides ing on the paternal allele is probably regulated by an hormones involved in activating pathways coupled to is targeted for destruction to the antisense RNA generated from SNURF/SNRPN58. Gs. Individuals with PHP-Ib, which is also maternally proteasome, a large cytosolic The most common molecular defects in PWS are inherited, show normal Gsα activity and only renal protein complex with several proteolytic activities. paternal deletions spanning the 15q11–q13 region parathyroid hormone resistance, with no other endo- (65–70% of cases) and maternal UPD (~20–30% of crine deficiencies64. CHROMOGRANINS cases). Paternally derived microdeletions of the ICR, The GNAS locus encodes three proteins and an A group of acidic, soluble, LOI at the ICR (de novo methylation of the pater- untranslated RNA from four alternative first exons secretory proteins that are produced by neurons and nal allele), and BALANCED TRANSLOCATIONS that disrupt that splice onto common exons 2–13 (FIG. 2c). NESP55, neuroendocrine cells. SNURF/SNRPN, account for the remainder51. Mouse generated from the most centromeric of the first exons, knockout studies indicate that Ndn59 and the region encodes a CHROMOGRANIN-like protein, XLαs encodes a 602 | AUGUST 2005 | VOLUME 6 www.nature.com/reviews/genetics © 2005 Nature Publishing Group
  • 7. REVIEWS Golgi-specific Gsα isoform, and exon 1 encodes Gsα. Three mechanisms give rise to TNDM: paternal UPD Transcripts derived from exon 1A do not seem to be of chromosome 6, paternal duplications of 6q24, and translated. Each alternative first exon is driven from its LOI at the TNDM CpG-island-associated ICR, which own promoter, and the first three promoters are dif- leads to increased PLAGL1 expression67. TNDM patients ferentially methylated. NESP55 is maternally expressed who lack chromosomal abnormalities demonstrate LOI and its promoter region is methylated on the paternal of the ICR, specifically loss of DNA methylation from allele. Gsα (exon 1) is preferentially expressed from the the maternal allele68. Because the methylated ICR acts as maternal allele, although its promoter region seems a repressor of PLAGL1 transcription, LOI has the same to be unmethylated on both alleles. Gsα is imprinted consequences as paternal UPD and 6q24 duplication. only in certain hormone-responsive tissues, such as the The TNDM locus has a much simpler organiza- renal proximal tubules, which is the probable cause of tion than those that are involved in BWS, PWS and the tissue-specific effects of these disorders. By con- AHO/PHP. However, this further emphasizes that most trast, XLαs, a NESP55 antisense transcript, and exon imprinted genes are found in clusters under the control 1A are all paternally expressed, and their promoters of regulatory regions that are often distant from the are methylated on the maternal allele64. genes themselves, and that this control is dependent The molecular defects that underlie AHO, PHP-Ia, on allele-specific DNA methylation. and PHP-Ib, are well characterized. AHO and PHP-Ia arise from the paternal and maternal transmission of A wider role of imprinting in disease. These examples Gsα mutations, respectively64. By contrast, PHP-Ib of diseases associated with aberrant imprinting are involves LOI at exon 1A (loss of maternal-specific probably only the tip of the iceberg. Unlike the global methylation), which occurs in all cases. Interestingly, genomic hypomethylation and widespread CpG island it was recently demonstrated that sporadic PHP-Ib hypermethylation seen in cancer and cancer-associated (the most common form) also involves imprinting LOI, which probably affects many imprinted regions, defects at the upstream DMRs, in the form of biallelic the DNA methylation defects in imprinting disorders methylation at NESP55 and hypomethylation at the are subtle. Numerous other pathologies demonstrate NESP antisense (NESPAS)/XLαs promoter region. By parent-of-origin effects or result from UPD, indicating contrast, familial PHP-Ib is associated with maternally that imprinted genes might be involved69. Interestingly, transmitted deletions within the nearby syntaxin 16 accumulating evidence from the diseases described (STX16) gene and LOI at the exon 1A DMR only65. above indicates that loss of normal CTCF function has NESP55, XLαs and exon 1A are probably regulated by an important role in imprinting disorders. Studies of antisense transcription and/or differential promoter the mechanism by which CTCF regulates DNA meth- methylation. However