Abstract
Even though the discovery of the term ‘epigenetics’ was in the 1940s, it has recently become one of the most promising and expanding fields to unravel the gene expression pattern in several diseases. The most well studied example is cancer, but other diseases like metabolic disorders, autism, or inflammation-associated diseases such as lung injury, autoimmune disease, asthma, and type-2 diabetes display aberrant gene expression and epigenetic regulation during their occurrence. The change in the epigenetic pattern of a gene may also alter gene function because of a change in the DNA status. Constant environmental pressure, lifestyle, as well as food habits are the other important parameters responsible for transgenerational inheritance of epigenetic traits. Discovery of epigenetic modifiers targeting DNA methylation and histone deacetylation enzymes could be an alternative source to treat or manipulate the pathogenesis of diseases. Particularly, the combination of epigenetic drugs such as 5-Aza-2-deoxycytidine (Aza) and trichostatin A (TSA) are well studied to reduce inflammation in an acute lung injury model. It is important to understand the epigenetic machinery and the function of its components in specific diseases to develop targeted epigenetic therapy. Moreover, it is equally critical to know the specific inhibitors other than the widely used pan inhibitors in clinical trials and explore their roles in regulating specific genes in a more defined way during infection.
Keywords: epigenetic modifier, DNA methyl transferase, Histone deacetylase, 5-Aza-2-deoxycytidine, trichostatin A, inflammation
Graphical abstract

1. Introduction
The study of epigenetics involves genetic control by factors other than an individual’s DNA sequence. Genes can be switched on or off, and transcription can be regulated through epigenetic changes. These phenomena are heritable, alternative states of gene activity that are not explained by mutation or normal developmental regulation [1]. Many human diseases, including various cancers are caused as a result of epigenetic changes [2]. Epigenetic research is now becoming a rapidly developing and exciting area in biology. Studies are available in cataloguing systematically different epigenetic modifications in various tissues during developmental and pathological situations [3]. The present understanding of the mechanisms underlying epigenetic phenomena and their predominance as contributors to the development of human disease has led to a greatly enhanced interest in epigenetic research.
Epigenetic mechanisms rely on the modifications of cytosine bases in the DNA and changes in the nucleosome positioning of histones as well. They are DNA-protein interactions, suppression of transposable element mobility, cellular differentiation, embryogenesis, X-chromosome inactivation, and genomic imprinting. In multicellular organisms, the ability of epigenetic marks persist during development and can potentially be transmitted to offspring which may be necessary for generating the large range of different phenotypes that arose from the same genotype [4]. For example, cloned animals with the identical genotype may develop a different disease pattern [5]. Monozygotic twins are identical at the DNA sequence level, but have different DNA methylation and histone modification profiles [6] that might affect the occurrence of diseases, including cancer [7] or autoimmune disorders [8]. Epigenetic modification is also a key factor in cellular differentiation during development [9]. This review mainly focuses on epigenetic changes such as chromatin dynamics, the establishment of methylation patterns on DNA, and the role of RNA and proteins during gene regulation processes. Moreover, we will discuss the role of environment on epigenetic changes during infection and inflammation.
2. Epigenetic modification causes different types of diseases
Epigenetic modifications have been well established in cancer [7] and occur in several other diseases, including diabetes [10], lupus [11], asthma [12, 13], and a variety of neurological disorders. Rett syndrome is the neuro-developmental disorder most intensely studied with regard to epigenetic changes. In this disease one can see mutations in methyl CpG binding protein 2 (MeCP2) that binds to methylated DNA [14]. Other mental retardation disorders such as α-thalassemia, X-linked syndrome, Rubinstein-Taybi syndrome and Coffin-Lowry syndrome are also linked to the disruption of genes involved due to epigenetic changes [15]. Recently, research has been started on Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease to explore the possible epigenetic changes [16]. Studies on Huntington’s disease showed that histone modifications are responsible for the development of the disease [17]. Aberrant DNA methylation, altered histone modifications, and chromatin modifications coordinate a reprogramming of immune T-cell response, dendritic cell function, macrophage activation, and a breach of airway epithelial barrier in early-life that commands asthma risk and severity in later life [18]. Epigenetic modifications are also responsible for altered gene expression that could predispose individuals to the diabetic phenotype during intrauterine and early postnatal development, as well as throughout adult life [10, 19, 20].
Several inherited disorders among humans are due to defect in genomic imprinting, and parent-specific monoallelic expression of a gene, such as Angelman’s syndrome, Prader–Willi syndrome, and Beckwith–Wiedemann syndrome [21]. Epigenetic changes can also introduce genetic instability and have a major role in the development of human cancer. There is an overall change in the DNA methylation pattern in cancers [22, 23]. For example, patients with sporadic colorectal cancers show methylation and silencing of the gene encoding MLH1a [2]. Epigenetic alterations due to DNA methylation by modifications in histone, such as histone methylation and acetylation are responsible for genomic instability and developing cancer [24, 25]. Furthermore, there are autosomal dominant disorders with overgrowth syndrome, like Sotos syndrome, caused by the change in the histone methylation pattern [26]. Recently, it has been found that autoimmune diseases may also be generated by several alterations in the epigenetic mechanism. For example, systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) have global hypomethylation in the cells they target at promoter regions of DNA [27]. The overall summary of different diseases caused by the dysfunction of the epigenetic changes is given in table 1. These studies clearly implicate that epigenetic changes occur not only in infection and inflammation, but also in other types of diseases. In the following sections, the epigenetic machineries and their components that are responsible for causing different types of diseases, including infection and inflammation will be discussed in this review.
Table 1.
Important enzymes and associated diseases involved in epigenetic modification
| Epigenetic modifiers | Enzyme responsible | Function | Epigenetic Modification | Diseases | Reference |
|---|---|---|---|---|---|
| DNA methyl transferase | DNMT1, DNMTA, DNMT3B, DNM3L | Maintenance and de novo methylation | Transcriptional repression and activation | Rett syndrome, | 14 |
| DNA hyper methylation | Diabetes | 20 | |||
| Global hypo methylation, hyper methylation of promoter. | Cancer | 22,23 | |||
| Defect in genomic imprinting | Prader–Willi syndrome (PWS) | 21 | |||
| Histone acetyl transferase | HATs and HDACs | Histone acetylation and deacetylation | Reduced histone acetylation | Rubinstein – Taybai Syndrome | 15 |
| Altered histone acetylation / deacetylation | Diabetes | 20 | |||
| Reduced HDAC expression and activity | Asthma | 13 | |||
| Reduced histone acetylation | Cancer | 26 | |||
| Decreased acetylation of histones | Huntington’s disease | 19 | |||
| Histone methylation | HMTs and HDMs | Histone methylation and demethylation | Low level of histone methylation | Cancer | 24 |
| Diminished level of methylation of histone | Sotos syndrome | 26 | |||
| Hyper methylation of histones | Huntington’s disease | 16 |
3. Epigenetics machinery
Epigenetic changes are defined as modifications of DNA or chromatin that do not involve alterations or deletion of the DNA sequence. Epigenetic modifications can be grouped into four main categories: DNA methylation, histone modifications, nucleosome positioning, and microRNA mediated (Figure 1). All of these modifications are functionally linked. DNA methylation is a major epigenetic modification of the genome that regulates crucial aspects of its function. Genomic methylation patterns in somatic differentiated cells are generally stable and heritable.
Figure 1. Key mechanisms for epigenetic modifications.

DNA methylation and histone modifications are the key epigenetic modifications that are equally influenced by environmental factors, diet, and inflammation or infection. DNA methylation occurs in CpG islands near the promoter regions of the genes and results in gene repression (heterochromatin). DNA demethylation also occurs in the CpG island results in gene activation (euchromatin). Histone modifications alter chromatin structure through acetylation (associated with gene activation) or deacetylation (associated with gene repression). The miRNAs binds to a specific 3′ untranslated region of an mRNA and inhibits its translation to protein. They are encoded within the genome, and their expression is regulated by other epigenetic mechanisms.
3.1 DNA methylation
DNA methylation, which involves methylation and demethylation of the cytosine residue in DNA, is the most widely known epigenetic modification. DNA methylation is an essential process important for development like embryogenesis, proliferation and differentiation [28]. This also plays a major role for the development of different germ line and somatic mutations that finally lead to the generation of different diseases including cancer [29]. DNA methylation occurs in CpG dinucleotides that are mostly associated near the promoters of genes, in the first exon, and near the 3′ end [29]. The CpG dinucleotides tend to cluster in regions called CpG islands [30] where mostly DNA methylation takes place. CpG island shores, a new concept, are highly conserved and differentially methylated regions which lie in close proximity (~2 kb) of CpG islands where most of the tissue-specific DNA methylation seem to occur [31]. It has also been seen that CpG island shores are the most (~70%) methylated regions involved during reprogramming [32]. CpG-island methylation is most commonly associated with gene repression, except in gene bodies where DNA methylation is associated with activation [33]. CpG islands remain unmethylated in normal active cells and germ line cells. However, there are some, ~6% of genes, that become methylated during the early phase of development or in differentiated tissues in a tissue-specific manner [34].
There are different mechanisms in which DNA methylation can directly block transcription by inhibiting the binding of specific transcription factors to its target sequence. DNA methylation also indirectly blocks transcription by recruiting methyl-CpG-binding proteins (MBD) and other repressive chromatin remodeling complexes to the methylated sites [21]. DNA methyltransferases (DNMTs) are enzymes that catalyze the transfer of a methyl group from S-adenosylmethionine to DNA [35]. DNMTs are also associated with histone modifying enzymes such as histone deacetylases (HDACs), histone methyltransferases (SUV(39)H1/2 and EZH2), and ATP dependent chromatin remodeling enzymes (hSNF2H and LSH) [35]. Five members of the DNMT family have been reported in mammals: DNMT1, DNMT2, DNMT3a, DNMT3b, and DNMT3L. Among them, only three enzymes namely DNMT1, DNMT3a, and DNMT3b, possess methyltransferase activity. DNMT3a and DNMT3b are considered as de novo DNMTs as they are catalytically active members of the DNMTs. Whereas, DNMT1 is a maintenance enzyme that methylates hemi-methylated CpG dinucleotides in the nascent strand of DNA after DNA replication and is transcribed during the S phase of the cell cycle. The functions of DNMTs are essential for maintaining DNA-methylation patterns in proliferating cells [5]. Defect in DNMTs has been implicated in multiple developmental abnormalities and are embryonically lethal at E14.5–18.5, demonstrating that de novo methylation is an essential process for mammalian development [36]. De novo DNMTs are highly expressed in embryonic stem (ES) cells and down regulated in differentiated cells [37]. DNMT3L, the third member of the DNMT3 family, is expressed during gametogenesis. Although catalytically inactive, it is required for establishing maternal genomic imprinting [38]. It also interacts and activates DNMT3a and DNMT3b and colocalizes with them in the nucleus [39]. Recently, small siRNA-mediated DNA methylation has also been described mainly in plants that recruit DNMTs to catalyze de novo DNA methylation of specific regions including not only gene promoters but also repetitive sequences but the exact mechanism is still unclear [40]. Further studies are needed to explore the role of different DNMTs in DNA methylation and gene regulation.
