Journal of Medicine and Medical Science Vol. 2(3) pp. 696-713, March 2011 Available online @http://www.interesjournals.org/JMMS Copyright © 2011 International Research Journals Review Epigenetics and its role in ageing and cancer Abhimanyu K. Jha*, Shailesh Kumar*, Mohsen Nikbakht, Vishal Sharma, Jagdeep Kaur§ Department of Biotechnology, Panjab University, Chandigarh, India *These authors contributed equally to this work Accepted 21 March 2011 Epigenetics refers to the change in gene expression without the change in the sequence of the gene. Epigenetics includes alternate phenotypic states that are not based on differences in genotype, but are generally stably maintained during cell division and are potentially reversible. A much more expanded view of epigenetics involves multiple mechanisms interacting to collectively establish alternate states of chromatin structure, histone modification, associated protein composition and transcriptional activity. Chromatin structure is not fixed. Instead, chromatin is a dynamic identity and is subject to extensive developmental, environmental and age-associated remodeling. In some cases, this remodeling appears to counter the ageing and age-associated diseases, such as cancer, and extend organismal lifespan. Advances in our understanding of chromatin structure, histone modification, transcriptional activity, promoter hypermethylation and global hypomethylation resulted in an increasingly integrated and expanded view of epigenetics. The study of epigenetics reveals how patterns of gene expression are passed from one cell to its descendants, how gene expression changes during the differentiation of one cell type into another, and how environmental factors can change the way genes are expressed. There are far-reaching implications of epigenetic research for human biology and diseases, including our understanding of cancer and ageing. Due to these developments, epigenetic therapy is expanding to include combinations of histone deacetylase inhibitors and DNA methyltransferase inhibitors. This review encompasses the different types of epigenetic changes, the interplay among them and the implications of these epigenetic changes in relation to cancer and ageing. Besides this, the development and perspective of epigenetic therapy has also been discussed in brief. Keywords: Epigenetics, histone deacetylase inhibitors, DNA methyltransferase inhibitors, cancer, histone modification, promoter hypermethylation. INTRODUCTION The central dogma of molecular biology states that the information embedded in the linear nucleotide sequence of DNA contains coding information for RNA and protein, as well as regulatory sequences that control the biology of DNA itself. The word "epigenetics" was coined by the developmental biologist C. H. Waddington in 1942 (Waddington et al., 1942) The Greek prefix "epi-" in the word "epigenetics" implies features that are "on top of" or "in addition to" genetics. Waddington proposed the epigenetic model to describe, the interaction of genes within a multicellular organism with their surroundings to produce a particular phenotype and this hypothesis was known as Conrad hypothesis. Holliday and Pugh proposed in 1975 that covalent chemical DNA § Corresponding author Email: jagsekhon@yahoo.com modifications, including methylation of cytosine-guanine (CpG) dinucleotides, were the molecular mechanisms behind Conrad’s hypothesis. Because all cells within an organism inherit the same DNA sequences, the process of cellular differentiation depend on pattern of epigenetic rather than genetic inheritance. Robin Holliday defined epigenetics as "the study of the mechanisms of temporal and spatial control of gene activity during the development of complex organisms" (Holliday et al.,1996). Thus, the word "epigenetic" can be used to describe any aspect other than DNA sequence that influences the development of an organism. The modern usage of the word "epigenetics" usually refers to heritable traits that do not involve changes to the underlying DNA sequence. Epigenetic regulation mediates adaptation to the environment by the genome lending plasticity that Jha et al. 697 translates into the presenting phenotype, particularly under “mismatched” environmental conditions (Godfrey et al., 2007). Epigenetic inheritance involves the transmission of information not encoded in DNA sequences from cell to daughter cell or from generation to generation. Covalent modifications of the DNA or its packaging histones are responsible for transmitting epigenetic information. Epigenetic modifications, such as acetylation, phosphorylation, methylation, ubiquitination, and ADP ribosylation, of the highly conserved core histones, H2A, H2B, H3, and H4, influence the genetic potential of DNA. The enormous regulatory potential of histone modification is illustrated in the vast array of epigenetic markers found throughout the genome. In addition, the modification of histones can cause a region of chromatin to undergo nuclear compartmentalization and, as such, specific epigenetic markers are nonrandomly distributed within interphase nuclei (Andrulis et al., 1998). But it can also be an important determinant of cellular senescence and organism ageing (Wilson et al.,1983; Issa 2003; Fraga et al., 2005 ). Now epigenetics in broad sense has changed to epigenomics that include the higher order of chromosome organization, Nucleosome formation and modification of histone tail like methylation, acetylation, phosphorylation and DNA methylation. Epigenomics is defined as a genome-wide approach to study epigenetics. This term encompasses whole-genome studies of epigenetic processes and the identification of the DNA sequences that specify where the epigenetic processes are targeted.The central goal of epigenomics is to define the DNA sequence features that in turn direct epigenetic processes. Possible mechanisms of epigenetic inheritance The best understood sequence-independent inheritance mechanism is that of DNA methylation, in which the maintenance methyltransferase DNMT1 specifically recognizes semi-methylated DNA and methylates the remaining strand. About 4% of the cytosines are usually methylated in mammalian genomic DNA and it has been shown that this methylation is essential for mouse development (Li et al., 1992). In addition, it has also been shown that DNA methylation plays an essential role in several epigenetic phenomena, including genomic imprinting, X-chromosome inactivation, and retro-element silencing. DNA methylation is known to interplay with other chromatin marks, such as histone modifications (Jones, 1998). Accurate transmission of the histone code through cell generations presents a paradox, because nucleosomes are not deposited in a semi-conservative manner during replication. Rather, ‘old’ histones are distributed randomly between the DNA molecules and the ‘gaps’ are filled with freshly synthesized (unmodified) histones, leading to a dilution of chromatin marks. It has been suggested that the chromatin code can then be reinstated by chromatin modifiers that are recruited to the remaining marks (Henikoff et al., 2004). It has also been proposed that the timing of locus replication might have a role in the maintenance of epigenetic states (Zhang et al., 2002). Nucleosome disruption and replacement are crucial activities that maintain epigenomes, but these highly dynamic processes have been difficult to study. A recent study described a direct method for measuring nucleosome turnover dynamics genome-wide. It concluded that nucleosome turnover is most rapid over active gene bodies, epigenetic regulatory elements, and replication origins in Drosophila cells. Nucleosomes turnover is faster at sites for trithorax-group than polycomb-group protein binding, suggesting that nucleosome turnover differences underlie their opposing activities and challenging models for epigenetic inheritance that rely on stability of histone marks. The results establish a general strategy for studying nucleosome dynamics and uncover nucleosome turnover differences across the genome that are likely to have functional importance for epigenome maintenance, gene regulation, and control of DNA replication (Deal et al., 2010). This is in contrast to another study which has clearly observed that silent histone modifications within large heterochromatic regions are maintained by copying modifications from neighboring preexisting histones without the need for H3-H4 splitting events (Xu et al., 2010). Semiconservative DNA replication ensures the faithful duplication of genetic information during cell divisions. However, how epigenetic information carried by histone modifications propagates through mitotic divisions remains elusive. To address this question, the DNA replication dependent nucleosome partition pattern should be clarified. Epigenetic mechanisms play a fundamental role in the interpretation of genetic information (Li et al., 2002). Depending on its particular epigenetic modification pattern, a gene can be expressed or silenced. Epigenetic modifications thus represent an integral mechanism for the control of complex gene expression patterns. One prominent example for an epigenetic control mechanism is the covalent modification of histones (Jenuwein et al., 2001). Epigenetic processes are natural and essential to many organism functions. Till now many types of epigenetic processes have been identified which include histone modifications, like acetylation, phosphorylation, ubiquitination and methylation and DNA methylation. The main types of epigenetic changes have been mentioned in Figure 1. The epigenome and histone modifications The work of Kornberg in 1974 proposed that chromatin is a repeating unit of 8-histone proteins and 200 bps of DNA wrapped around it. Later on the Nucleosome model 698 J. Med. Med. Sci. became the center of all molecular biology research to understand the script of life. Nucleosome is made up of four kinds of basic proteins –H1, H2A, H2B, H3 and H4. The amino acid sequencing of H4 reveals that this protein is highly conserved among species. The precise organization of chromatin is critical for many cellular processes, including transcription, replication, repair, recombination and chromosome segregation. Dynamic changes in chromatin structure are directly influenced by post-translational modifications of the amino-terminal tails of the histones (Luger et al., 1998). Packaging of DNA sequences of promoters region in nucleosomes prevents the initiation of transcription by bacterial and eukaryotic RNA polymerases in vitro (Knezetic et al.,1986, Lorch,1987). Nucleosomes exert a similar inhibitory effect upon transcription in vivo: turning off histone synthesis by genetic means in yeast, and consequent nucleosome loss, turns on transcription of all previously inactive genes tested (Han et al., 1988). The four core histones are present in equal amounts in the cell, whereas H1 is half as abundant as the other histones. This is in consistency with the finding that only one molecule of H1 is associated with each nucleosome (which contains two copies of each core histone). As they are closely associated with the negatively-charged DNA molecule, the histones have a high content of positivelycharged amino acids. It has been observed that greater than 20% of the residues in each histone are either lysine or arginine. Histone lysine methyltransferase G9a/KMT1C mediates H1.4K26 mono- and dimethylation in vitro and in vivo and thereby provides a recognition surface for the chromatin-binding proteins HP1 and L3MBTL1 (Trojer et al., 2009). Histone proteins are subjected to four major kinds of posttranslational modifications like methylation of arginine and lysine residues, phosphorylation of serine, lysine