CHAPTER
DNA methylation and chromatin modifications
2
Zahra Sepehri, Tasnim H. Beacon, Fadumo D.S. Osman, Sanzida Jahan, James R. Davie Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
1. Epigenetics and chromatin organization Epigenetic refers to a variety of processes that have heritable effects on gene expression programs without changes in DNA sequence [1,2]. Key players in epigenetic control are DNA methylation and histone modifications which, in concert with transcription factors, chromatin remodeling complexes, nuclear architecture and non-coding RNAs, define the chromatin structure of a gene and its transcriptional activity. Cellular differentiation is initiated and maintained by epigenetic mechanisms. Although epigenetic marks are established early during development and differentiation, adaptations occur throughout life in response to intrinsic (e.g. oncogene expression) and environmental stimuli (e.g. diet) and may lead to late life disease (e.g. cancer). Thus, the life of an individual is not only defined by his/her genome, but also by his/her numerous epigenomes, with different epigenomes being generated through development, not only during fetal development but also during the plastic phase of early childhood, and existing in different cell types. Moreover, epigenomes react to environmental influence including maternal care, diet, exposure to toxins and xenobiotics. Epigenetic responses to environmental stimuli may have long-term consequences, even affecting future generations. The task ahead of us, to decipher all normal epigenomes and dysfunctional epigenomes leading to the vast array of diseases and cancers is colossal. The basic repeating structural unit in chromatin is the nucleosome. The nucleosome consists of a histone octamer, arranged as an (H3eH4)2 tetramer and two H2A-H2B dimers, around which DNA is wrapped. The H1 histones bind to the DNA, the linker DNA that join the nucleosomes together [3]. The core histones (H2A, H2B, H3, H4) have a similar structure with a basic N terminal domain, a globular domain organized by the histone fold, and a C terminal tail. The N terminal tails emanate from the nucleosome in all directions and interact with linker DNA, nearby nucleosomes or with other proteins [4]. The histones undergo numerous reversible post-translational modifications (PTMs), including acetylation, methylation and phosphorylation [5]. Histone PTMs are added and removed from specific sites by a variety of enzymes [6]. Some histone PTMs (active marks) are associated with transcribed chromatin regions, while others (repressive marks) are present in silent regions. Histone acetylation and H3K4me3 are active gene marks, whereas H3K27me3 is a repressive mark. Histone PTMs function to alter chromatin structure and/or provide a “code” for recruitment or occlusion of Nutritional Epigenomics. https://doi.org/10.1016/B978-0-12-816843-1.00002-3 Copyright © 2019 Elsevier Inc. All rights reserved.
13
14
Chapter 2 DNA methylation and chromatin modifications
nonhistone chromosomal proteins to chromatin. The nucleosomal DNA may also be methylated (five methyl cytosine) [7]. Thus, the nucleosome serves multiple roles in regulating gene expression, a structural role and a role as a signaling module [8,9]. Adding to this complexity, most histones have variants, products of different genes [10]. Vertebrate histone variants are classified as replication-independent and replication-dependent. For example, H3.1 and H3.2 are replication-dependent, while H3.3 is replication-independent. Being synthesized throughout most of the cell cycle, the H3.3 variant is available to re-build nucleosomes displaced by transcription factors and the transcription machinery [11]. H3.3 is enriched in PTMs that are associated with transcriptional activity (for example, S28ph, K9ac, K14ac, K4me3) [12e14]. Mutations in H3.3 are associated with several cancers (e.g. pediatric glioblastoma) [15]. The structure and composition of a nucleosome depend to a certain extent on the DNA sequence associated with the core histones. A nucleosome with a DNA sequence that has binding sites for pioneer transcription factors and located in a tissue-specific enhancer may have an accessible configuration [16]. A pioneer transcription factor such as FoxA1 can bind to its binding site in a nucleosome context and in a repressive heterochromatin environment [17]. Nucleosomes associated with non-methylated CpG islands may be rich in H3K4me3. SETD1A/B, which catalyzes trimethylation of H3K4, has among its subunits, CXXC1 (also known as CFP1), a CpG island binding protein [18]. A nucleosome that has H3K4me3 attracts multiple lysine acetyltransferases (KATs) like HBO1 and SAGA which contain subunits that bind to K4me3. Thus, a nucleosome with H3K4me3 will have much different acetylation dynamics than a nucleosome next to it that does not have this modification [18e20]. Nucleosome structure is also radically changed during the process of transcription, which generates atypical nucleosomes known as U-shaped nucleosomes [21,22]. The open, U-shaped nucleosome will retain this altered configuration as long as its acetylation status remains high [23].
1.1 Three-dimensional structure of chromatin The organization of the genome plays an important role in the regulation of gene expression (Fig. 2.1). With the advancement of high-resolution techniques such as 3C and 3C based strategies (e.g. 3C, 4C, 5C, ChIA-PET and Hi-C analysis) [24], the 3D structure of chromatin has become an important model to address questions such as how the conformation of this structure controls interactions of regulatory elements, transcriptional activities and what factors/key players are involved in the maintenance of such conformation. High-resolution mapping of nucleosomes has allowed us to study misregulation of nucleosome positioning at the basic level of compaction of chromatin [25]. Chromatin is further organized into functional territories based on their state of condensation into euchromatin and heterochromatin. A higher level of compaction is required to fit the DNA and interacting proteins within the nucleus. As research advances to understand the underlying mechanism of how chromatin organization impacts gene regulation, major efforts have been made to understand the organization of these epigenetic compartments, map their interaction and determine their spatial arrangement [26]. Chromatin has different levels of spatial organization ranging from the nucleosome to domains. Current literature subcategorizes these spatial scales into three levels: large, intermediate and small. Compartmentalization and chromatin interactions are confirmed mainly by microscopic observations and by the Chromosome Conformation Capture (3C) technique. Microscopy is used to detect fluorescent probes used to track the spatial location of whole chromosomes and the relative positions of locus of interest, while 3C methods quantifies the interaction of two distal segments of the DNA
1. Epigenetics and chromatin organization
15
FIG. 2.1 Organization of Chromatin in the Interphase Nucleus. The chromosome territories in the interphase nucleus are composed of chromatin compartments (A/B). The A-compartments are active and are associated with the active marks and their catalyzing enzymes (KMTs, KDMS, KATs and HDACs). The B compartments are found near the nuclear lamina and contain the repressed mark and associated enzymes (PcG and JmJD). The compartments are further organized into TADs each individually regulating the genes and regulatory elements within the insulating loop. The TAD boundaries are the regions where the CTCF and cohesin complexes bind. Credit: James R. Davie.
based on ligation events that takes place between the chromatin fragments after the steps of fixation and digestion. The proximity relative to one another is then used to determine their genomic location [24]. These organizations are commonly termed A/B compartments, topologically associating domains (TADs), and chromatin loops [27] (Fig. 2.1). Segregating the nuclear organization into different scales has facilitated the study of individual chromatin components correlating their dimensional interaction and overarching function in gene regulation [28]. 3C techniques have been promising in providing insight into the 3D arrangement, while the Hi- C technique appreciates and quantifies the interactions leading to average chromosome conformations. Initially the compartments were defined as closed and open chromatin compartment. The Hi-C mapping was performed at 1 Mb and as low as 100 kb and was validated with 3D-Fish to confirm the proximity [27]. With the advent of higher resolution, the structural domains were classified into 2 major compartments in the interphase stage: A compartment and B compartment (Fig. 2.1). The A compartment houses active chromatin (H3K36me3), while the B compartment is associated with
16
Chapter 2 DNA methylation and chromatin modifications
inactive chromatin (H3K27me3). The active state is defined by transcriptional activity, associated histone modifications, and accessible DNA. Active chromatin domains are distinguished by their increased sensitivity to DNase I and association with highly acetylated histones [29e31]. Note that DNase I hypersensitive sites differ from that of DNase I sensitive regions. DNase I hypersensitivity marks regulatory elements in the genome that are typically nucleosome depleted and detected by techniques such as FAIRE and ATAC [29,32e34]. DNase I sensitivity encompasses much broader chromatin regions and correlates with histone acetylation status [35,36]. A/B compartments differ in chromatin composition and histone modifications, which identifies the compartment as active and inactive, respectively. The compartments stay constant in the genome throughout development [37]. However, changes to their associated histone modifications, DNAse I sensitivity or composition can lead to a change in their compartmentalization [38]. A pioneer transcription factor has the potential to convert a B to an A compartment [17]. The A compartment is organized into an inner ring-shaped structure, while the B-compartment is associated with the nuclear lamina and the edges of nucleoli within the nucleus. Single-cell Hi-C studies reveal the presence of these compartments in the only interphase cell. While chromosomes attain different conformations in different cells, the A/B compartments spatially segregate both within chromosomes and globally within nuclei [39,40]. At a more intermediate scale, self-interacting domains are seen which typically range in size from tens of kilobases to a few megabases usually comprising of a small number (e.g., 1e10) of genes. These interacting DNA sequences are commonly termed as ‘Topologically Associating Domains’ (TADs) but also are seen to be defined as sub-TADs, ‘contact domains’ and ‘insulated neighbourhoods’ based on their interaction and regulatory functions [37] (Fig. 2.1). TADs play an important role in domain organization. Experiments involving duplication, deletion or inversion of TADs or TAD boundaries have caused physiological malformations. Therefore, disruption of TADs can have significant effects on gene expression resulting in pathogenic phenotypes, indicating its importance in maintaining genomic architecture [41]. TADs function independently of the compartments [42]. The TAD boundaries interact more frequently with each other compared to the rest of the domain and result in a large fraction of peak (38%) when captured with Hi-C. This peak marks the binding of the CTCF and the cohesin complex [43]. The loop domain formed between CTCF and cohesin binding sites buffers the genes from the influence of enhancers residing in different domains [41,44]. The CTCF motifs of the insulating loop are always oppositely positioned marking the boundaries of the loops [39]. These boundaries are sensitive to DNAse I digestion, which indicates the low density of nucleosomes [45]. Therefore, altering the level of the nucleosome remodeling protein BRG1 was seen to change the density of nucleosomes at the boundary and affect boundary strength as well as CTCF binding [46]. While the loss of cohesin and CTCF was shown to have no effect on the compartmentalization of chromatin and the associated histone marks, it resulted in loss of TADs. TADs are segregated from each other by the formation of an insulating loop, which prevents the enhancer from one TAD to control the transcription of a gene in another TAD leading to insulated neighborhood where the gene and its regulatory elements are unaffected from external influence [47]. One exciting feature of the loop is its dynamic nature. It is observed to be present transiently while in a constant state of forming and collapsing resulting in an unstable structure [48]. One of the successful models describing the insulating loop formation is the ‘loop extrusion model’ where a loop extrusion factor (LEF) would bind to the chromatin and will initiate loop formation with the help of the boundary factors. The loop structure can incorporate additional LEF causing the formation of a secondary loop. The loop