it is still unclear how G s α ylation patterns in imprinted regions also indicate that exon 1 is regulated, as it is not differentially methylated it could have a much broader role in regulating DNA and no antisense transcript has been identified. The methylation patterns throughout the genome than is deleted region in STX16 might contain a cis-element currently appreciated70. that acts over a large distance to establish imprinting at the exon 1A DMR65. LOI at exon 1A (and biallelic Methylation and repeat-instability diseases Gsα expression) also occurs in a subset of growth- DNA methylation defects have been linked to members hormone-secreting pituitary adenomas, supporting of a large group of human diseases that are associated the idea that imprinting of each GNAS promoter is with repeat instability. Expansion of trinucleotide independently regulated66. repeats (TNRs) during gametogenesis leads to muta- tion or silencing of associated genes, with pathological Transient neonatal diabetes mellitus (TNDM). TNDM consequences71. The causes of TNR instability remain is a rare form of diabetes (occuring in ~1 in 400,000 largely unknown, although DNA polymerase slippage, births) that presents during the first few weeks after DNA repair mechanisms, transcription, nucleosome birth. Remission usually occurs after 3–6 months, but positioning and aberrant OKAZAKIFRAGMENT process- a significant proportion of patients develop diabetes ing within the repeat have been proposed72. Some of later in life67. The TNDM locus on chromosome 6q24 these diseases involve expansions of non-methylatable spans ~400 kb and contains three imprinted genes repeats, such as CAG in Huntington’s disease (HD) and — the paternally expressed genes hydatidiform mole several forms of spinocerebellar ataxia (SCA1, 2, 3, 6, associated and imprinted (HYMAI) and pleiomorphic 7, and 17), CTG in myotonic dystrophy type 1 (DM1) adenoma gene-like 1 (PLAGL1; also known as ZAC or and SCA8, and GAA in Friedreich’s ataxia (FRDA)71. LOT1), and the maternally expressed PLAGL1 anti- Expansion of methylatable CGG repeats occurs at sense transcript (ZAC-AS) (FIG. 2d). HYMAI gives rise to several sites that are known or likely to be involved an apparently untranslated RNA of unknown function, in human disease, and increased DNA methylation whereas PLAGL1 encodes a zinc finger protein that is a is thought to have a role in pathogenesis. In contrast putative tumour suppressor and transcriptional regula- to the TNR expansion diseases, facioscapulohumeral tor involved in modulating the cell cycle and apoptosis. muscular dystrophy (FSHD) is due to the contraction OKAZAKI FRAGMENTS Short pieces of DNA that are A putative ICR within the CpG island at the 5′-end of of a much larger repeat accompanied by hypomethyla- synthesized on the lagging HYMAI is methylated on the maternal allele and acts as tion. These diseases offer insights into possible mecha- strand at the replication fork. a transcriptional repressor when methylated68. nisms for the targeting of de novo DNA methylation. NATURE REVIEWS | GENETICS VOLUME 6 | AUGUST 2005 | 603 © 2005 Nature Publishing Group
  • 8. REVIEWS a FRAXA Xq27.3 CGG repeat The molecular basis of FRAXA lies in the highly ATG polymorphic CGG repeat within the 5′-untranslated 5′ 3′ region of FMR1 (FIG. 3a). Normally, 6–52 copies of the repeat are present, which increases to 52–200 copies TATA 5′ UTR FMR1 gene in the premutation state (now recognized as a separate disease called fragile X tremor/ataxia syndrome76), Repeat expansion and to more than 200 repeats in individuals with FRAXA73,77. Affected individuals show de novo methy- CGG repeat lation of the expanded CGG repeat and silencing of ATG FMR1 transcription77. DNA methylation is clearly an 5′ 3′ important mediator of silencing as DNMT inhibitors Targeting to DNA hairpins? reactivate transcription of the full-mutation allele, FMR1 hairpin RNA although FMRP is not produced78. DNMT1 Methylation? Other folate-sensitive fragile sites. The FRAXA locus RNAi Dicer machinery is one of more than 20 folate-sensitive fragile sites in the genome. CGG repeat expansion and hypermeth- ylation have been documented for several other such b FSHD 4q35 sites associated with genes (FRAXE, FRAXF, FRA10A, Repressor FRA11B and FRA16A) although only FRAXE has been ? ? ? complex firmly linked to human disease (nonspecific X-linked mental retardation)74,79. It is reasonable to assume 5′ 3′ that all folate-sensitive fragile sites similarly occur at ANT1 