Skip to main content
American Journal of Physiology - Heart and Circulatory Physiology logoLink to American Journal of Physiology - Heart and Circulatory Physiology
. 2020 Aug 21;319(4):H847–H865. doi: 10.1152/ajpheart.00382.2020

Transcriptional regulation by methyltransferases and their role in the heart: highlighting novel emerging functionality

Marta W Szulik 1,, Kathryn Davis 1, Anna Bakhtina 1, Presley Azarcon 1, Ryan Bia 1, Emilee Horiuchi 1, Sarah Franklin 1,2
PMCID: PMC7654657  PMID: 32822544

Abstract

Methyltransferases are a superfamily of enzymes that transfer methyl groups to proteins, nucleic acids, and small molecules. Traditionally, these enzymes have been shown to carry out a specific modification (mono-, di-, or trimethylation) on a single, or limited number of, amino acid(s). The largest subgroup of this family, protein methyltransferases, target arginine and lysine side chains of histone molecules to regulate gene expression. Although there is a large number of functional studies that have been performed on individual methyltransferases describing their methylation targets and effects on biological processes, no analyses exist describing the spatial distribution across tissues or their differential expression in the diseased heart. For this review, we performed tissue profiling in protein databases of 199 confirmed or putative methyltransferases to demonstrate the unique tissue-specific expression of these individual proteins. In addition, we examined transcript data sets from human heart failure patients and murine models of heart disease to identify 40 methyltransferases in humans and 15 in mice, which are differentially regulated in the heart, although many have never been functionally interrogated. Lastly, we focused our analysis on the largest subgroup, that of protein methyltransferases, and present a newly emerging phenomenon in which 16 of these enzymes have been shown to play dual roles in regulating transcription by maintaining the ability to both activate and repress transcription through methyltransferase-dependent or -independent mechanisms. Overall, this review highlights a novel paradigm shift in our understanding of the function of histone methyltransferases and correlates their expression in heart disease.

Keywords: epigenetics, heart disease, histone methyltransferase, posttranslational modification, transcriptional regulation

INTRODUCTION

Methyltransferases (MTs) are a large group of enzymes that catalyze the transfer of a methyl group from a donor molecule, most commonly S-adenosylmethionine (AdoMet), to a specific methyl acceptor. These enzymes (>90%) can be grouped into three major classes based on their structural features, which are also reflected by unique amino acid sequences. These classes have enabled the identification of previously unrecognized enzymes: class I enzymes contain a seven-strand twisted β-sheet structure (25), class II contain a SET-catalytic domain (199), and class III is exemplified by membrane-associated enzymes with multiple membrane-spanning regions (157). In addition, these MTs can also be classified on the basis of the target substrates they methylate: specifically, natural product methyltransferases, DNA/RNA methyltransferases, and protein methyltransferases. The first comprehensive compilation of the human methyltransferome was accomplished by Petrossian and Clarke (149), who employed bioinformatic approaches to interrogate the genome, ultimately categorizing 208 proteins as characterized or putative methyltransferases, including 38 previously unannotated proteins. We have updated this list of methyltransferases in this review by removing seven proteins that are no longer included in the Uniprot database (likely due to redundancy or lack of supporting data) and two additional proteins that have duplicate entries, leaving us with 199 total human methyltransferases. This included 86 protein MTs, 58 DNA/RNA MTs, 19 natural product MTs, and 36 with unknown methylation targets (Fig. 1). We have used this categorization as a baseline for this review, with one exception: in the 9 years since their analyses were published, 40 additional protein methyltransferases have been functionally characterized and, therefore, migrate from the “putative” to “characterized” category. Although there are many functional studies, which have been carried out on individual methyltransferases describing their methylation of unique targets and the subsequent effects on biological processes, no analyses exist describing the spatial distribution across tissues of this enzyme class or their differential expression in the diseased heart. For this review, we performed tissue profiling in publicly available protein databases of 199 confirmed or putative methyltransferases to demonstrate the ubiquitous or unique tissue-specific expression of these individual proteins. In addition, we examined transcript data sets from human heart failure patients and murine models of heart disease to identify methyltransferases, which have been shown to be differentially regulated in the heart. Lastly, we focused the remainder of the review on the largest functionally characterized subset: protein methyltransferases, the majority of which have reported roles in regulating gene expression. Interestingly, our analysis showed that 76 of the 86 protein methyltransferases have confirmed roles in methylating histone residues by targeting either lysine or arginine side chains, and present a newly emerging phenomenon in which 16 of these enzymes have been shown to play dual roles in regulating transcription by maintaining the ability to both activate and repress transcription.

Fig. 1.

Fig. 1.

Human methyltransferome. A: classification of all characterized (green) and putative (blue) methyltransferases in humans based on functional domains and bioinformatics predictions from a 2011 study by Petrussian and Clarke, which we have modified to reflect newly characterized protein methyltransferases (blue-green hash) over the last 9 years. B: a total of 86 protein methyltransferases have been hypothesized to exist in humans, 36 of these had been functionally validated in 2011, and an additional 40 protein methyltransferases have been functionally validated since then, bringing the total number of those characterized to 76 (88%). MT, methyltransferase.

EXPRESSION OF METHYLTRANSFERASES IN HUMAN TISSUE AND CELLS

To determine the extent of methyltransferase expression across a wide variety of mammalian tissues, we submitted our list of 199 methyltransferases [adapted from Petrossian et al. (149), as detailed above] to the Human Proteome Map database (92), which contains mass spectrometry-based (LC-MS/MS) protein expression data from 17 adult tissues, 6 primary hematopoietic cells, and 7 fetal tissues. We generated expression profiles for both characterized and putative subsets, shown in Fig. 2. While the Human Proteome Map contains direct evidence of translation for over 17,000 genes, covering >84% of the annotated protein-coding genes, it is important to recognize that because of the nature of biological mass spectrometry, low abundant proteins may escape detection in this data set. Additionally, as provided by the Human Proteome Map database, all samples used to generate these data were obtained from histologically normal samples, so the information provided in this data set is limited only to the basal human physiological state of tissue or cells. Interestingly, our analyses show a wide variety of expression patterns among methyltransferases with some being ubiquitously expressed across tissues, while others display a more restricted expression pattern found in only one or a few tissue types. Indeed, the restricted expression of most methyltransferases was somewhat surprising to us, because of their rather generic role in methylating proteins, DNA, and RNA found in virtually all cells; however, we expect that as we continue to learn about these proteins, the specific molecular mechanisms regulating their specificity in different cells will become more apparent. Of note, the following methyltransferases are absent from Fig. 2 because they are either not present in the Human Proteome Map database but are still maintained in the Uniprot database (A: BMT2, CSKMT, EEFAKMT1, EEF1AKMT2, EEFAKMT3, EEF2KMT, ETFBKMT, TEDC2; B: BUD23, CARNMT1, METTL26, METTL27, MRM3, SPOUT1, TRMT9B), or, were not identified in any of the tissue or cells listed (A: ASMT, HEMK1, KMT2B, METTL11B, METTL22, PRDM12, PRDM16, PRDM7, PRDM9, SETD4, TRDMT1; B: BCD1N3D, ERC2-IT1, FAM86B1, FAM86B2, FBLL1, METTL15P1, METTL21P1, METTL23, METTL8, FTSJ2 [MRM2], NSUN3, NSUN5D1, NSUNP2, SETD9). Specifically, in cardiac tissue, we observe expression of 88 MTs (51 characterized, 37 putative) in fetal heart and 54 in adult heart tissue (35 characterized, 19 putative). Of these MTs expressed in fetal or adult heart tissue 42 (27 characterized, 15 putative) were expressed at both developmental stages while 55 (29 characterized, 26 putative) displayed expression exclusive to one stage. Interestingly, there were several methyltransferases that were uniquely expressed in only 1 tissue/cell, including PRDM8, which is only expressed in retina, and FAM173B, which is only expressed in platelets. In this data set, only one methyltransferase, MECOM, shows exclusive expression to the heart (at any stage), although data from the literature suggest this protein is also expressed in hematopoietic cells (60, 122). While the expression of these methyltransferases in various tissues can be helpful to guide characterization of their role in regulating cellular processes in fetal or adult tissue, it is limited only to basal states, and it does not include information about expression changes during various stages of development or disease.

Fig. 2.

Fig. 2.

Expression of methyltransferases in human tissue and cells. The relative expression profiles of characterized (A) and putative (B) methyltransferases in human tissue and cells. Protein expression profiles were generated from the Human Proteome Map (92), which maintains mass spectrometry-based identification and normalized spectral counting-based quantification of proteins (listed under their corresponding gene identifier). Red indicates high expression, pink indicates low expression, white indicates no detectable expression. Green denotes fetal tissue, yellow denotes adult tissue, and blue denotes hematopoietic cells.

METHYLTRANSFERASES DIFFERENTIALLY EXPRESSED IN THE DISEASED HEART

To identify methyltransferases that are potentially involved in the pathophysiology of cardiac disease, regardless of whether their methylation target or specific role has been characterized, we analyzed publicly available data sets from five studies (three using human tissue and two using mouse tissue), which quantified changes in transcript abundance from cardiac tissue via RNA-sequencing to identify transcripts that are differentially expressed in human heart failure patients or mouse models of cardiac disease (Tables 1 and 2). This is not a comprehensive analysis of all data publicly available in humans and mice from individual studies but represents the most recent analysis of human and animal models in which large-scale global analysis was performed via RNA-sequencing, where the entire data set was available for us to examine. Information regarding the number of samples analyzed, sex, and region of the heart used for analysis from these studies can be found in Table 3. Each study applied different filters for determining significance within their data set, which are briefly described below. Ultimately, we included transcripts in this review that were found to be significant on the basis of the criteria of the individual publication (all of which included a P value ≤ 0.05). It is important to note that these data sets are a comparison of diseased to normal tissue where transcripts found to be differentially regulated during disease were included (those which were detected but showed no change in abundance were excluded in the original publications). Currently, there are no large-scale analyses performed at the protein level to quantify changes in methyltransferases in these same disease settings; therefore, protein data sets are not included in this analyses, but analysis of these data sets would be helpful to carry out in future investigations.

Table 1.

Transcripts of characterized or putative methyltransferases differentially expressed in humans with ischemic cardiomyopathy, nonischemic cardiomyopathy, or dilated cardiomyopathy

Yang et al. (197)
Sweet et al. (172)
Liu et al. (118)
Gene ID
(Uniprot)
ICM NICM DCM ICM DCM ICM
SMYD1 −1.7 −1.8 −1.6 −1.41
SMYD2 1.3
SMYD5 1.2
PRMT2 1.6 1.4
EZH1 1.3
SETD7 1.2 2.8 2.2
EHMT2 1.6 1.3
PRMT7 1.3 1.3
DOT1L −3.0 −3.8
SETD1B 2.7 −2.3
HNMT 1.9 1.8
RNMT 1.3 1.4 1.5 1.6
COMT 1.5 1.4
COQ3 −1.3 −1.2 −1.3
PNMT −2.3 −4.8
LCMT1 1.2
ICMT −1.3 −1.3
INMT 2.0 2.2 2.3
SETD3 −1.4 −1.5
AS3MT −13.3
GNMT −17.0 −26.8
NNMT −3.8
ASH1L 3.0
METTL21A −1.6 −1.7
PCMTD1 −1.4 −1.7 2.7
FAM173B 1.3
METTL5 1.3 1.4
NSUN6P2 1.6 1.8
FTSJ3 1.2 1.2 1.2
TRMT61B 1.7 1.5 1.5
ASMTL 1.4 1.3
TRMT1L 1.6 2.3
RRNAD1 1.3 1.6
FAM173A 1.5 2.1
EMG1 1.2 1.4
COMTD1 1.7 1.7
METTL9 1.2
ELP3 1.2
METTL23 1.4
METTL7B 2.6 2.5

Values indicate fold change for transcripts with a P ≤ 0.05. Negative value corresponds to downregulated transcripts and positive value to upregulated transcripts. Values without boldface indicate characterized methyltransferase transcripts, whereas boldface values indicate putative methyltransferase transcripts. DCM, dilated cardiomyopathy; ICM, ischemic cardiomyopathy; NICM, nonischemic cardiomyopathy.

Table 2.

Transcripts of characterized or putative methyltransferases differentially expressed in mouse models of cardiac disease

Lee et al. (105) Wang et al. (189)
Gene ID
(Uniprot)
TAC ISO
DOT1L −1.2
SETMAR −1.2
SETD6 −1.5
PRMT2 1.7
HNMT 1.7
COQ3 −1.5
INMT −4.4
AS3MT −1.7
NNMT 2.7 1.3
METTL1 −1.2
COMTD1 1.5
NSUN5 1.2
RSAD1 1.2
NRM 1.6
PCMTD2 1.5

Values indicate fold change for transcripts with a P ≤ 0.05. Negative value corresponds to downregulated transcripts and positive value to upregulated transcripts. Values without boldface denote transcripts of characterized methyltransferases, where boldface values denote transcripts of putative methyltransferases. ISO, isoproterenol infusion via mini-osmotic pump; TAC, transverse aortic constriction.

Table 3.

List of five transcriptome studies analyzed in this work with additional information on the number of samples analyzed, species, sex, and indication of what region of the heart was used for analysis

Publication n Value Sex Species Region of Heart Analyzed Notes
Yang et al. (197) Ischemic: n = 8
Nonischemic: n = 8
Nonfailing: n = 8
Ischemic: 8 M
Nonischemic: 3 F, 5 M
Nonfailing: 1 F, 7 M
Human left ventricle
Sweet et al. (172) Dilated: n = 37
Ischemic: n = 13
Nonfailing: n = 14
Dilated: 7 F, 30 M
Ischemic: 3 F, 10 M
Nonfailing: 3 F, 9 M
Human left ventricle Ischemic cohort had significantly greater proportion of patients taking statins, having coronary artery disease, hyperlipidemia, and diabetes mellitus.
Liu et al. (118) Dilated: n = 2
Ischemic: n = 1
Nonfailing: n = 3
n/a Human left ventricle
Lee et al. (105) 1 wk TAC: n = 6
1 wk Sham: n = 6
8 wk TAC: n = 4
8 wk Sham: n = 4
M Mouse left ventricle
Wang et al. (189) 104 ISO-treated strains, ~4 mice from each strain F Mouse left ventricle Fully controlled the age, environment, severity, and timing of cardiac injury

F, female; ISO, isoproterenol infusion via mini-osmotic pump; M, male; n/a, not applicable; TAC, transverse aortic constriction.

The first human study that we analyzed by Yang et al. (197) quantified transcripts in cardiac tissue from patients with either ischemic or nonischemic cardiomyopathy undergoing LVAD implantation and compared these to healthy donor tissue. In this study, 21 methyltransferases (14 characterized and 7 putative) were differentially expressed and considered significant on the basis of a fold change greater than 1.2 and P value < 0.5. The second study, by Sweet et al. (172) analyzed 64 human left ventricle tissue samples obtained at the time of heart transplantation that were categorized as dilated cardiomyopathy (37), ischemic (13), and nonfailing (14) hearts (172). Among the 2,993 differentially expressed transcripts, 18 were methyltransferases (9 characterized, 9 putative), which displayed significant changes in expression. The third human study from Liu et al. (118) reported gene expression signatures in heart tissue obtained from one patient with ischemia, two patients with diagnosed dilated cardiomyopathy, and three healthy controls. In their analysis of around 2,000 differentially regulated transcripts for both disease conditions, 10 were methyltransferases (all characterized).