acetylation and lysine ubiquitination (Kouzarids, 2007). The types of histone modifications have been shown in Table no.1. Different types of histone modifications at different sites are shown in Figure 2. Histone Methylation Histone methylation was first discovered more than thirty five years ago but very little was known about the impact of this biological phenomenon. The initial study was stressed on the pattern of methylation and its maintenance and relation with the kind of histone. It has been revealed that methylation of specific lysine residues in histone tails function as a stable epigenetic mark that directs particular biological functions, ranging from transcriptional regulation to heterochromatin assembly . There are three different types of methylation forms which occurs in lysine (me1,me2 and me3). There are three modifications, K9me3, K20me3 and K27me3, which are associated with repressed chromatin in many organisms. High-resolution ChIP seq in different mammalian cells gives a genome-wide snapshot of the modifications and reveals distinct patterns that reflect chromosome organization (Barski, 2007; Mikkelsen 2007). Their interaction with different histone binding proteins play a key role in transducing the pattern of modifications into a functional outcomes (Taverna et al., 2007 ). Previous studies showed that lysines 4, 9, 27 and 36 of H3 and lysine 20 in H4 can be mono, di or trimethylated (Van Holde, 1989). Histone lysine methylation appears to be a rather static process, and consequently it is generally viewed as an epigenetic mark rather than as a flexible regulatory signal (Jenuwein, 2001). In addition to lysines, certain arginines in histones H3 and H4 can be methylated (Davie et al., 2002) and this methylation can be correlated to gene activation (Bauer et. al., 2002). Several other histone methyltransferases (HMTs) have been characterized (Zhang et al., 2001) and it has become evident that specific methylation patterns correlate with gene activity. H3-K9 methylation has been observed to be primarily associated with heterochromatin (Noma et al., 2001; Lachner et al., 2001) whereas H3-K4 methylation (in higher eukaryotes) is observed in transcriptionally active regions(Noma et al., 2001; Strahl et al.,1999). Histone proteins assemble into nucleosomes, which function as DNA packaging units as well as transcriptional regulators. Methylation at lysine 9 (Lys-9) on histone H3 has recently been shown to be a marker of heterochromatin from yeast to mouse(Jenuweinet et al.,2002; Schneider et al., 2002). Lys-9 methylation is recognized by heterochromatin-associated proteins, such as HP-1, and is required to maintain heterochromatin. Studies have shown that modifications of histone H3 also contribute to euchromatin gene silencing by switching between Lys-9 acetylation and methylation (Taverna et al., 2007). Another histone modification, methylation of histone H3 Lys-4, localizes to sites of active transcription, and this modification may stimulate transcription. These different combinations of histone modifications at different residues may act synergistically or antagonistically to alter gene expression (Roh et al., 2006). H3 Lys-9 methylation is also closely related to DNA methylation and acts as an epigenetic mark of silencing in the tumor suppressor genes. Recent reports show that H3 Lys-9 methylation can be regulated by Suv39h-HP1independent pathways and occurs in facultative heterochromatin on the inactive X chromosome (Boggs et al. 2002). Methylation of histone H4 at lysine 20 (K20) has been implicated in transcriptional activation, gene silencing, heterochromatin formation, mitosis, and DNA repair. Even in newly synthesized histones, 90% of histone get methylated in 2 to 3 cell cycles and it can be concluded that it is required for normal mitosis and cell cycle progression, K20 methylation proceeds normally even in the colchicines treated cells (Pesavento et al., Jha et al. 699 Table 1. Modifications of histones Modification Acetylation ADP-ribosylation Histone (s) modified H3, H4 Core histone Methylation Phosphorylation H3, H4 H1 H3 Ubiquitination H2A, H4 H2A and H2B Effects/function Activation of gene Local disruption of chromatin structure to facilitate DNA repair Repression of transcription Chromatin condensation Gene activation, chromatin condensation, Nucleosome assembly Description of chromatin structure to facilitate transcription Figure 1. Main types of epigenetic changes 2008). Histone modification maps as ageing marks: Histone modifications also have a defined profile during ageing and cell transformation. For example, the trimethylation of H4-K20, which is enriched in differentiated cells (Biron et al., 2004) increases with age (Prokocimer et al., 2006) and is commonly reduced in cancer cells. The increase of trimethylated H4-K20 in aged-like cells has been associated with defects in the nuclear lamina (Shumaker et al., 2006). Histone Acetylation Acetylation neutralises the positive charge on the amino group of lysine and acts as a binding site for proteins containing a bromodomain (Guenther et al., 2007). There is a direct link between histone acetylation and active chromatin. It has been demonstrated directly that transcriptionally active genes carry acetylated core histones (Hebbes et al., 1988). In human T cells, there are more than 45,000 acetylation islands rich in H3K9acK14ac, many of which correspond to transcriptional regulatory elements, particularly enhancers and promoters of active genes (Brownell et al., 1996; Kuo et al.,1998). Acetylation of lysine residues on histones H3 and H4 leads to the formation of an open chromatin structure. Histone H4 acetylation distinguishes coding regions of the human genome from heterochromatin in a differentiationdependent but transcription independent manner. It has been concluded that nucleosomes containing acetylated H4 are scattered infrequently and possibly randomly through coding and adjacent regions and are essentially absent from heterochromatin. Induction of differentiation of HL-60 cells by exposure to dimethylsulfoxide or 12-otetradecanoylphorbol 13-acetate (TPA) did not alter the level of H4 acetylation within either the c-MYC or c-FOS genes or other coding regions, but did induce a transient increase in H4 acetylation within centric heterochromatin (Fukuda et al., 2006; O'Neill et al., 1996). Histone acetylation and phosphorylation are highly dynamic 700 J. Med. Med. Sci. Figure 2. Different types of histone modifications at different sites. processes with rapid turn-over rates. The connection between acetylation and transcription was established by the demonstration that yeast Gcn5 protein, a positive transcriptional regulator of many genes, has HAT activity (Parekh et al., 1999) and stimulation of transcription by Gcn5 requires the HAT activity (Grunstein, 1997). Histone acetylation by promoter-associated transcription factors is localized. For example, increased acetylation of H3 and H4, attributed to p300/CBP, was found upon viral infection in two to three nucleosomes surrounding the interferon-b promoter (Struhl, 1998). Histone acetylation also plays an important role in the activity-dependent regulation of sulfiredoxin and sestrin 2 which are neuroprotective antioxidant enzymes that reduce hydroperoxides and protect neurons against oxidative stress (Soriano et al., 2009). Jha et al. 701 Histone Deacetylation The connection between acetylation and transcription is further shown by the fact that deacetylation can cause repression. It has been discovered that many coactivators are HATs , proteins originally identified as corepressors which have now been shown to possess deacetylase activity (Grozinger et al., 1999 ; SantosRosa et al., 2005). The deacetylation–repression connection was most clearly demonstrated by the isolation of a human histone deacetylase, HDAC1, whose sequence was highly similar to that of a yeast negative regulatory protein Rpd3. Many additional deacetylases have been identified in yeast and human cells (Nowak et al., 2002). Histone acetylation tends to open up chromatin structure. Accordingly, histone acetyltransferase (HATs) tend to be transcriptional activators whereas histone deacetylases (HDACs) tend to be repressors. Many HAT genes are altered in some way in a variety of cancers (Cheung et al., 2000). For instance, the p300 HAT gene is mutated in a number of gastrointestinal tumours (Swarthout et al., 2009). On the other hand, alteration of HDAC genes in cancer seems to be far less common. However, despite of low incidence of genetic mutation in cancer, HDAC inhibitors are performing well in the clinic as anti-cancer drugs. Recent studies have shown that HDAC inhibition lead to Ubiquitin-Dependent Proteasomal Degradation of DNA Methyltransferase 1 in Human Breast Cancer Cells (Zhou et al., 2008). Histone Phosphorylation H1 histones play an important role in regulating higher order structure of chromatin and are potential regulators of gene expression. Phosphorylation of H1 on serine and threonine in their amino and carboxyl terminal tails occurs in vivo and alters their interaction with DNA. H1 phosphorylation destabilizes higher order chromatin structure which is thought to allow accessory factors to participate in replication, mitotic condensation, and gene activation. For instance, it has been observed that histone H3 thr 45 phosphorylation is a replicationassociated post-translational modification in S. cerevisiae (Baker et al., 2010). Quiescent fibroblasts treated with epidermal growth factor undergo rapid serine 10 phosphorylation which is coincident with the induction of early response genes such as c-FOS. This phosphorylation is catalyzed by the Rsk-2 kinase. The mechanism by which phosphorylation contributes to transcriptional activation is not well understood. The addition of negatively charged phosphate groups to histone tails neutralizes their basic charge and is considered to reduce their affinity for DNA. Furthermore, it has been observed that several acetyltransferases increased HAT activity on serine 10-phosphorylated substrates and mutation of serine 10 decreases activation of mGcn5-regulated genes (Clayton et al., 2000 ; Cheung et al., 2000). Thus, phosphorylation may contribute to transcriptional activation through the stimulation of HAT activity on the same histone tail. Indeed, phosphoacetylation of histone H3 on c-fos and cjun associated nucleosomes has been demonstrated upon gene activation (De Souza et al., 2000). Phosphorylation of H2A has also long been correlated with mitotic chromosome condensation, and in this case also serine 10 appears to play an important role. For instance, mutation of serine 10 Tetrahymena histones causes abnormal chromosomal condensation and defective chromosome separation during anaphase (Hsu et al., 2000). Phosphorylation of histone H3 is also known to occur after activation of DNA-damage signaling pathways. A conserved motif (ASQE, in the single-letter amino-acid code) found in the carboxyl terminus of yeast H2A and the mammalian H2A variant H2A.X is rapidly phosphorylated upon exposure to DNA-damaging agents (Downs et al., 2000; Rogakou et al., 1999) Serine 139 has been identified as the site for this modification, and its phosphorylation in response to damage is dependent on the phosphatidylinositol-3-OH kinase Mec1 in yeast. Mec1 dependent serine 139 phosphorylation is apparently required for efficient nonhomologous end-joining repair of DNA. This suggests that phosphorylation mediates an alteration of chromatin structure, which in turn facilitates repair. H3S10 and H3S28 are phosphorylated at mitosis which is a crucial part of the cell cycle. Any misregulation here is often associated with cancers. Indeed, the