FRG1 FRG2 D4Z4 (>11) CGG repeat expansions that are accompanied by DNA methylation. All fragile sites characterized so far that Repeat contraction have a genomic structure similar to the FMR1 gene (a and hypomethylation CGG repeat in the untranslated region and a nearby ? ? ? CpG island) show hypermethylation of both regions in the repeat-expanded state, and transcription of the 5′ 3′ associated gene is silenced74. ANT1 FRG1 FRG2 D4Z4 (1–10) Facioscapulohumeral muscular dystrophy. FSHD Figure 3 | DNA methylation and diseases associated with repeat instability. is an autosomal dominant disease that affects ~1 in a | Trinucleotide repeat instability associated with aberrant DNA methylation in FRAXA. A schematic representation of the 5′-end of the FMR1 gene is shown. The CGG repeat in 20,000 individuals (see online supplementary infor- FMR1 lies in the 5′ untranslated region (UTR), which is embedded in a hypomethylated CpG mation S3 (table)). The FSHD locus maps to 4q35 and island in the normal state. In fully affected individuals with large repeat expansions, the repeat the underlying molecular defect is well established, and CpG island become de novo methylated, resulting in gene silencing. Models for how the involving contraction of the polymorphic 3.3 kb D4Z4 repeat is targeted for methylation include the formation of unusual DNA structures that attract repeat. Unaffected individuals have 11–150 copies of DNA methyltransferase 1 (DNMT1), and the formation of hairpin RNA from the transcribed this repeat, whereas in most FSHD cases the repeat is repeat, which might be cleaved by Dicer and subsequently recruit the RNAi silencing machinery, including DNMTs. b | Facioscapulohumeral muscular dystrophy (FSHD) is caused reduced to 1–10 copies, although some patients (~5%) by contraction of the D4Z4 repeat on chromosome 4q35 and loss of DNA methylation. do not show repeat contraction. The abnormality that is Whereas repeat contraction is not always present in FSHD, repeat hypomethylation is an common to all FSHD patients is loss of DNA methyla- invariant feature. D4Z4 repeat contraction and hypomethylation are believed to cause the tion from D4Z4, indicating that aberrant hypomethy- abnormal expression of adjacent genes (FRG2 is considered a good candidate) or genes in lation is the main causal factor80. However, the genes other regions of the genome. One model has proposed that a repressor complex binds to the affected by this hypomethylation are unknown. repeats and regulates the expression of nearby genes, although the role of DNA methylation Most studies have focused on the idea that con- was not addressed in this study. ANT1, solute carrier family 25, also known as SLC25A4; FRG2, FSHD region gene 2 protein. traction and hypomethylation of D4Z4 alters the transcription of one or more genes that flank the repeat. Genes in the vicinity of D4Z4 (FIG. 3b) include Fragile X syndrome (FRAXA). FRAXA, an X-linked the adenine nucleotide translocator SLC25A4 (also disorder, is a common cause of inherited mental known as ANT1), the overexpression of which induces retardation and occurs in ~1 in 4000 males (see online apoptosis (characteristic of dystrophic muscle); FSHD supplementary information S3 (table))73,74. The disease region gene 1 (FRG1) which is ubiquitously expressed maps to the fragile X mental retardation 1 (FMR1) and might be involved in RNA processing; and FRG2 FOLATESENSITIVE gene at Xq27.3, which is also a FOLATESENSITIVE FRAGILE (which is closest to D4Z4), the function of which is FRAGILE SITE SITE. The FMR1 protein (FMRP) is an RNA-binding unknown but which has been shown to induce mor- A region of chromatin that fails protein expressed in many fetal and adult tissues, with phological changes in transfected myoblasts81. Results to compact normally during particularly high levels in brain and testis. It seems to from different laboratories have yielded conflicting mitosis and that can be observed after culturing cells in be involved in regulating translation and mRNA trans- data about the degree of overexpression of these genes media that is deficient in folic port, and is believed to regulate synaptic plasticity by in FSHD muscle, although FRG2 does seem to be a acid and thymidine. controlling translation at synapses75. promising candidate for the disease gene81–83. 604 | AUGUST 2005 | VOLUME 6 www.nature.com/reviews/genetics © 2005 Nature Publishing Group