In mouse models of cardiac disease, the first report that we analyzed by Lee et al. (105) examined transcripts from cardiac tissue in mice after transverse aortic constriction (TAC)-induced heart failure. Their data set identified a total of 1,478 differentially expressed transcripts in the diseased heart, 10 of which were methyltransferases (seven characterized and three putative). The second study by Wang et al. (190) used mini-osmotic pumps to infuse isoproterenol (ISO) in 104 inbred mouse strains over 21 days, to induce cardiac dysfunction and then quantified changes in transcripts. Their analysis identified 1,502 differentially expressed transcripts across all strains examined and included 6 methyltransferases (4 characterized, 2 putative).

Together, these five studies identified 40 methyltransferases (26 characterized, 14 putative) that are differentially expressed in human heart failure patients and 15 mouse-models of cardiac disease (10 characterized, 5 putative). Of the transcripts found to be differentially expressed, only eight of them were identified in both human patients and mouse models, five that display the same directional change in expression (DOT1L, PRMT2, HNMT, COQ3, and AS3MT) and three that display opposite changes in expression in different studies (NNMT, COMTD1, INMT). Of the methyltransferases that were identified collectively in these five studies, 12 of them have previously been examined in the heart and were shown to play a specific role in cardiac physiology and pathology [SMYD1 (181), SMYD2 (181), SMYD3 (181), EZH1 (5), SETD7 (49), EHMT2 (146) DOT1L (34, 138, 139), AS3MT (30, 48, 70), NNMT (116), COMT (74), PNMT (179), and LCMT1 (120)]. However, a majority of them (35) have never been examined within the heart (PRMT2, PRMT7, SETD1B, HNMT, RNMT, COQ3, ICMT, INMT, SETD3, GNMT, ASH1L, METTL21A, PCMTD1, FAM173B, METTL5, NSUN6P2, FTSJ3, TRMT61B, ASMTL, TRMT1L, RRNAD1, FAM173A, EMG1, COMTD1, METTL9, ELP3, METTL23, METTL7B, SETMAR, SETD6, METTL1, NSUN5, RSAD1, NRM, and PCMTD2). Therefore, future studies will be needed to determine their function in the cardiovascular system and to fully understand the variability in abundance of these enzymes across different conditions and species examined in Tables 1 and 2.

HISTONE METHYLTRANSFERASES IN TRANSCRIPTIONAL REGULATION

We next became interested in examining the largest characterized subset of methyltransferases, that of protein methyltransferases, 88% (76 of 86) of which have established roles in regulating gene expression through their ability to methylate lysine or arginine side chains on histone proteins. This subset is, therefore, the focus of the remainder of this review.

Historically, methylation of lysine residues was first reported in 1959 by Ambler and Rees (8), who described differences in the amino acid composition of flagellar proteins in Proteus vulgaris and Salmonella typhimurium, and showed the latter contained a new amino acid, ε-N-methyl lysine. Several years later, this modification (ε-N-methyl lysine) was identified on histone proteins (135) and was characterized as a posttranslational modification (occurring subsequent to peptide bond formation) (93). Subsequently, two studies initiated an interest in histone methylation and gene regulation, as it is studied today. First, Hwang and Bonner (82) reported that adding histones to DNA-dependent RNA polymerase attenuates RNA synthesis, and second, Allfrey et al. (7) discovered that histone residues can be acetylated and methylated, influencing RNA synthesis. The discovery of the first methyltransferases (the enzymes responsible for adding this modification) came soon after, for both arginine (142) and lysine (143) methyltransferase classes. However, with these discoveries in hand, it took more than two decades to decode the applicability of these methylation reactions and to provide evidence that the methylation of histones, indeed, plays a significant role in the regulation of gene expression (144). In addition, the discovery of the human SUV39H1 histone lysine methyltransferase spurred a new era of methylation research and influenced the growth of epigenetics by providing the very first link between histone methylation and the formation of higher-order chromatin structure, such as heterochromatin formation (155). It is now appreciated that both arginine and lysine residues on histones are substrates for methylation and that their side chain length (the longest side chains of all naturally occurring amino acids) and positive charge make them an attractive target for protein-protein interactions (15). Methylation of these residues enables them to contribute to changes in higher-order chromatin structure by modifying the charge state of histones and/or their interaction with DNA by influencing interactions with other hydrogen bond donors, which, in turn, regulates accessibility for transcription factors and other transcriptional machinery (72, 125). Moreover, histone methylation can influence the recruitment of proteins that specifically recognize, or “read”, these posttranslational modifications and their associated binding partners or inhibit binding of proteins to chromatin. In addition to being methylated or unmethylated, an additional level of complexity arises from the ability of these amino acids to be monomethylated, demethylated, or trimethylated on lysine or monomethylated or dimethylated (symmetrically or asymmetrically) on arginine; however, how this specificity is acquired to enable an enzyme to selectively participate in monomethylation, demethylation, or trimethylation is unknown, but it is an important area of research that will be necessary for us to fully understand the enzymatic function of these proteins. For many years, histone methylation, as opposed to acetylation or phosphorylation, was thought to be irreversible and biochemically stable. However, it was not until the discovery of the first histone demethylase in 2004 that this notion was challenged, and ever since then, we have continued to learn more about how this dynamic process of histone methylation influences a wide array of biological processes (75, 159).

Lysine Methylation

Methylation of lysine residues is regulated by lysine-specific methyltransferases (KMTs) primarily in an AdoMet-dependent manner. Except DOT1L, all lysine-specific histone methyltransferases share a SET [Su(var), Enhancer of Zeste, Trithorax] domain that is responsible for catalysis of AdoMet onto the target lysine, resulting in monomethylation demethylation, or trimethylation (abbreviated as me1, me2, and me3, respectively) (144). The established paradigm has been that histone lysine methylation is associated with either activation or repression of gene expression, depending on the location and degree of methylation. For example, all three methylation states of histone H3K4 mark active genes, while trimethylation of histone H4K20, in general, designates silenced genes. However, for some residues, the resulting biological consequence is not always straightforward; for instance, monomethylation of some residues leads to transcriptional activation (e.g., histone H3K9me1), while trimethylation at the same site leads to gene silencing (e.g., histone H3K9me3) (13). What dictates whether a methyltransferase will carry out monomethylation, dimethylation, or trimethylation of a specific residue is unknown, but it has been hypothesized to be influenced by protein binding partners or other posttranslational modifications. For example, DOT1L is known to regulate monomethylation, demethylation, and trimethylation of H3K79, which have opposing transcriptional functions (activation vs. repression), and while it is not known what influences DOT1L to carry out monomethylation versus trimethylation, it is known that this occurs in a nonprocessive manner (requiring the enzyme to dissociate from the substrate between each addition of a methyl group) (193). Even though the addition of a methyl group does not change the net charge of the protein, as it does with lysine acetylation, it does influence hydrophobicity, which, in turn, affects protein-protein interactions, including that of transcriptional repressors and activators. For instance, trimethylation of histone H3K4 recruits the chromatin remodeling proteins CHD1 and BPTF (63, 110), which opens the chromatin structure and prevents the binding of the repressive NuRD and INHAT complexes (140, 163). The most well-known sites of histone methylation occur on histone H3 (on lysine residues 4, 9, 14, 27, 36, and 79) and H4 (on lysine residues 20 and 59). However, there are also many other lysine residues on histone H1, H2A, and H2B that have been identified (79).

Apart from histones, lysine methylation also occurs on a wide variety of proteins involved in regulating transcription and chromatin structure, as well as other biological processes. Specifically, the methylation of nonhistone proteins is known to regulate protein-protein interactions, subcellular localization (e.g., nuclear localization of Hsp70), the promoter binding affinity of transcription factors at target genes (e.g., NF-κB activation by lysine methylation), various signaling pathways (NF-κB, MAPK, Wnt, ER, and p53) and DNA damage and stress response, among others, to influence the development and differentiation in various cell types (131). We will not discuss in detail the methylation of nonhistone proteins in this review, except when it is applicable to the regulation of gene expression by the methyltransferases described below.

Arginine Methylation

Arginine methylation is becoming increasingly more important in our understanding of the dynamic regulation of gene expression. Since the discovery of the first arginine methyltransferase decades ago, this family has expanded to include 11 members with known roles in various biological processes, including signaling, RNA splicing, DNA damage repair, and transcriptional regulation (90). Indeed, it has been estimated that roughly 0.5% of all arginine residues in mammalian cells are methylated, although a large majority of these are thought to occur on nonhistone proteins, in addition to the significant number of sites on histones, where arginine residues are known to be methylated (69).

Arginine methylation is carried out by protein arginine methyltransferases (PRMTs) that consist of two conserved domains (AdoMet binding- and COOH-terminal domains), which modify arginine residues in an AdoMet-depending manner. This results in three types of modifications (distinct from those generated by lysine methyltransferases). Specifically, monomethylation occurs on the terminal N atom that can only be carried out by PRMT7. The remaining family members can catalyze subsequent methylation reactions on either the same N atom to generate symmetric (s) dimethylation (via PRMT1, PRMT2, PRMT3, PRMT4 [also known as CARM1], PRMT6 and PRMT8) or on the adjacent N atom to generate asymmetric (a) dimethylation (via PRMT5, PRMT7, and PRMT9) (125). Ultimately, these methylation marks either activate or repress transcription directly or indirectly by recruiting other chromatin binding proteins or transcriptional regulators. In general, methylation of histone H3 and H4 on H4R3me1, H4R3me2a, H3R2me2s, H3R17me2a, or H3R26me2a results in transcriptional activation, while methylation on H3R2me2a, H3R8me2a, H3R8me2s, H3R38me2a, or H4R3me2s leads to transcriptional repression. For instance, PRMT1 has been shown to methylate histone H4R3me2a, which results in recruitment of the CBP/p300 complex, which, in turn, enhances acetylation of histone H3K9 and H3K14 and promotes binding of transcriptional machinery (14). On the other hand, histone H4R3me2s methylation by PRMT5 recruits DNMT3a to chromatin, which results in DNA methylation and gene silencing (211). Additional examples of arginine methylation regulating gene expression include the recruitment of PRMTs to the promoters of various transcription factors, including p53 (9), YY1 (156), and NF-κB (47), and their methylation of transcriptional coactivators (including CBP/p300 (43), RUNX1 (212), CERC (56), HIV Tat (27)), transcription elongation factors [SPT5 (100)], and chromatin structural proteins [HMGA (166)]. Similar to lysine methyltransferases, arginine methyltransferases have also been shown to methylate a wide variety of proteins, which are not involved in transcriptional regulation. Most prominently, they are known to methylate RNA binding proteins to regulate processing, folding, stabilization, localization, and translation of RNAs (40) and to regulate DNA repair (54), signal transduction (16, 102), or nuclear/cytoplasmic shuttling (168), although this review will focus only on their ability to regulate transcription.

The role of protein arginine methyltransferases in stem cell biology (23, 24, 208), development (76), and adult homeostasis (15, 98) is unquestionable; however, their role in human disease has only more recently emerged. Implications of arginine methylation to disease is most apparent in cancer, where it has been shown that PRMTs are usually upregulated, making them an attractive target for the development of small-molecule inhibitors (80). In addition, evidence now exists, suggesting a role in other human conditions, including neurodegenerative diseases, metabolic diseases, and aging (23, 73).

EMERGING MULTIFUNCTIONALITY OF METHYLTRANSFERASES IN TRANSCRIPTIONAL REGULATION

The classical dogma regarding histone methylation and gene expression suggests that each methyltransferase modifies a single and specific (nonrandom) amino acid residue and, thereby, influences gene expression in one direction, by either activating or repressing transcription (Fig. 3A). In this way, these enzymes were thought to maintain functional specificity for regulating transcription within the genome. However, as our understanding of methyltransferases has expanded over the years, we have come to recognize that the function of so-called “histone” methyltransferases is much broader than this. Indeed, evidence now exists showing that the same methyltransferase can modify different amino acids on histones to both activate and repress transcription (based on the residue modified), even in the same cell type (Fig. 3B). In addition, these enzymes have also been shown to regulate transcription by methylating additional, nonhistone proteins, including transcription factors, transcriptional machinery, and chromatin remodeling proteins (which regulate chromatin structure and accessibility, Fig. 3C). Lastly, in addition to these methyltransferase-dependent mechanisms (Me), many of these enzymes have also been shown to regulate transcription in a methyltransferase-independent (Me-I) manner, in which they interact with other proteins, but do not methylate them (Fig. 3D) or through a separate and currently unknown mechanism (Uk). Therefore, to systematically review the ability of methyltransferases to function through these mechanisms, we compiled data for all 76 known histone methyltransferases and specifically documented the manner by which they regulate gene expression. Our analysis identified 16 methyltransferases that maintain the ability to both activate and repress transcription by participating in at least two of the four modes of regulation mentioned above and shown in Fig. 3. These 16 enzymes (SMYD1, SMYD2, SMYD3, EHMT1/EHMT2 (G9a/GLP), EZH2, KMT2D, DOT1L, CARM1, PRMT5, PRMT6, ASH1L, SETDB2, SETD3, SETD6, and SET8) and the specific mechanisms by which they activate and repress transcription are listed in Table 4. It is important to note that this table is not comprehensive in listing all known methylation targets for these enzymes, but instead focuses on those by which they have been shown to regulate gene expression. In addition, of the 16 methyltransferases, 10 have been shown to play significant roles in cardiac physiology and pathology, as described in Table 5. An additional 6 enzymes (PRMT6, ASH1L, SETDB2, SETD3, SETD6 and SET8) have never been examined in the heart; thus, detailed information about their transcriptional regulation is not included below; however, information about their histone methylation targets and unique nonhistone targets and mode of regulation is included in Table 4. Interestingly, two of these methyltransferases (ASH1L and SETD3) with unknown roles in the cardiovascular system are expressed in both the developing and adult heart (Fig. 2); therefore, further studies would be helpful to elucidate their roles in the heart physiology and pathology.

Fig. 3.

Fig. 3.

Schematic diagram depicting mechanisms by which methyltransferases regulate gene expression. A: classically, histone methyltransferases (MT; yellow) were thought to only modify a single and specific amino acid (pink circle) and, thereby, maintain specificity for regulating gene expression in one direction within the genome. B: however, more recently, studies have shown that the same methyltransferase can modify multiple amino acids (purple circle) on histones to both activate and repress transcription, even within the same cell. In addition, the same methyltransferase can also regulate gene expression by methylating other chromatin binding proteins (C) or through methyltransferase-independent mechanisms (D) (typically as a component of a chromatin binding complex). MT, methyltransferase.

Table 4.