3.2 Histone modifications
Chromatin is the dynamic component of a cell in which DNA is wrapped around an octamer formed by the four core histones H2A, H2B, H3, and H4 to build a nucleosome [41]. Nucleosome is the fundamental unit of chromatin. DNA that is wrapped around the histone octamer in 1.65 turns is 147-bp segment in length and is separated from neighboring nucleosomes by ~50 bp of free DNA. Histone H1 is called the linker histone that binds to the linker DNA (DNA separating two histone complexes) and does not form part of the nucleosome [41]. The histone cores are mostly globular except for their N-terminal tails, which have no definite structure [42]. At the time of histone modifications, N-terminal tails of histones that protrude from their own nucleosome make contact with adjacent nucleosomes and undergo modifications that would affect the overall chromatin structure [43]. All histones are subjected to post-transcriptional or translational modifications. There are several post-transcriptional histone modifications that occur, including histone acetylation/methylation, phosphorylation, ubiquitination, SUMOylation, and ADP-ribosylation [44]. Histone modifications play key roles in most cellular functions that are involved in the manipulation and expression of DNA [41, 45]. Histones are therefore one of the important modifiers in epigenetic regulation. The different types of histone modifications will be discussed in the following subsections.
3.2.1. Histone acetylation/methylation
The chromatin present either into more loosen, and actively transcribed euchromatin or more tighten, transcriptionally inactive heterochromatin form. In general, euchromatin is characterized by high levels of acetylation and trimethylated H3K4, H3K36, and H3K79, and heterochromatin is characterized by low levels of acetylation and high levels of H3K9, H3K27, and H4K20 methylation [34]. Recent advance studies identified that histone modification levels are predictive for actively transcribed genes and are associated with high levels of H3K4me3, H3K27ac, H2BK5ac and H4K20me1in the promoter. Whereas, H3K79me1 and H4K20me1 are along the gene body [5]. In general, acetylation of histones is one of the important post-translational modifications where the lysine residue of histones is reversibly modified by two sets of enzymes–histones acetyl transferases (HATs) and histone deacetylases (HDACs).
The HATs use the cofactor acetyl CoA to catalyze the transfer of an acetyl group to the ε-amino group of lysine side chains of histones. This neutralizes the lysine’s positive charge and therefore loosens the interactions between histones and DNA. Thus, chromosomal domains are more accessible for the transcription factors to the gene promoters. Moreover, this unfolding of chromosomal domains also enhances the process of transcriptional elongation [46].
Two major types of HATs are responsible for histone acetylation: type-A and type-B. The type-B HATs mainly acetylate newly synthesized histone H4 at K5 and K12, but also certain sites within H3. However, they only acetylate free histones and not those already deposited into chromatin. Type-B pattern of acetylation is important for deposition of histones, is highly conserved, and generally found in the cytoplasm. Type-A HATs are more diverse and are classified into 3 groups according to amino acid sequence homology – GNAT, MYST, and CBP/p300 families [47]. Another important group of histone modifying enzymes, HDACs, are the predominant transcriptional repressor enzymes that act to reverse the effects of HATs and deacetylates lysine (K) to restores the positive charge of the lysine. There are four different classes of HDACs. Class I contain the enzymes HDACs 1, 2, 3, and 8, and class II contain the enzymes HDACs 4, 5, 6, 7, 9, and 10. Class IV has only a single member, HDAC11, while class III (referred to as sirtuins) are homologous to yeast scSir2. This latter class, in contrast to the other three classes, require a specific cofactor NAD+ for its activity [45]. Thus, the dynamic state of chromatin results from the crosstalk between HDACs and HATs. HATs and HDACs are therefore vital as they play a key-controlling role in developmental processes, and their deregulation has been linked to the progression of cancers (leukemia, colorectal and breast cancer), inflammatory diseases and different human disorders, like the Rubinstein–Taybi and Fragile X syndromes [48].
The next important histone modification is histone methylation. Histone methylation occurs on the side chains of lysine and arginine residues at histone tails of H3 and H4 [49]. Histone methyl transferases (HMTs) and histone demethylases (HDMs) are the two important factors that control or regulate DNA methylation through chromatin dependent transcription repression or activation. Histone methylation and demethylation modifications are essential and associated with either transcriptional repression or activation. This modification does not alter the charge of the histone protein, unlike acetylation and phosphorylation. In this modification, lysine may be mono-, di-, or tri-methylated, whereas arginine may be mono-, symmetrically or asymmetrically dimethylated [50]. Studies have shown that monomethylations of H3K27, H3K9, H4K20, H3K79 and H2BK5, and trimethylation of H3K4 (H3K4me3), H3K79me3 are responsible for gene activation, while H3K27me3, H3K9me3 are responsible for gene repression [51, 52]. It has been observed that histone methylation and other epigenetic regulation mechanisms are associated with aging and neurodegenerative diseases [53]. These studies further underscore that histone methylation is an important modification that takes place at all time.
3.2.2. Histone Phosphorylation
Primary sites for histone phosphorylation are the N-terminal tails, even though other regions within the core are also present. Histone tails are phosphorylated by different kinases and the phosphate groups are removed by phosphatases. This is a highly dynamic process that commonly takes place on serine, threonine, and tyrosine residues of histone tails [54]. As in the case of phosphorylation of H3Y41, which is phosphorylated by the non-receptor tyrosine kinase JAK2 [45]. Various cellular processes has been associated with phosphorylation of histones, including transcriptional regulation, apoptosis, cell cycle progression, DNA repair, chromosome condensation, and developmental gene regulation [55–57]. Phosphorylation of serine and threonine residues of the histone tails in general is associated with chromatin condensation during mitosis and meiosis. It has been found that C-terminal phosphorylation of Thr119 in histone H2A is linked to regulation of chromatin structure and function during mitosis [55]. Phosphorylation of H3 Ser10 (H3S10) has been observed – in chromatin compaction during mitosis. In addition, phosphorylation of H3S10 also plays a key role in transcriptional regulation of the NF-κB mediated pathway, including early genes like c-jun and c-fos [52]. Phosphorylation of H2A at Ser139 (γ-H2AX) is an important histone modification that plays a major role in DNA double-strand break by helping recruit DNA damage repair proteins to the site. Moreover, phosphorylation of γ-H2AX has been identified as one of the early events after DNA damage [58]. Thus, in parallel with other histone modifications, histone phosphorylation plays an important role in cells.
3.2.3. Ubiquitination and epigenetic modification
Histone ubiquitination was identified 28 years ago [59], but remains one of the least understood histone modifications. Until recently, it has been known that ubiquitination is a process that regulates a variety of cellular events, including protein degradation, stress response, cell-cycle, protein trafficking, endocytosis signaling, and transcriptional regulation [60]. Now, research on the role of ubiquitination on gene regulation is being considered as an important epigenetic modification and a new field of study. Ubiquitin is a 76 amino acid protein that can either direct the conjugated protein to proteasomal degradation or act as a modifier for protein function. Typically, ubiquitin receptors recognize a protein that is tagged with ubiquitin and remove it from the cell through the proteasomal degradation pathway. Otherwise, it modifies protein function by three-step enzymatic reaction, involving ubiquitin-activating, -conjugating and -ligating enzymes E1, E2 and E3, respectively. These enzymatic reactions conjugate either one (monoubiquitination) or multiple (polyubiquitination) ubiquitin moieties on the target polypeptides or the lysine residues in which ubiquitin attaches, depending on the substrate-specific combination of used E2 and E3 [61, 62]. More recent studies have revealed that reversible ubiquitylation of histone H2B by E2 and E3 has been associated in transcriptional activation and gene silencing. Ubiquitylation and subsequent deubiquitylation of H2B are both required for transcriptional elongation of certain stress-inducible genes, and removal of the ubiquitin tag from H2B enables the establishment of telomeric silencing through the association of Sir factors along with chromatin [63]. During transcription, ubiquitination of H2A is generally associated with gene silencing whereas ubiquitination of H2B has been related to both gene activation and silencing [61]. Ubiquitination of H3 and H4 are less abundant and as yet, no functional consequence has been assigned to this modification. It has been reported that murine HDAC6 is associated with two proteins, p97/VCP/Cdc48p, a homologue of yeast Cdc48p, and phospholipase A2-activating protein, a homologue of yeast UFD3 (ubiquitin fusion degradation protein 3) is involved in ubiquitination. A zinc-finger domain at the C-terminal of HDAC6, called “ubiquitin carboxyl-terminal hydrolase-like zinc finger” (ZnF-UBP) domain, signifies a possible link between histone acetylation and histone ubiquitination [64]. Moreover, studies have shown that H2B ubiquitination is upstream of H3-K4 methylation [65]. Aberration in the ubiquitin system plays a significant role in neurodegenerative diseases like Alzheimer’s, Parkinson’s, and Huntington’s disease, although the detailed role of epigenetic changes mediated by ubiquitination is poorly understood.