Aurora kinases that perform this H3 phosphorylation are implicated in cancer (Zhang et al., 2005). Histone Ubiquitination Modification of the N- and C-terminal tails of histones is thought to occur in patterns, which recruits specific effector proteins that alter chromatin structure and regulate gene expression. The addition and removal of histone modifications are thought to have antagonistic effects. Cul4–Ddb1, a Ring H2 ubiquitin ligase, plays an important role in many vital cellular processes including DNA replication, DNA repair and transcription (Saha et al., 2006) Of the four core histones, H2A and H2B have long been known to be modified by ubiquitin conjugation. Ubiquitination of histone H2B (uH2B) on lysine 120 (K120) in humans (Becker et al., 2002) and lysine 123 in yeast(O’Connel et al., 2007) has been correlated with increase in methylation of lysine 79 (K79) of histone H3 by K79-specific methyltransferase. Regulators of H2A and H2B ubiquitylation play roles in gene silencing (ubH2A) or in transcription initiation and elongation (ub-H2B) 702 J. Med. Med. Sci. (Oselu et al., 2006). In addition to participating in the cellular response to DNA damage, CUL4-mediated histone ubiquitination may regulate other aspects of chromatin function, including heterochromatin silencing (West et al., 1980). It has been proposed that uH2B may induce H3 K79 methylation directly, either by altering chromatin structure and therefore nucleosomal accessibility, or through the recruitment of enzymatic function (Robzyk et al., 2000). Compared with other histone modifications, ubiquitination involves the addition of a relatively large molecule that is two-thirds the mass of an individual histone. Due to the large size of this molecule, it has been proposed that ubiquitination should have an impact on chromatin structure. Chromatin fibers reconstituted with ubH2A molecules have similar properties to the control chromatin with regard to folding and sedimentation (Wang et al., 2006). Sumoylation Small ubiquitin-related modifier (SUMO) is the best characterized member of a growing family of ubiquitin-like proteins involved in posttranslational modifications. In mammals, there are three members of the SUMO protein family: SUMO-1, SUMO-2 (SMT3a), and SUMO-3 (SMT3b), which are implicated in partly overlapping, yet distinct functions (Sun et al., 2002). SUMO is covalently attached to other proteins through the activities of an enzyme cascade (E1–E2–E3) similar to that for ubiquitination. There is only one known SUMO-activating enzyme, E1 (a heterodimer of SAE1 and SAE2) and only one known SUMO-conjugating enzyme, E2 (UBC9). It has been proposed that histone sumoylation acts as a component of the group of modifications that appear to govern chromatin structure and function to regulate transcriptional repression and gene silencing (Jason et al., 2002). In Drosophila polytene chromosomes, the SUMO moiety was detected in many euchromatic sites and the chromocenter (Tatham et al., 2001) suggesting that histone (chromatin) sumoylation plays a role in both euchromatic transcriptional repression and heterochromatic gene silencing. It is reported that pontin, a component of chromatin-remodeling complexes, is SUMO-modified, and that SUMOylation of pontin is an active control mechanism for the transcriptional regulation of pontin on androgen-receptor target genes in prostate cancer cells (Kim et al., 2007). It has been hypothesized that SUMOylation is involved in the regulation of p53 in both protein stability and function by direct modification of p53 or in an indirect way via modulating the stability of MDM2. It has long been identified that p53 underwent SUMOylation on lysine 386 that increase transcriptional activity of p53 (Schreiber et al.,2002) and a necessary factor for full apoptotic activity of p53 (Lehembre et al., 2000). As SUMO E2, Ubc9 are only conjugating enzyme in SUMOylation so correlated with their involvement in cancer development and tumorigenesis. The increased expression of Ubc9 has been reported in several human ovarian cancer cell lines such as PA-1 and OVCAR-8 as well as in ovarian tumor tissues (Gostissa et al., 1999) human lung adenocarcinomas (Muller et al., 2000) and metastastic prostate cancer cell line, LNCaP (Mo et al., 2005). Chromatin remodeling Chromatin represents an important regulatory entity that provides a means of maintaining genome stability (e.g. by suppressing uncontrolled recombination or transposon mobilization) and allows the integration of multiple endogenous and exogenous signals at the single-gene level. Although a wide variety of proteins cooperate in reorganizing chromatin structure in response to these signals, the basic mechanisms seem to involve the covalent modification of histone tails and changes of nucleosome positioning that utilize ATP. Most of the key regulator complexes identified to date contain either one or both of these activities. These include not only protein complexes that are involved in the short-term regulation of gene activity but also components that affect long-term regulation and epigenetic inheritance, such as the Sir2 family of HDACs and various members of the Polycomb and Trithorax groups of proteins. Recent studies show that TF2I is involved in chromatin remodeling during embryonic stem cell differentiation but yet the exact role of these factors are yet to be revealed (Aleksandr et al., 2009). Dynamic chromatin remodeling utilizes several basic mechanisms, including covalent histone modifications, ATP-dependent chromatin remodeling, utilization of histone variants and DNA methylation to alter the accessibility of DNA. These mechanisms work either independently or in tandem to allow optimal chromatin remodeling for efficient transcriptional regulation, DNA replication and DNA-damage repair. ATP-dependent chromatin-remodeling enzymes and their functions ATP-dependent chromatin-remodeling enzymes, which are highly conserved in organisms from yeast to humans, are similar to the SNF2 (sucrose non-fermenting 2) family of DNA translocases and all of them contain a catalytic ATPase subunit (Doniels et al., 2002). These ATPase machineries utilize the energy of ATP hydrolysis to mobilize nucleosomes along DNA, evict histones off DNA or promote the exchange of histone variants, which in turn modulate DNA accessibility and alter nucleosomal structures (Kim et. al., 2006). Based on distinct domain structures, there are four well-characterized families of Jha et al. 703 mammalian chromatin remodeling ATPases: (a) SWI/SNF (switching defective/ sucrose non-fermenting) family: SWI/SNF remodeling complexes primarily disorganize and reorganize nucleosome positioning to promote accessibility for transcription-factor binding and gene activation. However, they also promote transcriptional- repressor binding and gene repression under certain conditions (Martens et al., 2003) (b) ISWI (imitation SWI) family: The ISWI remodeling complexes primarily organize and order nucleosome positioning to induce repression (Ooi et al., 2006) although they also mediate transcriptional activation and transcriptional elongation (Corona et al., 2004 ; Badenhorst et al., 2002; Morillon et. al., 2003) (c) NuRD (nucleosome remodeling and deacetylation)/ Mi-2/CHD (chromodomain, helicase, DNA binding) family NuRD/Mi-2/CHD remodeling complexes primarily mediate transcriptional repression in the nucleus (Shimono et al., 2005). However, they are also involved in transcriptional activation of rRNA in the nucleolus. (d) INO80 (inositol requiring 80) family (Bao et. al., 2007). The INO80 remodeling complexes appear to have both activating and repressive effects for a specific set of genes (Jonsson et al., 2004 ; Hassan et al., 2002). Both members of the SWI/SNF family of ATPases has BRM and BRG1 (BRM/SWI2-related gene 1), contain a C-terminal bromodomain that binds to acetylated histone tails (Hassan et al., 2002). ISWI family members, SNF2H and SNF2L, have a SANT (SWI3, ADA2, NCOR and TFIIIB’ DNA-binding domains) and a SLIDE (SANT-like ISWI) domain that mediate interaction with unmodified histone tails and linker DNA (Boyer et al., 2004). NuRD/Mi-2/CHD family members, CHDs 1–5, have unique tandem chromodomains that specifically recognize methylated histone tails (Flanagan et al., 2005). Different chromatin remodeling complexes have been mentioned in Table 2 (taken and modified after getting permission from the author and the publisher) (Kornberg et al., 1999). Besides HAT activities, ATP-dependent chromatin remodeling complexes have also been recently implicated in DNA repair processes. Demonstration was made a few years ago that Cockayne syndrome B protein (CSB) was required for coupling NER to transcription. CSB, a DNA-dependent ATPase of the SWI2/SNF2 family, has been shown to remodel chromatin substrates in vitro (Citterio et al., 2000). It has also been concluded that mammalian SWI/SNF complexes prevent DNA damage-induced apoptosis in part by facilitating efficient repair and thereby ensuring timely elimination of unrepaired DSBs that could otherwise lead to excessive prolongation of p53 activation (Park et al., 2009). It has been found that ATP dependent chromatin remodeling is highly affected by Inositol Polyphosphates. It has also been demonstrated that mutations in genes encoding inositol polyphosphate kinases that produce IP4, IP5, and IP6 impair transcription in vivo. These results provide a link between inositol polyphosphates, chromatin remodeling and gene expression (Shen et al., 2003). According to a recent study, structural studies of the RSC chromatin-remodeling complex prompts a proposal for the remodeling mechanism: RSC binding to the nucleosome releases the DNA from the histone surface and initiates DNA translocation (through one or a small number of DNA base pairs); ATP binding completes translocation and ATP hydrolysis resets the system. Binding energy thus plays a central role in the remodeling process. RSC may disrupt histone-DNA contacts by affecting histone octamer conformation and through extensive interaction with the DNA. Bulging of the DNA from the octamer surface is possible, and twisting is unavoidable, but neither is the basis of remodeling (Lorch et al., 2010). DNA Methylation DNA methylation occurs in bacteria, fungi, plants and animals, however its role varies widely among different organisms. DNA methylation might have evolved to protect bacterial genomes from invasion by foreign DNA. Thus, bacteria have devised a way to distinguish their own DNA from that of an invader; in the bacterial genome the sequence contains the methylated base, whereas in foreign DNA the same sequence is unmethylated and therefore digested by the restriction endonuclease (Sims et al., 2005). DNA methylation in mammalian cells occurs at the 5-position of cytosine within the CpG dinucleotide. CpG islands are the sites present in the promoter of most of the tumor suppressor genes, and hypermethylation at CpG island leads to silencing of the expression of these genes. The CpG islands have the following important characteristics: (i) G+C content of 0.50 or greater (ii) observed to expected CpG dinucleotide ratio of 0.60 or greater and (iii) both occurring within a sequence window of 200 bp or greater. CpG dinucleotides methylation in mammals represent the target for the covalent modification of DNA (Bao et al., 2007). This methyl group protrudes from the cytosine nucleotide into the major groove of the DNA and it displaces transcription factors that normally bind to the DNA (Kumar et al., 1994 ; Kim et al., 2003). Cell-typespecific cytosine methylation and histone-tail modifications could contribute to the differences in gene expression patterns between cell types. CpG island definition based