Methyltransferases capable of both activating and repressing transcription through methyltransferase-dependent (methylating histones or other proteins), methyltransferase-independent (binding but not methylating proteins), and/or uncharacterized mechanisms

Gene ID
(Uniprot)
Histone Residue Methylated Activation/Repression Mechanism of Regulation: Methylation (Me), Methylation-Independent (Me-I), Unknown (Uk) Activation/Repression
SMYD1 H3K4me3 (19, 175, 192) Activation (19, 175) Nppa (Uk) (65), Tgfβ3 (Uk) (65) Repression (65)
skNAC (Me) (19, 147, 213), TRB3 (Me) (154) Activation (19, 154)
SMYD2 H3K4me3 (3) Activation (3) RB (Me) (160), p53 (Me) (81) Repression (81, 160)
H3K36me2 (31) Repression (31) PARP1 (Me) (150), HSP90 (Me) (52) Activation (52, 150)
SMYD3 H3K4me2/3 (75) Activation (75, 183) MAP3K2 (Me) (124) Activation (124)
H4K20me3 (64, 186) Repression (64, 186)
EHMT1/EHMT2
(G9a/GLP)
H3K9me1/2 (113, 174) Both (108, 113, 174) G9a-p53 (Me-I) (152), G9a-CARM1-p300 (Me-I) (22) Activation (22, 152)
H3K27me1/2 (173) Both (173) Jarid1a (Me) (36) Repression (36)
EZH2 H3K27me1 (61) Activation (61) GATA4 (Me) (78) Repression (78)
H3K27me2/3 (61, 201, 210) Repression (61, 201, 210) Jarid2 (Me) (161), RelA/RelB (Me-I) (106), SWI/SNF (Me-I) (91) Activation (91, 106, 161)
KMT2D H3K4me1 (41, 66, 88, 104) Both (41, 66, 88, 104) CBP/p300 (Uk) (101), ERα (Me-I) Activation (101)
H3K4me2/3 (66, 104) Activation (66)
DOT1L H3K79me1/2 (193) Activation (193) ENL/MLLT1, AF4/MLLT2, AF9/MLLT3 and AF10/MLLT10 (Me-I) (21, 130, 133), RNAPII (Me-I) (94), c-Myc (Me-I) (44) Activation (21, 44, 94, 148)
H3K79me3 (193) Repression (193) Sir3 (Me-I) (62) Repression (62)
CARM1 H3R17me2a (69, 164, 194)
H3R26me2a (69, 164)
H3R42me2a (33, 69)
Activation (33, 69, 164, 194) Sox2 (Me) (209), CBP/p300 (Me) (136)
SRC-3 (Me) (59), GRIP1/p300 (Me) (107)
Activation (136, 209)
Repression (59, 107)
PRMT5 H3R2me2s (127) Activation (10, 15, 98, 127) hnRNP A1 (Me) (68) Activation (68)
H2AR3me2s (10, 15, 98, 176)
H3R8me2s (145, 190, 204)
H4R3me2s (190, 204)
Repression (10, 15, 98, 176)
Both (145, 190, 204)
PRMT6 H2AR11me2a (188)
H2AR29me2a (188)
Repression (188) HMGA1a (Me) (119, 128), DNA Pol b (Me) (54) Activation (54, 119, 128)
H3R2me2a (26, 126, 141) Both (26, 126, 141)
H3R42me2a (33) Activation (33)
ASH1L H3K4me3 (200)
H3K36me2 (202)
Activation (200)
Repression (202)
MMP (Me) (17) Repression (17)
SETDB2 H3K9me3 (96, 99, 162, 180) Repression (96, 99, 162, 180) Insig2 (Me-I) (158) Activation (158)
SETD3 H3K4me1/2 (55) H3K36m1/2 (55) Activation (55) Myod1 (Me) (55), p53 (Me) (1)
FoxM1 (Me) (46)
Activation (1, 55)
Repression (46)
SETD6 H2AZK7me1 (20) Repression (20) RelA (Me) (134), PLK-1 (Me) (58), DJ1 (Me-I) (37),
PAK4 (Me) (185)
Repression (37, 58, 134)
Activation (185)
SET8 H4K20me1 (87, 198) Both (87, 198) TWIST (Me) (196)
p53 (Me) (169)
Both (196)
Repression (169)

Currently, 16 of the 86 total protein methyltransferases have been shown to be capable of this dual functionality. Mechanism indicated in parenthesis: Me, methylation-dependent; Me-I, methylation-independent; Uk, unknown.

Table 5.

Methyltransferases with expanded functionality and their roles in the fetal and/or adult heart, as well as reported cardiac phenotype in zebrafish, mouse, or human

Methyltransferase Role in Fetal Heart Role in Adult Heart Cardiac Phenotype of KD/KO
SMYD1 Morphogenesis (71, 154); Myofibrillogenesis (154, 175); Cell proliferation (154) Inhibit hypertrophic growth (65) Zebrafish: Impaired myofibril formation (175), no heartbeat (86).
Mouse: Disrupted right ventricle formation and cardiomyocyte maturation (71, 154)
Human: Hypertrophic cardiomyopathy (57)
SMYD2 Dispensable (51); Myofibrillogenesis (187) No phenotype (51) Zebrafish: Impaired myofibril formation (187)
Mouse: Normal heart development (51)
SMYD3 Morphogenesis, maturation/proliferation of differentiated myocytes (67) Unknown Zebrafish: Abnormal looping of heart tube, pericardial edema (67)
EHMT1/EHMT2 (G9a/GLP) Morphogenesis (83) Inhibit hypertrophic growth, regulates cardiac homeostasis (146) Mouse: Neonatal lethality and severe cardiac defects (atrioventricular septal defects) (83, 146)
Human: Kleefstra syndrome (97)
EZH2 Cell proliferation (38); Apoptosis (77) Maintain cardiac homeostasis (50); Dispensable for cardiac regeneration (4) Mouse: Failure of myocardial compaction, hypertrabeculation, and ventricular and atrial septal defects (38)
KMT2D Cell differentiation, embryonic development, calcium handling/ion homeostasis (11) Unknown Zebrafish: Abnormal development of the atria and/or ventricle, prominent bulging of the myocardial wall (184)
Mouse: Embryonic lethal, disorganized interventricular septum (11)
Human: Kabuki syndrome (165, 191), congenital heart defects (203)
DOT1L Cell differentiation (34) Regulate growth/shape, maintains normal cardiac function (138) Mouse: Dilated cardiomyopathy, conduction abnormalities, increased mortality (138, 139)
CARM1 Unknown Regulates inflammation (117); Cardiac autophagy (109) Unknown
PRMT5 Dispensable for cell growth (207) Regulate hypertrophic growth via GATA4 (39); Muscle stem cell proliferation during regeneration/repair (207) Unknown
PRMT6 Unknown Unknown Unknown
ASH1L Unknown Unknown Unknown
SETDB2 Unknown Unknown Unknown
SETD3 Unknown Unknown Unknown
SETD6 Unknown Unknown Unknown
SET8 Unknown Unknown Unknown

KD/KD, knockdown or knockout.

The SMYD family of methyltransferases consists of five members that share a similar catalytic SET domain, which is split by a MYND domain thought to be involved in protein binding. SMYD1, a myocyte-specific lysine methyltransferase, was originally shown to methylate histone H3 at K4 (a known mark of gene activation), which was also thought to be the hallmark function of this family. However, since then, it has been demonstrated that SMYD enzymes can methylate several other unique histone residues and nonhistone proteins, with the ability to activate or repress transcription at different genomic loci. Specifically, SMYD1 can induce transcriptional activation of PGC1α through histone H3K4me3 (192), SMYD2 influences transcription of the Tacc2 gene by binding to its promoter (3), and SMYD3’s ability to dimethylate and trimethylate histone H3K4 results in recruitment of histone acetyltransferases and Sp1 to the promoter region of the androgen receptor (114). In contrast, these enzymes have been shown to repress transcription through histone and/or nonhistone methylation or methylation-independent mechanisms: SMYD1 has been shown to be a histone deacetylase-dependent transcriptional repressor (71) and can repress transcription of nppa and tgfβ3 in cardiomyocytes (65). SMYD2 can dimethylate histone H3K36 in vitro and repress transcription through interaction with the Sin3A histone deacetylase complex (31) and SMYD3 plays an important role in negative transcriptional regulation by trimethylating histone H4K20 (64). Regarding their methylation of nonhistone proteins, SMYD1 can regulate transcription by methylating skNAC (19) or TRB3 (154). SMYD2 methylates retinoblastoma in response to DNA damage (160) and p53 (81), both resulting in transcriptional repression, or SMYD2 methylates PARP1 in cancer cells (150) and Hsp90 in muscle cells to activate transcription (52, 187). SMYD3 plays a critical role in various forms of cancer by methylating MAP3K2, which results in activation of the Ras/Raf/MEK/ERK signaling pathway and tumorigenesis (124).

In the heart, the SMYD family has been shown to play key roles in the regulation of growth and differentiation (181). SMYD1, SMYD2, and SMYD3 have been shown to be involved in morphogenesis (67, 71, 154), SMYD1 and SMYD2 in myofibrillogenesis (86, 111, 187), and SMYD3 in the maturation of myocytes (67). Specifically, cardiac-specific deletion of SMYD1 in the mouse heart removes repression of developmental genes that leads to transcriptional remodeling, hypertrophy growth, and fulminate heart failure (65). The repressive transcriptional role of SMYD1 was examined by ChIP and luciferase reporter assays, where SMYD1 was shown to bind directly to the promoters of tgfb3 and nppa genes and inhibit their expression, independent of its methyltransferase activity toward histone H3K4me3, emphasizing an unknown mechanism for this transcriptional regulation (65). SMYD2 was shown to be a key regulator of myofibrillogenesis and sarcomere organization in zebrafish although knockout in the mouse heart gave no overt phenotype (51, 52). SMYD3 is necessary for the development of cardiac and skeletal muscle in zebrafish (67); however, the specific mechanism for its functionality in the heart was never reported.

G9a (also known as EHMT2 or KMT1C) is responsible for the majority of monomethylation and dimethylation of histone H3 at K9 in mammalian cells, and it has also been shown to methylate histone H3 on K27 (173). This methyltransferase uses its catalytic activity to efficiently repress developmentally regulated genes during embryonic stem cell differentiation (174), in addition to regulating T-cell differentiation (108) and muscle differentiation (113).

G9a and its coregulatory G9a-like protein/GLP (also known as EHMT1 or KMT1D) regulate transcription as either a homomeric or heteromeric complex; however, the heteromeric complex seems to be the functional form in vivo due to its increased stability (113). In addition, while G9a and GLP can both independently methylate histones and possess the same substrate specificity in vitro, when individually knocked down they cannot compensate for the levels of histone H3K9me2, further suggesting that they primarily function as a complex (113, 170). The G9a/GLP heterocomplex can also regulate transcription by partnering with cofactors, including the histone demethylase Jarid1a to repress gene expression during erythroid cell differentiation in a methyltransferase-dependent manner (36). Additionally, recent studies have demonstrated that the G9a/GLP heterocomplex can also regulate transcription through methyltransferase-independent mechanisms by either recruiting CARM1 and p300 to glucocorticoid receptor genes to activate transcription (22) or by interacting with p53 and influencing transcriptional activation in the absence of p53 methylation; however, the specific mechanism has not yet been established (152).

In the heart, in the conditional, cardiac-specific G9a knockout mice, G9a was shown to be an important regulator of cardiomyocyte homeostasis by mediating the repression of genes regulating cardiomyocyte function, including antihypertrophic genes through its methyltransferase activity on histone H3K9 and interaction with EZH2 and also by forming a complex with the transcription factor MEF2C (146). Additionally, Thienpont et al. (177) also demonstrated that loss of EHMT1 and EHMT2 enzymatic activity is sufficient to induce pathological cardiac hypertrophy in the adult cardiomyocyte and that activation of this pathway can protect against pathological hypertrophy in vivo and in vitro.

EZH2 (Enhancer of Zeste 2) is one of the most studied lysine methyltransferases. It belongs to the Polycomb group family, functions as an active catalytic subunit of the Polycomb Repressive Complex 2 (PRC2), and is responsible for the repression of many genes involved in development and cell differentiation (28, 29). EZH2 contains a SET domain that targets histone H3 K27. Initially, it was demonstrated that EZH2 plays a critical role in silencing tumor suppressor genes due to its trimethylation of histone H3K27 (123, 201). However, recently Ferrari et al. (61) reported that EZH2 and PRC2 complex can control all three stages of histone H3K27 methylation, and while dimethylation and trimethylation is associated with gene repression, H3K27 monomethylation was found to be enriched at the transcriptional start site of active genes. In addition to EZH2’s catalytic activity, there have been reports suggesting new noncanonical roles of this enzyme. Specifically, EZH2 has been shown to methylate GATA4, which lead to its transcriptional deactivation (78) or Jarid2, a known (1) cofactor of PRC2 that regulates chromatin structure by regulating H3K27me3 during cell differentiation (161). Moreover, several studies have reported that EZH2 can act independently of the PRC2 complex in a methyltransferase-independent manner. For example, EZH2 has been shown to activate NF-κB target gene expression in ER-negative basal-like breast cancer cells by interacting with RelA and RelB (106). Lastly, Kim et al. (91) demonstrated that when EZH2 interacts with the SWI-SNF complex, it can activate target genes via a PRC2-independent mechanism and can activate androgen receptor gene transcription by binding directly to its promoter (89).

Ablation of EZH2 in the fetal heart leads to decreased cardiomyocyte proliferation and increased apoptosis, and enzymatic inactivation of EZH2 in the fetal heart results in lethal congenital malformations or perinatal death (38, 77). However, in the neonatal heart, it has been shown to be dispensable for heart regeneration (4).

KMT2D (also known as MLL4 and MLL2 in humans) belongs to a family of histone lysine methyltransferases. It is ubiquitously expressed in adult tissues and plays an important role in embryonic development, cellular differentiation, metabolism, and tumor suppression. Additionally, it was implicated in human congenital heart disease and Kabuki syndrome (66). Molecularly it was first demonstrated that KMT2D plays an essential role in transcriptional activation due to histone H3K4 methylation; however, more recently, it has also been shown to negatively regulate gene expression. Specifically, KMT2D’s catalytic SET domain has been shown to methylate histone H3K4 generally at enhancer regions and maintain global levels of this methylation mark (104). More specifically, monomethylation of histone H3K4 by human KMT2D (MLL4) activates transcription of peroxisome proliferator-activated receptor γ in overnutrition-induced hepatic steatosis (88). KMT2D can also associate with WRAD, NCOA6, PTIP, PA1, and UTX demethylase in a multiprotein complex, where it is necessary to activate complex formation and stabilization of UTX during its histone H3K27 demethylation activity (45, 84). Additionally, KMT2D can interact with ERα in the presence of estrogen and regulate ERα transcriptional activity (129) or with CBP/p300 at enhancers during cell differentiation (101). Lastly, MLL4 in complex with MLL3 is known to monomethylate histone H3K4 at specific promoter regions, leading to conditional repression of muscle and inflammatory response genes in myoblasts (41).

It is well established that KMT2D is essential for cardiac cell differentiation and embryonic development (11). KMT2D is also implicated in cardiac disease, where it was first found to be differentially expressed in patients with congenital heart disease and later confirmed functionally using KM2TD knockout mouse that develop severe cardiac defects (11, 203). In addition, aberrant trimethylation by KMT2D can lead to Kabuki syndrome (137).