2. DNA methylation
17
eventually extends to reach the TAD boundaries and are temporarily stabilized [49]. The loop will then disrupt shortly following the dissociation of the LEF and boundary factors. This model is useful to predict the role of CTCF at the TAD boundaries while setting the stage to hypothesize models to understand the underlying mechanism of their function in maintaining the conformation of the 3D structure of the chromatin. To understand how the large and intermediate level structures complement each other, experiments have been designed to explore how altering one structure affected the other. While the loss of CTCF and cohesin-associated factors (Rad21 and Nipbl) led to the disruption of the TADs and loops, deletion of WAPL (cohesin antagonist) had the opposite effect. Meanwhile loss of CTCF and cohesin-associated factors had no effect on the A/B compartment structures, indicating TADs and A/B compartments to be independent structures [50]. Another important factor was the contribution of these structures in different species. Experiments have shown that mammals show strong CTCF/cohesin loop anchors at TAD boundaries [50], whereas in Drosophila CTCF occupancy at the TAD boundaries are comparatively less and are not usually in inverted orientation [28]. These structures can be concluded as species-dependent for the factors involved in domain formation. Although the variation in abundance of CTCF and cohesin has led to different theories in their role in gene regulation, different theories have been speculated on their function leading up to the 3D organization of the chromatin. On a small-scale chromatin organization, variations to the 3C methods have been applied to detect enhancer-promoter interaction, which functions to regulate transcription. To map the interactions of the regulatory region, techniques such as OCEAN-C have been a helpful tool to identify hubs of open chromatin regions (HOCIs) and their regulatory functions. The cells are fixed, digested, ligated, and sonicated to desired fragment sizes. Following the phenol-chloroform extraction, the biotinylated DNA is analyzed for sequencing. As the technique combines FAIRE-seq and Hi-C, it eliminates the limitations of the techniques by improving the background and only captures the peaks representing open chromatin interaction. Important HOCI interactions include promoter-enhancer, promoterpromoter, and enhancer-enhancer interactions. These interactions are key to study the changes in open chromatin conformations as well as the regulation of the transcription of genes localized in proximal and distal domains [51]. Overall, studying chromatin in different scales has greatly advanced our understanding of the 3D architecture of chromatin by defining compartments, TADs and loops. Studying the interactions of the regulatory elements provides a topological basis to correlate open chromatin conformations to transcriptional regulation [52]. Knowledge of CTCF and cohesin factors along with the individual chromatin structures have laid the foundation of the mammalian 3D genome to explore conformational changes and identify novel epigenetic regulators mediating gene expression.
2. DNA methylation DNA methylation is a crucial epigenetic modification found in the genome of various organisms. It is involved in development, differentiation, tissue-specific gene expression and cellular function. Moreover, it plays a fundamental role in epigenetic reprogramming, X-chromosome inactivation and genomic imprinting. Recently, an interesting discovery of the role of DNA methylation in individual CpG sites as an epigenetic age-predictor in both human and mice was made [53,54]. Despite the availability of such tools for predicting biological age, which is quite different from chronological age, a majority of people are not willing to examine CpG DNA methylation for age prediction.
18
Chapter 2 DNA methylation and chromatin modifications
However, this information is clinically relevant as a potential biomarker and in determining better therapeutic options for individuals according to their biological age. In mammals, methylation involves the addition of a methyl group at position five of the cytosine ring in the CpG dinucleotides by DNA methyltransferase enzymes (DNMTs) [55,56]. There are two classes of methyltransferases: the de novo methyltransferase (DNMT3a and DNMT3b) and the maintenance methyltransferase (DNMT1). The de novo methyltransferases establish the methylation mark onto the unmethylated CpG region whereas DNMT1 maintains the methylation through addition of methyl groups to hemi-methylated regions during replication. DNMT3l is a DNMT-related protein which binds H3 at lysine 4 (H3K4) and helps in guiding DNMT3a and DNMT3b to their target DNA region [57,58]. In addition to the enzymatic activity of DNMTs, these enzymes can contribute to gene inactivation through modifying the chromatin by binding to both histone deacetylase (HDAC) and lysine methyltransferases (SUV39h1/2 and G9; enzymes which methylate H3K9) [59]. Five-methylcytosine (5-mC) is predominantly found at CpG-rich sites but there are exceptions [60]. For example, human neurons and embryonic stem cells show intragenic methylation in a non-CpG context [61]. CpG islands, located usually at the 5’ end of genes, are generally unmethylated, and their methylation is more often restricted to genes which require stabilization for long-term silenced state such as imprinted genes and genes exclusively expressed in germ cells rather than somatic cells. The function of methylation varies with different genomic contexts [62,63] (Fig. 2.2). For example, methylation in the gene bodies supports gene expression and does not prevent transcriptional elongation. Recently, intragenic DNA methylation in vertebrates was shown to prevent cryptic transcription initiation within the coding region of expressed genes [64]. The intragenic methylation is catalyzed by DNMT3b, which is recruited to the gene body by H3K36me3 (catalyzed by SetD2) (Fig. 2.2). This mechanism involving crosstalk between SetD2, H3K36me3, DNMT3b and 5-mC is mediated by elongating RNA polymerase II [64]. By contrast, DNA methylation of promoter regions and transcription start sites blocks initiation and has been linked to gene silencing, either directly through blocking the binding of transcription factors [65] or indirectly through the involvement of methyl-CpG-binding proteins, which in turn recruit transcriptional repression and chromatin remodeling complexes [66] (Fig. 2.2). The methyl CpG-binding protein (MBP) family is responsible for identifying methylation including MBD1, MBD2, MBD4, MeCP2 and Kaiso. All the MBPs contain a methyl-CpG binding domain (MBD) that specifically recognizes methylated DNA [67] except for Kaiso, which depends on its zinc-finger (ZF) domain in the C terminus for methyl- CpG recognition [68]. Moreover, these proteins can function as methylation-dependent transcriptional repressors. Interestingly, not all transcription-factor binding is blocked by methylation as some factors like SP1 can bind strongly to methyl-CpG sequences and mediate gene expression through the passive removal (replication and enzyme independent demethylation) of 5-mC [69,70]. In the context of repetitive DNA sequences, methylation plays a repressive role by silencing transposable elements [71]. Similar effects are observed in methylated regulatory elements where the enhancer’s activity is reduced [72] and the binding of CTCFs is blocked [73]. However, for some enhancers, cytosine methylation is involved in maintaining an active enhancer state [74]. Also to note, several transcription factors, like SP1, bind to methylated DNA [75]. Overall, as the interpretation of 5-mC changes in a specific genomic and cellular context from another, its impact has gone beyond gene expression control to include genome stability and splicing regulation [61].
2. DNA methylation
19
FIG. 2.2 The Role of DNA Methylation in Gene Expression. (A) The CpG islands (CGI) within the promoter region and transcriptional start site (TSS) are normally unmethylated in active genes. The DNA methyltransferases (DNMT) are recruited by H3K36me3 to the gene body. Intragenic methylation (me) is positively correlated with active transcription and does not block the elongation process, but spurious transcription initiation is inhibited. (B) In inactive genes, methylation of promotors inhibits transcription initiation. The addition of methyl group is coupled with repressive chromatin modifications by DNMT-linked lysine methyltransferase (KMT) and histone deacetylase (HDAC). Additionally, methyl-binding domain (MBD) proteins recognize methylated regions and recruit chromatin remodeling corepressor complexes. Credit: James R. Davie.
2.1 DNA demethylation There are different pathways used in different tissues for the removal of 5-mC [76]. In mammals, 5-mC is passively demethylated during replication and cell division, while active demethylation occurs in cycling and non-replicating cells like neurons. In the oxidation pathway, 5-mC is oxidized by TET enzymes to 5-hmC which can be further oxidized to 5-formlycytosine (5-fC) and 5-carboxylcytosine (5-caC). 5-fC and 5-caC are cleaved by thymine DNA glycosylase (TDG) resulting in the involvement of the base excision repair (BER) machinery. On the other hand, the deamination pathway involves first the deamination of 5-mC by the activation-induced cytidine deaminase (AID)/apolipoprotein B mRNA editing enzyme, catalytic polypeptide (APOBEC) family producing a T:G mismatch identified by TDG which in turn creates an abasic site to be repaired by the BER machinery [76].
20
Chapter 2 DNA methylation and chromatin modifications
In 2009, the first member of the ten-eleven translocation family of methylcytosine dioxygenases (TET1) was discovered to be responsible for 5-mC oxidation and demethylation to 5-hydroxymethylcytosine (5-hmC) [77]. The TET family of enzymes is characterized as iron- and oxoglutarate-dependent enzymes with their respective binding domains. In contrast to DNMT1, the CXXC zinc-finger DNA-binding domain of TET1 and TET3 has the ability to bind both methylated and hydroxymethylated DNA regions [78]. Five-hmC levels in adult tissues are moderately low ranging between >0.1 % and w0.7% with the highest level in the tissues of the central nervous system [79]. Five-hmC does more than its role as a demethylation intermediate, and TET enzymes are more than their catalytic function. Hydroxymethylation is enriched in euchromatic regions within the gene bodies, TSS and upstream of TSS. Gene expression can be controlled either negatively or positively by 5-hmC and TETs. The presence of 5-hmC in gene bodies prevents the binding of the DNA methylation machinery and MBPs allowing for the activation of genes [80e82]. However, TET enzymes can play another role in silencing genes by their interaction with SIN3A co-repressor complex [83]. Furthermore, the presence of 5-hmC in the case of bivalent genes (transcriptionally poised genes) facilitates the recruitment and binding of polycomb repressive complex 2 (PRC2), which catalyzes the trimethylation of H3 at lysine 27, thus maintaining the repressed state of these genes [84].