3.2.4. SUMOylation
SUMOylation is the attachment of a small ubiquitin-related modifier (SUMO) moiety of around 11-kDa protein, covalently present at specific lysine residues in a variety of target proteins. SUMOylation cause modulation of protein functions by changing its stability, localization, or interaction with other proteins and has important roles in cell proliferation, differentiation, and apoptosis [66]. SUMOylation is an enzyme-catalyzed, multistep, reversible process that specifically targets proteins harboring a SUMO interaction motif [67, 68]. SUMOylation takes place mostly in transcription factors or transcription regulators and leads to transcriptional repression [69, 70]. A number of transcriptional repressors, such as the histone deacetylases HDAC1, HDAC4, HDAC6, and HDAC9 and the repressor C-terminal binding protein (CtBP), have shown to be subjected to sumoylation [71]. However, the precise molecular mechanisms of these repressions are not well known. In rheumatoid arthritis (RA), there is evidence that SUMO is overexpressed in synovial tissue and synovial fibroblasts (SF) [72]. Recent studies have shown that de-SUMOylation in rheumatoid arthritis synovial fibroblasts (RASFs) decreased the levels of histone acetylation, which later cause a reduction in the expression of certain matrix metaloproteinases and interleukins, thereby reducing the destructive potential of the fibroblasts [73]. Another study also showed that telomeric silencing in S. cerevisiae has been associated with decreased levels of acetylation where sumoylation of histones may enhance its silencing effect [74]. Sir2, one of the members of the conserved sirtuin family of NAD+-dependent histone deacetylase undergo sumoylation at several lysine residues by SUMO ligase Siz2, which therefore affects its function in telomere silencing [75]. Taken together, although there are some studies in this area, still it needs a better understanding of the underlying epigenetic impact of these modifications.
3.2.5. ADP-ribosylation
ADP-ribosylation of proteins is a cellular process in which the reversible post-translational modification of proteins are catalyzed by poly-ADP-ribose polymerases (PARPs), a family of nuclear enzymes that transfer and polymerize ADP-ribose units from NAD+ on to a variety of nuclear proteins to form a branched polymer known as poly-ADP-ribose (PAR). ADP-ribosylation therefore, plays an important role in a wide range of physiological and pathophysiological processes, including inter- and intracellular signaling. In recent years, it has been found that ADP-ribosylation also has nuclear functions, including histone modification, transcriptional regulation, DNA repair pathways, maintenance of genomic stability, telomere dynamics, cell differentiation and proliferation, and necrosis and apoptosis [76]. Until recently, there is less information on the direct regulation of epigenetic modifications by ADP-ribosylation. A growing body of evidence supports a role for PARP in heterochromatin organization through the establishment and maintenance of DNA methylation patterns. Few studies have shown that binding of PARP1, a major PARP, to the broken DNA ends induces significant PARylation, which results in recruitment and modification of the chromatin-associated proteins and therefore, changes the chromatin microenvironment [77]. Methyl CpG binding protein 2 (MeCP2) is a chromosomal protein that is greatly abundant within the brain and mutation of which causes the neurodevelopmental disorder Rett syndrome. This MeCP2 protein undergoes different post-translational modifications and recent studies have shown that it also undergoes polyADP-ribosylation mediated modifications. MeCP2 can bind to both the methylated and non-methylated regions of chromatin [78], although the exact molecular mechanism of this interaction is still unclear. Another study has revealed that endogenous MeCP2 from mouse brain shows polyADP-ribosylation, and deletion of the domains responsible for polyADP-ribosylation results in increased heterochromatin clustering. Furthermore, it was found that PARP1 deficiency increases the ability of MeCP2 to aggregate and to bind to pericentric heterochromatin [79]. A recent study has shown the role of ADP-ribosylation as an element of the histone code. Mass spectrometry and electron-transfer dissociation studies showed that K13 of histone H2A, K30 of H2B, K27 and K37 of H3 as well as K16 of H4 as ADP-ribose acceptor sites catalyzed by PARP1 [80]. Studies also showed that PARP1-dependent ribosylation of histones influences their subsequent methylation pattern, with the inhibition of H1 histone [81]. These data illustrate the possible role of ADP-ribosylation mediated chromatin modifications.
3.3 Nucleosome positioning
The nucleosome is the smallest unit of chromatin that takes part in DNA packaging as well as regulation of gene expression. Nucleosomes are a barrier to transcription, which impairs the binding of transcription factors to the DNA, as well as inhibit the transcriptional elongation by polymerases. In particular, the precise position of nucleosomes around the transcription start sites (TSSs) is important and influences the initiation of transcription. Promoter regions, just upstream of TSS at the 5′ end of a gene are mostly devoid of nucleosomes by poly-dA/dT sequences and are called “nucleosome free regions” (NFRs). NFRs are associated with active regulatory elements like enhancers, insulators, and silencers are profoundly important for transcription initiation. In the 5′ NFR, there are two highly positioned nucleosomes, termed the −1 and +1 nucleosomes, which are precisely localized. The −1 nucleosome is located upstream of the NFR, whereas +1 nucleosome is located downstream of the NFR in the yeast genome. Downstream of the +1 nucleosome, nucleosomes are less precisely localized and beyond ~1 kb from the TSS, there is an increased tendency for random nucleosome positions [82]. Nucleosomes strongly localized at the downstream of TSS at the 5′ end of a gene causes gene suppression [83]. If nucleosomes are displaced by even 30 bp at TSS, there are changes implicated in the activity of RNA polymerase II. Like 5′ ends, 3′ ends are also marked by distinct nucleosome positioning as well as an NFR region at the 3′ end. The 3′ NFR specifies the position where RNA polymerase II terminates transcription ([82]). Importantly, nucleosome positioning can also influence DNA methylation because DNMTs preferentially target nucleosome-bound DNA [27]. Methylated DNA is associated with condensed heterochromatin domains, where nucleosomes are strictly positioned, and thereby obstructing transcription. Recent studies have shown the connections between nucleosome positioning, DNA methylation, and other epigenetic factors during gene regulation [84]. Given above studies suggest that nucleosome positioning play an important role during gene regulation along with other epigenetic factors.
3.4. MicroRNA (miRNA)-mediated epigenetic modification
MiRNAs are non-coding RNAs, endogenously encoded in the genome and the size ranges from 17–25 nucleotides. The detailed biosynthesis of miRNA processing and gene regulation was previously discussed [85]. Sequential miRNAs processing have an important role in gene regulation and has done either by mRNA degradation or by translational repression [86]. MiRNAs are mostly located in the CpG island and therefore, are likely to be controlled by DNA methylation machinery. For example, miRNA like miR-127 was found to be expressed more in cancer cell lines after the treatment with Aza and 4 phenyl buteric acid (PBA) which lower DNA methylation levels and therefore open the chromatin structures to induce the expression of genes that had been silenced epigenetically [87]. It has been found that in colorectal cancer, there is an increased expression of miR-143 and reduced DNMT3A levels. Meanwhile, in lung cancer, the expression of miR-29 is increased and DNMT3B levels are decreased. In both cases, there is decreased cell growth and colony formation [88]. Another recent study has shown that miRNA-34b targets DNMT1, HDAC1, HDAC2 and HDAC4 in prostate cancer cell lines. More interestingly, miR-34b itself is epigenetically silenced by DNA methylation. The over expression of miR-34b in prostate cancer cells resulted in partial demethylation of miR-34b gene at an 5′ upstream sequence, as well as showed enrichment of trimethylated histone H3 lysine 4 (H3K4me3) mark for an active chromatin [89].
Regarding inflammation, a recent study has shown that miRNA 199 was found to be hyper methylated in chronic obstructive pulmonary disorder [90]. Furthermore, in obesity and metabolic diseases associated inflammation, the genes are epigenetically regulated by miRNAs. Similar to other diseases, miRNA primarily down regulates genes associated with proinflammatory pathways. For example, miRNA-146b-5 is decreased in monocytes from obese and type 2 diabetic mellitus patients. It has also been found that toll-like receptor 4 (TLR4)-induced inhibition of miRNA-107 is impaired in obesity, resulting in an increased inflammatory response in macrophage [91]. Moreover, miRNA-146a confines TLR-mediated signaling by blocking the signaling molecule TRAF6, whereas miRNA-155 down regulates the lipid phosphatase SHIP1, responsible for macrophage activation. MiRNA-132 binds to acetylcholine (ACH) mRNA and exerts its anti-inflammatory effects by reducing peripheral inflammation. Macrophages in culture when exposed to LPS leads to up-regulation of miRNA-155, which targets the mRNA for CCAAT/enhancer binding protein Beta (C/EBP Beta), is implicated in the regulation of pro-inflammatory cytokines during acute macrophage activation [92]. All of these studies therefore confirm that miRNA mediated epigenetic regulation of genes play an important role not only in cancer but also in inflammatory diseases.
3. Epigenetic mechanisms of infection and inflammation
The epigenome, the overall epigenetic state of an organism, is just as important as the genome to normal development. Importantly, environmental factors such as nutrients, pollution, toxins, infections, and hypoxia can have profound effects on the epigenetic signature and trigger susceptibility to diseases [92, 93] (Figure 2). For example, recently it has been found that the in-utero environment can also change the epigenome, with long-term adaptable phenotypes for gene regulation and age-associated diseases [94]. The studies by Bobetsis et al. showed that periodontal infection can lead to placental-fetal exposure and when coupled with fetal inflammatory response, leads to preterm delivery [95].
Figure 2. Environmental factors and epigenetic modulation in humans.

The figure demonstrates a sum of the various sources presents in the environment that regulates epigenetic parameters on humans. Among them, some might be advantageous for body health and performance such as, exercise, healthy dietary practice, others, like toxic chemicals, stress and misuse of drugs might be detrimental and impede body’s balance that manifest as a disease or psychological disorder. Long-term exposure to inflammation and immune system of the body could also trigger epigenetic alterations.
3.1. Epigenetics of environment associated infection and inflammation
Genome-wide epigenetic marks of DNA as well as chromatin modifications do not persist throughout life, but undergo defined, synchronized changes during different stages of development, predominantly in mammals. These alterations lead to the lineage- and tissue-specific expression of genes [96, 97]. Mostly in mammals and occasionally in plants, there is a change in epigenetic pattern after birth. Besides the developmental ontogeny of epigenetic modifications, the variation in epigenetic pattern is thought to be largely because of extrinsic or environmental factors and intrinsic factors, but their relative contributions remain largely unknown [98]. Recently, the possible impact of the environment on epigenetic regulation has drawn significant interest. To date, most studies done in ‘environmental epigenetics’ has been on DNA methylation, which is essential in development and is involved in processes such as genomic imprinting and silencing transposable elements [96, 99]. However, the role of environment in the change of the epigenome is not very straightforward. Developmental and pathological phenotypes could change the measured levels of epigenetic modifications at specific loci. Studies have been done to explore the role of environmental factors and diet in the regulation of genes by DNA methylation and covalent histone modifications. All of these studies are more nascent and work is still going on to understand the mechanism. Few experiments have been done to understand the role of stress on epigenetic changes and found that when laboratory rodents experience prenatal stress, including maternal separation, maternal care, abusive caregiving in infancy, juvenile social housing, and adult social stress, there are variations in DNA methylation and histone modification [100].