DOT1L (disruptor of telomeric silencing 1-like) is a unique lysine methyltransferase because instead of having a canonical SET catalytic domain, it contains a SET binding motif similar to arginine and DNA methyltransferases (193). Molecularly, DOT1L targets histone H3K79, which is located on the nucleosome surface, as opposed to the NH2-terminal tail where most posttranslational modifications are deposited. It catalyzes the monomethylation, demethylation, and trimethylation of K79, but must dissociate and reassociate again with the residue after each methylation event to add additional methyl groups. Monomethylation and dimethylation of histone H3K79 is associated with open chromatin and transcriptional activation, while H3K79me3 is a mark of transcriptional repression (193). DOT1L plays a critical role in transcriptional elongation, cell cycle progression, development, somatic reprogramming, and DNA damage repair. DOT1L was also found to play a role in tumorigenesis, as it interacts with the mixed lineage leukemia (MLL) fusion proteins ENL/MLLT1, AF4/MLLT2, AF9/MLLT3, and AF10/MLLT10 and forms a complex that is necessary for histone H3K79 methylation, euchromatin formation, and expression of downstream oncogenes and leukemogenesis (21, 130, 133). Additionally, Cho et al. (44) reported that DOT1L binds to the oncogene c-Myc before epigenetic activation of the epithelial-mesenchymal transition (EMT) regulators in breast cancer progression. DOT1L can interact directly with the CTD domain of RNA Polymerase II and recruit this enzyme to the promoter regions of genes to be transcribed (94). DOT1L was also implicated in telomeric silencing, where methylation of histone H3K79 prevents binding of Sir3 (62).

In the heart, DOTL1 has been shown to play a critical role during cardiomyocyte differentiation (34, 151). Additionally, cardiac-specific knockout of DOT1L in mice leads to downregulation of histone H3K79, the development of dilated cardiomyopathy, conduction abnormalities, and increased mortality (138).

CARM1 (coactivator-associated arginine methyltransferase 1, also known as PRMT4 or Protein Arginine Methyltransferase 4) was the first PRMT that was functionally linked to regulation of gene expression (195) and has been shown to methylate several arginine residues on histone H3, which lead to transcriptional activation, including R2, R17, R26, and R42 (although H3R2Me2a has only been shown to occur in vitro) (69, 90, 121, 164). Specifically, PRMT4 was found to asymmetrically dimethylate R17 on histone H3, which leads to recruitment of the transcription elongation-associated PAF1 complex that, in turn, activates a subset of genes, including targets of estrogen signaling (194). CARM1 can also methylate arginine residues on histone H3 in the core of the structure, including asymmetrical dimethylation of R42, which signals transcriptional activation by disrupting histone:DNA interactions (33). In addition, CARM1 can methylate several nonhistone substrates, including transcription factors (40, 209), coregulators (59, 136), and RNA polymerase II (171), which result in either positive or negative transcriptional regulation. Specifically, CARM1 methylation of Sox2 leads to transactivation in ESC cells (209), and methylation of CBP/p300 is required for the activity and stability of the transcriptional coactivator p/CIP/SRC-3 in hormone signaling (136). Conversely, methylating the GRIP1 binding domain of p300 by CARM1 blocks its interaction with GRIP1 and inhibits estrogen receptor-mediated transcription (107). PRMT4 can also repress transcription by methylating the steroid receptor coactivator (SRC-3), which leads to decreased ERα-mediated transcription and termination of hormone signaling by disassembling the SRC-3/CARM1 coactivator complex (59).

CARM1 has only been examined in two studies where CARM1’s stabilization, regulated by nuclear AMPK, can contribute to autophagy in the mature heart (109) and where it was shown to regulate inflammation in acute coronary syndrome (117); however, its regulatory role was dependent on miR-15a targeting.

PRMT5 (protein arginine methyltransferase 5) is known as a master epigenetic regulator, essential for cellular growth and development, and functions in a wide variety of cellular processes, including signal transduction, transcription, translation, and DNA recombination. Among the long list of PRMT5 substrates (167), we will focus only on those which provide evidence for its regulatory role in gene expression. Specifically, PRMT5 has been shown to regulate transcription by either modifying histone residues or by directly modulating the activity of several transcription factors to both activate and repress transcription depending on the arginine residue that is methylated and/or the promoter targeted. For instance, PRMT5-mediated symmetric dimethylation of histone H3R8 and histone H4R3 leads to silencing of several tumor suppressor genes [including Sp7, Rb, and Ptpro (6, 145, 190)] and immune response genes (182); however, methylation of these same residues can also activate transcription of Fgfr-3 and Eif4 (204) and adipogenic differentiation genes (103). PRMT5 can also methylate histone H3R2, which recruits Wdr5 and the MLL complex and stimulates histone H3K4 methylation and euchromatin maintenance (127). In association with Mep50, PRMT5 also methylates cytosolic histone H2AR3 to repress differentiation-specific genes in pluripotent ES cells (176). Lastly, PRMT5 can methylate the heterogeneous nuclear ribonucleoprotein hnRNP A1, which promotes its interaction with IRES RNA and results in IRES-dependent translation of various genes (Ccnd1, Cdkn1B, Myc, HIF1a, Mtif, and Mep50) (68).

PRMT5 is required for muscle stem cell differentiation and is important in adult muscle regeneration and repair, but shown to be dispensable for expansion of muscle progenitor cells during development (207, 208). In the heart, it can also regulate cardiac sodium channels through arginine methylation (18) and hypertrophic signaling by methylating GATA4 on three arginine residues, which inhibits the ability of GATA4 to promote transcriptional activation (39).

CONCLUDING REMARKS

Cardiovascular epigenetics defines heritable mechanisms that establish stable patterns of cardiac gene expression without modifying the base sequence of DNA. It is known that at the basis of many heart pathologies lie the vast spectrum of dynamic changes in cardiac gene expression that represent the molecular underpinnings driving morphological and physiological transformation of the myocardium, which also include both maladaptive and compensatory mechanisms aimed at mitigating disease-induced remodeling (32, 153). This includes any global changes in gene expression required for chromatin to undergo structural changes to silence or activate specific loci within the nucleus (178). Over the last decade there has been impressive progress made in our understanding of how environmental conditions affect cardiac gene expression through epigenetic mechanisms. We now know much more about the roles of chromatin modifications and chromatin regulators in the context of cardiac diseases (115). Specifically, several studies in murine models of cardiac hypertrophy have demonstrated that modulating key epigenetic factors, such as HDACs (85, 132, 205), BET domain-containing proteins (53), or specific transcription factors (112) can influence changes in gene expression to prevent pathological remodeling; however, we still lack the deep understanding of the mechanisms responsible for regulating chromatin structure during the pathology of heart disease. Among the many epigenetic mechanisms, which regulate heart physiology and pathology, several excellent reviews have recently highlighted DNA methylation, noncoding RNAs, structural remodeling of chromatin (2, 35, 95, 115), and histone posttranslational modifications (12) and can be found using these references, among others. However, the role of the enzymes that add or remove these posttranslational modifications have been less well studied, and a more global view (not focused on the heart) has been reviewed previously (42).

In recent years, the contribution of epigenetics to cardiac physiology has emerged as a key regulatory process in both cardiac development and disease and has recognized the involvement of methyltransferases as major players in transcriptional regulation. Specifically, protein methyltransferases have been linked to the development of congenital heart disease, cardiac hypertrophy and heart failure (206). For instance, aberrant trimethylation by KMT2D can lead to Kabuki syndrome (137) or inactivation of DOT1L in the mouse heart causes downregulation of H3K79 methylation that results in the development of dilated cardiomyopathy (138). However, we have only begun to scratch the surface in understanding the specific roles of lysine and arginine methyltransferases in the heart. This review highlights the many methyltransferases expressed in the heart, how their expression is differentially regulated during development or disease, and emphasizes that only a limited number of these enzymes have been functionally examined in any detail in this organ (more than one-third of these enzymes remain uncharacterized). Therefore, future studies will be needed to interrogate these enzymes to determine the implications to cardiac pathophysiology. Additional research that is focused on expanding our understanding of how epigenetic regulation is driven by these uncharacterized methyltransferases may unlock their therapeutic potential with new perspectives for translational research and new diagnostic tools, as well as novel strategies in drug design and discovery.

Indeed, we have much to learn about these enzymes, including the specific molecular mechanisms governing substrate specificity and the downstream effects of their catalytic activity. As highlighted in this review, there is now strong evidence to show that these enzymes have a much broader role in regulating transcription than has previously been appreciated. While their substrate specificity was traditionally thought to be engendered by the ability of one enzyme to methylate one amino acid residue in one manner (monomethylation, dimethylation, or trimethylation) to activate or repress transcription, the compilation of methyltransferase data in this review shows that these enzymes maintain the ability to both activate and repress transcription in a context-specific manner. Interestingly, some methyltransferases are very specific and exhibit this dual functionality with only one methylation target. For example, DOT1L is the only enzyme known to methylate K79 on histone H3, which, depending on the degree of methylation (monomethylation, demethylation, or trimethylation), can have a positive or negative effect on transcription. However, other enzymes, like G9a/GLP are more promiscuous and in addition to methylating multiple histone residues, they can also methylate other substrates within the nucleus or cytosol to positively or negatively regulate transcription. Given the growing amount of evidence, which continues to reveal new insights into the function of these enzymes, one can appreciate how little we know about their regulatory roles and how much is still to be discovered. And finally, we cannot forget about the substantial list of putative enzymes that have been classified on the basis of their unique methyltransferase domains but await functional characterization.

GRANTS

This work was supported by the National Institutes of Health Grants R01-HL-130424 and F32-HL-144034, American Heart Association Grant 20PRE35120356, and the Nora Eccles Harrison Treadwell Foundation Grant 10038331.

DISCLOSURES

No conflict of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

M.W.S., K.D., A.B., R.B., and S.F. prepared figures; M.W.S., K.D., and A.B. drafted manuscript; M.W.S., K.D., P.A., E.H., and S.F. edited and revised manuscript; M.W.S., K.D., A.B., P.A., R.B., E.H., and S.F. approved final version of manuscript.