2.2 Mitochondrial DNA methylation Unlike nuclear DNA, mitochondrial DNA (mtDNA) has a 16,569-base-pair circular structure and lacks histones. It consists of a heavy (H) strand and a light (L) strand, encoding 37 genes: 13 oxidative phosphorylation-related protein-encoding genes, 22 transfer RNAs and 2 ribosomal RNAs [85]. MtDNA is believed to be modified by both 5-mC and 5-hmC, however, the presence and function of such epigenetic modifications are still highly debated. In 2011, a mitochondrially targeted isoform of DNMT1 (mtDNMT1) was discovered and accounts for 1e2% of total DNMT1 transcripts. Hypoxiaresponsive transcription factors such as peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1a) and nuclear respiratory factor 1 (NRF1) are responsible for regulating the mtDNMT1 expression [86]. Moreover, other DNMTs and TET enzymes were identified to be present in the mitochondria in a tissue-type-dependent manner. For example, the localization of DNMT3A in excitable tissues such as skeletal and heart muscles [87]. DNA methylation and hydroxymethylation patterns are observed across the mitochondrial genome, particularly their main location in the D-loop region where all the major promoters are located. Interestingly, more non-CpG compared to CpG is methylated in the D-loop region [88]. To date, the findings supporting the functional and regulatory roles of mtDNA methylation are based on associations without confirmatory evidence on its mechanism of action. The changes in mitochondrial gene transcription as a result of mtDNMT1 upregulation are not random, but rather genespecific leading to the activation of ND1 and repression of ND6 [86]. Transcription of mtDNA is dependent on mainly three factors: mitochondrial transcription factor B2 (TFB2M), mitochondrial transcription factor A (TFAM) in addition to mitochondrial RNA polymerase (POLRMT). Methylation might influence the ability of TFAM to bind the promoter regions directly or by the recruitment of proteins that add post-transitional modifications to TFAM [89]. Further work is required to fully understand and determine the detailed mechanism as well as to identify the players regulating mitochondrial DNA methylation.
3. Histone modifications and their distribution in the genome
21
3. Histone modifications and their distribution in the genome Histones (H1, H2A, H2B, H3, H4) are basic proteins subject to a multitude of PTMs, which are mostly reversible [5,90]. Histone PTMs can affect almost all genomic events such as transcription, replication, recombination, DNA repair, kinetochore, chromatin remodeling and centromere formation [91]. Together with histone modifying enzymes and proteins that recognize specific histone PTMs, which are categorized as “Reader,” “Writer,” “Eraser,” “Effector” and “Presenter”, histone PTMs can regulate the transcriptional state [92,93]. Changes in histone PTMs can alter the active chromatin state into an inactive state and vice versa. Histone crosstalk is defined as a combination of histone PTMs that can code for transcriptional activation or repression in a context-dependent manner [94,95]. Histone PTM crosstalk can occur either cis or trans, involving events on the same histone tail or nearby histone tail within the same or neighboring nucleosome [96]. For example, it was demonstrated that serine 10 phosphorylation on H3 enhances the GCN5 mediated acetylation of H3 at lysine 14 [97]. Histone crosstalk can be initiated by preventing the nearby histone PTM. Indeed, H3 asymmetric di-methylation (H3R2me2a) was shown to prevent the MLL mediated formation of di- and tri-methylation of H3 lysine 4 (H3K4me3/H3K4me2) [98]. Interestingly, the presence of H3K4me3 prevents protein arginine methyltransferase (PRMT) 6-mediated H3R2me2a [98]. The advancement in the technology with tools such as chromatin immunoprecipitation (ChIP) and ChIP-sequencing (ChIP-seq) enables one to determine the crosstalk between different writers and readers or effector molecules. Lysine acetylation (all histones), lysine and arginine methylation (H3, H4, and H2B), serine and threonine phosphorylation (all histones), lysine ubiquitination (H2A, H2B), ADP- ribosylation at glutamine (H1) and sumoylation are some of the well-known histone PTMs [5]. Several of the histone marks are exclusively associated with active chromatin state (H3K4me3, H3K27ac), while others are with the inactive chromatin state (H3K27me3) [96].
3.1 Histone acetylation Histone acetylation was first reported by Vincent Allfrey and his group in 1964. Allfrey’s group described the dynamic and rapid histone acetylation using nuclei isolated from calf thymus [99]. Following on this study, they later reported that histone acetylation occurs on ε-amino lysine residue, and they also identified histone deacetylase (HDAC) activity in the nuclei. In 1978, both Dr. Davie and Dr. Allfrey reported for the very first time that n- butyrate acts as an HDAC inhibitor [100,101]. Hyperacetylation of H3 and H4 and to a lesser extent H2A and H2B were observed upon sodium butyrate treatment in the cell line investigated [100]. DNA sequences associated with the hyperacetylated histones showed increased DNase I sensitivity in HeLa and chicken erythrocyte cells [101]. The first report of a direct link between histone acetylation and transcriptionally active chromatin came from the study by Dr. Crane Robinson’s group using chicken erythrocytes [102]. In this study, the ChIP assay was used for the first time to demonstrate that acetylated histones are associated with transcriptionally active DNA sequences [102]. The relationship between histone acetylation and transcription became established after the discovery of KATs were co-activators [103]. KATs are categorized into four different groups; GCN5, MYST (SAS/MOZ), P300/CBP and SRC/p160 nuclear receptor coactivator family [104]. Acetylation of histone and non-histone proteins is catalyzed by KATs [105]. Dynamic and reversible histone acetylation is catalyzed by KATs and HDACs. The rate of histone acetylation can vary across the genomic regions with some regions having a faster rate of dynamic acetylation while some have slower or none [104].
22
Chapter 2 DNA methylation and chromatin modifications
Histone acetylation can prevent H1-mediated salt insolubility, facilitating solubility of the region at physiological salt concentration [35,106,107].
3.2 Histone lysine methylation Lysine and arginine located in the histone N-terminal tails are methylated by lysine methyltransferases (KMTs) or PRMTs [5]. Mono-, di- or tri-methylation of lysine and mono- or di-methylation of arginine can be distinguished as active or repressive chromatin marks [108,109]. H3K4me1 along with H3K27ac typically marks active enhancers. H3K27me3 is a strong repressive mark catalyzed by the polycomb complex [110]. With our current knowledge of the histone PTMs associated with regulatory elements such active enhancers, poised enhancers, upstream promoters and silenced genes, we can now read histone PTM tracks and have a good idea of the function of the associated DNA sequence [111]. H3K4me3 is an active mark found in the upstream promoter region and strongly positioned after the first exon of transcribed genes [112]. The mitotic inheritance (memory) of the transcriptional state of genes is dependent upon H3K4me3 [113]. Bryan Turner’s group noted that the H3K4me3 regions on mitotic chromosomes coincided with CpG islands, which are found at the promoter and 50 regions of genes [18,114]. H3K4me3 has several roles in the regulation of gene expression. H3K4me3 located in the 50 coding region of expressed genes interacts with the proteins involved in pre-mRNA splicing (reviewed in Ref. [18]). Although most genes have H3K4me3 limited to the promoter region and the 50 region of the gene body, a small subset of genes have broad H3K4me3 regions (called H3K4me3 buffer domains). The function of H3K4me3 buffer domain is to ensure transcriptional consistency of the gene, that is, the same transcription rate in each cell [113,115]. Of note, these genes with the H3K4me3 buffer domains have a role in cell identity [115]. The top 5% broadest H3K4me3 regions in a given cell type were most enriched for genes that had important functions for that specific cell type (for example, genes involved in muscle function in skeletal muscle or stem cell regulators in embryonic stem cells) [115]. In normal but not cancer cells, tumor suppressor genes have broad H3K4me3 peaks [116]. Importantly genes coding for transcription factors, which were critical for cell identity or cell fate, have H3K4me3 buffer domains in the relevant cell type/tissue. Through the identification of genes with the H3K4me3 buffer domains, Anne Brunet and colleagues found genes coding for transcription factors and non-coding RNA that were novel regulators of neural progenitor cells [115]. Knocking down the genes with the broadest H3K4me3 domains resulted in decreased neural progenitor cell proliferation and decreased neurogenesis. Lysine and arginine methylation of histones can serve either as a binding site or occlude the binding of other modifiers to the site and thereby play a crucial role in histone PTM mediated signaling events. Due to the existing signaling event, aberrant binding of the modifying enzymes can lead to diseased state as observed for several cancers [117e119]. EZ, SET1, SET2, SMYD, SUV39, SUV4-20, RIZ are among the major family of lysine methyltransferases [120]. S-adenosyl methionine (SAM) serves as methyl donor and co-factor for both KMTs and PRMTs [121].