Epigenetic states are also modified by the emotional health of an individual as studies were done with psychosis, psychotic disorders, and severe depression [101]. Anti-androgenic compound ‘vinclozolin’, an anti-fungal agent used in vineyards or chemical methoxychlor, an oestrogenic compound used as a pesticide, can cause reproductive abnormalities in laboratory animals. These groups of chemicals also have the potential to cause cancer in humans as well as reproductive abnormalities [102]. Several other human population studies report that the nutritional status of grandparents can have phenotypic consequences in their grandchildren. These trans-generational effects are not because of mutations but could be related to epigenetic inheritance [103, 104]. An effect of diet on the methylation status of DNA and thereby on the phenotype in humans comes from a recent study on patients with hyper-homocysteinaemia where there is an unbalanced DNA methylation status. Normal methylation is restored at the imprinted IGF2-H19 locus when folate is included in the diet because folate is needed for the methionine cycle, and therefore, for the synthesis of S-adenosylmethionine, the methyl donor for DNA methylation. Decrease of folate in the cells lead to an increase in S-adenosylhomocysteine, which is a powerful inhibitor of S-adenosylmethionine-dependent methyltransferases like DNMT1- and makes the cell have an increased level of methylation. [104, 105].
Recently, it has been reported that exercise has a positive effect, and a six-month exercise program can influence the genome wide methylation pattern in adipose tissue affecting adipocyte metabolism [106, 107]. Exposures to early life risk factors like gestational diabetes, maternal smoking, and maternal obesity are putative early-life predictors of adult cardiovascular disorders associated with epigenetic changes [108]. Recent studies depict that environment plays a crucial role in the global or local epigenetic changes in immune cells that are frequently observed in obesity and type-2 diabetes mellitus, which further perpetuate with changes in the phenotype and function of these immune cells. Diseases like obesity and metabolic disorders are linked to environmental factors in addition to the genetic factors. Furthermore, studies have shown that the environmental factors play a significant role in the expression of genes related to metabolic, endocrine and neural functions and therefore, reprogram immune cells to the diseases [91]. Taken together, studies discussed above suggest that environmental factors serve a potential role in epigenetic regulation of genes during different disease conditions.
3.2. Epigenetics of microbial mediated inflammation
Inflammation is a complex set of physiological responses to different noxious stimuli like infection, trauma, injury, toxins, and other imbalances [109]. An acute inflammatory response is associated with the recruitment of leukocytes, including the mast cells followed by macrophages from the blood to the damaged sites [109]. Whereas, in chronic inflammation, the condition is persistent and underlies many diseases, including periodontal disease and diabetes mellitus [110].
Macrophages are therefore one of the critical regulators in the inflammatory responses. Immediately after the microbial invasion, microbial products signal through extracellular and intracellular pattern-recognition receptors (PRRs) or sensors (e.g., Toll-like receptors, or TLRs, and nitric oxide dismutase or NOD proteins), which recognize small molecular sequences referred to as “pathogen-associated molecular patterns (PAMPs)”. Thus, signaling pathways are triggered which initiate inflammatory activation to produce histamine and pro-inflammatory cytokines like IL-1, TNFα, and IL-6. Inflammatory genes that encode for cytokines, chemokines, and other molecules responsible for inflammation that are found to be regulated by pro-inflammatory transcription factors, including nuclear factor kappa B (NF-κB) and activator protein (AP)-1, FOXP3, IRF, and STAT families. Transcription factors along with the inflammatory genes are regulated by epigenetic mechanisms including DNA methylation, histone modifications, and RNA-associated silencing by small non-coding RNAs [92, 111]. Thus, the body undergoes profound alterations in systemic homeostasis to support host defense.
3.3. DNA Methylation and Inflammation
DNA methylation is also an important mechanism of regulation of inflammatory genes. Recent studies have revealed that LPS cause down regulation of Thy-1 gene expression by activating HDACs via TLR4- signaling [112]. In disease like cystic fibrosis, TLR2 gene undergoes promoter hypo methylation, which is associated with enhanced pro-inflammatory response to bacterial peptidoglycan by the bronchial epithelial cells [113]. Intestinal homeostasis is maintained by methylation of DNA and acetylation of histone regulates TLR4 in intestinal epithelial cells [114]. Epigenetic modifications including DNA methylation and histone modifications play an important role in the regulation of TNFα and therefore inflammation associated with it [115]. Bacterial infections can alter the epigenetic machinery and therefore can alter the status of the genome which contribute host defense or pathogen load [116]. Microarray studies in mice explained that maternal health including infection by both bacteria ad virus contributes to changes in expression of key developmental genes in the placenta and therefore, linked to the development of the offspring [117]. Infection by Helicobacter pylori (HP) causes inflammation and induces alterations in the DNA methylation pattern in gastric epithelial cells showed that HP-induced DNA methylation in the Runx3 locus causes loss of expression in gastric epithelial cells responsible for the development of the disease [118].
3.4. Histone Methylation in Inflammation
Regulation of gene expression of CD4+ T cells and CD8+ T cells are primarily done by histone methylation, including H3K4me3, H3K9me2, H3K9me3, and H3K27me3 [119, 120]. Studies revealed regulatory regions of gene loci in T cells undergo dynamic and complex changes in histone methylation during their differentiation to distinct subsets of effector T cells [121]. Studies have shown that Jmjd3, a member of the Jumonji family, is expressed in macrophages when exposed to pathogens and pro-inflammatory cytokines, where it activates the polycomb-group (PcG) target genes and regulates their H3K27me3 levels and transcriptional activity. Some of the PcG proteins responsible for inflammation are subjected to aberrant DNA methylation. PcG proteins bind to the regulatory regions of target genes and recruit DNMTs for more efficient repression. Inflammation during sepsis also causes NF-κB/RelB- pathway mediated repression of pro-inflammatory genes inducing H3K9me3 [122]. Jmjd3 erases histone marks and has been found to regulate differentiation and cell identity in macrophages, thereby controls inflammation by reprogramming of the epigenome. Continuous exposure to IL-4 activates Jmjd3 and the release of H3K27me3 repressive marks from the STAT6 promoter. Activated STAT6 then positively regulates Jmjd3 by binding to its promoter. Removal of H3K27 methylation marks by Jmjd3 trigger expression of specific inflammatory genes [123]. These studies clearly indicate the dynamic role of histone methylation during inflammatory response.
3.5. Histone Acetylation and Inflammation
Genes are activated by the acetylation of histone by HATs and repressed by the activity of HDACs during inflammation caused by different agents like LPS, IL-1b, TNFα, etc. Many of these stimuli act through induction of pro-inflammatory transcription factors such as AP-1 and NF-κB, but other intracellular kinase pathways such as MAPKs and PI3K are also important [124]. Recent reports have shown the role of HDACs in the regulation of pro-inflammatory genes. For example, corticosteroids switch off inflammatory genes in asthma by the recruitment of HDAC2 to the promoter region NF-κB stimulated inflammatory gene complex and inhibition of HAT activity. There is a reduction in HDAC activity and HDAC2 expression in chronic obstructive pulmonary disease (COPD), a corticosteroid insensitive disease, where amplified inflammation and resistance to the actions of corticosteroids were observed [18]. CBP/p300 (cAMP-response element binding (CREB)- protein) has the intrinsic HAT activity and also recruit other HATs in the acetylation of histones at the promoter region of inflammatory genes (IL-1, IL-2, IL-8, and IL-12), thus regulating gene expression via transcriptional activation as well as displayed decreased HDAC activity. Transcription of both pro- and anti-inflammatory cytokines are regulated by the recruitment of HDACs to gene promoters via co-repressor complexes and transcription factors such as FOXP3, STATs, GATAs, ZEB1, and NF-κB [125]. In response to cytokine treatment, IKK-α, a member of IκB kinase complex binds to the NF-κB promoters with the aid of polymerase II and CBP, where it acetylates histone H3 at Lys9 and phosphorylates histone H3 at Ser10. [126, 127]. This is further responsible for the subsequent CBP-mediated acetylation of histone H3 on Lys14 ([128]. The p38 protein plays an important role in the induction of inflammatory and immune responses by enhancing the H3 phosphorylation marked promoters and thereby, enhance the recruitment to selected chromatin targets [129].
4. Clinical relevance of Epigenetic Modifiers
With the advancement of new diagnostic tools, it reveals that there are many diseases that are caused by epigenetic modifications. Development of ‘epigenetic therapies’ now has the immense potential towards the treatment of many diseases including cancer, cardiovascular disorders, and also inflammation. Recently, many agents have been found to modify epigenetic patterns by changing DNA methylation patterns or cause modification of histones. Many of these agents are currently being tested in clinical trials [2].
Two general types of HDAC inhibitors are commonly used for treatment, pan inhibitors that are broad acting and inhibitors that target a specific class of HDAC enzyme. DNA methyltransferase (DNMT) inhibitors, 5-azacytidine (clinical name Vidaza) and 5-Aza-2′deoxy-5-azacytidine (clinical name decitabine) are pan inhibitors and are now used to increase fetal hemoglobin production by causing hypo methylation of γ-globin genes in patients with sickle cell disease [130]. HDAC inhibitors have been found to be – a novel category of anti-cancer drugs. To date, the United States Food and Drug Administration have approved four HDAC inhibitors, Vorinostat, Romidepsin, Panobinostat, and Belinostat. More specifically, these HDAC inhibitors have the least severe side effects and are mostly used for hematologic cancers. More clinical trials are continuously expanding to address other types of cancer and also nonmalignant diseases. HDAC inhibitors are also used in various types of neurodegenerative diseases, inflammation disorders, and cardiovascular diseases [131]. Similar to Aza, Zebularine is another cytidine analog that inhibits DNMT1 activity. Although not yet FDA approved, this isoform has found good results in mouse models [132].