  • 9. REVIEWS There are also diverse models for how repeat con- number in FSHD could reduce the efficiency of RNAi traction and hypomethylation alter gene expression and targeting, eventually resulting in hypomethylation. give rise to FSHD. One possible mechanism involves The molecular defects that underlie these diseases the binding of a repressor complex to the D4Z4 repeat, are therefore consistent with the growing body of which is reduced as the repeat size decreases, causing evidence that the insulator factor CTCF and/or elevated expression of nearby genes (although this study RNAi are involved in regulating DNA methylation did not address the role of methylation)82. A looping patterns. model has also been proposed, whereby the D4Z4 repeat binds transcriptional regulatory factors and Specific defects of the methylation machinery loops over to distant genes, which might be well outside Human diseases that arise from mutations in the DNA the immediate vicinity of D4Z4. Repeat contraction and methylation machinery have provided important hypomethylation might disrupt the binding of crucial insights into the functional specialization of DNMTs. regulatory proteins or negatively affect the ability of the They have also shown that improper regulation of repeat to interact with other regulatory regions83. DNA methylation can occur in specific cell types, with pathological consequences. Is there a wider role for DNA methylation in diseases of repeat instability? Studies of repeat-instability dis- Systemic lupus erythematosus (SLE). SLE is an autoim- eases demonstrate that repetitive DNA, in the form of mune disease, with diverse clinical manifestations, that both naturally occurring repeats and pathologically is 8–10 times more frequent in females than in males expanded TNRs, is efficiently targeted by the methyla- (~1 in 2000 in the overall population) (see online sup- tion machinery. Why only some expanded TNRs are plementary information S3 (table)). Common features targeted for de novo methylation remains unknown. It in all cases are autoantibody production and the pres- might simply be the availability of a methylatable site ence of antibodies against nuclear components, such as within or near the repeat expansion that makes CGG DNA, chromatin factors and small nuclear ribonuclear rather than CTG or CAG TNRs attractive to the meth- protein particles92. There is also evidence for defective ylation machinery. However, several lines of evidence T-cell function, which provides the main connection indicate that the silencing machinery might be recruited with DNA methylation93. to expanded TNRs, regardless of their sequence. For T cells are believed to drive the autoantibody RNAI example, a CpG island adjacent to the CTG repeat response in SLE, and this has been proposed to result The process whereby double- involved in DM1 (which also contains binding sites for from loss of DNA methylation in these cells. The stranded RNAs are cleaved into CTCF) becomes hypermethylated in the severe con- genomes of SLE T cells are globally hypomethylated 21-23 nucleotide duplexes termed small interfering RNAs, genital form of the disease84,85. In both DM1 and FRDA, (15–20% reduction) and DNMT1 levels are reduced94. leading to inhibition of the associated genes (DMPK and FRDA, respectively) Treatment of normal CD4 T CELLS with the DNA methy- + expression of genes that contain are downregulated, and the expanded repeat adopts a lation inhibitor 5-aza-2′-deoxycytidine renders them a complementary sequence. repressive chromatin structure86. This indicates that autoreactive, and ADOPTIVE TRANSFER of these cells TNR expansions might commonly be targeted for causes an SLE-like disease in mice95. Hypomethylated DICER A ribonuclease that processes silencing at the chromatin level, and that additional T cells are also able to promiscuously kill SYNGENEIC dsRNAs to ~21 nucleotide factors might influence whether DNA methylation MACROPHAGES; this increased apoptosis, combined with siRNAs (for RNAi) or excises also occurs. In addition, DNA methylation stabilizes reduced clearance of apoptotic material, is likely to microRNAs from their hairpin expanded repeats in model systems regardless of have a role in inducing the anti-DNA antibodies that precursors. whether they contain a CpG site87,88. characterise SLE. In addition, several genes relevant to CD4+ T CELL Other aspects that might influence methylation the SLE phenotype are methylated in normal T cells Also known as a helper T cell. targeting include whether the repeats involved are but are demethylated by both 5-aza-2′-deoxycytidine