3.3 PRMTs and histone arginine methylation Eleven mammalian PRMTs have been discovered and reported to date. Mono and dimethylation of arginine are catalyzed by three classes of PRMT enzymes. PRMTs catalyze arginine methylation by
3. Histone modifications and their distribution in the genome
23
using molecule of S-adenosyl-L-methionine (AdoMet) to form asymmetric (u-NG,NG-dimethylarginine or ADMA) or symmetric (u-NG,N0 G-dimethylarginine or SDMA) or monomethylarginines (MMA). Type I PRMTs, which asymmetrically dimethylate arginine, include PRMT1, 3, 4, 6 and 8, with PRMT1 accounting for most of the type I PRMT activity. PRMT5 and 9 are involved in symmetric dimethylation of arginine and belong to type II, while PRMT7, a type III methyltransferase, contributes to monomethylation of arginine [122]. Therefore, PRMTs have a wide range of substrate specificity. Arginine methylation of proteins and histones catalyzed by PRMTs can be either symmetrical or asymmetrical, and they are categorized based on this chemical feature [123]. Similar to lysine methylation, arginine methylation can contribute to activate or repress the chromatin state in a contextdependent manner [124]. H4R3me2a, an active chromatin mark, is catalyzed by PRMT1, whereas PRMT5 is responsible for the symmetric dimethylation of H4R3 (repressive mark). PRMT6 is the major methyltransferase responsible for the genesis of H3R2me2a (repressive mark) in vivo. H3R2me2s, formed by PRMT5, recruits WDR5, which is a subunit of several co-activator SET1/MLL complexes that produce H3K4me3 (an active mark) [125,126]. Other histone arginine methylations are listed below. H3R17me2a: This arginine modification is catalyzed by PRMT4/CARM1 and is associated with transcriptional activation. This mark is located at the upstream promoter of several genes [119,127,128]. Moreover, it was shown that prior acetylation of H3K18 and K23 promotes H3R17 methylation and activation of estrogen-responsive gene TFF1 [129,130]. H3R26me2a: PRMT4/CARM1 produces H3R26me2a, which is a less studied mark. Similar to H3R17me2a, this mark was also found associated with the upstream promoter region of target genes [131]. H3R2me2a: PRMT6 produces H3R2me2a. This mark has an intriguing feature as it can antagonize H3K4me3 through blocking the binding of WDR5 to the site [132]. This mark is associated with repressive chromatin and found in pericentromeric regions [133]. H3R2me2s: This mark is associated with transcriptionally active chromatin regions and is generated by PRMT5. H3R2me2s recruits WDR5, which is found in complexes such as MLL, SET1A, and SET1B, which catalyze H3K4me3. Therefore, H3R2me2s is located with H3K4me3 [134]. H4R3me2a and H2AR3me2a: PRMT1 is involved in the generation of these two modifications. H4R3me2a was found associated with the upstream promoter regions of several genes [135e138]. H4R3me2a was reported to facilitate the subsequent acetylation of H4 at Lys 8 and 12 and of H3 at Lys 9 and 14 by p300, and therefore H4R3me2a can be considered as an active mark that recruits the KATs, p300 and PCAF [135,139]. Acetylation of H4 Lys 5 results in reduced arginine methylation by PRMT1 but increased activity by PRMT5 [140,141]. Highly acetylated H4 that has H4K5ac is strongly detrimental to PRMT1 mediated H4R3 methylation. In contrast, acetylation of H4 at K16 is a better substrate for PRMT1 than unmodified H4 [141]. This is an interesting observation as H4K16ac is involved in decondensing chromatin [142]. H4R3me2s and H2AR3me2s: PRMT5 generates the repressive marks, H4R3me2s and H2AR3me2s. H4R3me2s was found associated with the upstream promoter region of several silenced genes and with imprinting control regions [143,144]. H3R8me2s: This is a repressive chromatin mark generated by PRMT5 [143]. Prior acetylation was shown to prevent H3R8me2s [145].
24
Chapter 2 DNA methylation and chromatin modifications
3.4 Arginine demethylation Protein arginine deiminase (PAD) family of enzymes catalyze citrullination from the amino acid arginine. To date, PADs, PAD1-4 and PAD6, have been identified [146]. PADs replace the ketamine (¼NH) group of arginine to keto group (¼ O), thereby resulting in no net charge from the positively charged arginine. This change in charge due to citrullination alters the structure and function of the protein as well affect the binding of protein interacting partners [147]. It was reported that both PAD2 and PAD4 could catalyze citrullination on a histone tail, albeit it is PAD4 which is involved in citrullination of monomethyl arginine [148,149]. Symmetric and asymmetric mono and dimethylation of arginine H3/H4 were reported to be catalyzed by Jumonji domain-containing 6 protein (JMJD6) [150]. However, later it was shown that JMJD6 is involved only in the demethylation of mono- and di-methyl H4 arginine residues [151]. Although JMJD6 has been reported as a candidate for demethylation of arginine, there is still a lack of sufficient biochemical evidence for that. Moreover, demethylation of H3R2 has not yet been detected.
4. Metabolism and epigenetics Metabolism is a set of life-saving chemical modifications in the cells [152]. Every cell has a set of responses to changes in its environment. These responses depend on various factors such as cell type and duration of changes, to name a few. Overall, nutrition as an environmental input modulates cell metabolism, and in turn cell metabolism regulates epigenetic processes [153]. More precisely, the dynamic range of physiological concentrations of commensurate intermediates in metabolism will characterize the “kinetics” of crosstalk between metabolism and chromatin [154] (Fig. 2.3).
4.1 Metabolism and chromatin modifications Enzymes that are responsible for histone and DNA modifications need metabolites such as acetyl or methyl groups to do their enzymatic reactions specifically and efficiently. Availability of these metabolites and their localization in the cell play major roles in the activity of the enzymes regulating DNA and histone modifications. For example, efficiency of KATs depends on local subcellular acetylCoA concentration, which is produced during catabolism and anabolism [152,153]. In a nutrient favorable situation, that is, there is a lot of acetyl-CoA precursors entailing glucose, galactose or ethanol, adenosine triphosphate citrate lyase will convert glucose derived citrate into acetyl-CoA in the nucleus [152]. On the other hand, class III HDACs (the sirtuins) deacetylate histones and non-histone proteins dependent on the concentration of available local nicotinamide adenine dinucleotide (NADþ). The Sirtuin family of HDACs (SIRTs) “sense” the positive effects of caloric restriction on physiology of the cell and are a key controller in mitochondrial energy metabolism, senescence of the cell, inflammation and most importantly tumorigenesis [153]. Yet, they are NAD dependent [152]. When fasting happens, available NADþ and activity of SIRT1 are high. In contrast when energy resources are high, NADþ would be converted to NADH and consequently its concentration would decrease. In aging and diabetes, NAD levels significantly decrease [152]. Actually, SIRTs are the only HDACs that are dependent on an endogenous metabolite such as NAD [153]. Although SIRT6 has high affinity to bind to NAD, its dependency to NAD is less. SIRT6 specifically deacetylates pericentric
4. Metabolism and epigenetics
25
FIG. 2.3 Interactions Among Environment, Cell Metabolism and Epigenetics. There are dynamic interactions among environmental inputs (such as nutrition, O2), cell metabolism pathways and epigenetic changes. NADþ, involved in redox reactions as electron carrier, is a coenzyme for class III HDACs (sirtuins). Acetyl CoA, which is an oxidizing reagent that conveys carbon atoms in TCA cycle, is a coenzyme for KATs. bOHB, ketone body in starvation and exercise, is an endogenous HDAC inhibitor. SAM, donor of methyl group, is coenzyme for DNMTs and HMTs. Credit: James R. Davie.
heterochromatin H3K18ac [155]. SIRT6 also deacetylates H3K9ac and H3K56ac resulting in repression of transcription of hypoxic inducible factor (HIF)-a driven glycolytic gene [152]. Another interesting possibility about links between nutrition and histone PTMs is that the ketone body b-hydroxybutyrate e product of breakage of fatty acids due to starvation or long time caloric restriction and consequently activation of fatty acid b oxidation - acts as an endogenous HDAC inhibitor [152,153,156,157]. This would lead to increased H3 acetylation (Lys9 and Lys14), and transcription of genes that are controlled with FOXO3a. Other studies confirmed that caloric restriction e mostly low carbohydrate diets-would lead to ketogenesis and this is associated with class I HDACs (yeast, Caenorhabditis elegans, Drosophila) in the extension of life span along with protection against reactive oxygen species (ROS). It seems that this metabolite controlled HDAC is a process that is involved in lots of cell events and not just longevity [153]. SAM is a donor of the methyl group for both DNA and histone methylation. Reduction of threonine, a metabolic precursor for the genesis of SAM, results in reduced levels of H3K4me2 and H3K4me3 but not H3K4me1 [158]. It is important to note that loss of SAM will reduce the activities of the KMTs, PRMTs and DNMTs but unless there is an event removing the histone or DNA modification, the modification will appear unaffected. TET enzymes need substrates and cofactors such as a-ketoglutarate (a-KG), Fe2þ and O2 for stepwise oxidation reactions [152,159]. More interestingly, a-KG, which is normally derived from
26
Chapter 2 DNA methylation and chromatin modifications
either glucose or glutamine anabolism [160], plays a key role in the TCA cycle, amino acid synthesis and nitrogen transport. However, in any situation in which high levels of the inhibitor of F2- and a-KG-dependent dioxygenase are present as a result of dysregulative mutations, certain types of cancer would occur [161].
4.2 Hypoxia-induced epigenetic changes Hypoxia is defined as inadequate available oxygen (O2) for the demands of target tissues. Demethylation enzymes such as LSD1, JHDMs, JMJDs, JARIDs, UTX, and TETs need oxygen as a substrate and/or cofactor [154]. When tissue metabolism exceeds metabolic supplies, adaptation and maladaptation processes will start. These processes occur to prevent cell death. In the presence of enough O2, cells oxidize glucose to provide adenosine triphosphate (ATP) as the source of energy. In hypoxia, glucose is converted to lactate. This switch from oxidative to glycolytic metabolism is mediated by hypoxiainducible factor 1 (HIF-1) [162]. Hypoxia might occur as chronic, intermittent, and acute. Adaptive responses to chronic hypoxia are due to binding of HIF-1 to a hypoxia-response element (HRE) located near the target gene. This HIF-1-dependent gene regulation is inherently sensitive to cytosine methylation by DNA methyltransferases. The changes in DNA methylation alter gene expression patterns, in some cases irreversibly and last long after hypoxia is resolved [163]. Living in low barometric pressure at high altitude such as in the mountains is one of the best examples of chronic exposure to hypoxia [164]. Preeclampsia is a pathologic situation of chronic hypoxia and changes in gene expression due to disruption in DNA methylation [165]. In the tumor context, hypoxia is responsible for about half of DNA hypermethylation in solid tumors [166,167] and triggers initiating tumor occurrence in cells through epigenetic changes [168,169]. In cancer cells, there is often a lower expression of TET. However, the low oxygen levels in hypoxia also reduce the activity of the TET enzymes. Together, the low protein levels and reduced activity of the TET enzymes may alter DNA methylation and expression of some genes [167,168,170e173]. Sub-acute intermittent hypoxic exposure may result in reversible epigenetic adaptations, while chronic hypoxia leads to irreversible epigenetic adaptations [164]. One example of long-term intermittent hypoxia is obstructive sleep apnea (OSA). OSA is a common clinical problem that affects one in 5 (approximately 20%) healthy individuals and 40%e70% of obese people [174] and even children [175]. Long-term chronic intermediate hypoxia increased DNA methylation and down-regulation of anti-oxidant genes, leading to persistent cardiorespiratory abnormalities [176,177]. The expression of the anti-oxidant genes was restored after decitabine treatment. This treatment also normalized cardiorespiratory function [176]. There are fewer studies about short-term acute nonlethal hypoxia situation. A study on hypoxia induced epigenetic reprogramming in hippocampal neurons showed that acute hypoxia causes upregulation in the genes more than downregulation. In addition, promoters and CpG islands remained hypomethylated several days after hypoxic exposure. It supports the idea that whole genome methylation reprogramming is correlated with gene expression days after sub-lethal hypoxic stress [178].