Three naturally occurring small molecules such as curcumin, anacardic acid, and garcinol have been found to have HAT inhibitory activity. Curcumin, the active ingredient in turmeric, is in phase II trials in advanced pancreatic, breast, and colorectal cancer (NCT00094445, NCT01740323, NCT01490996). HMT and HDM inhibitors, such as BIX-01294, chaetocin, and UNCO224, DZNep, are primarily in a preclinical stage of development. However, the HDM inhibitor phenelzine sulfate, a monoamine oxidase A inhibitor, is in phase II trials in combination with docetaxel in prostate cancer (NCT01253642) (Figure 3, 4). Recent studies have suggested that epigenetic modulation with chromatin-modifying agents can induce stemness and dedifferentiation and increase developmental plasticity. For instance, valproic acid (VPA), an HDAC inhibitor, has been shown to promote self-renewal/expansion of hematopoietic stem cells and facilitate the generation of induced pluripotent stem cells (iPSC) [133]. More recent studies show that the epigenetic reprogramming of adult somatic cells into pluripotent stem cells could be a safe method to generate iPSCs and may provide an attractive source of stem cells for regenerative medicine [134]. Recently, studies have shown that combinations of epigenetic modifiers are also found to play an important role in the reduction of inflammation. A single dose of combinatorial administration of Aza+TSA after the onset of acute lung injury (ALI) has been found to significantly attenuate lung vascular hyper permeability and inflammatory lung injury. Moreover, the combinatorial treatment with Aza+TSA reduces inflammation and promotes anti-inflammatory M2 macrophages during ALI [135, 136]. These two studies clearly demonstrated that epigenetic modifiers have a therapeutic potential for patients with sepsis-induced vascular injury and inflammation.
Figure 3. Clinical trials in aberrant DNA methylations associated with disease outcome.

This diagram represents the drugs mostly used in different phases of clinical trial that are used to alter DNA methylation associates with diseases in an individual. Aberrant DNA methylation is associated with gene repression in the promoter and activation in the gene body results in the manifestation of various diseases, including a variety of cancers and neurodegenerative diseases. Several molecules have been screened for altering the DNA methylation status associated with the disease outcome and are currently in different phases of clinical trial.
Figure 4. Clinical trials in aberrant histone modifications associated with disease outcome.

Histone modifications by HATs lead to active chromatin state associated with the activation of different genes, including inflammatory genes or genes responsible for neuronal functions. HDACs are responsible for repressed chromatin and therefore, transcriptionally inactive state of genes. Aberrant expression of HDACs might also shut down different essential genes responsible to maintain normal function of the body and thereby results with diseased state. Thus, various small molecules and drugs are now under screening and trials to use them clinically to modify the altered epigenetic state and reduce the diseased condition.
5. Summary and Future perspective
Previously for several decades, genetics was key to understand human diseases, but in recent years, epigenetics has taken the most important place to describe human diseases where environment, lifestyle, and diet play important roles in epigenetic modulation. Changes in the epigenetic landscape give rise to several significant human diseases, and the focus is to find therapies that can reverse gene expression. But despite the promise of epigenetic therapy, the clinical use of these agents may cause the nonspecific activation of genes and transposable elements in normal cells, as well as potential mutagenicity and carcinogenicity are the major concerns regarding their clinical use. Still, it is good that trials are mainly on life-threatening diseases like cancer where the target is to hit abnormally methylated CpG island with a single drug. Most commonly, drugs used in the clinical trial are inhibitors of DNMTs and/or HDACs, which has a broad spectrum of inhibition rather than specific activity. This could result in wide range change in the epigenetic pattern that could also upregulate several other genes including diseased genes or proto-oncogenes. Therefore, it is important to focus on individual inhibitors that will have more specific inhibition of a particular gene or a set of genes that will result in a more specific inhibition. At the end, methylation of CpG islands increases with age and could be important for the development of chronic diseases like inflammation in addition to cancer. Drugs and lifestyle changes could bring reversionor slow gradual epigenetic silencing.
6. Concluding Remarks
The study of epigenetics, therefore, has profound clinical significance. Understanding the role of epigenetic aberrations in disease progressions still is primitive. Future studies are needed to understand the relationship between epigenetic mechanisms, gene expression and the environment, and more emphasis is to be given on clinical trials of novel epigenetic therapies directed at a wide variety of diseases. Moreover, it is important to decrease the risk of epigenetic instability and epigenetic abnormalities that may be caused by broad-spectrum inhibitors later in life. Therefore, recent studies are focusing on identifying disease specific inhibitors rather than pan inhibitors. The promise for epigenetic therapies specific for diseases is as an essential treatment goal needed to be fulfilled. In this review, we have summarized the investigations that are critical to understand the complex epigenetic mechanisms to provide new ways for treating diseases associated with infection and inflammation.
Supplementary Material
Highlights.
Epigenetic state plays an important role in disease manifestation.
DNA methylation, Histone modification, nucleosomal positioning and miRNA, mediate major epigenetic regulation.
Environmental factors are also responsible for associate alteration of the epigenetic state.
Targeting the epigenetic machinery is a newer approach to change the altered epigenetic state.
Acknowledgments
This work was supported, in part, by American Heart Association Grant-in-Aid 16GRNT30950010 and National Institutes of Health COBRE grant P20GM104936 (to JR).
Abbreviations
- Aza
5-Aza 2 deoxycytidine
- TSA
Trichostatin A
- MeCP2
Methyl CpG binding protein 2
- SLE
Systemic lupus erythematosus
- RA
Rheumatoid arthritis
- DNMTs
DNA methyltransferases
- HDACs
Histone deacetylases
- HAT
Histones acetyl transferases
- HMT
Histone methyl transferases
- HDM
Histone demethylases
- MLH1
MutL homolog 1
- CBP
CREB-binding protein
- RASF
Rheumatoid arthritis synovial fibroblasts
- PBA
Phenyl butyric acid
- FOXO3
Forkhead box O3
- IRF1
Interferon regulatory factor 1
- STAT
Signal transducer and activator of transcription
- LPS
Lipopolysaccharide
- PcG
Polycomb-group proteins
- MAPK
Mitogen-activated protein kinases
- PI3K
Phosphatidylinositol-4,5-bisphosphate 3-kinase
- ZEB1
Zinc finger E-box-binding homeobox 1
- SUMO
Small ubiquitin-related modifier
- TSS
Transcription start sites
- NFR
Nucleosome free region
- PRRs
Pattern-recognition receptors
- PAMP
Pathogen-associated molecular patterns
- NOD
Nitric oxide dismutase
- APR
acute phase response
- NF-κB
Nuclear factor kappaB
- AP
Activator protein
- Jmjd
Jumonji family
- COPD
Chronic obstructive pulmonary disease
- PARP
Poly-ADP-ribose polymerases
- ACH
Acetylcholine
- TLR4
Toll-like receptor 4
- EBP
Enhancer binding protein
- TNFα
Tumor necrosis factor alpha
- HP
Helicobacter pylori
- CREB
cAMP-response element binding
- VPA
Valproic acid
- iPSCs
Induced pluripotent stem cells
- ALI
Acute lung injury
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Pikaard CS. The epigenetics of nucleolar dominance. Trends in genetics : TIG. 2000;16:495–500. doi: 10.1016/s0168-9525(00)02113-2. [DOI] [PubMed] [Google Scholar]
- 2.Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429:457–463. doi: 10.1038/nature02625. [DOI] [PubMed] [Google Scholar]
- 3.Reik W, Santos F, Dean W. Mammalian epigenomics: reprogramming the genome for development and therapy. Theriogenology. 2003;59:21–32. doi: 10.1016/s0093-691x(02)01269-4. [DOI] [PubMed] [Google Scholar]
- 4.Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31:27–36. doi: 10.1093/carcin/bgp220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Portela A, Esteller M. Epigenetic modifications and human disease. Nature biotechnology. 2010;28:1057–1068. doi: 10.1038/nbt.1685. [DOI] [PubMed] [Google Scholar]
- 6.Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suner D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M. Epigenetic differences arise during the lifetime of monozygotic twins. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:10604–10609. doi: 10.1073/pnas.0500398102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lund AH, van Lohuizen M. Epigenetics and cancer. Genes & development. 2004;18:2315–2335. doi: 10.1101/gad.1232504. [DOI] [PubMed] [Google Scholar]
- 8.Hewagama A, Richardson B. The genetics and epigenetics of autoimmune diseases. Journal of autoimmunity. 2009;33:3–11. doi: 10.1016/j.jaut.2009.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fedoriw A, Mugford J, Magnuson T. Genomic imprinting and epigenetic control of development. Cold Spring Harbor perspectives in biology. 2012;4:a008136. doi: 10.1101/cshperspect.a008136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Keating ST, El-Osta A. Epigenetic changes in diabetes. Clinical genetics. 2013;84:1–10. doi: 10.1111/cge.12121. [DOI] [PubMed] [Google Scholar]
- 11.Ballestar E, Esteller M, Richardson BC. The epigenetic face of systemic lupus erythematosus. Journal of immunology (Baltimore, Md: 1950) 2006;176:7143–7147. doi: 10.4049/jimmunol.176.12.7143. [DOI] [PubMed] [Google Scholar]