on sequence composition identifies these elements at the promoter sites of approximately half of the genes in the human genome (Ioshikhes et al., 2000) most of which are expressed in most or all tissues, hence they have been designed as ‘housekeeping’ genes. The dynamic nature of cytosine methylation becomes especially evident during tumorigenesis in which methylation is decreased genome-wide, whereas the CpG islands at promoters of tumour-suppressor genes acquire methylation, which leads to their silencing 704 J. Med. Med. Sci. and subsequent tumour progression. Hypermethylation is also linked to chromosomal instability, a common phenomenon in human tumours (Lengauer et al., 1998) which has been observed in mice with hypomethylated genomes due to engineered methyltransferase deficiencies (Gaudet et al., 2003). Although CpG islands account for only about 1% of the genome and for 15% of the total genomic CpG sites, these regions contain over 50% of the unmethylated CpG dinucleotides. There are about 45,000 CpG islands, most of which reside within or near the promoters or first exons of genes and are unmethylated in normal cells, with the exception of CpG islands on the inactive X chromosome in females (Arber et al., 1969). CpG sites have been shown to act as hot-spots for germline mutations, contributing to 30% of all point mutations in the germ line and for acquired somatic mutations that lead to cancer. For example, methylated CpG sites in the TP53 coding region contribute to as many as 50% of all inactivating mutations in colon cancer and 25% of cancers (Hendrich et al., 1999). Cellular DNA methylation patterns is established by a complex interplay of at least three independent DNA methyltransferases: DNMT1, DNMT3A and DNMT3B. The first methyltransferase to be discovered was DNMT1. Pioneering work has established that DNMT1 has a 10– 40-fold preference for hemimethylated DNA (Pradhan et al.,1999; Pradhan et al., 1997). By providing both enhanced transcriptional control and protection against mutation, the methyl-CpG binding proteins could have facilitated the expansion of the methylated DNA compartment within the evolving vertebrate genome. MBD2 and MBD3 are the only vertebrate methyl-CpG binding proteins and in mammals, MBD2 and MBD3 genes have an identical genomic structure, differing only in the sizes of their introns, and they encode proteins that are 70% identical (Baylin et al., 2002). The DNMT protein motif is evolutionarily ancient, occurring in all known DNA methyltransferases from bacteria to plants and humans (Baylin et al., 2000). The animals, in which DNA methylation is predominantly associated with transcriptional repression, the presence or absence of DNA methylation and of the DNMTs varies, as does the apparent use of DNA methylation within animal genomes (Bird et al., 2002). CpG islands are associated with at least half of all cellular genes and are normally methylation-free. Dense methylation of cytosine residues within islands results in strong and heritable transcriptional silencing. Such silencing normally occurs almost solely at genes subject to genomic imprinting or to X chromosome inactivation. Aberrant methylation of CpG islands associated with tumor suppressor genes has been proposed to contribute to carcinogenesis (Antequera et al., 1993). In addition to carcinogensis and genomic imprinting, DNA methylation has also been found to regulate memory formation and synaptic plasticity in the adult rat hippocampus (Miller et al., 2008). The understanding of chromatin with respect to the components that specify for states of gene expression is growing rapidly, and this knowledge is establishing a base from which abnormal as well as normal gene expression events can be understood. In this regard, an especially active field in cancer research is concerned with patterns of aberrant gene promoter hypermethylation that have been associated with loss of transcription of a growing list of genes in virtually every type of human cancer (Greenblatt et al., 1994). Several mechanisms have been proposed to account for transcriptional repression by DNA methylation. The first mechanism involves direct interference with the binding of specific transcription factors to their recognition sites in their respective promoters. Several transcription factors, including AP-2, c-Myc/Myn, the cyclic AMPdependent activator CREB, E2F and NFkB, recognize sequences that contain CpG residues, and binding of each has been shown to be inhibited by methylation (Baylin et al., 1998). The second mode of repression involves a direct binding of specific transcriptional repressors to methylated DNA. Hypomethylation is the second kind of methylation defect that is observed in a wide variety of malignancies (Jones et al., 1999). It is common in solid tumors such as metastatic hepatocellular cancer, cervical cancer, prostate tumors, and also in hematologic malignancies such as B-cell chronic lymphocytic leukemia. The global hypomethylation seen in a number of cancers, such as breast, cervical, and brain, show a progressive increase with the grade of malignancy (Kim et al., 1994). A mutation of DNMT3b has been found in patients with immunodeficiency, centromeric instability, and facial abnormalities, which causes the instability of the chromatin (Okano et. al., 1999). Hypomethylation has been hypothesized to contribute to oncogenesis by activation of oncogenes such as cMYC and H-RAS or by activation of latent retrotransposons (Alves et al., 1996) or by chromosome instability (Tuck-Muller et al., 2000). Much attention in the methylation field has focussed on CpG islands, primarily because of the propensity of such sequences to become aberrantly hypermethylated in tumours, resulting in the transcriptional silencing of the associated gene (Kochanek et al., 1995; Jones et al., 1998). Tumor cells have less methylation than normal cells, and this loss appears to occur primarily from parasitic and repetitive DNAs, which are usually heavily methylated.The connection between CpG methylation and transcriptional silencing in vertebrates has been recognized for more than two decades (Tate et al., 1993). The local cytosine methylation of a particular sequence could directly interfere with transcription-factor binding (Wade et al., 1999). Chromatin assembly facilitates the repression of methylated DNA. Methyl-CpG binding proteins, including MECP2, associate with co-repressor complexes that include histone deacetylases (Nan et al., Jha et al. 705 1998). Two important additional links between DNA methylation and chromatin structure have recently come to light. First, DNMT1 forms a complex with Rb, E2F1, and HDAC1 and represses transcription from E2F responsive promoters (Robertson et al., 2000). The second link between chromatin structure and methylation comes from patients with mutations in a putative ATPdependent chromatin-remodelling factor of the SNF2 family, termed ATR-X (Gibbons et al., 2000). a lower percentage of methylated CpG than other vertebrates (20%), shares the same global methylation distribution and has a similar density of methylation (Tweedie et al., 1997). This is significant because methylation density is a factor in methylation dependent silencing. In all the substantially methylated invertebrates tested the distribution of methylated bases is quite different from that of vertebrates. Rather than global distribution with increased distances between methylated sites, the genome is separated into alternating compartments of methylated and unmethylated DNA (Reik et al., 2001). Methyltransferase recognition sequences Methylation in most animals occurs at cytosines within the sequence CpG with additional low levels of non-CpG methylation reported in some species. Plants are additionally methylated extensively at CpNpG sequences. However there are exceptions to these general rules: CpT was recently identified as the preferred recognition sequence for Drosophila methylation (Lyko et al., 2004) and non-CpG methylation is common in methylated fungi (Selker, 1997). Methylation changes during development Methylation patterns can also get altered during the course of development. For instance in mammals there is loss of methylation in early development and then the pattern is established again (Egger et al., 2004) whereas methylation in Drosophila is only present during early development. Changes also occur locally with loss of methylation at some sites in some tissues; such local demethylation often correlates with expression. Methylation levels DNA methylation and ageing The percentage of methylated cytosines varies substantially between species from no detectable methylation (e.g. the nematode Caenorhabditis elegans, the flat worm Schistosoma mansoni and the yeast species, Schizosaccharomyces pombe and Saccharomyces cerevisiae) to very high levels in typical vertebrates (60–90% of all CpGs methylated) and most plants. It is assumed that methylation has been lost in some lineages, but the details of which and when remain incomplete because of the scarcity of data. All invertebrates tested have either no methylation or some intermediate level of methylation. Direct comparison between species is complicated by the fact that there are several different ways to estimate methylation levels. These methods differ in sensitivity. To complicate matters further, some methods test only a subset of cytosines and the levels of methyl cytosine are alternatively reported as the percentage of all bases, percentage of all cytosines, or fraction of the subset of sites tested (Jablanka et al., 1995). Distribution of methylated sites Vertebrate genomes are globally methylated i.e. methylated cytosines are found over the entire genome for short (nearly 1-kb) stretches. This unmethylated DNA, the CpG island fraction, accounts for around 1% of the genome and frequently coincides with promoter regions. It is interesting to note that the lamprey, although having The previous studies found a pattern of low global DNA methylation levels in many aged mammalian tissues.The great fidelity with which DNA methylation patterns in mammals are inherited after each cell division is ensured by the DNA methyltransferases (DNMTs). However, the ageing cell undergoes a DNA methylation drift. Early studies showed that global DNA methylation decreases during ageing in many tissue types and it was subsequently observed that mammalian fibroblasts cultured to senescence increasingly lost DNA methylation (Wilson et al., 1983). The loss of global DNA methylation during ageing is probably mainly the result of the passive demethylation of heterochromatic DNA as a consequence of a progressive loss of DNMT1 efficacy and/or erroneous targeting of the enzyme by other cofactors (Casillas et al., 2003). Several specific regions of the genomic DNA become hypermethylated during ageing. New findings demonstrated a widespread and tissue specific age-related DNA methylation changes in mice. A surprisingly high rate of hyper- and hypomethylation as a function of age in normal mouse small intestine tissues and a strong tissue-specificity to the process has also been demonstrated. It has been concluded that epigenetic deregulation is a common feature of ageing in mammals (Maegawa et al., 2010). Normal ageing cells and tissues show a progressive loss of 5-methylcytosine content, primarily within DNA repeated sequences, as well as in potential gene regulatory areas . In addition, selected genes show 706 J. Med. Med. Sci. progressive age-related increases in promoter methylation, which, once a critical methylation density is reached, have the potential to permanently silence gene expression. These changes are highly mosaic within a given tissue and introduce a high degree of epigenetic