5. Concluding remarks There is still much to learn about the plethora of histone PTMs and how these modifications crosstalk with each other and with DNA modifications. Several of the histone PTMs play a role in altering
Acknowledgments
27
chromatin structure (such as H4K16ac); however, many do not. The latter serve important roles in attracting or repelling the binding of non-histone proteins and/or chromatin modifying enzymes (the readers). There is much to be worked out in order to understand how the readers recognize the histone and DNA modifications and what the roles of the reader once bound are. Metabolism plays a major role in regulating histone and DNA modifications involved in epigenetic processes [179]. Metabolism is impacted in many disease states particularly in cancer [180,181]. The mitochondria dynamic network “communicates” with the nucleus in the regulation of metabolism and epigenetic processes; there is much to be learned about this relationship and how it is altered in disease. Each cell may have up to 10,000 mitochondria and each mitochondrion may have up to ten circular mitochondrial DNAs. How do mitochondria numbers and network play into the dynamic web of events involving mitochondria, nucleus, metabolism and epigenetic processes? How does exercise fit into this picture? Clearly, the external (diet, environment in which we live) and internal environments (metabolic enzymes, hypoxia) will impact epigenetic programming and could potentially have long term, transgenerational consequences. As we will all find out one day, our metabolism changes as we age. NAD levels drop [182] and as a consequence, epigenetic processes do not perform with the same efficiency as they did when we were younger. With the decline of NAD, acetylation of histone and non-histone proteins may increase due to reduced activity of the sirtuins. The increased acetylation of histone and non-histone proteins will have consequences on their activities and cellular levels. In cases where the acetylated sites are also ubiquitinated, the acetylation event may prevent turnover of the protein by the proteasome [183]. For example, SIRT1 deacetylates acetylated HDAC1 [184,185]. Acetylated HDAC1 is not active and trans-represses the activity of HDAC2. Acetylation of p53 increases the stability and activity of this tumor suppressor by suppressing ubiquitination. HDAC1 and SIRT1 deacetylate acetylated p53, regulating its activity and levels. Loss of SIRT1 results in the accumulation of acetylated HDAC1 and acetylated p53 [184]. The genomic levels of the replication-independent histone variants, such as H3.3 increase with age and with the elevated levels of H3.3 comes an increase in the PTMs associated with this variant [186]. With old age, DNA methylation of the genome declines [187]. Neurological disorders, which escalate with age, are a consequence of deregulation in the epigenetic processes. As many players in these epigenetic processes are druggable, refinement of our drug arsenal may counter neural decline during normal aging [188]. As our understanding of the mechanisms regulating metabolism and epigenetic processes matures, we will realize the importance of our lifestyle choices such as diet and regular exercise in healthy living and healthy aging.
Acknowledgments This work is supported by an Environments, Genes and Chronic Disease Canadian Institutes for Health Research Team Grant (144626), a Natural Sciences and Engineering Research Council of Canada Grant (RGPIN-201705927), and a CancerCare Manitoba Foundation Grant (761020234). Ms Beacon and Dr. Sepehri were funded by the Graduate Enhancement of Tri- Council Stipends (GETS) through the University of Manitoba. Ms Osman was funded by CancerCare Manitoba/Children’s Hospital Research Institute of Manitoba Master’s Studentship. Dr. Sepehri thanks the Zabol University of Medical Sciences for the opportunity to study abroad in Dr. Davie’s research group.
28
Chapter 2 DNA methylation and chromatin modifications
References [1] Liyanage VR, Jarmasz JS, Murugeshan N, Del Bigio MR, Rastegar M, Davie JR. DNA modifications: function and applications in normal and disease states. Biology 2014;3(4):670e723. [2] Delcuve GP, Khan DH, Liyanage VRB, Jahan S, Rastegar M, Kirshenbaum LA, et al. Epigenetics: chromatin organization and function. In: Backs J, McKinsey T, editors. Epigenetics in cardiac disease. 1 ed. Switzerland: Springer International Publishing; 2017. p. 1e35. [3] White AE, Hieb AR, Luger K. A quantitative investigation of linker histone interactions with nucleosomes and chromatin. Sci Rep 2016;6:19122. [4] Luger K, Richmond TJ. The histone tails of the nucleosome. Curr Opin Genet Dev 1998;8(2):140e6. [5] Huang H, Sabari BR, Garcia BA, Allis CD, Zhao Y. SnapShot: histone modifications. Cell 2014;159(2): 458e458.e1. [6] Allis CD, Berger SL, Cote J, Dent S, Jenuwien T, Kouzarides T, et al. New nomenclature for chromatinmodifying enzymes. Cell 2007;131(4):633e6. [7] Ball DJ, Gross DS, Garrard WT. 5-methylcytosine is localized in nucleosomes that contain histone H1. Proc Natl Acad Sci USA 1983;80(18):5490e4. [8] Turner BM. The adjustable nucleosome: an epigenetic signaling module. Trends Genet 2012;28(9): 436e44. [9] Turner BM. Nucleosome signalling; an evolving concept. Biochim Biophys Acta 2014;1839(8):623e6. [10] Maze I, Noh KM, Soshnev AA, Allis CD. Every amino acid matters: essential contributions of histone variants to mammalian development and disease. Nat Rev Genet 2014;15(4):259e71. [11] Kang J, Kim YW, Kim A. Histone variants H3.3 and H2A.Z are incorporated into the beta-globin locus during transcription activation via different mechanisms. Biochim Biophys Acta 2018;1861(7):637e46. [12] McKittrick E, Gafken PR, Ahmad K, Henikoff S. Histone H3.3 is enriched in covalent modifications associated with active chromatin. Proc Natl Acad Sci USA 2004;101(6):1525e30. [13] Josefowicz SZ, Shimada M, Armache A, Li CH, Miller RM, Lin S, et al. Chromatin kinases act on transcription factors and histone tails in regulation of inducible transcription. Mol Cell 2016;64(2):347e61. [14] Sun JM, Chen HY, Espino PS, Davie JR. Phosphorylated serine 28 of histone H3 is associated with destabilized nucleosomes in transcribed chromatin. Nucleic Acids Res 2007;35(19):6640e7. [15] Shi L, Wen H, Shi X. The histone variant H3.3 in transcriptional regulation and human disease. J Mol Biol 2017;429(13):1934e45. [16] Iwafuchi-Doi M, Donahue G, Kakumanu A, Watts JA, Mahony S, Pugh BF, et al. The pioneer transcription factor FoxA maintains an accessible nucleosome configuration at enhancers for tissue-specific gene activation. Mol Cell 2016;62(1):79e91. [17] Mayran A, Drouin J. Pioneer transcription factors shape the epigenetic landscape. J Biol Chem 2018; 293(36):13795e804. [18] Davie JR, Xu W, Delcuve GP. Histone H3K4 trimethylation: dynamic interplay with pre-mRNA splicing. Biochem Cell Biol 2016;94(1):1e11. [19] Khan DH, Gonzalez C, Cooper C, Sun JM, Chen HY, Healy S, et al. RNA-dependent dynamic histone acetylation regulates MCL1 alternative splicing. Nucleic Acids Res 2014;42(3):1656e70. [20] Khan DH, Gonzalez C, Tailor N, Hamedani MK, Leygue E, Davie JR. Dynamic histone acetylation of H3K4me3 nucleosome regulates MCL1 pre-mRNA splicing. J Cell Physiol 2016;231(10):2196e204. [21] Bazett-Jones DP, Mendez E, Czarnota GJ, Ottensmeyer FP, Allfrey VG. Visualization and analysis of unfolded nucleosomes associated with transcribing chromatin. Nucleic Acids Res 1996;24(2):321e9. [22] Locklear Jr L, Ridsdale JA, Bazett-Jones DP, Davie JR. Ultrastructure of transcriptionally competent chromatin. Nucleic Acids Res 1990;18(23):7015e24.