- 12.Ho SM. Environmental epigenetics of asthma: an update. The Journal of allergy and clinical immunology. 2010;126:453–465. doi: 10.1016/j.jaci.2010.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bhavsar P, Ahmad T, Adcock IM. The role of histone deacetylases in asthma and allergic diseases. The Journal of allergy and clinical immunology. 2008;121:580–584. doi: 10.1016/j.jaci.2007.12.1156. [DOI] [PubMed] [Google Scholar]
- 14.Hansen JC, Ghosh RP, Woodcock CL. Binding of the Rett syndrome protein, MeCP2, to methylated and unmethylated DNA and chromatin. IUBMB life. 2010;62:732–738. doi: 10.1002/iub.386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Park E, Kim Y, Ryu H, Kowall NW, Lee J, Ryu H. Epigenetic mechanisms of Rubinstein-Taybi syndrome. Neuromolecular medicine. 2014;16:16–24. doi: 10.1007/s12017-013-8285-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Urdinguio RG, Sanchez-Mut JV, Esteller M. Epigenetic mechanisms in neurological diseases: genes, syndromes, and therapies. Lancet neurology. 2009;8:1056–1072. doi: 10.1016/S1474-4422(09)70262-5. [DOI] [PubMed] [Google Scholar]
- 17.Sadri-Vakili G, Cha JH. Mechanisms of disease: Histone modifications in Huntington’s disease, Nature clinical practice. Neurology. 2006;2:330–338. doi: 10.1038/ncpneuro0199. [DOI] [PubMed] [Google Scholar]
- 18.Adcock IM, Ito K, Barnes PJ. Histone deacetylation: an important mechanism in inflammatory lung diseases. Copd. 2005;2:445–455. doi: 10.1080/15412550500346683. [DOI] [PubMed] [Google Scholar]
- 19.Guo CL, Liu L, Jia YD, Zhao XY, Zhou Q, Wang L. A novel variant of Oct3/4 gene in mouse embryonic stem cells. Stem cell research. 2012;9:69–76. doi: 10.1016/j.scr.2012.04.004. [DOI] [PubMed] [Google Scholar]
- 20.Gray SG, De Meyts P. Role of histone and transcription factor acetylation in diabetes pathogenesis. Diabetes/metabolism research and reviews. 2005;21:416–433. doi: 10.1002/dmrr.559. [DOI] [PubMed] [Google Scholar]
- 21.Robertson KD. DNA methylation and human disease. Nature reviews Genetics. 2005;6:597–610. doi: 10.1038/nrg1655. [DOI] [PubMed] [Google Scholar]
- 22.Baylin SB. DNA methylation and gene silencing in cancer, Nature clinical practice. Oncology. 2005;2(Suppl 1):S4–11. doi: 10.1038/ncponc0354. [DOI] [PubMed] [Google Scholar]
- 23.Ehrlich M. DNA hypomethylation in cancer cells. Epigenomics. 2009;1:239–259. doi: 10.2217/epi.09.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Varier RA, Timmers HT. Histone lysine methylation and demethylation pathways in cancer. Biochimica et biophysica acta. 2011;1815:75–89. doi: 10.1016/j.bbcan.2010.10.002. [DOI] [PubMed] [Google Scholar]
- 25.Glozak MA, Seto E. Histone deacetylases and cancer. Oncogene. 2007;26:5420–5432. doi: 10.1038/sj.onc.1210610. [DOI] [PubMed] [Google Scholar]
- 26.Berdasco M, Ropero S, Setien F, Fraga MF, Lapunzina P, Losson R, Alaminos M, Cheung NK, Rahman N, Esteller M. Epigenetic inactivation of the Sotos overgrowth syndrome gene histone methyltransferase NSD1 in human neuroblastoma and glioma. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:21830–21835. doi: 10.1073/pnas.0906831106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Quintero-Ronderos P, Montoya-Ortiz G. Epigenetics and autoimmune diseases. Autoimmune diseases. 2012;2012:593720. doi: 10.1155/2012/593720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhang Z, Zhang R. Epigenetics in autoimmune diseases: Pathogenesis and prospects for therapy. Autoimmunity reviews. 2015;14:854–863. doi: 10.1016/j.autrev.2015.05.008. [DOI] [PubMed] [Google Scholar]
- 29.Jones PA, Takai D. The role of DNA methylation in mammalian epigenetics. Science (New York, NY) 2001;293:1068–1070. doi: 10.1126/science.1063852. [DOI] [PubMed] [Google Scholar]
- 30.Zeng J, Nagrajan HK, Yi SV. Fundamental diversity of human CpG islands at multiple biological levels. Epigenetics : official journal of the DNA Methylation Society. 2014;9:483–491. doi: 10.4161/epi.27654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Esteller M. Non-coding RNAs in human disease. Nature reviews Genetics. 2011;12:861–874. doi: 10.1038/nrg3074. [DOI] [PubMed] [Google Scholar]
- 32.Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, Herb B, Ladd-Acosta C, Rho J, Loewer S, Miller J, Schlaeger T, Daley GQ, Feinberg AP. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nature genetics. 2009;41:1350–1353. doi: 10.1038/ng.471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Jin B, Li Y, Robertson KD. DNA Methylation: Superior or Subordinate in the Epigenetic Hierarchy? Genes & cancer. 2011;2:607–617. doi: 10.1177/1947601910393957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kanwal R, Gupta S. Epigenetic modifications in cancer. Clinical genetics. 2012;81:303–311. doi: 10.1111/j.1399-0004.2011.01809.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gopalakrishnan S, Van Emburgh BO, Robertson KD. DNA methylation in development and human disease. Mutation research. 2008;647:30–38. doi: 10.1016/j.mrfmmm.2008.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li E. Chromatin modification and epigenetic reprogramming in mammalian development, Nature reviews. Genetics. 2002;3:662–673. doi: 10.1038/nrg887. [DOI] [PubMed] [Google Scholar]
- 37.Portela A, Esteller M. Epigenetic modifications and human disease. Nat Biotech. 2010;28:1057–1068. doi: 10.1038/nbt.1685. [DOI] [PubMed] [Google Scholar]
- 38.Bourc’his D, Xu GL, Lin CS, Bollman B, Bestor TH. Dnmt3L and the establishment of maternal genomic imprints. Science (New York, NY) 2001;294:2536–2539. doi: 10.1126/science.1065848. [DOI] [PubMed] [Google Scholar]
- 39.Chen ZX, Mann JR, Hsieh CL, Riggs AD, Chedin F. Physical and functional interactions between the human DNMT3L protein and members of the de novo methyltransferase family. Journal of cellular biochemistry. 2005;95:902–917. doi: 10.1002/jcb.20447. [DOI] [PubMed] [Google Scholar]
- 40.Mosher RA, Melnyk CW. siRNAs and DNA methylation: seedy epigenetics. Trends in plant science. 2010;15:204–210. doi: 10.1016/j.tplants.2010.01.002. [DOI] [PubMed] [Google Scholar]
- 41.Lennartsson A, Ekwall K. Histone modification patterns and epigenetic codes. Biochimica et biophysica acta. 2009;1790:863–868. doi: 10.1016/j.bbagen.2008.12.006. [DOI] [PubMed] [Google Scholar]
- 42.Ramakrishnan V. Histone structure and the organization of the nucleosome. Annual review of biophysics and biomolecular structure. 1997;26:83–112. doi: 10.1146/annurev.biophys.26.1.83. [DOI] [PubMed] [Google Scholar]
- 43.Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature. 1997;389:251–260. doi: 10.1038/38444. [DOI] [PubMed] [Google Scholar]
- 44.Sadakierska-Chudy A, Filip M. A comprehensive view of the epigenetic landscape. Part II: Histone post-translational modification, nucleosome level, and chromatin regulation by ncRNAs. Neurotoxicity research. 2015;27:172–197. doi: 10.1007/s12640-014-9508-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell research. 2011;21:381–395. doi: 10.1038/cr.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yang XJ, Seto E. HATs and HDACs: from structure, function and regulation to novel strategies for therapy and prevention. Oncogene. 2007;26:5310–5318. doi: 10.1038/sj.onc.1210599. [DOI] [PubMed] [Google Scholar]
- 47.Hodawadekar SC, Marmorstein R. Chemistry of acetyl transfer by histone modifying enzymes: structure, mechanism and implications for effector design. Oncogene. 2007;26:5528–5540. doi: 10.1038/sj.onc.1210619. [DOI] [PubMed] [Google Scholar]
- 48.Timmermann S, Lehrmann H, Polesskaya A, Harel-Bellan A. Histone acetylation and disease. Cellular and molecular life sciences : CMLS. 2001;58:728–736. doi: 10.1007/PL00000896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wang Y, Jia S. Degrees make all the difference: the multifunctionality of histone H4 lysine 20 methylation. Epigenetics : official journal of the DNA Methylation Society. 2009;4:273–276. doi: 10.4161/epi.4.5.9212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ng SS, Yue WW, Oppermann U, Klose RJ. Dynamic protein methylation in chromatin biology. Cellular and molecular life sciences : CMLS. 2009;66:407–422. doi: 10.1007/s00018-008-8303-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–837. doi: 10.1016/j.cell.2007.05.009. [DOI] [PubMed] [Google Scholar]
- 52.Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005. [DOI] [PubMed] [Google Scholar]
- 53.Kramer JM. Epigenetic regulation of memory: implications in human cognitive disorders. Biomolecular concepts. 2013;4:1–12. doi: 10.1515/bmc-2012-0026. [DOI] [PubMed] [Google Scholar]
- 54.Oki M, Aihara H, Ito T. Role of histone phosphorylation in chromatin dynamics and its implications in diseases. Sub-cellular biochemistry. 2007;41:319–336. [PubMed] [Google Scholar]
- 55.Cruickshank MN, Besant P, Ulgiati D. The impact of histone post-translational modifications on developmental gene regulation. Amino acids. 2010;39:1087–1105. doi: 10.1007/s00726-010-0530-6. [DOI] [PubMed] [Google Scholar]
- 56.Cheung P, Allis CD, Sassone-Corsi P. Signaling to chromatin through histone modifications. Cell. 2000;103:263–271. doi: 10.1016/s0092-8674(00)00118-5. [DOI] [PubMed] [Google Scholar]
- 57.Banerjee T, Chakravarti D. A peek into the complex realm of histone phosphorylation. Molecular and cellular biology. 2011;31:4858–4873. doi: 10.1128/MCB.05631-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Rossetto D, Avvakumov N, Cote J. Histone phosphorylation: a chromatin modification involved in diverse nuclear events. Epigenetics : official journal of the DNA Methylation Society. 2012;7:1098–1108. doi: 10.4161/epi.21975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Goldknopf IL, Taylor CW, Baum RM, Yeoman LC, Olson MO, Prestayko AW, Busch H. Isolation and characterization of protein A24, a “histone-like” non-histone chromosomal protein. The Journal of biological chemistry. 1975;250:7182–7187. [PubMed] [Google Scholar]