Initiates both antibody transcribed, as they are in FRAXA and DM1, or and in SLE patients96,97. An alternative hypothesis production by B cells and transcribed and translated, as in HD, where there is for the role of DNA methylation in SLE involves the stimulates the activation of other immune cells, such as no evidence of disease-associated hypermethylation89. hypomethylation of endogenous retroviruses (HERV) macrophages, after recognizing Another factor might be the propensity of a repeat to and the re-expression of viral proteins. Antibodies to a portion of a protein antigen form DNA hairpins, which have been shown to be pre- HERV-encoded proteins have been detected in SLE on the surface of an antigen ferred substrates for DNMT190. The finding that FMR1 patients, and peptides derived from these proteins presenting cell. mRNA also forms hairpin structures, and the discovery cause aberrant CD4+ T-cell responses98. ADOPTIVE TRANSFER that they are substrates for the RNAI -associated DICER ribo- Exposure to several DNA methylation inhibitors The process of conferring nuclease provides a possible alternative explanation. also induces a lupus-like disease in humans, provid- immunity to an individual by Components of the histone-modification machinery ing further support for a role of aberrant DNA methy- transferring cells or serum from that cause transcriptional repression are recruited to lation in SLE93,99. DNMT1 is upregulated in normal another individual that has been immunized with a specific repeats by the RNAi pathway, which probably also T cells following stimulation by signals transmitted antigen. recruits the DNA methylation machinery, although through the extracellular signal-regulated kinase this has not been demonstrated directly (see later for (ERK) pathway, which is impaired in lupus T cells. SYNGENEIC MACROPHAGES a further discussion)13,91. In support of this model, The drug hydralazine is thought to cause hypomethy- macrophages (immune cells that engulf foreign particles) the affected repeats and/or adjacent sequences adopt lation in T cells and an SLE-like disease in humans that are transferred between tightly packed chromatin structures in FRAXA, DM1 by inhibiting this pathway. Impaired ERK signalling genetically identical mice. and FRDA78,86. Conversely, reduction in D4Z4 repeat probably contributes to reduced DNMT1 levels and NATURE REVIEWS | GENETICS VOLUME 6 | AUGUST 2005 | 605 © 2005 Nature Publishing Group
  • 10. REVIEWS hypomethylation of SLE T cells 100 . SLE therefore toid cell lines, indicating a defect in differentiation demonstrates not only that DNA methylation defects or activation. However, these experiments did not can be restricted to only one or a few cell types, but provide obvious candidate genes to explain the devel- also that DNA methylation is regulated differently, opmental and neurological aspects of ICF syndrome. or responds to different stimuli, depending on cell Interestingly, genes with altered expression in ICF type. cells are not hypomethylated. This implies either that DNMT3B regulates gene expression independently of Immunodeficiency, centromeric instability and its ability to methylate DNA, or that the affected genes facial anomalies (ICF) syndrome. ICF syndrome is show altered expression owing to downstream effects an extremely rare autosomal recessive disease that of other genes, such as transcription factors, which is characterized by profound immunodeficiency, and might be activated by hypomethylation113. The former which results from an absence or significant reduction idea is supported by recent findings that DNMT3B of at least two immunoglobulin isotypes. Other more interacts with histone deacetylases (HDACs), histone variable features include impaired cellular immunity methylases and chromatin-remodelling enzymes114,115. and unusual facial features101,102. All ICF patients show In addition, DNMT3B can profoundly influence neu- marked hypomethylation and decondensation of peri- ronal differentiation of multipotent cells in a manner centromeric heterochromatin on chromosomes 1 and that depends on its ability to interact with HDACs, but 16, and, to a lesser extent, chromosome 9 (see online not on its ability to methylate DNA116. Both methyla- supplementary information S3 (table)). This abnor- tion-related and unrelated defects might therefore be mality is observed almost exclusively in ICF B cells or important in ICF syndrome. in LYMPHOBLASTOID CELL LINES that have been stimulated with