References
29
[23] Walia H, Chen HY, Sun JM, Holth LT, Davie JR. Histone acetylation is required to maintain the unfolded nucleosome structure associated with transcribing DNA. J Biol Chem 1998;273(23):14516e22. [24] Han J, Zhang Z, Wang K. 3C and 3C-based techniques: the powerful tools for spatial genome organization deciphering. Mol Cytogenet 2018;11:21. [25] Jiang C, Pugh BF. Nucleosome positioning and gene regulation: advances through genomics. Nat Rev Genet 2009;10(3):161e72. [26] Dekker J, Rippe K, Dekker M, Kleckner N. Capturing chromosome conformation. Science 2002; 295(5558):1306e11. [27] Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 2009; 326(5950):289e93. [28] Serizay J, Ahringer J. Genome organization at different scales: nature, formation and function. Curr Opin Cell Biol 2018;52:145e53. [29] Barkess G, West AG. Chromatin insulator elements: establishing barriers to set heterochromatin boundaries. Epigenomics 2012;4(1):67e80. [30] Stalder J, Larsen A, Engel JD, Dolan M, Groudine M, Weintraub H. Tissue-specific DNA cleavages in the globin chromatin domain introduced by DNAase I. Cell 1980;20(2):451e60. [31] Tanay A, Cavalli G. Chromosomal domains: epigenetic contexts and functional implications of genomic compartmentalization. Curr Opin Genet Dev 2013;23(2):197e203. [32] Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr Protoc Mol Biol 2015;109:21 9 1e9. [33] Simon JM, Giresi PG, Davis IJ, Lieb JD. Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat Protoc 2012;7(2):256e67. [34] Elgin SC. The formation and function of DNase I hypersensitive sites in the process of gene activation. J Biol Chem 1988;263(36):19259e62. [35] Jahan S, Xu W, He S, Gonzalez C, Delcuve GP, Davie JR. The chicken erythrocyte epigenome. Epigenet Chromatin 2016;9:19. [36] Hebbes TR, Clayton AL, Thorne AW, Crane-Robinson C. Core histone hyperacetylation co-maps with generalized DNase I sensitivity in the chicken beta-globin chromosomal domain. EMBO J 1994;13(8): 1823e30. [37] Evans KJ, Huang N, Stempor P, Chesney MA, Down TA, Ahringer J. Stable Caenorhabditis elegans chromatin domains separate broadly expressed and developmentally regulated genes. Proc Natl Acad Sci USA 2016;113(45):E7020e9. [38] Wijchers PJ, Krijger PHL, Geeven G, Zhu Y, Denker A, Verstegen M, et al. Cause and consequence of tethering a SubTAD to different nuclear compartments. Mol Cell 2016;61(3):461e73. [39] Rowley MJ, Nichols MH, Lyu X, Ando-Kuri M, Rivera ISM, Hermetz K, et al. Evolutionarily conserved principles predict 3D chromatin organization. Mol Cell 2017;67(5):837e852 e7. [40] Carelli FN, Sharma G, Ahringer J. Broad chromatin domains: an important facet of genome regulation. Bioessays 2017;39(12). [41] Lupianez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 2015;161(5):1012e25. [42] Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 2012;485(7398):376e80. [43] Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 2014;159(7):1665e80. [44] Zadeh G, Aldape K. Bringing IDH into the fold. Cancer Cell 2016;29(2):139e40.
30
Chapter 2 DNA methylation and chromatin modifications
[45] Stadler MR, Haines JE, Eisen MB. Convergence of topological domain boundaries, insulators, and polytene interbands revealed by high-resolution mapping of chromatin contacts in the early Drosophila melanogaster embryo. Elife 2017;6. [46] Barutcu AR, Lajoie BR, Fritz AJ, McCord RP, Nickerson JA, van Wijnen AJ, et al. SMARCA4 regulates gene expression and higher-order chromatin structure in proliferating mammary epithelial cells. Genome Res 2016;26(9):1188e201. [47] Nora EP, Goloborodko A, Valton AL, Gibcus JH, Uebersohn A, Abdennur N, et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 2017; 169(5):930e44 e22. [48] Flyamer IM, Gassler J, Imakaev M, Brandao HB, Ulianov SV, Abdennur N, et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 2017;544(7648):110e4. [49] Fudenberg G, Imakaev M, Lu C, Goloborodko A, Abdennur N, Mirny LA. formation of chromosomal domains by loop extrusion. Cell Rep 2016;15(9):2038e49. [50] Rao SSP, Huang SC, Glenn St Hilaire B, Engreitz JM, Perez EM, Kieffer-Kwon KR, et al. Cohesin loss eliminates all loop domains. Cell 2017;171(2):305e20 e24. [51] Li T, Jia L, Cao Y, Chen Q, Li C. OCEAN-C: mapping hubs of open chromatin interactions across the genome reveals gene regulatory networks. Genome Biol 2018;19(1):54. [52] Li G, Ruan X, Auerbach RK, Sandhu KS, Zheng M, Wang P, et al. Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell 2012;148(1e2):84e98. [53] Han Y, Eipel M, Franzen J, Sakk V, Dethmers-Ausema B, Yndriago L, et al. Epigenetic age-predictor for mice based on three CpG sites. Elife 2018;7. [54] Weidner CI, Lin Q, Koch CM, Eisele L, Beier F, Ziegler P, et al. Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol 2014;15(2):R24. [55] Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Science 1975; 187(4173):226e32. [56] Riggs AD. X inactivation, differentiation, and DNA methylation. Cytogenet Cell Genet 1975;14(1):9e25. [57] Ooi SK, Qiu C, Bernstein E, Li K, Jia D, Yang Z, et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 2007;448(7154):714e7. [58] Suetake I, Shinozaki F, Miyagawa J, Takeshima H, Tajima S. DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J Biol Chem 2004;279(26):27816e23. [59] Geiman TM, Sankpal UT, Robertson AK, Zhao Y, Zhao Y, Robertson KD. DNMT3B interacts with hSNF2H chromatin remodeling enzyme, HDACs 1 and 2, and components of the histone methylation system. Biochem Biophys Res Commun 2004;318(2):544e55. [60] Ndlovu MN, Denis H, Fuks F. Exposing the DNA methylome iceberg. Trends Biochem Sci 2011;36(7): 381e7. [61] Laurent L, Wong E, Li G, Huynh T, Tsirigos A, Ong CT, et al. Dynamic changes in the human methylome during differentiation. Genome Res 2010;20(3):320e31. [62] Jones PA. The DNA methylation paradox. Trends Genet 1999;15(1):34e7. [63] Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 2012; 13(7):484e92. [64] Neri F, Rapelli S, Krepelova A, Incarnato D, Parlato C, Basile G, et al. Intragenic DNA methylation prevents spurious transcription initiation. Nature 2017;543(7643):72e7. [65] Watt F, Molloy PL. Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter. Genes Dev 1988;2(9):1136e43. [66] Boyes J, Bird A. DNA methylation inhibits transcription indirectly via a methyl-CpG binding protein. Cell 1991;64(6):1123e34.
References
31
[67] Hendrich B, Bird A. Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol Cell Biol 1998;18(11):6538e47. [68] Prokhortchouk A, Hendrich B, Jorgensen H, Ruzov A, Wilm M, Georgiev G, et al. The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev 2001;15(13):1613e8. [69] Harrington MA, Jones PA, Imagawa M, Karin M. Cytosine methylation does not affect binding of transcription factor Sp1. Proc Natl Acad Sci USA 1988;85(7):2066e70. [70] Matsuo K, Silke J, Georgiev O, Marti P, Giovannini N, Rungger D. An embryonic demethylation mechanism involving binding of transcription factors to replicating DNA. EMBO J 1998;17(5):1446e53. [71] Yoder JA, Walsh CP, Bestor TH. Cytosine methylation and the ecology of intragenomic parasites. Trends Genet 1997;13(8):335e40. [72] Schmidl C, Klug M, Boeld TJ, Andreesen R, Hoffmann P, Edinger M, et al. Lineage-specific DNA methylation in T cells correlates with histone methylation and enhancer activity. Genome Res 2009;19(7): 1165e74. [73] Bell AC, Felsenfeld G. Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature 2000;405(6785):482e5. [74] Charlet J, Duymich CE, Lay FD, Mundbjerg K, Dalsgaard Sorensen K, Liang G, et al. Bivalent regions of cytosine methylation and H3K27 acetylation suggest an active role for DNA methylation at enhancers. Mol Cell 2016;62(3):422e31. [75] Mahe EA, Madigou T, Salbert G. Reading cytosine modifications within chromatin. Transcription 2018; 9(4):240e7. [76] Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 2010; 11(9):607e20. [77] Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009;324(5929):930e5. [78] Zhang H, Zhang X, Clark E, Mulcahey M, Huang S, Shi YG. TET1 is a DNA-binding protein that modulates DNA methylation and gene transcription via hydroxylation of 5-methylcytosine. Cell Res 2010; 20(12):1390e3. [79] Globisch D, Munzel M, Muller M, Michalakis S, Wagner M, Koch S, et al. Tissue distribution of 5-hydroxymethylcytosine and search for active demethylation intermediates. PLoS One 2010;5(12):e15367. [80] Valinluck V, Tsai HH, Rogstad DK, Burdzy A, Bird A, Sowers LC. Oxidative damage to methyl-CpG sequences inhibits the binding of the methyl-CpG binding domain (MBD) of methyl-CpG binding protein 2 (MeCP2). Nucleic Acids Res 2004;32(14):4100e8. [81] Greco CM, Kunderfranco P, Rubino M, Larcher V, Carullo P, Anselmo A, et al. DNA hydroxymethylation controls cardiomyocyte gene expression in development and hypertrophy. Nat Commun 2016;7:12418. [82] Branco MR, Ficz G, Reik W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat Rev Genet 2011;13(1):7e13. [83] Williams K, Christensen J, Pedersen MT, Johansen JV, Cloos PA, Rappsilber J, et al. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 2011;473(7347):343e8. [84] Wu H, D’Alessio AC, Ito S, Xia K, Wang Z, Cui K, et al. Dual functions of Tet1 in transcriptional regulation in mouse embryonic stem cells. Nature 2011;473(7347):389e93. [85] Iacobazzi V, Castegna A, Infantino V, Andria G. Mitochondrial DNA methylation as a next-generation biomarker and diagnostic tool. Mol Genet Metabol 2013;110(1e2):25e34. [86] Shock LS, Thakkar PV, Peterson EJ, Moran RG, Taylor SM. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc Natl Acad Sci USA 2011; 108(9):3630e5. [87] Wong M, Gertz B, Chestnut BA, Martin LJ. Mitochondrial DNMT3A and DNA methylation in skeletal muscle and CNS of transgenic mouse models of ALS. Front Cell Neurosci 2013;7:279.