- 60.Pickart CM. Mechanisms underlying ubiquitination. Annual review of biochemistry. 2001;70:503–533. doi: 10.1146/annurev.biochem.70.1.503. [DOI] [PubMed] [Google Scholar]
- 61.Dinant C, Houtsmuller AB, Vermeulen W. Chromatin structure and DNA damage repair. Epigenetics & chromatin. 2008;1:9. doi: 10.1186/1756-8935-1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhang Y. Transcriptional regulation by histone ubiquitination and deubiquitination. Genes & development. 2003;17:2733–2740. doi: 10.1101/gad.1156403. [DOI] [PubMed] [Google Scholar]
- 63.Wright DE, Wang CY, Kao CF. Histone ubiquitylation and chromatin dynamics. Frontiers in bioscience (Landmark edition) 2012;17:1051–1078. doi: 10.2741/3973. [DOI] [PubMed] [Google Scholar]
- 64.Hard RL, Liu J, Shen J, Zhou P, Pei D. HDAC6 and Ubp-M BUZ domains recognize specific C-terminal sequences of proteins. Biochemistry. 2010;49:10737–10746. doi: 10.1021/bi101014s. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sun ZW, Allis CD. Ubiquitination of histone H2B regulates H3 methylation and gene silencing in yeast. Nature. 2002;418:104–108. doi: 10.1038/nature00883. [DOI] [PubMed] [Google Scholar]
- 66.Verger A, Perdomo J, Crossley M. Modification with SUMO. A role in transcriptional regulation. EMBO reports. 2003;4:137–142. doi: 10.1038/sj.embor.embor738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Stielow B, Sapetschnig A, Kruger I, Kunert N, Brehm A, Boutros M, Suske G. Identification of SUMO-dependent chromatin-associated transcriptional repression components by a genome-wide RNAi screen. Molecular cell. 2008;29:742–754. doi: 10.1016/j.molcel.2007.12.032. [DOI] [PubMed] [Google Scholar]
- 68.Liu B, Shuai K. Regulation of the sumoylation system in gene expression. Current opinion in cell biology. 2008;20:288–293. doi: 10.1016/j.ceb.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yang SH, Sharrocks AD. SUMO promotes HDAC-mediated transcriptional repression. Molecular cell. 2004;13:611–617. doi: 10.1016/s1097-2765(04)00060-7. [DOI] [PubMed] [Google Scholar]
- 70.Ling Y, Sankpal UT, Robertson AK, McNally JG, Karpova T, Robertson KD. Modification of de novo DNA methyltransferase 3a (Dnmt3a) by SUMO-1 modulates its interaction with histone deacetylases (HDACs) and its capacity to repress transcription. Nucleic acids research. 2004;32:598–610. doi: 10.1093/nar/gkh195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Perdomo J, Verger A, Turner J, Crossley M. Role for SUMO modification in facilitating transcriptional repression by BKLF. Molecular and cellular biology. 2005;25:1549–1559. doi: 10.1128/MCB.25.4.1549-1559.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Meinecke I, Cinski A, Baier A, Peters MA, Dankbar B, Wille A, Drynda A, Mendoza H, Gay RE, Hay RT, Ink B, Gay S, Pap T. Modification of nuclear PML protein by SUMO-1 regulates Fas-induced apoptosis in rheumatoid arthritis synovial fibroblasts. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:5073–5078. doi: 10.1073/pnas.0608773104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Strietholt S, Maurer B, Peters MA, Pap T, Gay S. Epigenetic modifications in rheumatoid arthritis. Arthritis research & therapy. 2008;10:219. doi: 10.1186/ar2500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Nathan D, Ingvarsdottir K, Sterner DE, Bylebyl GR, Dokmanovic M, Dorsey JA, Whelan KA, Krsmanovic M, Lane WS, Meluh PB, Johnson ES, Berger SL. Histone sumoylation is a negative regulator in Saccharomyces cerevisiae and shows dynamic interplay with positive-acting histone modifications. Genes & development. 2006;20:966–976. doi: 10.1101/gad.1404206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hannan A, Abraham NM, Goyal S, Jamir I, Priyakumar UD, Mishra K. Sumoylation of Sir2 differentially regulates transcriptional silencing in yeast. Nucleic acids research. 2015;43:10213–10226. doi: 10.1093/nar/gkv842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hassa PO, Haenni SS, Elser M, Hottiger MO. Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going? Microbiology and molecular biology reviews : MMBR. 2006;70:789–829. doi: 10.1128/MMBR.00040-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Tallis M, Morra R, Barkauskaite E, Ahel I. Poly(ADP-ribosyl)ation in regulation of chromatin structure and the DNA damage response. Chromosoma. 2014;123:79–90. doi: 10.1007/s00412-013-0442-9. [DOI] [PubMed] [Google Scholar]
- 78.Ausio J. MeCP2 and the enigmatic organization of brain chromatin. Implications for depression and cocaine addiction. Clinical epigenetics. 2016;8:58. doi: 10.1186/s13148-016-0214-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Becker A, Zhang P, Allmann L, Meilinger D, Bertulat B, Eck D, Hofstaetter M, Bartolomei G, Hottiger MO, Schreiber V, Leonhardt H, Cardoso MC. Poly(ADP-ribosyl)ation of Methyl CpG Binding Domain Protein 2 Regulates Chromatin Structure. The Journal of biological chemistry. 2016;291:4873–4881. doi: 10.1074/jbc.M115.698357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Messner S, Altmeyer M, Zhao H, Pozivil A, Roschitzki B, Gehrig P, Rutishauser D, Huang D, Caflisch A, Hottiger MO. PARP1 ADP-ribosylates lysine residues of the core histone tails. Nucleic acids research. 2010;38:6350–6362. doi: 10.1093/nar/gkq463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kassner I, Barandun M, Fey M, Rosenthal F, Hottiger MO. Crosstalk between SET7/9-dependent methylation and ARTD1-mediated ADP-ribosylation of histone H1.4. Epigenetics & chromatin. 2013;6:1. doi: 10.1186/1756-8935-6-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Jansen A, Verstrepen KJ. Nucleosome Positioning in Saccharomyces cerevisiae. Microbiology and Molecular Biology Reviews : MMBR. 2011;75:301–320. doi: 10.1128/MMBR.00046-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Radman-Livaja M, Rando OJ. Nucleosome positioning: how is it established, and why does it matter? Developmental biology. 2010;339:258–266. doi: 10.1016/j.ydbio.2009.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Teif VB, Beshnova DA, Vainshtein Y, Marth C, Mallm JP, Hofer T, Rippe K. Nucleosome repositioning links DNA (de)methylation and differential CTCF binding during stem cell development. Genome research. 2014;24:1285–1295. doi: 10.1101/gr.164418.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Samanta S, Balasubramanian S, Rajasingh S, Patel U, Dhanasekaran A, Dawn B, Rajasingh J. MicroRNA: A new therapeutic strategy for cardiovascular diseases. Trends in cardiovascular medicine. 2016;26:407–419. doi: 10.1016/j.tcm.2016.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S, Kim VN. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425:415–419. doi: 10.1038/nature01957. [DOI] [PubMed] [Google Scholar]
- 87.Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, Jones PA. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 2006;9:435–443. doi: 10.1016/j.ccr.2006.04.020. [DOI] [PubMed] [Google Scholar]
- 88.Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nature biotechnology. 2010;28:1069–1078. doi: 10.1038/nbt.1678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Majid S, Dar AA, Saini S, Shahryari V, Arora S, Zaman MS, Chang I, Yamamura S, Tanaka Y, Chiyomaru T, Deng G, Dahiya R. miRNA-34b inhibits prostate cancer through demethylation, active chromatin modifications, and AKT pathways. Clinical cancer research : an official journal of the American Association for Cancer Research. 2013;19:73–84. doi: 10.1158/1078-0432.CCR-12-2952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hassan T, Carroll TP, Buckley PG, Cummins R, O’Neill SJ, McElvaney NG, Greene CM. miR-199a-5p silencing regulates the unfolded protein response in chronic obstructive pulmonary disease and alpha1-antitrypsin deficiency. American journal of respiratory and critical care medicine. 2014;189:263–273. doi: 10.1164/rccm.201306-1151OC. [DOI] [PubMed] [Google Scholar]
- 91.Raghuraman S, Donkin I, Versteyhe S, Barres R, Simar D. The Emerging Role of Epigenetics in Inflammation and Immunometabolism. Trends in endocrinology and metabolism: TEM. 2016 doi: 10.1016/j.tem.2016.06.008. [DOI] [PubMed] [Google Scholar]
- 92.Bayarsaihan D. Epigenetic mechanisms in inflammation. Journal of dental research. 2011;90:9–17. doi: 10.1177/0022034510378683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Barros SP, Offenbacher S. Epigenetics: connecting environment and genotype to phenotype and disease. Journal of dental research. 2009;88:400–408. doi: 10.1177/0022034509335868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ho SM, Johnson A, Tarapore P, Janakiram V, Zhang X, Leung YK. Environmental Epigenetics and Its Implication on Disease Risk and Health, ILAR. journal / National Research Council, Institute of Laboratory Animal Resources. 2012;53:289–305. doi: 10.1093/ilar.53.3-4.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Bobetsis YA, Barros SP, Offenbacher S. Exploring the relationship between periodontal disease and pregnancy complications. Journal of the American Dental Association (1939) 2006;137(Suppl):7S–13S. doi: 10.14219/jada.archive.2006.0403. [DOI] [PubMed] [Google Scholar]
- 96.Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447:425–432. doi: 10.1038/nature05918. [DOI] [PubMed] [Google Scholar]
- 97.Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247–257. doi: 10.1016/s0092-8674(00)81656-6. [DOI] [PubMed] [Google Scholar]
- 98.Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nature reviews. Genetics. 2011;13:97–109. doi: 10.1038/nrg3142. [DOI] [PubMed] [Google Scholar]