variability in ageing cells (Calvanese et al., 2009). The ageing associated epigenetic changes are shown in Table 3. DNA methylation in cancer DNA methylation was the first epigenetic alteration to be observed in cancer cells. Hypermethylation of CpG islands at tumour suppressor genes switches off these genes, whereas global hypomethylation leads to genome instability and inappropriate activation of oncogenes and transposable elements (Feinberg et al., 2004). It appears that genomic DNA methylation levels, which are maintained by DNMT enzymes, are delicately balanced within cells. The over-expression of DNMTs is linked to cancer in humans, and their deletion from animals is lethal (Rodenhiser et al., 2006). Furthermore, methylcytosine is capable of spontaneously mutating in vivo by deamination to give thymine. Indeed, 37% of somatic p53 gene mutations (and 58% of germ-line mutations) occur at methyl CpGs, and these mutations are strongly implicated in the cause of cancer (Rideout et al., 1990). The importance of epigenetic alterations in cancer progression was shown years ago when methylation of the 50 CpG-island of the p16/CDKN2A gene was proved to be responsible for its transcriptional silencing in 20–40% of most common cancers (Herman et al., 1995 ; Merlo et al., 1995). Several cancer susceptibility genes, including BRCA1 and VHL, which cause familial forms of breast and kidney cancer, respectively, are silenced by methylation in a significant percentage of sporadic forms of the respective tumor types. Fifteen percent of sporadic breast cancers harbor methylated BRCA1 genes and their gene-expression profiles are identical to those of tumors from inherited families in which BRCA1 is mutated; both are completely distinct from those of other breast-cancer types (Jones et al., 2002). Several groups have been developing array-based methods for genome-wide detection of methylation or other epigenetic alterations such as histone modifications (Callinan et al., 2006 ; Wu et al., 2006). Interplay between DNA methylation and histone modifications It is important to note that there is a direct link between DNA methylation and histone modifications. A number of proteins involved in DNA methylation (e.g. DNMTs and MBDs) directly interact with histone modifying enzymes such as histone methyltransferases (HMTs) and histone deacetylases (HDACs). The growing evidence for dynamic inter/intra-regulation of these modifications, position and modification-specific protein interactions, and biochemical/biophysical interaction between modifications has strengthened the ‘histone code’ hypothesis, in which histone modifications are integral to regulating the expression of the genome (Strahl et al., 2000). There are now several examples of modification patterns and sequences that relate to gene activation, some of which occur on the same histone tail or on the same amino acid. Thus, if ubiquitination/sumoylation of histones function to activate/repress, respectively, it will be interesting to determine whether they occur on the same lysine residues and whether , in a simple reciprocal fashion, oppose one another's activity. In fact, it is now believed that DNA methylation and histone methylation are tied together in a loop where one modification is dependent on the other. Altering this relationship will almost certainly have severe consequences on the epigenome and chromatin organization. Thus most, if not all, factors that affect DNA methylation levels also affect histone modifications. For instance, it appears that H3K9 methylation and DNA methylation are linked (Fuks, 2005). In mammals, DNA methyltransferases interact with Suv39h H3K9 methyltransferases and loss of H3K9 methylation inSuv39h-knockout embryonic stem cells decreases Dnmt3b-dependent CpG methylation at major centromeric satellites (Lehnertz et al., 2003). Methyl-CpG-binding proteins may recruit histone deacetylase complexes to deacetylate histone tails so that the tails become suitable for serving as substrates for methylation. In contrast with this sequential process, MBD-containing HMTs may bind directly to methylated DNA to methylate histone tails. Alternatively, it is also possible that chromodomain-containing proteins bind to methylated histone tails and recruit DNA methyltransferase (DNMT) to methylate adjacent CpG sequences. Irrespective of the sequence of events, it is likely that a concerted action of HMT and HDAC complexes may play an important role in methylated DNA silencing (Zhang et al., 2001). In contrast to the above predictons it has been also observed that transcription of mouse DNA methyltransferase 1 (DNMT 1) is regulated by both E2FRb-HDAC dependent and -independent pathways. It has been identified that the promoter region and major transcription start sites of mouse Dnmt1 and found two important cis-elements within the core promoter region. One is an E2F binding site, and the other is a binding site for an as yet unidentifed factor. Point mutations in the two cis-elements decreased promoter activity in both nontransformed and transformed cells. Thus, both sites play a critical role in regulation of DNMT 1 transcription in proliferating cells (Hiromichi et al., 2003). As DNA methylation is found to be linked to histone Jha et al. 707 Table 2. Chromatin-Remodeling Complexes (taken and modified after obtaining permission from the author and publisher). Complex Organism ATPase Mass (MDa) No. of Subunits S. cerevisiae S. cerevisie D. melanogaster H. sapiens H. sapiens H. sapiens Swi2/Snf2 Sth1 Brahma Hbrm CHD4 BRG1 2 1 2 2 1.5 2 11 15 ND 10 18 10 S. cerevisiae S. cerevisiae D. melanogaster D. melanogaster D. melanogaster H. sapiens ISWI1 ISWI2 ISWI ISWI ISWI hISWI 0.4 0.3 0.5 0.7 0.2 0.5 4 2 4 5 4 2 Xenopus H.sapiens Mi-2 Mi-2 ------ 6 7 SWI/SNF family SWI/SNF RSC Brahma h SWI/SNF NRD h SWI/SNF ISWI family I SWI1 I SWI2 NURF CHRAC ACF RSF Mi-2/CHD family Mi-2 NuRD Table 3. Ageing associated epigenetic changes in different tissues of different species [taken after obtaining permission from the author and publisher (Calvanese et al., 2009)]. 708 J. Med. Med. Sci. deacetylation in the same manner, methylation of histone H4 by arginine methyltransferase PRMT1 is essential in vivo for many subsequent histone modifications knocking out of PRMT1 gene leads to a domain-wide loss of histone acetylation on both histones H3 and H4, as well as an increase in H3 Lys9 and Lys27 methylation, both marks associated with inactive chromatin (Huang et al., 2005). Epigenetic Therapy Epigenetic therapy, the use of drugs to correct epigenetic defects, is a new and rapidly developing area of pharmacology. Because so many diseases, such as cancer, involve epigenetic changes, it seems reasonable to try to counteract these modifications with epigenetic treatments. These changes seem an ideal target because they are by nature reversible, unlike DNA sequence mutations. The most popular of these treatments aim to alter either DNA methylation or histone acetylation. The emerging use of drugs that modulate epigenetic alterations, including the hypomethylating agents and histone deacetylase inhibitors, is an exciting advance for cancer treatment. These agents have shown great promise in the treatment of several hematologic malignancies, especially myelodysplastic syndromes, acute myeloid leukemia, and cutaneous T-cell lymphoma. The potential reversibility of epigenetic states offers an exciting opportunity for novel cancer drugs that can reactivate epigenetically silenced tumor-suppressor genes (Esteller et al., 2005; Jha et al., 2010). Blocking either DNA methyltransferase or histone deacetylase activity could potentially inhibit or reverse the process of epigenetic silencing. DNA methyltransferases and histone deacetylases are the two major drug targets for epigenetic inhibition to date, for instance histone deacetylase inhibitors induce apoptosis in peripheral blood lymphocytes along with histone H4 acetylation and the expression of the linker histone variant, H1. Histone deacetylase inhibitors induce apoptosis in peripheral blood lymphocytes along with histone H4 acetylation and the expression of the linker histone variant, H18 (Sourlingas et al., 2001). Vorinostat/SAHA, has been approved by the FDA for use as second-line therapy in patients with cutaneous Tcell lymphoma (CTCL). The other agents include (1) sodium phenylbutyrate; (2) MS-275; (3) valproic acid; (4) depsipeptide (FK228); (5) LBH-589; and (6) CI-994 (Mann et al., 2007 ; Duvic et al., 2007). The histone deacetylase inhibitor valproic acid inhibits cancer cell proliferation via down-regulation of the alzheimer amyloid precursor protein. Based on these observations, the data suggest that APP down-regulation via HDAC inhibition provides a novel mechanism for pancreatic and colon cancer therapy (Venkataramani et al., 2010). Histone deacetylase inhibitors suppress inflammatory activation of rheumatoid arthritis patient’s synovial macrophages and tissues (Chen et al., 2001). It has been proposed that HDAC inhibition promotes neuronal outgrowth and counteracts growth cone collapse through CBP/p300 and P/CAF-dependent p53 acetylation (Gaub et al., 2010). The most broadly used DNA methyltransferase inhibitor, 5-aza-2’-deoxycytidine (5-aza-CdR), clinically referred to as decitabine, has been shown to be have toxic effects aside from its demethylating properties and has been found to be mutagenic in vivo (Jackson et al., 1997). Moreover, 5-aza-CdR has been shown to be capable of transcriptionally activating genes with unmethylated promoters (Soengas et al., 2001) which leads to increased acetylation and H3 lysine 4 methylation (Nguyen et al., 2002) suggesting this drug can induce chromatin remodeling independently of its effects on cytosine methylation. The commonly used drugs targeting methylation are azacytidine (5azacytidine), decitabine (5-aza-2’-deoxycytidine), fazarabine (1-β-D-arabinofurasonyl-5-azacytosine), and dihydro-5-azacytidine (Goffin et al., 2002). These are all derivatives of deoxycytidine with some modification at the fifth position in the pyrimidine ring. Other drugs include zebularine and antisense oligodeoxynucleotides. Dietary phytochemicals particularly catechol-containing polyphenols were shown to inhibit DNMT and reactivate epigenetically silenced genes (Fang et al., 2003). Certain dietary polyphenols, such as (–)-epigallocatechin 3gallate (EGCG) from green tea and genistein from soybean, have recently been demonstrated to inhibit DNA methyltransferases (DNMT) in vitro. This inhibitory activity is associated with the demethylation of the CpG islands in the promoters and the reactivation of methylationsilenced genes such as p16INK4a, retinoic acid receptor ß, O6-methylguanine methyltransferase, human mutL homolog 1, and glutathione S-transferase- , (Millar et al., 1999 ; Jarrard et al.,1998; Izbicka et al., 1999; Nakayama et. al., 2000 ; Kinoshita et al., 2000 Sasak et al., 2002; Jarrard et al., 1997 et al., Chi et al., 1997). These activities have been observed in human esophageal, colon, prostate, and mammary cancer cell lines, and the activity can be enhanced by the presence of histone deacetylase inhibitors or by a longer-term treatment (Fang et al., 2007). The combined inhibition of DNA methylation and histone acetylation enhances gene reexpression and drug sensitivity in vivo (Steele et al., 2009). Curcumin and one of its major metabolites, tetrahydrocurcumin can inhibit M. SssI, a DNMT1 analog, activity (Liu et al., 2009). Several phytochemicals inhibit the DNA methyltransferase activity with betanin being the weakest while rosmarinic and ellagic acids were the most potent modulators (up to 88% inhibition) (Paluszczak et al., 2010). Curcumin and genistein cause reversal of