REFERENCES

  • 1.Abaev-Schneiderman E, Admoni-Elisha L, Levy D. SETD3 is a positive regulator of DNA-damage-induced apoptosis. Cell Death Dis 10: 74, 2019. doi: 10.1038/s41419-019-1328-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Abi Khalil C. The emerging role of epigenetics in cardiovascular disease. Ther Adv Chronic Dis 5: 178–187, 2014. doi: 10.1177/2040622314529325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Abu-Farha M, Lambert JP, Al-Madhoun AS, Elisma F, Skerjanc IS, Figeys D. The tale of two domains: proteomics and genomics analysis of SMYD2, a new histone methyltransferase. Mol Cell Proteomics 7: 560–572, 2008. doi: 10.1074/mcp.M700271-MCP200. [DOI] [PubMed] [Google Scholar]
  • 4.Ahmed A, Wang T, Delgado-Olguin P. Ezh2 is not required for cardiac regeneration in neonatal mice. PLoS One 13: e0192238, 2018. doi: 10.1371/journal.pone.0192238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ai S, Yu X, Li Y, Peng Y, Li C, Yue Y, Tao G, Li C, Pu WT, He A. Divergent requirements for EZH1 in heart development versus regeneration. Circ Res 121: 106–112, 2017. doi: 10.1161/CIRCRESAHA.117.311212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Alinari L, Mahasenan KV, Yan F, Karkhanis V, Chung JH, Smith EM, Quinion C, Smith PL, Kim L, Patton JT, Lapalombella R, Yu B, Wu Y, Roy S, De Leo A, Pileri S, Agostinelli C, Ayers L, Bradner JE, Chen-Kiang S, Elemento O, Motiwala T, Majumder S, Byrd JC, Jacob S, Sif S, Li C, Baiocchi RA. Selective inhibition of protein arginine methyltransferase 5 blocks initiation and maintenance of B-cell transformation. Blood 125: 2530–2543, 2015. doi: 10.1182/blood-2014-12-619783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci USA 51: 786–794, 1964. doi: 10.1073/pnas.51.5.786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ambler RP, Rees MW. Epsilon-N-methyl-lysine in bacterial flagellar protein. Nature 184: 56–57, 1959. doi: 10.1038/184056b0. [DOI] [PubMed] [Google Scholar]
  • 9.An W, Kim J, Roeder RG. Ordered cooperative functions of PRMT1, p300, and CARM1 in transcriptional activation by p53. Cell 117: 735–748, 2004. doi: 10.1016/j.cell.2004.05.009. [DOI] [PubMed] [Google Scholar]
  • 10.Ancelin K, Lange UC, Hajkova P, Schneider R, Bannister AJ, Kouzarides T, Surani MA. Blimp1 associates with Prmt5 and directs histone arginine methylation in mouse germ cells. Nat Cell Biol 8: 623–630, 2006. doi: 10.1038/ncb1413. [DOI] [PubMed] [Google Scholar]
  • 11.Ang SY, Uebersohn A, Spencer CI, Huang Y, Lee JE, Ge K, Bruneau BG. KMT2D regulates specific programs in heart development via histone H3 lysine 4 di-methylation. Development 143: 810–821, 2016. doi: 10.1242/dev.132688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res 21: 381–395, 2011. doi: 10.1038/cr.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.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 129: 823–837, 2007. doi: 10.1016/j.cell.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 14.Beacon TH, Xu W, Davie JR. Genomic landscape of transcriptionally active histone arginine methylation marks, H3R2me2s and H4R3me2a, relative to nucleosome depleted regions. Gene 742: 144593, 2020. doi: 10.1016/j.gene.2020.144593. [DOI] [PubMed] [Google Scholar]
  • 15.Bedford MT, Clarke SG. Protein arginine methylation in mammals: who, what, and why. Mol Cell 33: 1–13, 2009. doi: 10.1016/j.molcel.2008.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bedford MT, Richard S. Arginine methylation an emerging regulator of protein function. Mol Cell 18: 263–272, 2005. doi: 10.1016/j.molcel.2005.04.003. [DOI] [PubMed] [Google Scholar]
  • 17.Bei Y, Tianqian H, Fanyuan Y, Haiyun L, Xueyang L, Jing Y, Chenglin W, Ling Y. ASH1L suppresses matrix metalloproteinase through mitogen-activated protein kinase signaling pathway in pulpitis. J Endod 43: 306–314.e2, 2017. doi: 10.1016/j.joen.2016.10.020. [DOI] [PubMed] [Google Scholar]
  • 18.Beltran-Alvarez P, Espejo A, Schmauder R, Beltran C, Mrowka R, Linke T, Batlle M, Pérez-Villa F, Pérez GJ, Scornik FS, Benndorf K, Pagans S, Zimmer T, Brugada R. Protein arginine methyl transferases-3 and -5 increase cell surface expression of cardiac sodium channel. FEBS Lett 587: 3159–3165, 2013. doi: 10.1016/j.febslet.2013.07.043. [DOI] [PubMed] [Google Scholar]
  • 19.Berkholz J, Orgeur M, Stricker S, Munz B. skNAC and Smyd1 in transcriptional control. Exp Cell Res 336: 182–191, 2015. doi: 10.1016/j.yexcr.2015.06.019. [DOI] [PubMed] [Google Scholar]
  • 20.Binda O, Sevilla A, LeRoy G, Lemischka IR, Garcia BA, Richard S. SETD6 monomethylates H2AZ on lysine 7 and is required for the maintenance of embryonic stem cell self-renewal. Epigenetics 8: 177–183, 2013. doi: 10.4161/epi.23416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bitoun E, Oliver PL, Davies KE. The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling. Hum Mol Genet 16: 92–106, 2007. doi: 10.1093/hmg/ddl444. [DOI] [PubMed] [Google Scholar]
  • 22.Bittencourt D, Wu DY, Jeong KW, Gerke DS, Herviou L, Ianculescu I, Chodankar R, Siegmund KD, Stallcup MR. G9a functions as a molecular scaffold for assembly of transcriptional coactivators on a subset of glucocorticoid receptor target genes. Proc Natl Acad Sci USA 109: 19673–19678, 2012. doi: 10.1073/pnas.1211803109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Blanc RS, Richard S. Arginine methylation: the coming of age. Mol Cell 65: 8–24, 2017. doi: 10.1016/j.molcel.2016.11.003. [DOI] [PubMed] [Google Scholar]
  • 24.Blanc RS, Vogel G, Li X, Yu Z, Li S, Richard S. Arginine methylation by PRMT1 regulates muscle stem cell fate. Mol Cell Biol 37: e00457-16, 2017. doi: 10.1128/MCB.00457-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Boriack-Sjodin PA, Swinger KK. Protein methyltransferases: a distinct, diverse, and dynamic family of enzymes. Biochemistry 55: 1557–1569, 2016. doi: 10.1021/acs.biochem.5b01129. [DOI] [PubMed] [Google Scholar]
  • 26.Bouchard C, Sahu P, Meixner M, Nötzold RR, Rust MB, Kremmer E, Feederle R, Hart-Smith G, Finkernagel F, Bartkuhn M, Savai Pullamsetti S, Nist A, Stiewe T, Philipsen S, Bauer UM. Genomic location of PRMT6-dependent H3R2 methylation is linked to the transcriptional outcome of associated genes. Cell Reports 24: 3339–3352, 2018. doi: 10.1016/j.celrep.2018.08.052. [DOI] [PubMed] [Google Scholar]
  • 27.Boulanger MC, Liang C, Russell RS, Lin R, Bedford MT, Wainberg MA, Richard S. Methylation of Tat by PRMT6 regulates human immunodeficiency virus type 1 gene expression. J Virol 79: 124–131, 2005. doi: 10.1128/JVI.79.1.124-131.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, Levine SS, Wernig M, Tajonar A, Ray MK, Bell GW, Otte AP, Vidal M, Gifford DK, Young RA, Jaenisch R. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 441: 349–353, 2006. doi: 10.1038/nature04733. [DOI] [PubMed] [Google Scholar]
  • 29.Bracken AP, Dietrich N, Pasini D, Hansen KH, Helin K. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev 20: 1123–1136, 2006. doi: 10.1101/gad.381706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Broka D, Ditzel E, Quach S, Camenisch TD. Methylation of inorganic arsenic by murine fetal tissue explants. Drug Chem Toxicol 39: 279–283, 2016. doi: 10.3109/01480545.2015.1092043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Brown MA, Sims RJ 3rd, Gottlieb PD, Tucker PW. Identification and characterization of Smyd2: a split SET/MYND domain-containing histone H3 lysine 36-specific methyltransferase that interacts with the Sin3 histone deacetylase complex. Mol Cancer 5: 26, 2006. doi: 10.1186/1476-4598-5-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Buttrick PM, Kaplan M, Leinwand LA, Scheuer J. Alterations in gene expression in the rat heart after chronic pathological and physiological loads. J Mol Cell Cardiol 26: 61–67, 1994. doi: 10.1006/jmcc.1994.1008. [DOI] [PubMed] [Google Scholar]
  • 33.Casadio F, Lu X, Pollock SB, LeRoy G, Garcia BA, Muir TW, Roeder RG, Allis CD. H3R42me2a is a histone modification with positive transcriptional effects. Proc Natl Acad Sci USA 110: 14894–14899, 2013. doi: 10.1073/pnas.1312925110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cattaneo P, Kunderfranco P, Greco C, Guffanti A, Stirparo GG, Rusconi F, Rizzi R, Di Pasquale E, Locatelli SL, Latronico MV, Bearzi C, Papait R, Condorelli G. DOT1L-mediated H3K79me2 modification critically regulates gene expression during cardiomyocyte differentiation. Cell Death Differ 23: 555–564, 2016. doi: 10.1038/cdd.2014.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chang CP, Bruneau BG. Epigenetics and cardiovascular development. Annu Rev Physiol 74: 41–68, 2012. doi: 10.1146/annurev-physiol-020911-153242. [DOI] [PubMed] [Google Scholar]
  • 36.Chaturvedi CP, Somasundaram B, Singh K, Carpenedo RL, Stanford WL, Dilworth FJ, Brand M. Maintenance of gene silencing by the coordinate action of the H3K9 methyltransferase G9a/KMT1C and the H3K4 demethylase Jarid1a/KDM5A. Proc Natl Acad Sci USA 109: 18845–18850, 2012. doi: 10.1073/pnas.1213951109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chen A, Feldman M, Vershinin Z, Levy D. SETD6 is a negative regulator of oxidative stress response. Biochim Biophys Acta 1859: 420–427, 2016. doi: 10.1016/j.bbagrm.2016.01.003. [DOI] [PubMed] [Google Scholar]
  • 38.Chen L, Ma Y, Kim EY, Yu W, Schwartz RJ, Qian L, Wang J. Conditional ablation of Ezh2 in murine hearts reveals its essential roles in endocardial cushion formation, cardiomyocyte proliferation and survival. PLoS One 7: e31005, 2012. doi: 10.1371/journal.pone.0031005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chen M, Yi B, Sun J. Inhibition of cardiomyocyte hypertrophy by protein arginine methyltransferase 5. J Biol Chem 289: 24325–24335, 2014. doi: 10.1074/jbc.M114.577494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Cheng D, Côté J, Shaaban S, Bedford MT. The arginine methyltransferase CARM1 regulates the coupling of transcription and mRNA processing. Mol Cell 25: 71–83, 2007. doi: 10.1016/j.molcel.2006.11.019. [DOI] [PubMed] [Google Scholar]
  • 41.Cheng J, Blum R, Bowman C, Hu D, Shilatifard A, Shen S, Dynlacht BD. A role for H3K4 monomethylation in gene repression and partitioning of chromatin readers. Mol Cell 53: 979–992, 2014. doi: 10.1016/j.molcel.2014.02.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Cheng X, Blumenthal RM. Coordinated chromatin control: structural and functional linkage of DNA and histone methylation. Biochemistry 49: 2999–3008, 2010. doi: 10.1021/bi100213t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chevillard-Briet M, Trouche D, Vandel L. Control of CBP co-activating activity by arginine methylation. EMBO J 21: 5457–5466, 2002. doi: 10.1093/emboj/cdf548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cho MH, Park JH, Choi HJ, Park MK, Won HY, Park YJ, Lee CH, Oh SH, Song YS, Kim HS, Oh YH, Lee JY, Kong G. DOT1L cooperates with the c-Myc-p300 complex to epigenetically derepress CDH1 transcription factors in breast cancer progression. Nat Commun 6: 7821, 2015. doi: 10.1038/ncomms8821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Cho YW, Hong T, Hong S, Guo H, Yu H, Kim D, Guszczynski T, Dressler GR, Copeland TD, Kalkum M, Ge K. PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J Biol Chem 282: 20395–20406, 2007. doi: 10.1074/jbc.M701574200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cohn O, Feldman M, Weil L, Kublanovsky M, Levy D. Chromatin associated SETD3 negatively regulates VEGF expression. Sci Rep 6: 37115, 2016. doi: 10.1038/srep37115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Covic M, Hassa PO, Saccani S, Buerki C, Meier NI, Lombardi C, Imhof R, Bedford MT, Natoli G, Hottiger MO. Arginine methyltransferase CARM1 is a promoter-specific regulator of NF-kappaB-dependent gene expression. EMBO J 24: 85–96, 2005. doi: 10.1038/sj.emboj.7600500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Currier JM, Douillet C, Drobná Z, Stýblo M. Oxidation state specific analysis of arsenic species in tissues of wild-type and arsenic (+3 oxidation state) methyltransferase-knockout mice. J Environ Sci (China) 49: 104–112, 2016. doi: 10.1016/j.jes.2016.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dang Y, Ma X, Li Y, Hao Q, Xie Y, Zhang Q, Zhang F, Qi X. Inhibition of SETD7 protects cardiomyocytes against hypoxia/reoxygenation-induced injury through regulating Keap1/Nrf2 signaling. Biomed Pharmacother 106: 842–849, 2018. doi: 10.1016/j.biopha.2018.07.007. [DOI] [PubMed] [Google Scholar]
  • 50.Delgado-Olguín P, Huang Y, Li X, Christodoulou D, Seidman CE, Seidman JG, Tarakhovsky A, Bruneau BG. Epigenetic repression of cardiac progenitor gene expression by Ezh2 is required for postnatal cardiac homeostasis. Nat Genet 44: 343–347, 2012. doi: 10.1038/ng.1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Diehl F, Brown MA, van Amerongen MJ, Novoyatleva T, Wietelmann A, Harriss J, Ferrazzi F, Böttger T, Harvey RP, Tucker PW, Engel FB. Cardiac deletion of Smyd2 is dispensable for mouse heart development. PLoS One 5: e9748, 2010. doi: 10.1371/journal.pone.0009748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Donlin LT, Andresen C, Just S, Rudensky E, Pappas CT, Kruger M, Jacobs EY, Unger A, Zieseniss A, Dobenecker MW, Voelkel T, Chait BT, Gregorio CC, Rottbauer W, Tarakhovsky A, Linke WA. Smyd2 controls cytoplasmic lysine methylation of Hsp90 and myofilament organization. Genes Dev 26: 114–119, 2012. doi: 10.1101/gad.177758.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Duan Q, McMahon S, Anand P, Shah H, Thomas S, Salunga HT, Huang Y, Zhang R, Sahadevan A, Lemieux ME, Brown JD, Srivastava D, Bradner JE, McKinsey TA, Haldar SM. BET bromodomain inhibition suppresses innate inflammatory and profibrotic transcriptional networks in heart failure. Sci Transl Med 9: eaah5084, 2017. doi: 10.1126/scitranslmed.aah5084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.El-Andaloussi N, Valovka T, Toueille M, Steinacher R, Focke F, Gehrig P, Covic M, Hassa PO, Schär P, Hübscher U, Hottiger MO. Arginine methylation regulates DNA polymerase beta. Mol Cell 22: 51–62, 2006. doi: 10.1016/j.molcel.2006.02.013. [DOI] [PubMed] [Google Scholar]
  • 55.Eom GH, Kim KB, Kim JH, Kim JY, Kim JR, Kee HJ, Kim DW, Choe N, Park HJ, Son HJ, Choi SY, Kook H, Seo SB. Histone methyltransferase SETD3 regulates muscle differentiation. J Biol Chem 286: 34733–34742, 2011. doi: 10.1074/jbc.M110.203307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Fabbrizio E, El Messaoudi S, Polanowska J, Paul C, Cook JR, Lee JH, Negre V, Rousset M, Pestka S, Le Cam A, Sardet C. Negative regulation of transcription by the type II arginine methyltransferase PRMT5. EMBO Rep 3: 641–645, 2002. doi: 10.1093/embo-reports/kvf136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fan LL, Ding DB, Huang H, Chen YQ, Jin JY, Xia K, Xiang R. A de novo mutation of SMYD1 (p.F272L) is responsible for hypertrophic cardiomyopathy in a Chinese patient. Clin Chem Lab Med 57: 532–539, 2019. doi: 10.1515/cclm-2018-0578. [DOI] [PubMed] [Google Scholar]