- 99.Law JA, Jacobsen SE. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nature reviews. Genetics. 2010;11:204–220. doi: 10.1038/nrg2719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gudsnuk K, Champagne FA. Epigenetic influence of stress and the social environment. ILAR journal / National Research Council, Institute of Laboratory Animal Resources. 2012;53:279–288. doi: 10.1093/ilar.53.3-4.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Pishva E, Drukker M, Viechtbauer W, Decoster J, Collip D, van Winkel R, Wichers M, Jacobs N, Thiery E, Derom C, Geschwind N, van den Hove D, Lataster T, Myin-Germeys I, van Os J, Rutten BP, Kenis G. Epigenetic genes and emotional reactivity to daily life events: a multi-step gene-environment interaction study. PloS one. 2014;9:e100935. doi: 10.1371/journal.pone.0100935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, Hauser R, Prins GS, Soto AM, Zoeller RT, Gore AC. Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocrine reviews. 2009;30:293–342. doi: 10.1210/er.2009-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. European journal of human genetics : EJHG. 2002;10:682–688. doi: 10.1038/sj.ejhg.5200859. [DOI] [PubMed] [Google Scholar]
- 104.Vadlamudi S, Kalhan SC, Patel MS. Persistence of metabolic consequences in the progeny of rats fed a HC formula in their early postnatal life. The American journal of physiology. 1995;269:E731–738. doi: 10.1152/ajpendo.1995.269.4.E731. [DOI] [PubMed] [Google Scholar]
- 105.McKay JA, Williams EA, Mathers JC. Folate and DNA methylation during in utero development and aging. Biochemical Society transactions. 2004;32:1006–1007. doi: 10.1042/BST0321006. [DOI] [PubMed] [Google Scholar]
- 106.Ronn T, Volkov P, Davegardh C, Dayeh T, Hall E, Olsson AH, Nilsson E, Tornberg A, Dekker Nitert M, Eriksson KF, Jones HA, Groop L, Ling C. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS genetics. 2013;9:e1003572. doi: 10.1371/journal.pgen.1003572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Horsburgh S, Robson-Ansley P, Adams R, Smith C. Exercise and inflammation-related epigenetic modifications: focus on DNA methylation. Exercise immunology review. 2015;21:26–41. [PubMed] [Google Scholar]
- 108.Sun C, Burgner DP, Ponsonby AL, Saffery R, Huang RC, Vuillermin PJ, Cheung M, Craig JM. Effects of early-life environment and epigenetics on cardiovascular disease risk in children: highlighting the role of twin studies. Pediatric research. 2013;73:523–530. doi: 10.1038/pr.2013.6. [DOI] [PubMed] [Google Scholar]
- 109.Nathan C. Points of control in inflammation. Nature. 2002;420:846–852. doi: 10.1038/nature01320. [DOI] [PubMed] [Google Scholar]
- 110.Dunning T. Periodontal disease–the overlooked diabetes complication. Nephrology nursing journal : journal of the American Nephrology Nurses’ Association. 2009;36:489–495. quiz 496. [PubMed] [Google Scholar]
- 111.Foster SL, Medzhitov R. Gene-specific control of the TLR-induced inflammatory response. Clinical immunology (Orlando, Fla) 2009;130:7–15. doi: 10.1016/j.clim.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Xing S, Nie F, Xu Q, Deng Y, Li W, Yang Z, Zhao X, Zhu P, Wang X, Gao Y, He Z. HDAC is essential for epigenetic regulation of Thy-1 gene expression during LPS/TLR4-mediated proliferation of lung fibroblasts. Laboratory investigation; a journal of technical methods and pathology. 2015;95:1105–1116. doi: 10.1038/labinvest.2015.97. [DOI] [PubMed] [Google Scholar]
- 113.Shuto T, Furuta T, Oba M, Xu H, Li JD, Cheung J, Gruenert DC, Uehara A, Suico MA, Okiyoneda T, Kai H. Promoter hypomethylation of Toll-like receptor-2 gene is associated with increased proinflammatory response toward bacterial peptidoglycan in cystic fibrosis bronchial epithelial cells. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2006;20:782–784. doi: 10.1096/fj.05-4934fje. [DOI] [PubMed] [Google Scholar]
- 114.Takahashi K, Sugi Y, Hosono A, Kaminogawa S. Epigenetic regulation of TLR4 gene expression in intestinal epithelial cells for the maintenance of intestinal homeostasis. Journal of immunology (Baltimore, Md.: 1950) 2009;183:6522–6529. doi: 10.4049/jimmunol.0901271. [DOI] [PubMed] [Google Scholar]
- 115.Sullivan KE, Reddy AB, Dietzmann K, Suriano AR, Kocieda VP, Stewart M, Bhatia M. Epigenetic regulation of tumor necrosis factor alpha. Molecular and cellular biology. 2007;27:5147–5160. doi: 10.1128/MCB.02429-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Bierne H, Hamon M, Cossart P. Epigenetics and Bacterial Infections. Cold Spring Harbor perspectives in medicine. 2012;2:a010272. doi: 10.1101/cshperspect.a010272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Oskvig DB, Elkahloun AG, Johnson KR, Phillips TM, Herkenham M. Maternal immune activation by LPS selectively alters specific gene expression profiles of interneuron migration and oxidative stress in the fetus without triggering a fetal immune response. Brain, behavior, and immunity. 2012;26:623–634. doi: 10.1016/j.bbi.2012.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Niwa T, Tsukamoto T, Toyoda T, Mori A, Tanaka H, Maekita T, Ichinose M, Tatematsu M, Ushijima T. Inflammatory processes triggered by Helicobacter pylori infection cause aberrant DNA methylation in gastric epithelial cells. Cancer research. 2010;70:1430–1440. doi: 10.1158/0008-5472.CAN-09-2755. [DOI] [PubMed] [Google Scholar]
- 119.Chang S, Aune TM. Dynamic changes in histone-methylation ‘marks’ across the locus encoding interferon-gamma during the differentiation of T helper type 2 cells. Nature immunology. 2007;8:723–731. doi: 10.1038/ni1473. [DOI] [PubMed] [Google Scholar]
- 120.Wei G, Wei L, Zhu J, Zang C, Hu-Li J, Yao Z, Cui K, Kanno Y, Roh TY, Watford WT, Schones DE, Peng W, Sun HW, Paul WE, O’Shea JJ, Zhao K. Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity. 2009;30:155–167. doi: 10.1016/j.immuni.2008.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Madrigal-Matute J, Rotllan N, Aranda JF, Fernandez-Hernando C. MicroRNAs and atherosclerosis. Current atherosclerosis reports. 2013;15:322. doi: 10.1007/s11883-013-0322-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.De Santa F, Totaro MG, Prosperini E, Notarbartolo S, Testa G, Natoli G. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell. 2007;130:1083–1094. doi: 10.1016/j.cell.2007.08.019. [DOI] [PubMed] [Google Scholar]
- 123.Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, Carson WFt, Cavassani KA, Li X, Lukacs NW, Hogaboam CM, Dou Y, Kunkel SL. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009;114:3244–3254. doi: 10.1182/blood-2009-04-217620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Adcock IM, Lee KY. Abnormal histone acetylase and deacetylase expression and function in lung inflammation. Inflammation research : official journal of the European Histamine Research Society… [et al.] 2006;55:311–321. doi: 10.1007/s00011-006-0081-1. [DOI] [PubMed] [Google Scholar]
- 125.Villagra A, Sotomayor EM, Seto E. Histone deacetylases and the immunological network: implications in cancer and inflammation. Oncogene. 2010;29:157–173. doi: 10.1038/onc.2009.334. [DOI] [PubMed] [Google Scholar]
- 126.Ghosh S, Karin M. Missing pieces in the NF-kappaB puzzle. Cell. 2002;109(Suppl):S81–96. doi: 10.1016/s0092-8674(02)00703-1. [DOI] [PubMed] [Google Scholar]
- 127.Yang J, Park Y, Zhang H, Xu X, Laine GA, Dellsperger KC, Zhang C. Feed-forward signaling of TNF-alpha and NF-kappaB via IKK-beta pathway contributes to insulin resistance and coronary arteriolar dysfunction in type 2 diabetic mice. American journal of physiology Heart and circulatory physiology. 2009;296:H1850–1858. doi: 10.1152/ajpheart.01199.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Yamamoto Y, Verma UN, Prajapati S, Kwak YT, Gaynor RB. Histone H3 phosphorylation by IKK-alpha is critical for cytokine-induced gene expression. Nature. 2003;423:655–659. doi: 10.1038/nature01576. [DOI] [PubMed] [Google Scholar]
- 129.Saccani S, Pantano S, Natoli G. p38-Dependent marking of inflammatory genes for increased NF-kappa B recruitment. Nature immunology. 2002;3:69–75. doi: 10.1038/ni748. [DOI] [PubMed] [Google Scholar]
- 130.Mabaera R, Greene MR, Richardson CA, Conine SJ, Kozul CD, Lowrey CH. Neither DNA hypomethylation nor changes in the kinetics of erythroid differentiation explain 5-azacytidine’s ability to induce human fetal hemoglobin. Blood. 2008;111:411–420. doi: 10.1182/blood-2007-06-093948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yoon S, Eom GH. HDAC and HDAC Inhibitor: From Cancer to Cardiovascular Diseases. Chonnam medical journal. 2016;52:1–11. doi: 10.4068/cmj.2016.52.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Heerboth S, Lapinska K, Snyder N, Leary M, Rollinson S, Sarkar S. Use of epigenetic drugs in disease: an overview. Genetics & epigenetics. 2014;6:9–19. doi: 10.4137/GEG.S12270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Omer D, Harari-Steinberg O, Buzhor E, Metsuyanim S, Pleniceanu O, Zundelevich A, Gal-Yam EN, Dekel B. Chromatin-modifying agents reactivate embryonic renal stem/progenitor genes in human adult kidney epithelial cells but abrogate dedifferentiation and stemness. Cellular reprogramming. 2013;15:281–292. doi: 10.1089/cell.2012.0087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Rajasingh J. Reprogramming of somatic cells. Progress in molecular biology and translational science. 2012;111:51–82. doi: 10.1016/B978-0-12-398459-3.00003-4. [DOI] [PubMed] [Google Scholar]
- 135.Thangavel J, Malik AB, Elias HK, Rajasingh S, Simpson AD, Sundivakkam PK, Vogel SM, Xuan YT, Dawn B, Rajasingh J. Combinatorial therapy with acetylation and methylation modifiers attenuates lung vascular hyperpermeability in endotoxemia-induced mouse inflammatory lung injury. The American journal of pathology. 2014;184:2237–2249. doi: 10.1016/j.ajpath.2014.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Thangavel J, Samanta S, Rajasingh S, Barani B, Xuan YT, Dawn B, Rajasingh J. Epigenetic modifiers reduce inflammation and modulate macrophage phenotype during endotoxemia-induced acute lung injury. Journal of cell science. 2015;128:3094–3105. doi: 10.1242/jcs.170258. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