hypermethylation and reactivation of RARβ2 gene in SiHa cell line (a squamous cervical cancer cell line) (Jha Jha et al. 709 et al., 2010). Histone deacetylase (HDAC) inhibitors like trichostatin A, SAHA (Suberoylanilide hydroxamic acid) etc. are also being tried as potential chemotherapeutic agents. However, epigenetic therapy has its limitations, such as the fact that both DNMT as well as HDAC inhibitors may activate oncogenes due to lack of specificity, resulting in accelerated tumour progression. Moreover, epigenetic states, once corrected, may revert back to the original state because of the reversible nature of DNA methylation patterns. Indeed, combinations of DNA methyltransferase and histone deacetylase inhibitors appear to synergize effectively in the reactivation of epigenetically silenced genes (Shi et al., 2003 ; Thiagalingam et al., 2003). Reports suggest that reduced histone acetylation or H3K4me2 methylation and increased dimethyl-H3-K9 methylation play a critical role in the maintenance of promoter DNA methylation associated RASSF1A gene silencing in prostate cancer (Kawamoto et al., 2007). Using biological and statistical criteria, four hypermethylated genes CDKN2B, MLF-1, PCDH8, HOXD8 and four hypomethylated genes CD37, HDAC1, NOTCH1 and CDK5 were identified, where aberrant methylation was associated with inverse changes in mRNA levels. Prominent and aberrant promoter methylation in Mantle Cell Lymphoma (MCL) suggests that differentially methylated genes can be targeted for therapeutic benefit in MCL (Leshchenko et al., 2010). Combination trials are underway to test this concept in the clinic. Caution in using epigenetic therapy is necessary because epigenetic processes and changes are so widespread. To be successful, epigenetic treatments must be selective to irregular cells; otherwise, activating gene transcription in normal cells could make them cancerous, so the treatments could cause the very disorders they are trying to counteract. CONCLUSIONS Epigenetic changes like histone modifications and DNA methylation play an important role in several of the biological processes like cancer, ageing and development. Epigenetics has reached a new level of maturity over the past few years, with many findings highlighting the intimate link between DNA methylation and histone modifications. As a result of these discoveries, we have begun to unlock the long-standing mystery of how CpG methylation patterns are established. Histone deacetylation and H3K9 methylation appear to pave the way for CpG methylation. In addition, evidence suggests that the DNA methylation associates with histone deacetylation and H3K9 methylation to generate a selfpropagating cycle that promotes transcriptional repression. Yet our understanding of the interplay between these epigenetic modifications is still incomplete. A major challenge is to uncover the mutual reinforcements of repression and the different states of covalent histone and DNA modification required to silence specific genomic regions in specific cases of epigenetic regulation. An important question is whether DNA methylation interacts with other histone modifications besides deacetylation and H3K9 methylation. The findings described in this review point to a possible connection with H3K27 and H4K20me3 methylation. Further studies are going on to confirm, if the links between CpG methylation and H3K27 in addition to H4K20me3 methylation are strong enough to support the proposed hypotheses. There is tremendous potential in epigenetics. We might have just begun to uncover the link between histone and DNA modifications. Epigenetic therapy is a new and rapidly developing area in pharmacology. To date, most trials of epigenetic drugs have been conducted to evaluate their effects on cancers, many of which have shown promising results. The past few years have seen many exciting discoveries and undoubtedly, many more are yet to come. The future of this field is having a vast potential and it is expected that it would solve many of the biological mysteries. ACKNOWLEDGEMENTS The authors acknowledge the financial assistance provided by CSIR (Council of Scientific and Industrial Research), India to AKJ. REFERENCES Aleksandr M, Nyam-Osor C, Badam ED, ashzeveg B (2009). "TF2I is involved in chromatin remodeling during embryonic stem cell differentiation" Paper presented at the annual meeting of the Connecticut's Stem Cell Research International Symposium, Omni Hotel, New Haven, CT, Mar 23 Alves G, Tatro A, Fanning T (1996). Differential methylation of human LINE-1 retrotransposons in malignant cells. Gene 176: 39-44 Andrulis ED, Neiman AM, Zappulla DC, Sternglanz R (1998). Perinuclear localization of chromatin facilitates transcriptional silencing. Nature 394:592–595 Antequera F, Bird A (1993). Number of CpG islands and genes in human and mouse. Proc Natl Acad Sci USA 901: 1995-1999 Arber W, Linn S, (1969). DNA modification and restriction. Annu Rev Biochem. 38: 467–500 Badenhorst P, Voas M, Rebay I, Wu C (2002). Biological functions of the ISWI chromatin remodeling complex NURF. Genes & Dev. 16: 3186-3198 Baker SP, Phillips J,Anderson S, Qiu Q, Shabanowitz J, Smith MM, Yates RJ, Hunt DF, Grant PA (2010). Histone H3 Thr 45 phosphorylation is a replication-associated post-translational modification in S cerevisiae. Nat Cell Biol. 12: 294–298 Bao Y, Shen X (2007). SnapShot, chromatin remodelling complexes. Cell 129: 632e1–632e2 Bao Y, Shen X (2007). INO80 subfamily of chromatin remodelling complexes. Mutat Res. 618: 18–29 Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K (2007). High-resolution profiling of histone methylations in the human genome. Cell 129: 823- 837 710 J. Med. Med. Sci. Bauer UM, Daujat S, Nielsen SJ, Nightingale K, Kouzarides T (2002). Methylation at arginine 17 of histone H3 is linked to gene activation. EMBO Rep 3: 39-44 Baylin S, Bestors TH (2002). Altered methylation patterns in cancer cell genomes, Cause or consequence . Cancer Cell 1:229-305 Baylin SB, Herman JG, (2000). DNA hypermethylation in tumorigenesis, epigenetics joins genetics. Trends Genet. 16:168–174 Baylin SB, Herman JG, Herman JR, Vertino PM, Issa JP (1998). Alterations in DNA methylation, a fundamental aspect of neoplasia. Adv Cancer Res. 72: 141–196 Becker PB, Horz W (2002). ATP-dependent nucleosome remodeling .Annu Rev Biochem. 71: 247–273 Bird A (2002). DNA methylation patterns and epigenetic memory .Genes Dev. 16: 6–21 Biron L, McManus KJ, Hu N, Hendzel MJ, Underhil DA (2004). Distinct dynamics and distribution of histone methyl-lysine derivatives in mouse development. Dev Biol. 276: 337–351. Boggs BA, Cheung P, Heard E, Spector DL, Chinault AC, Allis CD (2002). Differentially methylated forms of histone H3 show unique association patterns with inactive human X chromosome. Nat Genet. 30: 73–76 Boyer LA, Latek RR, Peterson CL (2004). The SANT domain, a unique histone-tailbinding module? Nat Rev Mol Cell Biol. 5: 158–163 Brownell JE, Zhou J, Ranalli T, Kobayashi R, Edmondson DG, Roth SY, Allis CD (1996). Tetrahymena histone acetyltransferase A, a homolog to yeast Gcn5p linking histone acetylation to gene activation. Cell 84: 843–851 Callinan PA, Feinberg AP (2006). The emerging science of epigenomics. Hum Mol Genet. 1: R95-R101 Calvanese V, Lara E , Kahn A, Fraga MF (2009). The role of epigenetics in aging and age-related diseases. Ageing Res Rev. 8: 268-276 Casillas MAJ, Lopatina N, Andrews LG, Tollefsbol TO (2003). Transcriptional control of the DNA methyltransferases is altered in aging and neoplastically-transformed human fibroblasts. Mol Cell Biochem. 252: 33–43 Chen H, Liu J, Zhao CQ, Diwan B A, Merrick BA, Waalkes MP (2001). Association of c-myc overexpression and hyperproliferation with arsenite-induced malignant transformation .Toxicol Appl Pharmacol.175: 260–268 Cheung P, Tanner KG, Cheung WL, Sassone-Corsi P, Denu JM, Allis CD (2000). Synergistic coupling of histone H3 phosphorylation and acetylation in response to epidermal growth factor stimulation.Mol Cell. 5: 905-915 Cheung P, Allis CD, Sassone-Corsi P (2000). Signaling to chromatin Chi SG, DeVere White RW, Muenzer JT, Gumerlock PH (1997). Frequent alteration of CDKN2, p16(INK4A),MTS1 expression in human primary prostate carcinomas. Clin Cancer Res. 3: 1889–1897. Citterio E, Van Den Boom V, Schnitzler G, Kanaar R, Bonte E, Kingston RE, Hoeijmakers JH, Vermuelen W (2000). ATPdependent chromatin remodeling by the Cockayne syndrome B DNA repair transcription coupling factor. Mol Cell Biol. 20: 7643– 7653 Clayton AL, Rose S, Barratt MJ, Mahadevan LC (2000). Phosphoacetylation of histone H3 on c-fos and c-jun-associated nucleosomes upon gene activation. EMBO J. 19: 3714-3726 Corona DF, Tamkun JW (2004). Multiple roles for ISWI in transcription, chromosome organization and DNA replication. Biochim Biophys Acta. 1677: 113–119 Davie JK, Dent SY (2002). Transcriptional control, an activating role for arginine methylation.Curr Biol, 12:R59-R61 De Souza CP, Osmani AH, Wu LP, Spotts JL, Osmani SA (2000). Mitotic histone H3 phosphorylation by the NIMA kinase in Aspergillus nidulans. Cell 102: 293-302 Deal RB, Henikoff JD, Henikoff S (2010). Genome-Wide Kinetics of Nucleosome Turnover Determined by Metabolic Labeling of Histones. Sci. 328: 1161 – 1164. Doniels SAL, Nimri CF, Stoner GD, Lubet RA, You M (2002). Differential gene expression in human lung adenocarcinomas and squamous cell carcinomas. Clin Cancer Res 8 , 1127−1138 Downs JA, Lowndes NF, Jackson SP (2000). A role for Saccharomyces cerevisiae histone H2A in DNA repair. Nature 408: 1001- 1004 Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, Chiao JH, Reilly JF, Ricker JL, Richon VM, Frankel SR (2007). Phase 2 trial of oral vorinostat (suberoylanilide hydroxamicacid, SAHA). for refractory cutaneous T-cell lymphoma (CTCL). Blood 109: 31–9 Egger G, Liang G, Aparicio A, Jones PA (2004). Epigenetics in human disease and prospects for epigenetic therapy. Nature 429: 457-463 Esteller M (2005). DNA methylation and cancer therapy, new developments and expectations. Curr Opin Oncol. 17: 55–60 Fang MZ, Wang Y, Ai N, Hou Z, Sun Y, Lu H, Welsh W, Yang CS (2003). Tea polyphenols (-).-Epigallocatechin –3-gallate inhibits DNA Methyltransferase and reactivates methylation silenced genes in cancer cell lines. Cancer Res. 63: 7563-7570 Feinberg AP, Tycko B (2004). The history of cancer epigenetics . Nat Rev Cancer 4:143-153 Flanagan JF, Mi LZ, Chruszcz M, Cymborowski M, Clines KL, Kim Y, Minor W, Rastinejad F, Khorasanizadeh S (2005). Double chromodomains cooperate to recognize the methylated histone H3 tail. Nature 438: 1181–1185 Fraga MF, Ballestar E, Paz MF, Ropero S , Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, BoixChornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector DT, Wu YZ, Plass C, Esteller M (2005). Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA 102: 10604–10609 Fuks F (2005). DNA methylation and histone modifications, teaming up to silence genes. Curr Opin Genet Dev. 15: 490-495 Fukuda H, Sano N, Muto S, Horikoshi M (2006). Simple histone acetylation plays a complex role in the regulation of gene expression. Brief Fun Gen Prot. 5: 190-208 Gama-Sosa MA, Midgett RM, Slagel VA, Githens S, Kuo KC, Gehrke CW, Ehrlich M, (1983). Tissue-specific differences in DNA methylation in various mammals Biochem Biophys Acta. 740:212-19 Gaub P, Tedeschi A, Puttagunta R, Nguyen T, Schmandke A, Giovanni DS (2010). HDAC inhibition promotes neuronal outgrowth and counteracts growth cone collapse through CBPp300 and PCAFdependent p53 acetylation. Cell Death & Different. 