32
Chapter 2 DNA methylation and chromatin modifications
[88] Bellizzi D, D’Aquila P, Scafone T, Giordano M, Riso V, Riccio A, et al. The control region of mitochondrial DNA shows an unusual CpG and non-CpG methylation pattern. DNA Res 2013;20(6):537e47. [89] Ngo HB, Kaiser JT, Chan DC. The mitochondrial transcription and packaging factor Tfam imposes a U-turn on mitochondrial DNA. Nat Struct Mol Biol 2011;18(11):1290e6. [90] Sabari BR, Zhang D, Allis CD, Zhao Y. Metabolic regulation of gene expression through histone acylations. Nat Rev Mol Cell Biol 2017;18(2):90e101. [91] Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell 2007;128(4):707e19. [92] Musselman CA, Lalonde ME, Cote J, Kutateladze TG. Perceiving the epigenetic landscape through histone readers. Nat Struct Mol Biol 2012;19(12):1218e27. [93] Tarakhovsky A. Tools and landscapes of epigenetics. Nat Immunol 2010;11(7):565e8. [94] Lee JS, Smith E, Shilatifard A. The language of histone crosstalk. Cell 2010;142(5):682e5. [95] Soshnev AA, Josefowicz SZ, Allis CD. Greater than the sum of parts: complexity of the dynamic epigenome. Mol Cell 2016;62(5):681e94. [96] Zhang T, Cooper S, Brockdorff N. The interplay of histone modifications e writers that read. EMBO Rep 2015;16(11):1467e81. [97] Cheung P, Tanner KG, Cheung WL, Sassone-Corsi P, Denu JM, Allis CD. Synergistic coupling of histone H3 phosphorylation and acetylation in response to epidermal growth factor stimulation. Mol Cell 2000; 5(6):905e15. [98] Guccione E, Bassi C, Casadio F, Martinato F, Cesaroni M, Schuchlautz H, et al. Methylation of histone H3R2 by PRMT6 and H3K4 by an MLL complex are mutually exclusive. Nature 2007;449(7164):933e7. [99] Allfrey V, Faulkner R, Mirsky A. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci Unit States Am 1964;51(5):786e94. [100] Candido EP, Reeves R, Davie JR. Sodium butyrate inhibits histone deacetylation in cultured cells. Cell 1978;14(1):105e13. [101] Vidali G, Boffa LC, Bradbury EM, Allfrey VG. Butyrate suppression of histone deacetylation leads to accumulation of multiacetylated forms of histones H3 and H4 and increased DNase I sensitivity of the associated DNA sequences. Proc Natl Acad Sci USA 1978;75(5):2239e43. [102] Hebbes TR, Thorne AW, Crane-Robinson C. A direct link between core histone acetylation and transcriptionally active chromatin. EMBO J 1988;7(5):1395e402. [103] Brownell JE, Allis CD. An activity gel assay detects a single, catalytically active histone acetyltransferase subunit in Tetrahymena macronuclei. Proc Natl Acad Sci USA 1995;92(14):6364e8. [104] Davie JR, Spencer VA. Control of histone modifications. J Cell Biochem 1999;(Suppl. 32e33):141e8. [105] Davie JR, Chadee DN. Regulation and regulatory parameters of histone modifications. J Cell Biochem Suppl 1998;30e31:203e13. [106] Spencer VA, Davie JR. Role of covalent modifications of histones in regulating gene expression. Gene 1999;240(1):1e12. [107] Ridsdale JA, Hendzel MJ, Delcuve GP, Davie JR. Histone acetylation alters the capacity of the H1 histones to condense transcriptionally active/competent chromatin. J Biol Chem 1990;265(9):5150e6. [108] Schneider R, Bannister AJ, Myers FA, Thorne AW, Crane-Robinson C, Kouzarides T. Histone H3 lysine 4 methylation patterns in higher eukaryotic genes. Nat Cell Biol 2004;6(1):73e7. [109] Jackson JP, Johnson L, Jasencakova Z, Zhang X, PerezBurgos L, Singh PB, et al. Dimethylation of histone H3 lysine 9 is a critical mark for DNA methylation and gene silencing in Arabidopsis thaliana. Chromosoma 2004;112(6):308e15. [110] Schuettengruber B, Bourbon HM, Di Croce L, Cavalli G. Genome regulation by polycomb and trithorax: 70 Years and counting. Cell 2017;171(1):34e57. [111] Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein CB, et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 2011;473(7345):43e9.
References
33
[112] Bieberstein NI, Carrillo Oesterreich F, Straube K, Neugebauer KM. First exon length controls active chromatin signatures and transcription. Cell Rep 2012;2(1):62e8. [113] Muramoto T, Muller I, Thomas G, Melvin A, Chubb JR. Methylation of H3K4 Is required for inheritance of active transcriptional states. Curr Biol 2010;20(5):397e406. [114] Terrenoire E, McRonald F, Halsall JA, Page P, Illingworth RS, Taylor AM, et al. Immunostaining of modified histones defines high-level features of the human metaphase epigenome. Genome Biol 2010; 11(11):R110. [115] Benayoun BA, Pollina EA, Ucar D, Mahmoudi S, Karra K, Wong ED, et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell 2014;158(3):673e88. [116] Chen K, Chen Z, Wu D, Zhang L, Lin X, Su J, et al. Broad H3K4me3 is associated with increased transcription elongation and enhancer activity at tumor-suppressor genes. Nat Genet 2015;47(10):1149e57. [117] Zhang K, Dent SY. Histone modifying enzymes and cancer: going beyond histones. J Cell Biochem 2005; 96(6):1137e48. [118] Wu SC, Zhang Y. Minireview: role of protein methylation and demethylation in nuclear hormone signaling. Mol Endocrinol 2009;23(9):1323e34. [119] Yang Y, Bedford MT. Protein arginine methyltransferases and cancer. Nat Rev Canc 2013;13(1):37e50. [120] Feng Q, Wang H, Ng HH, Erdjument-Bromage H, Tempst P, Struhl K, et al. Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain. Curr Biol 2002;12(12):1052e8. [121] Helin K, Dhanak D. Chromatin proteins and modifications as drug targets. Nature 2013;502(7472):480e8. [122] Zurita-Lopez CI, Sandberg T, Kelly R, Clarke SG. Human protein arginine methyltransferase 7 (PRMT7) is a type III enzyme forming omega-NG-monomethylated arginine residues. J Biol Chem 2012;287(11): 7859e70. [123] Jahan S, Davie JR. Protein arginine methyltransferases (PRMTs): role in chromatin organization. Adv Biol Regul 2015;57:173e84. [124] Chen D, Ma H, Hong H, Koh SS, Huang SM, Schurter BT, et al. Regulation of transcription by a protein methyltransferase. Science 1999;284(5423):2174e7. [125] Binda O. On your histone mark. SET, methylate! Epigenetics 2013;8(5):457e63. [126] Chiang K, Zielinska AE, Shaaban AM, Sanchez-Bailon MP, Jarrold J, Clarke TL, et al. PRMT5 is a critical regulator of breast cancer stem cell function via histone methylation and FOXP1 expression. Cell Rep 2017; 21(12):3498e513. [127] Cheng X, Collins RE, Zhang X. Structural and sequence motifs of protein (histone) methylation enzymes. Annu Rev Biophys Biomol Struct 2005;34:267e94. [128] Selvi BR, Batta K, Kishore AH, Mantelingu K, Varier RA, Balasubramanyam K, et al. Identification of a novel inhibitor of coactivator-associated arginine methyltransferase 1 (CARM1)-mediated methylation of histone H3 Arg-17. J Biol Chem 2010;285(10):7143e52. [129] Daujat S, Bauer UM, Shah V, Turner B, Berger S, Kouzarides T. Crosstalk between CARM1 methylation and CBP acetylation on histone H3. Curr Biol 2002;12(24):2090e7. [130] Molina-Serrano D, Schiza V, Kirmizis A. Cross-talk among epigenetic modifications: lessons from histone arginine methylation. Biochem Soc Trans 2013;41(3):751e9. [131] El Messaoudi S, Fabbrizio E, Rodriguez C, Chuchana P, Fauquier L, Cheng D, et al. Coactivator-associated arginine methyltransferase 1 (CARM1) is a positive regulator of the Cyclin E1 gene. Proc Natl Acad Sci USA 2006;103(36):13351e6. [132] Hyllus D, Stein C, Schnabel K, Schiltz E, Imhof A, Dou Y, et al. PRMT6-mediated methylation of R2 in histone H3 antagonizes H3 K4 trimethylation. Genes Dev 2007;21(24):3369e80. [133] Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell 2007;129(4):823e37.