  • 58.Feldman M, Vershinin Z, Goliand I, Elia N, Levy D. The methyltransferase SETD6 regulates mitotic progression through PLK1 methylation. Proc Natl Acad Sci USA 116: 1235–1240, 2019. doi: 10.1073/pnas.1804407116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Feng Q, Yi P, Wong J, O’Malley BW. Signaling within a coactivator complex: methylation of SRC-3/AIB1 is a molecular switch for complex disassembly. Mol Cell Biol 26: 7846–7857, 2006. doi: 10.1128/MCB.00568-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Fenouille N, Bassil CF, Ben-Sahra I, Benajiba L, Alexe G, Ramos A, Pikman Y, Conway AS, Burgess MR, Li Q, Luciano F, Auberger P, Galinsky I, DeAngelo DJ, Stone RM, Zhang Y, Perkins AS, Shannon K, Hemann MT, Puissant A, Stegmaier K. The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia. Nat Med 23: 301–313, 2017. doi: 10.1038/nm.4283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ferrari KJ, Scelfo A, Jammula S, Cuomo A, Barozzi I, Stützer A, Fischle W, Bonaldi T, Pasini D. Polycomb-dependent H3K27me1 and H3K27me2 regulate active transcription and enhancer fidelity. Mol Cell 53: 49–62, 2014. doi: 10.1016/j.molcel.2013.10.030. [DOI] [PubMed] [Google Scholar]
  • 62.Fingerman IM, Li HC, Briggs SD. A charge-based interaction between histone H4 and Dot1 is required for H3K79 methylation and telomere silencing: identification of a new trans-histone pathway. Genes Dev 21: 2018–2029, 2007. doi: 10.1101/gad.1560607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Flanagan JF, Mi LZ, Chruszcz M, Cymborowski M, Clines KL, Kim Y, Minor W, Rastinejad F, Khorasanizadeh S. Double chromodomains cooperate to recognize the methylated histone H3 tail. Nature 438: 1181–1185, 2005. doi: 10.1038/nature04290. [DOI] [PubMed] [Google Scholar]
  • 64.Foreman KW, Brown M, Park F, Emtage S, Harriss J, Das C, Zhu L, Crew A, Arnold L, Shaaban S, Tucker P. Structural and functional profiling of the human histone methyltransferase SMYD3. PLoS One 6: e22290, 2011. doi: 10.1371/journal.pone.0022290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Franklin S, Kimball T, Rasmussen TL, Rosa-Garrido M, Chen H, Tran T, Miller MR, Gray R, Jiang S, Ren S, Wang Y, Tucker HO, Vondriska TM. The chromatin-binding protein Smyd1 restricts adult mammalian heart growth. Am J Physiol Heart Circ Physiol 311: H1234–H1247, 2016. doi: 10.1152/ajpheart.00235.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Froimchuk E, Jang Y, Ge K. Histone H3 lysine 4 methyltransferase KMT2D. Gene 627: 337–342, 2017. doi: 10.1016/j.gene.2017.06.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fujii T, Tsunesumi S, Yamaguchi K, Watanabe S, Furukawa Y. Smyd3 is required for the development of cardiac and skeletal muscle in zebrafish. PLoS One 6: e23491, 2011. doi: 10.1371/journal.pone.0023491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gao G, Dhar S, Bedford MT. PRMT5 regulates IRES-dependent translation via methylation of hnRNP A1. Nucleic Acids Res 45: 4359–4369, 2017. doi: 10.1093/nar/gkw1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gayatri S, Bedford MT. Readers of histone methylarginine marks. Biochim Biophys Acta 1839: 702–710, 2014. doi: 10.1016/j.bbagrm.2014.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gong G, O’Bryant SE. Low-level arsenic exposure, AS3MT gene polymorphism and cardiovascular diseases in rural Texas counties. Environ Res 113: 52–57, 2012. doi: 10.1016/j.envres.2012.01.003. [DOI] [PubMed] [Google Scholar]
  • 71.Gottlieb PD, Pierce SA, Sims RJ 3rd, Yamagishi H, Weihe EK, Harriss JV, Maika SD, Kuziel WA, King HL, Olson EN, Nakagawa O, Srivastava D. Bop encodes a muscle-restricted protein containing MYND and SET domains and is essential for cardiac differentiation and morphogenesis. Nat Genet 31: 25–32, 2002. doi: 10.1038/ng866. [DOI] [PubMed] [Google Scholar]
  • 72.Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet 13: 343–357, 2012. doi: 10.1038/nrg3173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Guccione E, Richard S. The regulation, functions and clinical relevance of arginine methylation. Nat Rev Mol Cell Biol 20: 642–657, 2019. doi: 10.1038/s41580-019-0155-x. [DOI] [PubMed] [Google Scholar]
  • 74.Hall KT, Battinelli E, Chasman DI, Ridker PM, Psaty BM, Rotter JI, Kaptchuk TJ, Tracy RP, Wassel CL, Mukamal KJ. Catechol-O-methyltransferase and cardiovascular disease: MESA. J Am Heart Assoc 8: e014986, 2019. doi: 10.1161/JAHA.119.014986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hamamoto R, Furukawa Y, Morita M, Iimura Y, Silva FP, Li M, Yagyu R, Nakamura Y. SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat Cell Biol 6: 731–740, 2004. doi: 10.1038/ncb1151. [DOI] [PubMed] [Google Scholar]
  • 76.Hashimoto M, Murata K, Ishida J, Kanou A, Kasuya Y, Fukamizu A. Severe hypomyelination and developmental defects are caused in mice lacking protein arginine methyltransferase 1 (PRMT1) in the central nervous system. J Biol Chem 291: 2237–2245, 2016. doi: 10.1074/jbc.M115.684514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.He A, Ma Q, Cao J, von Gise A, Zhou P, Xie H, Zhang B, Hsing M, Christodoulou DC, Cahan P, Daley GQ, Kong SW, Orkin SH, Seidman CE, Seidman JG, Pu WT. Polycomb repressive complex 2 regulates normal development of the mouse heart. Circ Res 110: 406–415, 2012. doi: 10.1161/CIRCRESAHA.111.252205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.He A, Shen X, Ma Q, Cao J, von Gise A, Zhou P, Wang G, Marquez VE, Orkin SH, Pu WT. PRC2 directly methylates GATA4 and represses its transcriptional activity. Genes Dev 26: 37–42, 2012. doi: 10.1101/gad.173930.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.He H, Lehming N. Global effects of histone modifications. Brief Funct Genomics Proteomics 2: 234–243, 2003. doi: 10.1093/bfgp/2.3.234. [DOI] [PubMed] [Google Scholar]
  • 80.Hu H, Qian K, Ho MC, Zheng YG. Small molecule inhibitors of protein arginine methyltransferases. Expert Opin Investig Drugs 25: 335–358, 2016. doi: 10.1517/13543784.2016.1144747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Huang J, Perez-Burgos L, Placek BJ, Sengupta R, Richter M, Dorsey JA, Kubicek S, Opravil S, Jenuwein T, Berger SL. Repression of p53 activity by Smyd2-mediated methylation. Nature 444: 629–632, 2006. doi: 10.1038/nature05287. [DOI] [PubMed] [Google Scholar]
  • 82.Huang RC, Bonner J. Histone, a suppressor of chromosomal RNA synthesis. Proc Natl Acad Sci USA 48: 1216–1222, 1962. doi: 10.1073/pnas.48.7.1216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Inagawa M, Nakajima K, Makino T, Ogawa S, Kojima M, Ito S, Ikenishi A, Hayashi T, Schwartz RJ, Nakamura K, Obayashi T, Tachibana M, Shinkai Y, Maeda K, Miyagawa-Tomita S, Takeuchi T. Histone H3 lysine 9 methyltransferases, G9a and GLP are essential for cardiac morphogenesis. Mech Dev 130: 519–531, 2013. doi: 10.1016/j.mod.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 84.Jang Y, Wang C, Zhuang L, Liu C, Ge K. H3K4 methyltransferase activity is required for MLL4 protein stability. J Mol Biol 429: 2046–2054, 2017. doi: 10.1016/j.jmb.2016.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Jung H, Lee E, Kim I, Song JH, Kim GJ. Histone deacetylase inhibition has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy. Physiol Res 68: 727–737, 2019. doi: 10.33549/physiolres.934110. [DOI] [PubMed] [Google Scholar]
  • 86.Just S, Meder B, Berger IM, Etard C, Trano N, Patzel E, Hassel D, Marquart S, Dahme T, Vogel B, Fishman MC, Katus HA, Strähle U, Rottbauer W. The myosin-interacting protein SMYD1 is essential for sarcomere organization. J Cell Sci 124: 3127–3136, 2011. doi: 10.1242/jcs.084772. [DOI] [PubMed] [Google Scholar]
  • 87.Kalakonda N, Fischle W, Boccuni P, Gurvich N, Hoya-Arias R, Zhao X, Miyata Y, Macgrogan D, Zhang J, Sims JK, Rice JC, Nimer SD. Histone H4 lysine 20 monomethylation promotes transcriptional repression by L3MBTL1. Oncogene 27: 4293–4304, 2008. doi: 10.1038/onc.2008.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kim DH, Kim J, Kwon JS, Sandhu J, Tontonoz P, Lee SK, Lee S, Lee JW. Critical roles of the histone methyltransferase MLL4/KMT2D in murine hepatic steatosis directed by ABL1 and PPARγ2. Cell Reports 17: 1671–1682, 2016. doi: 10.1016/j.celrep.2016.10.023. [DOI] [PubMed] [Google Scholar]
  • 89.Kim J, Lee Y, Lu X, Song B, Fong KW, Cao Q, Licht JD, Zhao JC, Yu J. Polycomb- and methylation-independent roles of EZH2 as a transcription activator. Cell Reports 25: 2808–2820.e4, 2018. doi: 10.1016/j.celrep.2018.11.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kim JH, Yoo BC, Yang WS, Kim E, Hong S, Cho JY. The role of protein arginine methyltransferases in inflammatory responses. Mediators Inflamm 2016: 4028353, 2016. doi: 10.1155/2016/4028353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kim KH, Kim W, Howard TP, Vazquez F, Tsherniak A, Wu JN, Wang W, Haswell JR, Walensky LD, Hahn WC, Orkin SH, Roberts CW. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat Med 21: 1491–1496, 2015. doi: 10.1038/nm.3968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kim MS, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Chaerkady R, Madugundu AK, Kelkar DS, Isserlin R, Jain S, Thomas JK, Muthusamy B, Leal-Rojas P, Kumar P, Sahasrabuddhe NA, Balakrishnan L, Advani J, George B, Renuse S, Selvan LD, Patil AH, Nanjappa V, Radhakrishnan A, Prasad S, Subbannayya T, Raju R, Kumar M, Sreenivasamurthy SK, Marimuthu A, Sathe GJ, Chavan S, Datta KK, Subbannayya Y, Sahu A, Yelamanchi SD, Jayaram S, Rajagopalan P, Sharma J, Murthy KR, Syed N, Goel R, Khan AA, Ahmad S, Dey G, Mudgal K, Chatterjee A, Huang TC, Zhong J, Wu X, Shaw PG, Freed D, Zahari MS, Mukherjee KK, Shankar S, Mahadevan A, Lam H, Mitchell CJ, Shankar SK, Satishchandra P, Schroeder JT, Sirdeshmukh R, Maitra A, Leach SD, Drake CG, Halushka MK, Prasad TS, Hruban RH, Kerr CL, Bader GD, Iacobuzio-Donahue CA, Gowda H, Pandey A. A draft map of the human proteome. Nature 509: 575–581, 2014. doi: 10.1038/nature13302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kim S, Paik WK. Studies on the origin of epsilon-N-methyl-l-lysine in protein. J Biol Chem 240: 4629–4634, 1965. [PubMed] [Google Scholar]
  • 94.Kim SK, Jung I, Lee H, Kang K, Kim M, Jeong K, Kwon CS, Han YM, Kim YS, Kim D, Lee D. Human histone H3K79 methyltransferase DOT1L methyltransferase binds actively transcribing RNA polymerase II to regulate gene expression. J Biol Chem 287: 39698–39709, 2012. doi: 10.1074/jbc.M112.384057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kim SY, Morales CR, Gillette TG, Hill JA. Epigenetic regulation in heart failure. Curr Opin Cardiol 31: 255–265, 2016. doi: 10.1097/HCO.0000000000000276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kimura H. Histone modifications for human epigenome analysis. J Hum Genet 58: 439–445, 2013. doi: 10.1038/jhg.2013.66. [DOI] [PubMed] [Google Scholar]
  • 97.Kleefstra T, Brunner HG, Amiel J, Oudakker AR, Nillesen WM, Magee A, Geneviève D, Cormier-Daire V, van Esch H, Fryns JP, Hamel BC, Sistermans EA, de Vries BB, van Bokhoven H. Loss-of-function mutations in euchromatin histone methyl transferase 1 (EHMT1) cause the 9q34 subtelomeric deletion syndrome. Am J Hum Genet 79: 370–377, 2006. doi: 10.1086/505693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Krause CD, Yang ZH, Kim YS, Lee JH, Cook JR, Pestka S. Protein arginine methyltransferases: evolution and assessment of their pharmacological and therapeutic potential. Pharmacol Ther 113: 50–87, 2007. doi: 10.1016/j.pharmthera.2006.06.007. [DOI] [PubMed] [Google Scholar]
  • 99.Kroetz DN, Allen RM, Schaller MA, Cavallaro C, Ito T, Kunkel SL. Type I Interferon-induced epigenetic regulation of macrophages suppresses innate and adaptive immunity in acute respiratory viral infection. PLoS Pathog 11: e1005338, 2015. doi: 10.1371/journal.ppat.1005338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kwak YT, Guo J, Prajapati S, Park KJ, Surabhi RM, Miller B, Gehrig P, Gaynor RB. Methylation of SPT5 regulates its interaction with RNA polymerase II and transcriptional elongation properties. Mol Cell 11: 1055–1066, 2003. doi: 10.1016/S1097-2765(03)00101-1. [DOI] [PubMed] [Google Scholar]
  • 101.Lai B, Lee JE, Jang Y, Wang L, Peng W, Ge K. MLL3/MLL4 are required for CBP/p300 binding on enhancers and super-enhancer formation in brown adipogenesis. Nucleic Acids Res 45: 6388–6403, 2017. doi: 10.1093/nar/gkx234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Le Romancer M, Treilleux I, Leconte N, Robin-Lespinasse Y, Sentis S, Bouchekioua-Bouzaghou K, Goddard S, Gobert-Gosse S, Corbo L. Regulation of estrogen rapid signaling through arginine methylation by PRMT1. Mol Cell 31: 212–221, 2008. doi: 10.1016/j.molcel.2008.05.025. [DOI] [PubMed] [Google Scholar]
  • 103.LeBlanc SE, Konda S, Wu Q, Hu YJ, Oslowski CM, Sif S, Imbalzano AN. Protein arginine methyltransferase 5 (Prmt5) promotes gene expression of peroxisome proliferator-activated receptor γ2 (PPARγ2) and its target genes during adipogenesis. Mol Endocrinol 26: 583–597, 2012. doi: 10.1210/me.2011-1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lee JE, Wang C, Xu S, Cho YW, Wang L, Feng X, Baldridge A, Sartorelli V, Zhuang L, Peng W, Ge K. H3K4 mono- and di-methyltransferase MLL4 is required for enhancer activation during cell differentiation. eLife 2: e01503, 2013. doi: 10.7554/eLife.01503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lee JH, Gao C, Peng G, Greer C, Ren S, Wang Y, Xiao X. Analysis of transcriptome complexity through RNA sequencing in normal and failing murine hearts. Circ Res 109: 1332–1341, 2011. doi: 10.1161/CIRCRESAHA.111.249433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lee ST, Li Z, Wu Z, Aau M, Guan P, Karuturi RK, Liou YC, Yu Q. Context-specific regulation of NF-κB target gene expression by EZH2 in breast cancers. Mol Cell 43: 798–810, 2011. doi: 10.1016/j.molcel.2011.08.011. [DOI] [PubMed] [Google Scholar]
  • 107.Lee YH, Coonrod SA, Kraus WL, Jelinek MA, Stallcup MR. Regulation of coactivator complex assembly and function by protein arginine methylation and demethylimination. Proc Natl Acad Sci USA 102: 3611–3616, 2005. doi: 10.1073/pnas.0407159102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lehnertz B, Northrop JP, Antignano F, Burrows K, Hadidi S, Mullaly SC, Rossi FM, Zaph C. Activating and inhibitory functions for the histone lysine methyltransferase G9a in T helper cell differentiation and function. J Exp Med 207: 915–922, 2010. doi: 10.1084/jem.20100363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Li C, Yu L, Xue H, Yang Z, Yin Y, Zhang B, Chen M, Ma H. Nuclear AMPK regulated CARM1 stabilization impacts autophagy in aged heart. Biochem Biophys Res Commun 486: 398–405, 2017. doi: 10.1016/j.bbrc.2017.03.053. [DOI] [PubMed] [Google Scholar]