17: 1392-1408 Gaudet F , Hodgson G, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H, Jaenisch R (2003). Induction of tumors in mice by genomic hypomethylation. Science 300: 489–492 Gibbons RJ, McDowell TL, Raman S, O'Rourke DM, Garrick D, Ayyub H, Higgs DR (2000). Mutations in ATRX, encoding a SWISNF-like protein, cause diverse changes in the patterns of DNA methylation. Nature Genet. 24: 368–371 Godfrey KM, Lillycrop KA, Burdge GC, Gluckman PD, Hanson MA (2007). Epigenetic mechanisms and the mismatch concept of the developmental origins of health and disease Pediatr Res. 61:5–10 Goffin J, Eisenhauer E, (2002). DNA methyltransferase inhibitorsstate of the art. Ann Oncol. 13: 1699-1716 Gostissa M, Hengstermann A, Fogal V, Sandy P, Schwarz SE (1999). Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. EMBO J. 18: 6462−6471 Greenblatt MS, Bennett WP, Hollstein M, Harris CC (1994). Mutations in the p53 tumor suppressor gene, Clues to cancer etiology and molecular pathogenesis. Cancer Res. 54: 4855–4878 Grozinger CM, Hassig CA, Schreiber SL (1999). Three proteins define a class of human histone deacetylases related to yeast hda1p. Proc Natl Acad Sci USA 96: 4868–4873 Grunstein M (1997). Histone acetylation in chromatin structure and transcription. Nature 389: 349–352 Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007). A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130: 77-88 Han M, Grunstein M (1988). Nucleosome loss activates yeast downstream promoters in vivo Cell 55: 1137–1145 Jha et al. 711 Hassan AH, Prochasson P, Neely KE, Galasinski SC, Chandy M, Carrozza MJ, Workman JL (2002). Function and selectivity of bromodomains in anchoring chromatin-modifying complexes to promoter nucleosomes. Cell 111: 369–379 Hebbes TR, Thorne AW, Crane-Robinson C (1988). A direct link between core histone acetylation and transcriptionally active chromatin. The EMBO Journ. 7: 1395- 1402 Hendrich B, McQueen CAH, McQueen DCH, Chambers D (1999). Genomic structure and chromosomal mapping of the murine and human Mbd1,Mbd2, Mbd3 and Mbd4 genes. Mamm Genome 10: 906–912 Henikoff S, Furuyama T, Ahmad K (2004). Histone variants, nucleosome assembly and epigenetic inheritance. Trends Genet. 20: 320–326 Herman JG, Merlo A, Mao L, Lapidus RG, Issa JP, Davidson NE, Sidransky D, Baylin SB (1995). Inactivation of the CDKN2p16MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res. 55: 4525-4530: Holliday R (1996). Mechanisms for the control of gene activity during development Biol Rev Cambr Philos Soc. 5:431-471 Hsu JY, Sun ZW, Li X, Reuben M, Tatchell K, Bishop DK, Grushcow JM, Brame CJ, Caldwell JA, Hunt DF (2000). Mitotic phosphorylation of histone H3 is governed by Ipl1aurora kinase and Glc7PP1 phosphatase in budding yeast and nematodes. Cell 102: 279-291 Huang S, Litt M, Felsenfeld G, Esteller M (2005). Methylation of histone H4 by arginine methyltransferase PRMT1 is essential in vivo for many subsequent histone modifications. Genes & Dev. 19: 18851893 Ioshikhes IP, Zhang MQ (2000). Large-scale human promoter mapping using CpG islands. Nature Genet. 26: 61–63 Issa JP (2003). Age-related epigenetic changes and the immune system. Clin Immunol. 109: 103–1086 Izbicka E, MacDonald JR, Davidson K, Lawrence RA, Gomez L, Von Hoff D (1999). 5:6-Dihydro-5_-azacytidine (DHAC). restores androgen responsiveness in androgen-insensitive prostate cancer cells. Anticancer Res. 19: 1285–91 Jablanka E, Regev A (1995) . Gene number, methylation and biological complexity. Trends Genet. 11: 383–384 Jackson-Grusby L, Laird PW, Magge SN, Moeller BJ and Jaenisch R (1997). Mutagenicity of 5-aza-2¢-deoxycytidine is mediated by the mammalian DNA methyltransferase. Proc Natl Acad Sci USA 94: 4681-4685 Jaenisch R, Bird A (2003). Epigenetic regulation of gene expression, how the genome integrates intrinsic and environmental signals. Nature Genet. 33: S245–S254 Jarrard DF, Bova GS, Ewing CM, Pin SS, Nguyen SH, Baylin SB, Cairns P, Sidransky D, Herman JG, and Isaacs WB (1997). Deletional, mutational, and methylation analyses of CDKN2 (p 16MTS1). in primary and metastatic prostate cancer Genes Chromosomes. Cancer 19: 90–6 Jarrard DF, Kinoshita H, Shi Y, Sandefur C, Hoff D, Meisner LF, Chang C, Herman JG, Isaacs WB, Nassif N (1998). Methylation of the androgen receptor promoter CpG island is associated with loss of androgen receptor expression in prostate cancer cells. Cancer Res. 58: 5310–4 Jason L, Moore S, Lewis J, Lindsey G, Ausio J (2002). Histone ubiquitination, a tagging tail unfolds? Bioessays 24: 166–174 Jenuwein T, Allis CD (2002). Translating the histone code. Science 293: 1074–1080 Jenuwein T (2001). Re-SET-ting heterochromatin by histone methyltransferases. Trends Cell Biol. 11: 266-273 Jenuwein T, Allis CD (2001). Translating the histone code. Science 293: 1074-80 Jha AK, Nikbakht M, Parashar G, Shrivastava A, Capalash N, Kaur J (2010). Reversal of hypermethylation and reactivation of RARβ2 gene by natural compounds in cervical cancer cell lines. Folia Biologica (Praha). 56: 195-200 Jha AK, Nikbakht M, Capalash N, Kaur J (2010). DNA methylation inhibitors: Role in cancer therapy SAJOSPS 11(1):131 Jones PA, Baylin SB (2002). The fundamental role of epigenetic events in cancer. Nat Rev Genet. 3: 415-428 Jones PA, Laird PW (1999). Cancer epigenetics comes of age. Nature Genet. 21: 163–166. Jones PL,Veenstra GJC, Wade PA, Vermaak D, Kass SU, Landsberger N, Strouboulis J, Wolffe AP (1998). Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nature Genet. 19: 187–191. Jones P (1998). Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription Nature Genet.19: 187–191 Jonsson ZO, Jha S , Wohlschlegel JA, Dutta A (2004). Rvb1p, Rvb2p recruit Arp5p and assemble a functional Ino80 chromatin remodeling complex. Mol Cell 16: 465–477. Kawamoto K, Okino ST, Place RF, Urakami S, Hirata H, Kikuno N, Kawakami T Tanaka Y, Pookot D, Chen Z (2007). Epigenetic Modifications of RASSF1A Gene through Chromatin Remodeling in Prostate cancer. Clinical Cancer Research 13: 2541. Kim J, Kollhoff A, Bergmann A, Stubbs L (2003). Methylation-sensitive binding of transcription factor YY1 to an insulator sequence within the paternally expressed imprinted gene. Hum Mol Genet. 12 :233–245 Kim JH, Choi HJ, Kim B, Kim MH, Lee JH, Kim HJ, Choi B, Kim MH, Kim JM, Lee KIS, Lee MH, Choi SJ, Kim KI, Kim SI, Chung CH, Baek SH, (2006). Roles of sumoylation of a reptin chromatinremodelling complex in cancer metastasis. Nat Cell Biol 8: 631–639 Kim JH, Lee JM, Nam HJ, Choi HJ, Yang JW, Lee JS, Kim MH, Kim S, Chung CH, Kim K, Baek SH (2007). SUMOylation of pontin chromatin-remodeling complex reveals a signal integration code in prostate cancer cells. PNAS 104: 20793–20798. Kim, YI, Giuliano, A, Hatch, KD (1994). Global DNA hypomethylation increases progressively in cervical dysplasia and carcinoma. Cancer 74: 893- 899 Kimura H, Nakamura T, Ogawa T, Tanaka S, Shiota K (2003). Transcription of mouse DNA methyltransferase 1 (DNMT1). is regulated by both E2F-Rb-HDAC-dependent and -independent pathways. Nucleic Acids Res. 31: 3101-13 Kinoshita H, Shi Y, Sandefur C, Meisner LF, Chang C, Choon A, Reznikoff CR, Bova SG, Friedl A, Jarrard DJ (2000). Methylation of the androgen receptor minimal promoter silences transcription in human prostate cancer. Cancer Res. 60: 3623-30 Knezetic JA, Luse DS (1986). The presence of nucleosomes on a DNA template prevents initiation by RNA polymerase II in vitro. Cell 45: 95–104 Kochanek S, Renz D, Doerfler W (1995). Transcriptional silencing of human Alu sequences and inhibition of protein binding in the B box regulatory elements by 5’-CG-3’ methylation. FEBS Lett. 360: 115– 120 Kornberg RD, and Lorch Y (1999). Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome. Cell 98 ,285–294 Kouzarids T (2007). Chromatin Modifications and their functions. Cell 128: 693-705 Kumar S, Cheng X, Klimasauskas SMS, Posfai J, Roberts RJ, Wilson GG (1994). The DNA (cytosine-5). methyltransferases. Nucleic Acids Res. 22: 1–10 Kuo MH, Zhou J, Jambeck P, Churchill MEA, Allis CD (1998). Histone acetyltransferase activity of yeast Gcn5p is required for the activation of target genes in vivo. Genes Dev. 12: 627–639 Lachner M, O’Carroll D, Rea S, Mechtler K, Jenuwein T (2001). Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410: 116-120 Lehembre F, Badenhorst P, Muller S, Travers A, Schweisguth F, Dejean A (2000). Covalent modification of the transcriptional repressor tramtrack by the ubiquitin-related smt3 protein in Drosophila. Mol Cell Biol. 20: 1072–1082 Lehnertz B, Ueda Y, Derijck AA, Braunschweig U, Perez-Burgos L, Kubicek S, Chen T, Li E, Jenuwein T, Peters AH (2003). Suv39hmediated histone H3 lysine 9 methylation directs DNA methylation to major satellite repeats at pericentric heterochromatin. Curr Biol. 13: 1192-1200 Lengauer C, Kinzler KW, Vogelstein B (1998). Genetic instabilities in human cancers. Nature 396: 643–649 Leshchenko VV, Kuo PY, Shaknovich R, Yang DT, Gellen T, Weniger MA, Rafiq S, Suh S, Goy A, Wilson W, Verma A, Braunschweig I, Muthusamy N, Kahl BS, Byrd JC, Wiestne Ar, 712 J. Med. Med. Sci. Melnick A, Parekh S (2010). Genome wide DNA methylation analysis reveals novel targets for drug development in mantle cell lymphoma. Blood 116 : 1025-1034 Li E (2002). Chromatin modification and epigenetic reprogramming in mammalian development. Nat Rev Genet. 3: 662-73 Li E, Bestor TH, Jaenisch R (1992). Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69: 915-26 Liu Z, Xie Z, Jones W, Pavlovicz RE, Liu S,Yu J, Pui-kai L, Lin J, Fuchs JR, Marcucci G, Li C, Chan KK (2009). Curcumin is a potent DNA hypomethylation agent. Bioorganic & Medicinal Chemistry Letters 19: 3706-709 Lorch Y, LaPointe JW, Kornberg RD (1987). Nucleosomes inhibit the initiation of transcription but allow chain elongation with the displacement of histones. Cell 49: 203–210 Lorch Y, Maier-Davis B, Kornberg RD (2010). Mechanism of chromatin remodeling PNAS. 107: 3458-3462 Luger K, Richmond TJ (1998). The histone tails of the nucleosome. Currn Opin Genet Dev. 8:140-146 Lyko F, Ramsahoye BH, Jaenisch R (2004). DNA methylation in Drosophila melanogaster. Nature 8 : 538–540 Maegawa S, Hinkal G, Kim SH, Shen L, Zhang L, Zhang J, Zhang N, Liang S, Lawrence A, Donehower Issa JJ (2010). Widespread and tissue specific age-related DNA methylation changes in mice.Genome Res. 20: 332–340 Mann BS, Johnson JR, He K, Sridhara R, Abraham S, Booth BP, Verbois L, Morse DE, Jee JM, Pope S, Harapanhalli RS, Dagher R, Farrell A, Justice R, Pazdur R (2007). Vorinostat for treatment of cutaneous manifestationsof advanced primary cutaneous T-cell lymphoma. Clin Cancer Res. 13: 2318–22 Martens JA, Winston F (2003). Recent advances in understanding chromatin remodeling by Swi/Snf complexes. Curr. Opin. Genet. Dev. 13: 136–142. Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC, Baylin SB, Sidransky D (1995). CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16 CDKN2MTS1 in human cancers. Nat Med. 1: 686-692 Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, Alvarez P, Brockman W, Kim TK, Koche RP (2007). Genomewide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448: 553-560 Millar DS, Ow KK, Paul CL, Russell PJ, Molloy PL, Clark SJ (1999). Detailed methylation analysis of the glutathione S-transferase pi (GSTP1). gene in prostate cancer. Oncogene 18: 1313–24 Miller CA, Campbell SL, Sweatt D, (2008). Brief Report DNA methylation and histone acetylation work in concert to regulate memory formation and synaptic plasticity . Neur Learn Mem. 89: 599-603 Mo YY, Moschos SJ (2005). Targeting Ubc9 for cancer therapy. Expert Opin Ther Targets 9: 1203−1216 Morillon A, Karabetsou N, O'Sullivan J, Nicholas K, Nicholas P, Jane M (2003). Isw1 chromatin remodeling ATPase coordinates transcription elongation and termination by RNA polymerase II. Cell 115: 425–435 Muller S, Berger M, Lehembre F, Seeler JS, Haupt Y (2000). c-Jun and p53 activity is modulated by SUMO-1 modification.J Biol Chem. 275: 13321−13329 Nakayama T, Watanabe M, Suzuki H, Toyota M, Sekita N, Hirokawa Y, Mizokami A, Ito H, Yatani R, Shiraishi T (2000). Epigenetic regulation of androgen receptor gene expression in human prostate cancers. Lab Invest. 