34
Chapter 2 DNA methylation and chromatin modifications
[134] Migliori V, Muller J, Phalke S, Low D, Bezzi M, Mok WC, et al. Symmetric dimethylation of H3R2 is a newly identified histone mark that supports euchromatin maintenance. Nat Struct Mol Biol 2012;19(2): 136e44. [135] Huang S, Litt M, Felsenfeld G. Methylation of histone H4 by arginine methyltransferase PRMT1 is essential in vivo for many subsequent histone modifications. Genes Dev 2005;19(16):1885e93. [136] Xie Y, Ke S, Ouyang N, He J, Xie W, Bedford MT, et al. Epigenetic regulation of transcriptional activity of pregnane X receptor by protein arginine methyltransferase 1. J Biol Chem 2009;284(14):9199e205. [137] Kleinschmidt MA, Streubel G, Samans B, Krause M, Bauer UM. The protein arginine methyltransferases CARM1 and PRMT1 cooperate in gene regulation. Nucleic Acids Res 2008;36(10):3202e13. [138] Denis H, Deplus R, Putmans P, Yamada M, Metivier R, Fuks F. Functional connection between deimination and deacetylation of histones. Mol Cell Biol 2009;29(18):4982e93. [139] Li X, Hu X, Patel B, Zhou Z, Liang S, Ybarra R, et al. H4R3 methylation facilitates beta-globin transcription by regulating histone acetyltransferase binding and H3 acetylation. Blood 2010;115(10): 2028e37. [140] Fulton MD, Zhang J, He M, Ho MC, Zheng YG. Intricate effects of alpha-amino and lysine modifications on arginine methylation of the N-terminal tail of histone H4. Biochemistry 2017;56(28):3539e48. [141] Feng Y, Wang J, Asher S, Hoang L, Guardiani C, Ivanov I, et al. Histone H4 acetylation differentially modulates arginine methylation by an in Cis mechanism. J Biol Chem 2011;286(23):20323e34. [142] Shogren-Knaak M, Ishii H, Sun JM, Pazin MJ, Davie JR, Peterson CL. Histone H4-K16 acetylation controls chromatin structure and protein interactions. Science 2006;311(5762):844e7. [143] Pal S, Vishwanath SN, Erdjument-Bromage H, Tempst P, Sif S. Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol Cell Biol 2004;24(21):9630e45. [144] Girardot M, Hirasawa R, Kacem S, Fritsch L, Pontis J, Kota SK, et al. PRMT5-mediated histone H4 arginine-3 symmetrical dimethylation marks chromatin at G þ C-rich regions of the mouse genome. Nucleic Acids Res 2014;42(1):235e48. [145] Di Lorenzo A, Bedford MT. Histone arginine methylation. FEBS Lett 2011;585(13):2024e31. [146] Vossenaar ER, Zendman AJ, van Venrooij WJ, Pruijn GJ. PAD, a growing family of citrullinating enzymes: genes, features and involvement in disease. Bioessays 2003;25(11):1106e18. [147] Witalison EE, Thompson PR, Hofseth LJ. Protein arginine deiminases and associated citrullination: physiological functions and diseases associated with dysregulation. Curr Drug Targets 2015;16(7):700e10. [148] Zhang X, Bolt M, Guertin MJ, Chen W, Zhang S, Cherrington BD, et al. Peptidylarginine deiminase 2-catalyzed histone H3 arginine 26 citrullination facilitates estrogen receptor alpha target gene activation. Proc Natl Acad Sci USA 2012;109(33):13331e6. [149] Wang Y, Wysocka J, Sayegh J, Lee YH, Perlin JR, Leonelli L, et al. Human PAD4 regulates histone arginine methylation levels via demethylimination. Science 2004;306(5694):279e83. [150] Chang B, Chen Y, Zhao Y, Bruick RK. JMJD6 is a histone arginine demethylase. Science 2007;318(5849): 444e7. [151] Liu W, Ma Q, Wong K, Li W, Ohgi K, Zhang J, et al. Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release. Cell 2013;155(7):1581e95. [152] Etchegaray JP, Mostoslavsky R. Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes. Mol Cell 2016;62(5):695e711. [153] Sassone-Corsi P. Physiology. When metabolism and epigenetics converge. Science 2013;339(6116): 148e50. [154] Reid MA, Dai Z, Locasale JW. The impact of cellular metabolism on chromatin dynamics and epigenetics. Nat Cell Biol 2017;19(11):1298e306.
References
35
[155] Tasselli L, Xi Y, Zheng W, Tennen RI, Odrowaz Z, Simeoni F, et al. SIRT6 deacetylates H3K18ac at pericentric chromatin to prevent mitotic errors and cellular senescence. Nat Struct Mol Biol 2016;23(5): 434e40. [156] Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P, Ng CP, et al. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell Metabol 2017;26(3):547e557 e8. [157] Newman JC, Verdin E. Beta-hydroxybutyrate: a signaling metabolite. Annu Rev Nutr 2017;37:51e76. [158] Shyh-Chang N, Locasale JW, Lyssiotis CA, Zheng Y, Teo RY, Ratanasirintrawoot S, et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 2013;339(6116):222e6. [159] Yin X, Xu Y. Structure and function of TET enzymes. Adv Exp Med Biol 2016;945:275e302. [160] Lu C, Thompson CB. Metabolic regulation of epigenetics. Cell Metabol 2012;16(1):9e17. [161] Janke R, Dodson AE, Rine J. Metabolism and epigenetics. Annu Rev Cell Dev Biol 2015;31:473e96. [162] Semenza GL. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Canc Biol 2009; 19(1):12e6. [163] Veron V, Marandel L, Liu J, Velez EJ, Lepais O, Panserat S, et al. DNA methylation of the promoter region of bnip3 and bnip3l genes induced by metabolic programming. BMC Genomics 2018;19(1):677. [164] Nanduri J, Semenza GL, Prabhakar NR. Epigenetic changes by DNA methylation in chronic and intermittent hypoxia. Am J Physiol Lung Cell Mol Physiol 2017;313(6):L1096e100. [165] Xiong J, Ding N, Gao T, Wang Y, Guo W, Zhang H, et al. Hypermethylation of endoplasmic reticulum disulfide oxidase 1alpha leads to trophoblast cell apoptosis through endoplasmic reticulum stress in preeclampsia. J Cell Biochem 2018;119(10):8588e99. [166] Ramachandran S, Ient J, Gottgens EL, Krieg AJ, Hammond EM. Epigenetic therapy for solid tumors: highlighting the impact of tumor hypoxia. Genes 2015;6(4):935e56. [167] Thienpont B, Steinbacher J, Zhao H, D’Anna F, Kuchnio A, Ploumakis A, et al. Tumour hypoxia causes DNA hypermethylation by reducing TET activity. Nature 2016;537(7618):63e8. [168] Wu MZ, Chen SF, Nieh S, Benner C, Ger LP, Jan CI, et al. Hypoxia drives breast tumor malignancy through a TET-TNFalpha-p38-MAPK signaling Axis. Cancer Res 2015;75(18):3912e24. [169] Nam HY, Chandrashekar DS, Kundu A, Shelar S, Kho EY, Sonpavde G, et al. Integrative epigenetic and gene expression analysis of renal tumor progression to metastasis. Mol Cancer Res 2019;17(1):84e96. [170] Alivand MR, Soheili ZS, Pornour M, Solali S, Sabouni F. Novel epigenetic controlling of hypoxia pathway related to overexpression and promoter hypomethylation of TET1 and TET2 in RPE cells. J Cell Biochem 2017;118(10):3193e204. [171] Dales JP, Garcia S, Meunier-Carpentier S, Andrac-Meyer L, Haddad O, Lavaut MN, et al. Overexpression of hypoxia-inducible factor HIF-1alpha predicts early relapse in breast cancer: retrospective study in a series of 745 patients. Int J Cancer 2005;116(5):734e9. [172] Wang Z, Long H, Chang C, Zhao M, Lu Q. Crosstalk between metabolism and epigenetic modifications in autoimmune diseases: a comprehensive overview. Cell Mol Life Sci 2018;75(18):3353e69. [173] Ahani-Nahayati M, Solali S, Shams Asenjan K, Movassaghpour Akbari AA, Talebi M, Zadi Heydarabad M, et al. Promoter methylation status of survival-related genes in MOLT- 4 cells Co-cultured with bone marrow mesenchymal stem cells under hypoxic conditions. Cell J 2018;20(2):188e94. [174] Framnes SN, Arble DM. The bidirectional relationship between obstructive sleep apnea and metabolic disease. Front Endocrinol 2018;9:440. [175] Xu H, Li X, Zheng X, Xia Y, Fu Y, Li X, et al. Pediatric obstructive sleep apnea is associated with changes in the oral microbiome and urinary metabolomics profile: a pilot study. J Clin Sleep Med 2018;14(9): 1559e67. [176] Nanduri J, Peng YJ, Wang N, Khan SA, Semenza GL, Kumar GK, et al. Epigenetic regulation of redox state mediates persistent cardiorespiratory abnormalities after long-term intermittent hypoxia. J Physiol 2017; 595(1):63e77.
36
Chapter 2 DNA methylation and chromatin modifications
[177] Nanduri J, Prabhakar NR. Developmental programming of O(2) sensing by neonatal intermittent hypoxia via epigenetic mechanisms. Respir Physiol Neurobiol 2013;185(1):105e9. [178] Hartley I, Elkhoury FF, Heon Shin J, Xie B, Gu X, Gao Y, et al. Long-lasting changes in DNA methylation following short-term hypoxic exposure in primary hippocampal neuronal cultures. PLoS One 2013;8(10): e77859. [179] Schvartzman JM, Thompson CB, Finley LWS. Metabolic regulation of chromatin modifications and gene expression. J Cell Biol 2018;217(7):2247e59. [180] Weyandt JD, Thompson CB, Giaccia AJ, Rathmell WK. Metabolic alterations in cancer and their potential as therapeutic targets. Am Soc Clin Oncol Educ Book 2017;37:825e32. [181] Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 2012;21(3):297e308. [182] Schultz MB, Sinclair DA. Why NAD(þ) declines during aging: it’s destroyed. Cell Metabol 2016;23(6): 965e6. [183] Koumbadinga GA, Mahmood N, Lei L, Kan Y, Cao W, Lobo VG, et al. Increased stability of heterogeneous ribonucleoproteins by a deacetylase inhibitor. Biochim Biophys Acta 2015;1849(8):1095e103. [184] Yang H, Yan B, Liao D, Huang S, Qiu Y. Acetylation of HDAC1 and degradation of SIRT1 form a positive feedback loop to regulate p53 acetylation during heat-shock stress. Cell Death Dis 2015;6:e1747. [185] Dobbin MM, Madabhushi R, Pan L, Chen Y, Kim D, Gao J, et al. SIRT1 collaborates with ATM and HDAC1 to maintain genomic stability in neurons. Nat Neurosci 2013;16(8):1008e15. [186] Tvardovskiy A, Schwammle V, Kempf SJ, Rogowska-Wrzesinska A, Jensen ON. Accumulation of histone variant H3.3 with age is associated with profound changes in the histone methylation landscape. Nucleic Acids Res 2017;45(16):9272e89. [187] Heyn H, Li N, Ferreira HJ, Moran S, Pisano DG, Gomez A, et al. Distinct DNA methylomes of newborns and centenarians. Proc Natl Acad Sci USA 2012;109(26):10522e7. [188] Delgado-Morales R, Agis-Balboa RC, Esteller M, Berdasco M. Epigenetic mechanisms during ageing and neurogenesis as novel therapeutic avenues in human brain disorders. Clin Epigenet 2017;9:67.