  • 110.Li H, Ilin S, Wang W, Duncan EM, Wysocka J, Allis CD, Patel DJ. Molecular basis for site-specific read-out of histone H3K4me3 by the BPTF PHD finger of NURF. Nature 442: 91–95, 2006. doi: 10.1038/nature04802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Li H, Xu J, Bian YH, Rotllant P, Shen T, Chu W, Zhang J, Schneider M, Du SJ. Smyd1b_tv1, a key regulator of sarcomere assembly, is localized on the M-line of skeletal muscle fibers. PLoS One 6: e28524, 2011. doi: 10.1371/journal.pone.0028524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Li Y, Li Z, Zhang C, Li P, Wu Y, Wang C, Bond Lau W, Ma XL, Du J. Cardiac fibroblast-specific activating transcription factor 3 protects against heart failure by suppressing MAP2K3-p38 signaling. Circulation 135: 2041–2057, 2017. doi: 10.1161/CIRCULATIONAHA.116.024599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ling BM, Bharathy N, Chung TK, Kok WK, Li S, Tan YH, Rao VK, Gopinadhan S, Sartorelli V, Walsh MJ, Taneja R. Lysine methyltransferase G9a methylates the transcription factor MyoD and regulates skeletal muscle differentiation. Proc Natl Acad Sci USA 109: 841–846, 2012. doi: 10.1073/pnas.1111628109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Liu C, Wang C, Wang K, Liu L, Shen Q, Yan K, Sun X, Chen J, Liu J, Ren H, Liu H, Xu Z, Hu S, Xu D, Fan Y. SMYD3 as an oncogenic driver in prostate cancer by stimulation of androgen receptor transcription. J Natl Cancer Inst 105: 1719–1728, 2013. doi: 10.1093/jnci/djt304. [DOI] [PubMed] [Google Scholar]
  • 115.Liu CF, Tang WH. Epigenetics in cardiac hypertrophy and heart failure. JACC Basic Transl Sci 4: 976–993, 2019. doi: 10.1016/j.jacbts.2019.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Liu M, He A, Chu J, Chen C, Zhang S, He Y, Tao W, Lu M, Hua M, Ju W, Fang Z. Serum N1-methylnicotinamide is associated with left ventricular systolic dysfunction in Chinese. Sci Rep 8: 8581, 2018. doi: 10.1038/s41598-018-26956-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Liu X, Wang L, Li H, Lu X, Hu Y, Yang X, Huang C, Gu D. Coactivator-associated arginine methyltransferase 1 targeted by miR-15a regulates inflammation in acute coronary syndrome. Atherosclerosis 233: 349–356, 2014. doi: 10.1016/j.atherosclerosis.2014.01.039. [DOI] [PubMed] [Google Scholar]
  • 118.Liu Y, Morley M, Brandimarto J, Hannenhalli S, Hu Y, Ashley EA, Tang WH, Moravec CS, Margulies KB, Cappola TP, Li M; MAGNet Consortium . RNA-Seq identifies novel myocardial gene expression signatures of heart failure. Genomics 105: 83–89, 2015. doi: 10.1016/j.ygeno.2014.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lo Sardo A, Altamura S, Pegoraro S, Maurizio E, Sgarra R, Manfioletti G. Identification and characterization of new molecular partners for the protein arginine methyltransferase 6 (PRMT6). PLoS One 8: e53750, 2013. [Erratum in PLoS One 8, 2013.] doi: 10.1371/journal.pone.0053750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Longman MR, Ranieri A, Avkiran M, Snabaitis AK. Regulation of PP2AC carboxylmethylation and cellular localisation by inhibitory class G-protein coupled receptors in cardiomyocytes. PLoS One 9: e86234, 2014. doi: 10.1371/journal.pone.0086234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lupien M, Eeckhoute J, Meyer CA, Krum SA, Rhodes DR, Liu XS, Brown M. Coactivator function defines the active estrogen receptor α cistrome. Mol Cell Biol 29: 3413–3423, 2009. doi: 10.1128/MCB.00020-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Maicas M, Vázquez I, Alis R, Marcotegui N, Urquiza L, Cortés-Lavaud X, Cristóbal I, García-Sánchez MA, Odero MD. The MDS and EVI1 complex locus (MECOM) isoforms regulate their own transcription and have different roles in the transformation of hematopoietic stem and progenitor cells. Biochim Biophys Acta Gene Regul Mech 1860: 721–729, 2017. doi: 10.1016/j.bbagrm.2017.03.007. [DOI] [PubMed] [Google Scholar]
  • 123.Mäkelä PH, Eskola J, Peltola H, Takala AK, Käyhty H. Clinical experience with Haemophilus influenzae type b conjugate vaccines. Pediatrics 85: 651–653, 1990. [PubMed] [Google Scholar]
  • 124.Mazur PK, Reynoird N, Khatri P, Jansen PW, Wilkinson AW, Liu S, Barbash O, Van Aller GS, Huddleston M, Dhanak D, Tummino PJ, Kruger RG, Garcia BA, Butte AJ, Vermeulen M, Sage J, Gozani O. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature 510: 283–287, 2014. doi: 10.1038/nature13320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.McBride AE, Silver PA. State of the arg: protein methylation at arginine comes of age. Cell 106: 5–8, 2001. doi: 10.1016/S0092-8674(01)00423-8. [DOI] [PubMed] [Google Scholar]
  • 126.Michaud-Levesque J, Richard S. Thrombospondin-1 is a transcriptional repression target of PRMT6. J Biol Chem 284: 21338–21346, 2009. doi: 10.1074/jbc.M109.005322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Migliori V, Müller J, Phalke S, Low D, Bezzi M, Mok WC, Sahu SK, Gunaratne J, Capasso P, Bassi C, Cecatiello V, De Marco A, Blackstock W, Kuznetsov V, Amati B, Mapelli M, Guccione E. Symmetric dimethylation of H3R2 is a newly identified histone mark that supports euchromatin maintenance. Nat Struct Mol Biol 19: 136–144, 2012. doi: 10.1038/nsmb.2209. [DOI] [PubMed] [Google Scholar]
  • 128.Miranda TB, Webb KJ, Edberg DD, Reeves R, Clarke S. Protein arginine methyltransferase 6 specifically methylates the nonhistone chromatin protein HMGA1a. Biochem Biophys Res Commun 336: 831–835, 2005. doi: 10.1016/j.bbrc.2005.08.179. [DOI] [PubMed] [Google Scholar]
  • 129.Mo R, Rao SM, Zhu YJ. Identification of the MLL2 complex as a coactivator for estrogen receptor alpha. J Biol Chem 281: 15714–15720, 2006. doi: 10.1074/jbc.M513245200. [DOI] [PubMed] [Google Scholar]
  • 130.Mohan M, Herz HM, Takahashi YH, Lin C, Lai KC, Zhang Y, Washburn MP, Florens L, Shilatifard A. Linking H3K79 trimethylation to Wnt signaling through a novel Dot1-containing complex (DotCom). Genes Dev 24: 574–589, 2010. doi: 10.1101/gad.1898410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Moore KE, Gozani O. An unexpected journey: lysine methylation across the proteome. Biochim Biophys Acta 1839: 1395–1403, 2014. doi: 10.1016/j.bbagrm.2014.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Morales CR, Li DL, Pedrozo Z, May HI, Jiang N, Kyrychenko V, Cho GW, Kim SY, Wang ZV, Rotter D, Rothermel BA, Schneider JW, Lavandero S, Gillette TG, Hill JA. Inhibition of class I histone deacetylases blunts cardiac hypertrophy through TSC2-dependent mTOR repression. Sci Signal 9: ra34, 2016. doi: 10.1126/scisignal.aad5736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Mueller D, Bach C, Zeisig D, Garcia-Cuellar MP, Monroe S, Sreekumar A, Zhou R, Nesvizhskii A, Chinnaiyan A, Hess JL, Slany RK. A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood 110: 4445–4454, 2007. doi: 10.1182/blood-2007-05-090514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Mukherjee N, Cardenas E, Bedolla R, Ghosh R. SETD6 regulates NF-κB signaling in urothelial cell survival: Implications for bladder cancer. Oncotarget 8: 15114–15125, 2017. doi: 10.18632/oncotarget.14750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Murray K. The occurrence of epsilon-N-methyl lysine in histones. Biochemistry 3: 10–15, 1964. doi: 10.1021/bi00889a003. [DOI] [PubMed] [Google Scholar]
  • 136.Naeem H, Cheng D, Zhao Q, Underhill C, Tini M, Bedford MT, Torchia J. The activity and stability of the transcriptional coactivator p/CIP/SRC-3 are regulated by CARM1-dependent methylation. Mol Cell Biol 27: 120–134, 2007. doi: 10.1128/MCB.00815-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ng SB, Bigham AW, Buckingham KJ, Hannibal MC, McMillin MJ, Gildersleeve HI, Beck AE, Tabor HK, Cooper GM, Mefford HC, Lee C, Turner EH, Smith JD, Rieder MJ, Yoshiura K, Matsumoto N, Ohta T, Niikawa N, Nickerson DA, Bamshad MJ, Shendure J. Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nat Genet 42: 790–793, 2010. doi: 10.1038/ng.646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Nguyen AT, Xiao B, Neppl RL, Kallin EM, Li J, Chen T, Wang DZ, Xiao X, Zhang Y. DOT1L regulates dystrophin expression and is critical for cardiac function. Genes Dev 25: 263–274, 2011. doi: 10.1101/gad.2018511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Nguyen AT, Zhang Y. The diverse functions of Dot1 and H3K79 methylation. Genes Dev 25: 1345–1358, 2011. doi: 10.1101/gad.2057811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Nishioka K, Chuikov S, Sarma K, Erdjument-Bromage H, Allis CD, Tempst P, Reinberg D. Set9, a novel histone H3 methyltransferase that facilitates transcription by precluding histone tail modifications required for heterochromatin formation. Genes Dev 16: 479–489, 2002. doi: 10.1101/gad.967202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Okuno K, Akiyama Y, Shimada S, Nakagawa M, Tanioka T, Inokuchi M, Yamaoka S, Kojima K, Tanaka S. Asymmetric dimethylation at histone H3 arginine 2 by PRMT6 in gastric cancer progression. Carcinogenesis 40: 15–26, 2019. doi: 10.1093/carcin/bgy147. [DOI] [PubMed] [Google Scholar]
  • 142.Paik WK, Kim S. Protein methylase I. Purification and properties of the enzyme. J Biol Chem 243: 2108–2114, 1968. [PubMed] [Google Scholar]
  • 143.Paik WK, Kim S. Solubilization and partial purification of protein methylase 3 from calf thymus nuclei. J Biol Chem 245: 6010–6015, 1970. [PubMed] [Google Scholar]
  • 144.Paik WK, Paik DC, Kim S. Historical review: the field of protein methylation. Trends Biochem Sci 32: 146–152, 2007. doi: 10.1016/j.tibs.2007.01.006. [DOI] [PubMed] [Google Scholar]
  • 145.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 24: 9630–9645, 2004. doi: 10.1128/MCB.24.21.9630-9645.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Papait R, Serio S, Pagiatakis C, Rusconi F, Carullo P, Mazzola M, Salvarani N, Miragoli M, Condorelli G. Histone methyltransferase G9a is required for cardiomyocyte homeostasis and hypertrophy. Circulation 136: 1233–1246, 2017. doi: 10.1161/CIRCULATIONAHA.117.028561. [DOI] [PubMed] [Google Scholar]
  • 147.Park CY, Pierce SA, von Drehle M, Ivey KN, Morgan JA, Blau HM, Srivastava D. skNAC, a Smyd1-interacting transcription factor, is involved in cardiac development and skeletal muscle growth and regeneration. Proc Natl Acad Sci USA 107: 20750–20755, 2010. doi: 10.1073/pnas.1013493107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Park G, Gong Z, Chen J, Kim JE. Characterization of the DOT1L network: implications of diverse roles for DOT1L. Protein J 29: 213–223, 2010. doi: 10.1007/s10930-010-9242-8. [DOI] [PubMed] [Google Scholar]
  • 149.Petrossian TC, Clarke SG. Uncovering the human methyltransferasome. Mol Cell Proteomics 10: 000976, 2011. doi: 10.1074/mcp.M110.000976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Piao L, Kang D, Suzuki T, Masuda A, Dohmae N, Nakamura Y, Hamamoto R. The histone methyltransferase SMYD2 methylates PARP1 and promotes poly(ADP-ribosyl)ation activity in cancer cells. Neoplasia 16: 257–264.e2, 2014. doi: 10.1016/j.neo.2014.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Pursani V, Bhartiya D, Tanavde V, Bashir M, Sampath P. Transcriptional activator DOT1L putatively regulates human embryonic stem cell differentiation into the cardiac lineage. Stem Cell Res Ther 9: 97, 2018. doi: 10.1186/s13287-018-0810-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Rada M, Vasileva E, Lezina L, Marouco D, Antonov AV, Macip S, Melino G, Barlev NA. Human EHMT2/G9a activates p53 through methylation-independent mechanism. Oncogene 36: 922–932, 2017. doi: 10.1038/onc.2016.258. [DOI] [PubMed] [Google Scholar]
  • 153.Rajabi M, Kassiotis C, Razeghi P, Taegtmeyer H. Return to the fetal gene program protects the stressed heart: a strong hypothesis. Heart Fail Rev 12: 331–343, 2007. doi: 10.1007/s10741-007-9034-1. [DOI] [PubMed] [Google Scholar]
  • 154.Rasmussen TL, Ma Y, Park CY, Harriss J, Pierce SA, Dekker JD, Valenzuela N, Srivastava D, Schwartz RJ, Stewart MD, Tucker HO. Smyd1 facilitates heart development by antagonizing oxidative and ER stress responses. PLoS One 10: e0121765, 2015. doi: 10.1371/journal.pone.0121765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Rea S, Eisenhaber F, O’Carroll D, Strahl BD, Sun ZW, Schmid M, Opravil S, Mechtler K, Ponting CP, Allis CD, Jenuwein T. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature 406: 593–599, 2000. doi: 10.1038/35020506. [DOI] [PubMed] [Google Scholar]
  • 156.Rezai-Zadeh N, Zhang X, Namour F, Fejer G, Wen YD, Yao YL, Gyory I, Wright K, Seto E. Targeted recruitment of a histone H4-specific methyltransferase by the transcription factor YY1. Genes Dev 17: 1019–1029, 2003. doi: 10.1101/gad.1068003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Romano JD, Michaelis S. Topological and mutational analysis of Saccharomyces cerevisiae Ste14p, founding member of the isoprenylcysteine carboxyl methyltransferase family. Mol Biol Cell 12: 1957–1971, 2001. doi: 10.1091/mbc.12.7.1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Roqueta-Rivera M, Esquejo RM, Phelan PE, Sandor K, Daniel B, Foufelle F, Ding J, Li X, Khorasanizadeh S, Osborne TF. SETDB2 links glucocorticoid to lipid metabolism through Insig2a regulation. Cell Metab 24: 474–484, 2016. doi: 10.1016/j.cmet.2016.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Ruthenburg AJ, Li H, Patel DJ, Allis CD. Multivalent engagement of chromatin modifications by linked binding modules. Nat Rev Mol Cell Biol 8: 983–994, 2007. doi: 10.1038/nrm2298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Saddic LA, West LE, Aslanian A, Yates JR III, Rubin SM, Gozani O, Sage J. Methylation of the retinoblastoma tumor suppressor by SMYD2. J Biol Chem 285: 37733–37740, 2010. doi: 10.1074/jbc.M110.137612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Sanulli S, Justin N, Teissandier A, Ancelin K, Portoso M, Caron M, Michaud A, Lombard B, da Rocha ST, Offer J, Loew D, Servant N, Wassef M, Burlina F, Gamblin SJ, Heard E, Margueron R. Jarid2 methylation via the PRC2 complex regulates H3K27me3 deposition during cell differentiation. Mol Cell 57: 769–783, 2015. doi: 10.1016/j.molcel.2014.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Schliehe C, Flynn EK, Vilagos B, Richson U, Swaminathan S, Bosnjak B, Bauer L, Kandasamy RK, Griesshammer IM, Kosack L, Schmitz F, Litvak V, Sissons J, Lercher A, Bhattacharya A, Khamina K, Trivett AL, Tessarollo L, Mesteri I, Hladik A, Merkler D, Kubicek S, Knapp S, Epstein MM, Symer DE, Aderem A, Bergthaler A. The methyltransferase Setdb2 mediates virus-induced susceptibility to bacterial superinfection. Nat Immunol 16: 67–74, 2015. doi: 10.1038/ni.3046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Schneider R, Bannister AJ, Weise C, Kouzarides T. Direct binding of INHAT to H3 tails disrupted by modifications. J Biol Chem 279: 23859–23862, 2004. doi: 10.1074/jbc.C400151200. [DOI] [PubMed] [Google Scholar]