mitogen101,103,104. ATRX: a connection with chromatin remodelling The molecular defect in 60–70% of ICF patients is Alpha-thalassemia/mental retardation syndrome, mutation of the DNMT3B gene at 20q11.2, leading to X-linked (ATRX) is a very rare X-linked disease that defective methylation11,105,106. Patients without detect- occurs in <1 in 100,000 males. It is characterized by able DNMT3B mutations might have abnormalities in severe mental retardation and genital abnormalities, other regions of the gene that have not been exam- with alpha-thalassemia in many, but not all, patients. ined (such as the promoter and 5′ and 3′ UTRs), or The affected gene, ATRX, is located at Xq13 (see online might arise from mutations in a gene that regulates supplementary information S3 (table)) and encodes DNMT3B. Mutations in DNMT3B are usually het- an ATP-dependent chromatin-remodelling protein of erozygous and target the catalytic domain. Complete the chromodomain and helicase-like domains (CHD) loss-of-function of the encoded enzyme is probably subclass of SNF2LIKE PROTEINS117. embryonic lethal in humans, as it is in mice11, and sev- Functional domains of ATRX include a PHD zinc eral of the catalytic domain mutations examined so far finger-like motif at its N-terminus (homologous to do not result in complete loss of enzyme activity105,107. the PHD motifs in DNMT3A and DNMT3B), and a The clinical variability of the disease might therefore helicase domain at its C-terminus. Mutations in ATRX be related to the level of residual DNMT3B activity. are clustered in these two domains and are thought to It was recently suggested that different methylation impair its nuclear localization, protein–protein inter- defects are present in ICF patients with and without actions, or chromatin-remodelling functions118–120. DNMT3B mutations, but the molecular basis behind ATRX localizes to pericentromeric heterochromatin these differences is not known108. and PROMYELOCYTIC LEUKEMIA PML NUCLEAR BODIES in a The instability of repeat-rich pericentromeric cell-cycle-dependent manner, and exists in a complex heterochromatin in this disease is probably due to with the transcriptional regulatory death-associated its hypomethylation in ICF cells. Loss of DNA meth- protein 6 (DAXX)120–122. It is therefore thought to LYMPHOBLASTOID CELL LINE ylation in ICF syndrome is region specific and affects regulate transcription, although its target genes and Immortalized B cell line created by infecting primary B cells mainly the pericentromeric satellite 2 and 3 repeats the nature of the regulation (positive or negative) are with Epstein-Barr virus. and non-pericentromeric regions that include the unknown. inactive X chromosome, cancer–testis genes, and Interestingly, ATRX patients also have DNA- SNF2LIKE PROTEINS other sporadic repeats (such as NBL2 and the tar- methylation defects. These are diverse, and include ATP-dependent chromatin remodelling enzymes that get in FSHD, D4Z4)109–111. This indicates that these both aberrant hypermethylation (at the DYZ2 contain a region homologous to regions are bona-fide targets for DNMT3B. Satellite Y-chromosome repeat) and hypomethylation events an extended family of proteins 2 and 3 hypomethylation is common in cancer cells, (at ribosomal DNA repeats), but no overall change in that include known RNA and and although no elevated cancer incidence has been 5-methylcytosine levels123. It remains unclear, how- DNA helicases. reported in ICF patients, the small number of these ever, if methylation abnormalities directly contribute PML BODIES patients and their short lifespan make it possible that a to the ATRX phenotype. There have been important Dot-like structures in the slightly increased incidence has gone undetected112. advances in recent years in our understanding of the nucleus of most mammalian The role of DNMT3B mutations in the invariant direct and indirect connections between the DNA cells. These were originally immune defects in ICF syndrome is far less clear. methylation and chromatin-remodelling machin- defined by the localization of the PML protein, which is Microarray analyses have revealed that many genes eries, and the identification of DNA methylation involved in transcriptional involved in immune function and B-cell immunoglob- defects in ATRX patients has reinforced this link in regulation. ulin production are deregulated in ICF lymphoblas- human cells124. 606 | AUGUST 2005 | VOLUME 6 www.nature.com/reviews/genetics © 2005 Nature Publishing Group