80: 1789–1796 Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN, Bird A (1998). Transcriptional repression by the methyl-CpG binding protein MeCP2 invloves a histone deacetylase complex. Nature 393: 386–389 Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN, Bird A (1998). MBD2 is a transcriptional repressor belonging to the MeCP1 histone deacetylase complex. Nature Genet. 23: 58–61 Nguyen CT, Weisenberger DJ, Velicescu M, Gonzales FA, Lin JC, Liang G, Jones PA (2002). Histone H3-lysine 9 methylation is associated with aberrant gene silencing in cancer cells and is rapidly reversed by 5-aza-2- deoxycytidine. Cancer Res. 62, 6456–6461 Noma K, Allis CD, Grewal SI (2001). Transitions in distinct histone H3 methylation patterns at the heterochromatin domain boundaries. Science 293: 1150-1155 Nowak SJ, Corces VG (2000). Phosphorylation of histone H3 correlates with transcriptionally active loci. Genes Dev. 14: 30033013. O’Connell B, Harper JW (2007). Ubiquitin proteasome system (UPS). what can chromatin do for you? Curr Opin Cell Biol. 19: 206–214 Okano M, Bell DW, Haber DA (1999). DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99: 247-257 O'Neill LR, Turner BM (1995). Histone H4 acetylation distinguishes coding regions of the human genome from heterochromatin in a differentiation-dependent but transcriptionindependent manner. The EMBO Journ.14: 3946-3957 Ooi L, Belyaev ND, Miyake K, Wood IC, Buckley NJ (2006). BRG1 chromatin remodeling activity is required for efficient chromatin binding by repressor element 1-silencing transcription factor (REST). and facilitates REST-mediated repression. J Biol Chem. 281: 3897438980 Oselu MA (2006). Regulation of histone H2A and H2B ubiquitylation. Brief Functi Genom Prot. 5: 179-189 Paluszczak J, Krajka-Ku´zniak V, Baer-Dubowska WB (2010). The effect of dietary polyphenols on the epigenetic regulation of gene expression in MCF7 breast cancer cells. Toxicology Letters 192: 119–125 Parekh BS, Maniatis T (1999). Virus infection leads to localized hyperacetylation of histones H3 and H4 at the IFN-b promoter. Mol Cell 3: 125–129 Park J, Park E, Hur S, Kim S, Kwon J (2009). Mammalian SWI/SNF chromatin remodeling complexes are required to prevent apoptosis after DNA damage. DNA repair 8: 29–39 Pesavento JJ, Yang H, Kelleher NL, Mizzen AC (2008). Certain and Progressive Methylation of Histone H4 at Lysine 20 during the Cell Cycle. Mol. Cell Biol. 28: 468–486. Pradhan S, Bacolla A, Wells RD, Roberts RJ (1999). Recombinant human DNA (cytosine-5)-methyltransferase I Expression, purification, and comparison of de novo and maintenance methylation. J Biol Chem.274 : 33002–33010 Pradhan S, Dale T, Sha M, Benner J, Hornstra L, Li E, Rudolf J, Roberts RJ (1997). Baculovirus-mediated expression and characterization of the full-length murine DNA methyltransferase.Nucleic Acids Res. 25: 4666–4673. Prokocimer M (2006). The nuclear lamina and its proposed roles in tumorigenesis, projection on the hematologic malignancies and future targeted therapy. J Struct Biol. 155: 351–360 Reik W, Dean W, Walter J (2001). Epigenetic reprogramming in mammalian development. Science 293: 1089–1093 Rideout WM, Coetzee GA, Olumi, AF, Jones PA (1990). 5Methylcytosine as an endogenous mutagen in the human LDL receptor and p53 genes. Science 249: 1288-1290 Robertson KD, Ait-Si-Ali S, Yokochi T, Wade PA, Jones PL, Wolffe AP (2000). DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F responsive promoters. Nature Genet. 25: 338–342 Robzyk K, Recht J, Osley MA (2000). Rad6-dependent ubiquitination of histone H2B in yeast. Science 287: 501–504 Rodenhiser D, Mann M, (2006). Epigenetics and human disease, translating basic biology into clinical applications. CMAJ. 174: 341348 Rogakou EP, Boon C, Redon C, Bonner WM (1999). Megabase chromatin domains involved in DNA double-strand breaks in vivo. J Cell Biol. 146: 905-916 Roh TY, Cuddapah S, Cui K, Zhao K (2006). The genomic landscape of histone modifications in human T cells. Proc Natl Acad Sci USA 103: 15782-15787 Saha A, Wittmeyer J, Cairns BR (2006). Chromatin remodelling, the industrial revolution of DNA around histones. Nat Rev Mol Cell Biol. 7: 437–447 Jha et al. 713 Santos-Rosa H, Caldas C (2005). Chromatin modifier enzymes, the histone code and cancer. Eur J Cancer 41: 2381-2402 Sasaki M, Tanaka Y, Perinchery G, Sasaki M, Tanaka Y, Perinchery G, Dharia A, Kotcherguina I, Fujimoto S, Choon A, Reznikoff CR, Bova SG, Friedl A, Jarrard DJ (2002). Methylation and inactivation of estrogen, progesterone, and androgen receptors in prostate cancer. J Natl Cancer Inst. (Bethesda) 94: 384–90 109. Schneider R, Bannistr AJ, Kouzarides T (2002). Unsafe SETs, histone lysine methyltransferases and cancer Trends Biochem Sci 27: 396-402 Schreiber SL, Bernstein BE (2002). Signaling network model of chromatin Cell 111: 771–778 Selker EU (1997). Epigenetic phenomena in filamentous fungi, useful paradigms or repeat-induced confusion? Trends Genet.13: 296–301 Shen X, Xiao H, Ranallo R, Wu WH, Wu, C (2003). Modulation of ATP Dependent Chromatin-Remodeling Complexes by Inositol Polyphosphates.Science 299: 112 – 114 Shi H, Wei SH, Leu YW, Rahmatpanah F, Liu JC, Yan PS, Nephew KP, Huang TH (2003). Triple analysis of the cancer epigenome, an integrated microarray system for assessing gene expression, DNA methylation and histone acetylation.Cancer Res. 63: 2164–2171 Shimono K, Shimono Y, Shimokata K , Ishiguro N, Takahashi M (2005). Microspherule protein 1: Mi-2beta, and RET finger protein associate in the nucleolus and up-regulate ribosomal gene transcription. J Biol Chem. 280: 39436–39447 Shumaker SK, Dechat T, Kohlmaier A, Adam SA, Bozovsky MR, Erdos MR, Eriksson M, Goldman AE, Khuon S, Collins FS, Jenuwein T, Goldman RD (2006). Mutant nuclear lamin A leads to progressive alterations of epigenetic control in premature aging. Proc Natl Acad Sci U S A 103: 8703–8708 Sims RJ, Chen CF,Santos-Rosa H, Kouzarides T , Patel SS, Reinberg D (2005). Human but not yeast CHD1 binds directly and selectively to histone H3 methylated at lysine 4 via its tandem chromodomains. J Biol Chem. 280: 41789–41792 Soengas MS, Capodieci P, Polsky D, Mora J, Esteller M, Opitz-Araya X, McCombie R, Herman JG, Gerald WL, Lazebnik YA, Cordón-Cardó C, Lowe SW (2001)., Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature 409: 207–211 Soriano FX, Papadia S, Bell KFS, Hardingham GE (2009). Role of histone acetylation in the activity-dependent regulation of sulfiredoxin and sestrin 2. Epigenetics 4: 152–158 Sourlingas TG, Tsapali DS, Kaldis AD and KE Sekeri-PataryaS (2001). Histone deacetylase inhibitors induce apoptosis in peripheral blood lymphocytes along with histone H4 acetylation and the expression of the linker histone variant, H1o. Eur. J. Cell. Biol. 80: 726-732. Steele N, Finn P, Brown R, Plumb JA (2009). Combined inhibition of DNA methylation and histone acetylation enhances gene reexpression and drug sensitivity in vivo .Journ Cancer 100:758 – 763 Strahl BD, Allis CD (2000). The language of covalent histone modifications Nature 403: 41- 45 Strahl BD, Ohba R, Cook RG, Allis CD (1999). Methylation of histone H3 at lysine 4 is highly conserved and correlates with transcriptionally active nuclei in Tetrahymena. Proc Natl Acad Sci USA 96: 14967-14972 Struhl K (1998). Histone acetylation and transcriptional regulatory mechanisms. Genes Dev 12: 599–606 Sun ZW, Allis CD (2002). Ubiquitylation of histone H2B regulates H3 methylationand gene silencing in yeast. Nature 418: 104–108 Swarthout J, Bagga S, Shivakumar S, Cao SL (2009). CancerResearch, Epigenetic Mechanisms Sigma Life Science, Sigma-Aldrich Understanding Epigenetics and its role in cancer development can help scientists to discover ways to deal with the disease. Saturday, August 01 Tate PH, Bird AP (1993). Effects of DNA methylation on DNA binding proteins and gene expression. Curr Opin Genet Dev. 3: 226–231 Tatham MH, Jaffray E, Vaughan OA, Desterro JM, Botting CH, Naismith JH, Hay RT (2001). Polymeric chains of SUMO-2 and SUMO-3 are conjugated to protein substrates by SAE1: SAE2 and Ubc9. J Biol Chem. 276: 35368–35374 Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ (2007). How chromatin-binding modules interpret histone modifications, lessons from professional pocket pickers. Nat Struct Mol Biol. 14: 1025-1040 Taverna SD, Li H, Ruthenburg AJ, Allis CD, Patel DJ, (2007). How chromatin-binding modules interpret histone modifications, lessons from professional pocket pickers. Nat Struct Mol Biol. 14: 1025-1040 Thiagalingam S, Cheng KH, Lee HJ, Mineva N, Thiagalingam A, Ponte, JF (2000). Histone deacetylases, Unique players in shaping the epigenetic histone code Ann N Y Acad Sci.983: 84-100 through histone modifications. Cell 103: 263-271 Trojer T, Zhang J, Yonezawa M, Schmidt A, Zheng H, Jenuwein T, Reinberg D (2009). Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine methyltransferase G9aKMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins. J Biol Chem. 284: 8395–8405 Tuck-Muller CM, Narayan A, Tsien F, Smeets D, Sawyer J, Fiala ES, Sohn O, Ehrlich M (2000). DNA hypomethylation and unusual chromosome instability in cell lines from ICF syndrome patients .Cytogenet Cell Genet. 89:121-128 Tweedie S, Charlton J, Clark V, Bird AP (1997). Methylation of genomes and genes at the invertebrate-vertebrate boundary. Mol Cell Biol. 17: 1469–1475 Van Holde KE (1989). Chromatin.Springer Verlag, New York Venkataramani V, Rossner C, Iffland L, Schweyer S, Tamboli I, Walter J, Wirths O, Thomas A, Bayer TA (2010). The histone deacetylase inhibitor valproic acid inhibits cancer cell proliferation via down-regulation of the alzheimer amyloid precursor protein.The Journ Biolog Chem. 285: 10678-10689 Waddington CH (1942). The epigenotype. Endeav. 1: 18-20 Wade PA, Gegonne PL, Jones E, Ballestar F, Wolffe AP (1999). The Mi-2 complex couples DNA methylation to chromatin remodelling and histone deacetylation. Nature Genet. 23: 62–66 Wang H, Zhai L, Xu J (2006). Histone H3 and H4 Ubiquitylation by the CUL4-DDB-ROC1 Ubiquitin Ligase Facilitates Cellular Response to DNA Damage. Molecular Cell 22: 383–394 West MHP, Bonner WM (1980). Histone 2B can be modified by the attachment of ubiquitin Nucleic Acids Res. 8 : 4671–4680 Wilson VL, Jones PA, (1983). DNA methylation decreases in aging but not in immortal cells. Science 220: 1055–1057 Wilson VL, Jones PA, (1983). DNA methylation decreases in aging but not in immortal cells Science 220: 1055–1057 Wu J, Smith LT, Plass C, Huang TH (2006). ChIP-chip comes of age for genome-wide functional analysis. Cancer Res. 66: 6899- 6902 Xu M, Long C, Chen X, Huang XC, Chen S (2010). Partitioning of Histone H3-H4 Tetramers During DNA Replication–Dependent Chromatin Assembly .Science 328: 94 – 98. Zhang Yi, Reinberg D (2001). Transcription regulation by histone methylation, interplay between different covalent modifications of the core histone tails, Genes & Dev. 15: 2343-2360. Zhang K, Dent SY (2005). Histone modifying enzymes and cancer, going beyond histones J Cell Biochem. 96: 1137-1144. Zhang Y, Reinberg D (2001). Transcription regulation by histone methylation, interplay between different covalent modifications of the core histone tails. Genes Dev. 15:2343-2360. Zhang J, Xu F, Hashimshony T, Keshet I, Cedar H (2002). Establishment of transcriptional competence in early and late S phase. Nature 420: 198–202. Zhou Q, Agoston AT, Atadja P, Nelson W G,Nancy E (2008). Ubiquitin-Dependent Proteasomal Degradation of DNA Methyltransferase 1 in Human Breast Cancer Cells. Mol Cancer Res. 6: 873