  • 164.Schurter BT, Koh SS, Chen D, Bunick GJ, Harp JM, Hanson BL, Henschen-Edman A, Mackay DR, Stallcup MR, Aswad DW. Methylation of histone H3 by coactivator-associated arginine methyltransferase 1. Biochemistry 40: 5747–5756, 2001. doi: 10.1021/bi002631b. [DOI] [PubMed] [Google Scholar]
  • 165.Sen R, Lencer E, Geiger EA, Jones KL, Shaikh TH, Artinger K. The role of KMT2D and KDM6A in cardiac development: A cross-species analysis in humans, mice, and zebrafish. bioRxiv April 4, 2020. doi: 10.1101/2020.04.03.024646. [DOI]
  • 166.Sgarra R, Diana F, Bellarosa C, Dekleva V, Rustighi A, Toller M, Manfioletti G, Giancotti V. During apoptosis of tumor cells HMGA1a protein undergoes methylation: identification of the modification site by mass spectrometry. Biochemistry 42: 3575–3585, 2003. doi: 10.1021/bi027338l. [DOI] [PubMed] [Google Scholar]
  • 167.Shailesh H, Zakaria ZZ, Baiocchi R, Sif S. Protein arginine methyltransferase 5 (PRMT5) dysregulation in cancer. Oncotarget 9: 36705–36718, 2018. doi: 10.18632/oncotarget.26404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Shen EC, Henry MF, Weiss VH, Valentini SR, Silver PA, Lee MS. Arginine methylation facilitates the nuclear export of hnRNP proteins. Genes Dev 12: 679–691, 1998. doi: 10.1101/gad.12.5.679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Shi X, Kachirskaia I, Yamaguchi H, West LE, Wen H, Wang EW, Dutta S, Appella E, Gozani O. Modulation of p53 function by SET8-mediated methylation at lysine 382. Mol Cell 27: 636–646, 2007. doi: 10.1016/j.molcel.2007.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Shinkai Y, Tachibana M. H3K9 methyltransferase G9a and the related molecule GLP. Genes Dev 25: 781–788, 2011. doi: 10.1101/gad.2027411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Sims RJ III, Rojas LA, Beck DB, Bonasio R, Schüller R, Drury WJ III, Eick D, Reinberg D. The C-terminal domain of RNA polymerase II is modified by site-specific methylation. Science 332: 99–103, 2011. doi: 10.1126/science.1202663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Sweet ME, Cocciolo A, Slavov D, Jones KL, Sweet JR, Graw SL, Reece TB, Ambardekar AV, Bristow MR, Mestroni L, Taylor MR. Transcriptome analysis of human heart failure reveals dysregulated cell adhesion in dilated cardiomyopathy and activated immune pathways in ischemic heart failure. BMC Genomics 19: 812, 2018. doi: 10.1186/s12864-018-5213-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Tachibana M, Sugimoto K, Fukushima T, Shinkai Y. Set domain-containing protein, G9a, is a novel lysine-preferring mammalian histone methyltransferase with hyperactivity and specific selectivity to lysines 9 and 27 of histone H3. J Biol Chem 276: 25309–25317, 2001. doi: 10.1074/jbc.M101914200. [DOI] [PubMed] [Google Scholar]
  • 174.Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, Fukuda M, Takeda N, Niida H, Kato H, Shinkai Y. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev 16: 1779–1791, 2002. doi: 10.1101/gad.989402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tan X, Rotllant J, Li H, De Deyne P, Du SJ. SmyD1, a histone methyltransferase, is required for myofibril organization and muscle contraction in zebrafish embryos. Proc Natl Acad Sci USA 103: 2713–2718, 2006. doi: 10.1073/pnas.0509503103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Tee WW, Pardo M, Theunissen TW, Yu L, Choudhary JS, Hajkova P, Surani MA. Prmt5 is essential for early mouse development and acts in the cytoplasm to maintain ES cell pluripotency. Genes Dev 24: 2772–2777, 2010. doi: 10.1101/gad.606110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Thienpont B, Aronsen JM, Robinson EL, Okkenhaug H, Loche E, Ferrini A, Brien P, Alkass K, Tomasso A, Agrawal A, Bergmann O, Sjaastad I, Reik W, Roderick HL. The H3K9 dimethyltransferases EHMT1/2 protect against pathological cardiac hypertrophy. J Clin Invest 127: 335–348, 2017. doi: 10.1172/JCI88353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT, Haugen E, Sheffield NC, Stergachis AB, Wang H, Vernot B, Garg K, John S, Sandstrom R, Bates D, Boatman L, Canfield TK, Diegel M, Dunn D, Ebersol AK, Frum T, Giste E, Johnson AK, Johnson EM, Kutyavin T, Lajoie B, Lee BK, Lee K, London D, Lotakis D, Neph S, Neri F, Nguyen ED, Qu H, Reynolds AP, Roach V, Safi A, Sanchez ME, Sanyal A, Shafer A, Simon JM, Song L, Vong S, Weaver M, Yan Y, Zhang Z, Zhang Z, Lenhard B, Tewari M, Dorschner MO, Hansen RS, Navas PA, Stamatoyannopoulos G, Iyer VR, Lieb JD, Sunyaev SR, Akey JM, Sabo PJ, Kaul R, Furey TS, Dekker J, Crawford GE, Stamatoyannopoulos JA. The accessible chromatin landscape of the human genome. Nature 489: 75–82, 2012. doi: 10.1038/nature11232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tillinger A, Bruderova V, Kubovcakova L, Zeman M, Kopacek J, Novakova M, Kvetnansky R, Krizanova O. Gene expression of the phenylethanolamine N-methyltransferase is differently modulated in cardiac atria and ventricles. Gen Physiol Biophys 25: 355–364, 2006. [PubMed] [Google Scholar]
  • 180.Torrano J, Al Emran A, Hammerlindl H, Schaider H. Emerging roles of H3K9me3, SETDB1 and SETDB2 in therapy-induced cellular reprogramming. Clin Epigenetics 11: 43, 2019. doi: 10.1186/s13148-019-0644-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Tracy C, Warren JS, Szulik M, Wang L, Garcia J, Makaju A, Russell K, Miller M, Franklin S. The Smyd family of methyltransferases: role in cardiac and skeletal muscle physiology and pathology. Curr Opin Physiol 1: 140–152, 2018. doi: 10.1016/j.cophys.2017.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Tsutsui T, Fukasawa R, Shinmyouzu K, Nakagawa R, Tobe K, Tanaka A, Ohkuma Y. Mediator complex recruits epigenetic regulators via its two cyclin-dependent kinase subunits to repress transcription of immune response genes. J Biol Chem 288: 20955–20965, 2013. doi: 10.1074/jbc.M113.486746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Van Aller GS, Reynoird N, Barbash O, Huddleston M, Liu S, Zmoos AF, McDevitt P, Sinnamon R, Le B, Mas G, Annan R, Sage J, Garcia BA, Tummino PJ, Gozani O, Kruger RG. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation. Epigenetics 7: 340–343, 2012. doi: 10.4161/epi.19506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Van Laarhoven PM, Neitzel LR, Quintana AM, Geiger EA, Zackai EH, Clouthier DE, Artinger KB, Ming JE, Shaikh TH. Kabuki syndrome genes KMT2D and KDM6A: functional analyses demonstrate critical roles in craniofacial, heart and brain development. Hum Mol Genet 24: 4443–4453, 2015. doi: 10.1093/hmg/ddv180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Vershinin Z, Feldman M, Chen A, Levy D. PAK4 methylation by SETD6 promotes the activation of the Wnt/β-catenin pathway. J Biol Chem 291: 6786–6795, 2016. doi: 10.1074/jbc.M115.697292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Vieira FQ, Costa-Pinheiro P, Almeida-Rios D, Graça I, Monteiro-Reis S, Simões-Sousa S, Carneiro I, Sousa EJ, Godinho MI, Baltazar F, Henrique R, Jerónimo C. SMYD3 contributes to a more aggressive phenotype of prostate cancer and targets cyclin D2 through H4K20me3. Oncotarget 6: 13644–13657, 2015. doi: 10.18632/oncotarget.3767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Voelkel T, Andresen C, Unger A, Just S, Rottbauer W, Linke WA. Lysine methyltransferase Smyd2 regulates Hsp90-mediated protection of the sarcomeric titin springs and cardiac function. Biochim Biophys Acta 1833: 812–822, 2013. doi: 10.1016/j.bbamcr.2012.09.012. [DOI] [PubMed] [Google Scholar]
  • 188.Waldmann T, Izzo A, Kamieniarz K, Richter F, Vogler C, Sarg B, Lindner H, Young NL, Mittler G, Garcia BA, Schneider R. Methylation of H2AR29 is a novel repressive PRMT6 target. Epigenetics Chromatin 4: 11, 2011. doi: 10.1186/1756-8935-4-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Wang JJ, Rau C, Avetisyan R, Ren S, Romay MC, Stolin G, Gong KW, Wang Y, Lusis AJ. Genetic dissection of cardiac remodeling in an isoproterenol-induced heart failure mouse model. PLoS Genet 12: e1006038, 2016. doi: 10.1371/journal.pgen.1006038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Wang L, Pal S, Sif S. Protein arginine methyltransferase 5 suppresses the transcription of the RB family of tumor suppressors in leukemia and lymphoma cells. Mol Cell Biol 28: 6262–6277, 2008. doi: 10.1128/MCB.00923-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Wang YR, Xu NX, Wang J, Wang XM. Kabuki syndrome: review of the clinical features, diagnosis and epigenetic mechanisms. World J Pediatr 15: 528–535, 2019. doi: 10.1007/s12519-019-00309-4. [DOI] [PubMed] [Google Scholar]
  • 192.Warren JS, Tracy CM, Miller MR, Makaju A, Szulik MW, Oka SI, Yuzyuk TN, Cox JE, Kumar A, Lozier BK, Wang L, Llana JG, Sabry AD, Cawley KM, Barton DW, Han YH, Boudina S, Fiehn O, Tucker HO, Zaitsev AV, Franklin S. Histone methyltransferase Smyd1 regulates mitochondrial energetics in the heart. Proc Natl Acad Sci USA 115: E7871–E7880, 2018. doi: 10.1073/pnas.1800680115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Wood K, Tellier M, Murphy S. DOT1L and H3K79 methylation in transcription and genomic stability. Biomolecules 8: 11, 2018. doi: 10.3390/biom8010011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Wu J, Xu W. Histone H3R17me2a mark recruits human RNA polymerase-associated factor 1 complex to activate transcription. Proc Natl Acad Sci USA 109: 5675–5680, 2012. doi: 10.1073/pnas.1114905109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Wysocka J, Allis CD, Coonrod S. Histone arginine methylation and its dynamic regulation. Front Biosci 11: 344–355, 2006. doi: 10.2741/1802. [DOI] [PubMed] [Google Scholar]
  • 196.Yang F, Sun L, Li Q, Han X, Lei L, Zhang H, Shang Y. SET8 promotes epithelial-mesenchymal transition and confers TWIST dual transcriptional activities. EMBO J 31: 110–123, 2012. doi: 10.1038/emboj.2011.364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Yang KC, Yamada KA, Patel AY, Topkara VK, George I, Cheema FH, Ewald GA, Mann DL, Nerbonne JM. Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support. Circulation 129: 1009–1021, 2014. doi: 10.1161/CIRCULATIONAHA.113.003863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Yao L, Li Y, Du F, Han X, Li X, Niu Y, Ren S, Sun Y. Histone H4 Lys 20 methyltransferase SET8 promotes androgen receptor-mediated transcription activation in prostate cancer. Biochem Biophys Res Commun 450: 692–696, 2014. doi: 10.1016/j.bbrc.2014.06.033. [DOI] [PubMed] [Google Scholar]
  • 199.Yeates TO. Structures of SET domain proteins: protein lysine methyltransferases make their mark. Cell 111: 5–7, 2002. doi: 10.1016/S0092-8674(02)01010-3. [DOI] [PubMed] [Google Scholar]
  • 200.Yin B, Yu F, Wang C, Li B, Liu M, Ye L. Epigenetic control of mesenchymal stem cell fate decision via histone methyltransferase Ash1l. Stem Cells 37: 115–127, 2019. doi: 10.1002/stem.2918. [DOI] [PubMed] [Google Scholar]
  • 201.Yu J, Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, Wang X, Wu L, Li J, Hu M, Gong Y, Cheng H, Laxman B, Vellaichamy A, Shankar S, Li Y, Dhanasekaran SM, Morey R, Barrette T, Lonigro RJ, Tomlins SA, Varambally S, Qin ZS, Chinnaiyan AM. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell 17: 443–454, 2010. doi: 10.1016/j.ccr.2010.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Yuan W, Xu M, Huang C, Liu N, Chen S, Zhu B. H3K36 methylation antagonizes PRC2-mediated H3K27 methylation. J Biol Chem 286: 7983–7989, 2011. doi: 10.1074/jbc.M110.194027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Zaidi S, Choi M, Wakimoto H, Ma L, Jiang J, Overton JD, Romano-Adesman A, Bjornson RD, Breitbart RE, Brown KK, Carriero NJ, Cheung YH, Deanfield J, DePalma S, Fakhro KA, Glessner J, Hakonarson H, Italia MJ, Kaltman JR, Kaski J, Kim R, Kline JK, Lee T, Leipzig J, Lopez A, Mane SM, Mitchell LE, Newburger JW, Parfenov M, Pe’er I, Porter G, Roberts AE, Sachidanandam R, Sanders SJ, Seiden HS, State MW, Subramanian S, Tikhonova IR, Wang W, Warburton D, White PS, Williams IA, Zhao H, Seidman JG, Brueckner M, Chung WK, Gelb BD, Goldmuntz E, Seidman CE, Lifton RP. De novo mutations in histone-modifying genes in congenital heart disease. Nature 498: 220–223, 2013. doi: 10.1038/nature12141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Zhang B, Dong S, Zhu R, Hu C, Hou J, Li Y, Zhao Q, Shao X, Bu Q, Li H, Wu Y, Cen X, Zhao Y. Targeting protein arginine methyltransferase 5 inhibits colorectal cancer growth by decreasing arginine methylation of eIF4E and FGFR3. Oncotarget 6: 22799–22811, 2015. doi: 10.18632/oncotarget.4332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Zhang CL, McKinsey TA, Chang S, Antos CL, Hill JA, Olson EN. Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell 110: 479–488, 2002. doi: 10.1016/S0092-8674(02)00861-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Zhang QJ, Liu ZP. Histone methylations in heart development, congenital and adult heart diseases. Epigenomics 7: 321–330, 2015. doi: 10.2217/epi.14.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Zhang T, Braun T, Zhou Y. Prmt5 differentiates developmental vs regenerative myogenesis. Oncotarget 6: 35153–35154, 2015. doi: 10.18632/oncotarget.6207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Zhang T, Günther S, Looso M, Künne C, Krüger M, Kim J, Zhou Y, Braun T. Prmt5 is a regulator of muscle stem cell expansion in adult mice. Nat Commun 6: 7140, 2015. doi: 10.1038/ncomms8140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Zhao HY, Zhang YJ, Dai H, Zhang Y, Shen YF. CARM1 mediates modulation of Sox2. PLoS One 6: e27026, 2011. doi: 10.1371/journal.pone.0027026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Zhao JC, Yu J, Runkle C, Wu L, Hu M, Wu D, Liu JS, Wang Q, Qin ZS, Yu J. Cooperation between Polycomb and androgen receptor during oncogenic transformation. Genome Res 22: 322–331, 2012. doi: 10.1101/gr.131508.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Zhao Q, Rank G, Tan YT, Li H, Moritz RL, Simpson RJ, Cerruti L, Curtis DJ, Patel DJ, Allis CD, Cunningham JM, Jane SM. PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing. Nat Struct Mol Biol 16: 304–311, 2009. doi: 10.1038/nsmb.1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Zhao X, Jankovic V, Gural A, Huang G, Pardanani A, Menendez S, Zhang J, Dunne R, Xiao A, Erdjument-Bromage H, Allis CD, Tempst P, Nimer SD. Methylation of RUNX1 by PRMT1 abrogates SIN3A binding and potentiates its transcriptional activity. Genes Dev 22: 640–653, 2008. doi: 10.1101/gad.1632608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Zhu L. Functional characterization of Smyd1, a methyltransferase essential for heart and skeletal muscle development. 2006, https://repositories.lib.utexas.edu/handle/2152/2967.

Articles from American Journal of Physiology - Heart and Circulatory Physiology are provided here courtesy of American Physiological Society

RESOURCES