Histone lysine methylation and congenital heart disease: From bench to bedside (Review)

  • Authors:
    • Xin Yi
    • Xuejun Jiang
    • Xiaoyan Li
    • Ding-Sheng Jiang
  • View Affiliations

  • Published online on: August 30, 2017     https://doi.org/10.3892/ijmm.2017.3115
  • Pages: 953-964
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Histone post-translational modifications (PTM) as one of the key epigenetic regulatory mechanisms that plays critical role in various biological processes, including regulating chromatin structure dynamics and gene expression. Histone lysine methyltransferase contributes to the establishment and maintenance of differential histone methylation status, which can recognize histone methylated sites and build an association between these modifications and their downstream processes. Recently, it was found that abnormalities in the histone lysine methylation level or pattern may lead to the occurrence of many types of cardiovascular diseases, such as congenital heart disease (CHD). In order to provide new theoretical basis and targets for the treatment of CHD from the view of developmental biology and genetics, this review discusses and elaborates on the association between histone lysine methylation modifications and CHD.

1. Introduction

Congenital heart disease (CHD) involves the anatomical structure abnormality caused by the formation of obstacles or the abnormal development of the heart and great vessels during the period of embryonic development, or a group of congenital malformations with actual or potential influence on heart function arising from the open tunnels which should have self-closed after childbirth. CHD mostly occurs during 2–8 weeks after impregnation, and it is the most common cardiovascular malformation affecting children; it severely affects the health of infants and young children (14). At present, CHD is regarded as a multigene disease influenced by the environment and heredity; however, the pathogenesis of the disease and the underlying molecular mechanisms and interations between genes remain unclear (58). Heart development is a very complex process, involving the expression of numerous genes at specific time points in the process of embryonic development, and it is regulated by many transcription factors (9,10). The realization of this process is not only determined by gene sequences, but is also largely generated by the transformation of epigenetics. In addition, an increasing number of studies have found that children with CHD have an extremely low occurrence rate of gene mutation, which can only explain a small number of CHD cases, as there is no pathogenic gene transformation for the majority of CHD cases (1113). Some recent studies have found that 'epigenetics' may very likely participate or play an important role in the occurrence of CHD (1416).

Epigenetics suggests that DNA sequence undergoes no transformation, but the gene expression occurs by heritable transformation, which is the other heritable material transformations in the cells apart from the heritage information and with stable heredity in the process of cell development and proliferation (1719). Epigenetics is mainly the reversible and heritable transformation of gene function with the DNA sequences of the nucleus unchanged, and these transformations include DNA modifications (such as methylation), various kinds of histone modifications (such as methylation, acetylation, ubiquitination and phosphorylation), chromatin remodeling, non-encoding RNA regulation (2026). In the field of 'epigenetics', by transforming the chromatin structure with the help of related enzymes and interacting with other regulator protein, histone modifications regulate gene expression, and influence occurrence and development of diseases, which is also known as 'the second heritage code', mainly including methylation, acetylation, phosphorylation, ubiquitin, adenosine, small ubiquitin related modification, ADP ribosylation and proline isomerization (21,27,28). Histone methylation is one of the most common histone modifications, and mainly includes arginine methylation and lysine methylation (2931). Comparatively speaking, histone methylation modification transforms loosen or agglutination state of chromatin mainly through influencing the affinity of histone and DNA, regulates gene expression by influencing the affinity of other transcription factors and structural gene promoters. Thus, it can be seen that histone methylation modification has the gene expression regulatory function similar to the DNA genetic code, and plays an important role in the process of growth and development (3234).

In recent years, studies have shown that histone lysine methylation is not only closely related to tumor occurrence and development, developmental defects, senile dementia, cardiac hypertrophy and other clinical diseases, but also participates in the occurrence of CHD, influencing the development of heart structure and CHD candidate gene expression (3545). For this reason, this review aims to summarize the new progress of CHD epigenetic mechanism research from the aspect of histone lysine methylation modification, in order to provide a new scientific basis for the prevention and treatment of CHD.

2. Histone methylation

Histone methylation involves the methylation occurring at histone H3 and the N-terminal of H4 arginine (Arg) or lysine (Lys) residues, catalyzed by histone methyltransferase. The function of histone methylation is mainly reflected in heterochromatin forming, genomic imprinting, X chromosome inactivation and transcriptional control (4652). Apart from histone methyltransferase, the histone demethylase is also found (53,54). At first, it was considered that the histone methylation effect was stable and irreversible; however, the existence of methyltransferase renders the process of histone methylation more dynamical.

Modification sites of histone methylation

Histone methylation can occur on of Lys and Arg histone residues. Lysine residues can be single, double and triple methylated, while arginine residues can be single and double methylated (5557). This varying degree of methylation largely increases the complexity of histone modifications and regulator gene expressions (29,58). Methylation action sites are in the N atoms at the side chains of Lys and Arg. Lys locus 4, 9, 27, 36 and 79 of histone H3 (H3K4, H3K9, H3K27, H3K36 and H3K79), Lys locus 20 of histone H4 (H4K20), Arg locus 2, 17, 26 of histone H3 (H3R2, H3R17 and H3R26) and Arg locus 3 of histone H4 (H4R3) are the common loci of methylation (Fig. 1). Studies have shown that histone Arg methylation is a comparatively dynamical mark, and is related to gene activation; however, Arg methylation lacking H3 and H4 is related to gene silencing (5961). On the contrary, Lys methylation seems to be a stable mark of gene expression regulation. For example, H3K4 methylation is related to gene activation, while H3K9 and H3K27 methylation are related to gene silencing (38,6266). In addition, H4K20 methylation is related to gene silencing, H3K36 and H3K79 are related to gene activation (6772).

Related enzymes of histone methylation

Arginine methylation occurs at the equal locus of histone H3R2/R17/R26 and H4R3, and plays a promoting effect on gene expression (60). When the methylation process occurs, histone arginine methyltransferase or protein arginine methyltransferase (PRMT), as the collaborative activity factor, is recruited into the promoter region of the target gene and thus activates gene expression (7375). The PRMT of catalyzing histone arginine methyltransferase includes two categories: the first type of PRMT being catalyzed to form single methylation arginine and unsymmetrical double methylation arginine, and the second type of PRMT being catalyzed to form single methylation arginine and symmetrical double methylation arginine (76,77). The family of PRMT includes PRMTl, PRMT3, RMTI/HMT, PRMT4/CAMRl and PRMT5. K4, K9, K27, K36, K79 of histone H3 and K20 of H4 all can be methylated. The methylation level is regulated and executed by a type of methyltransferase with SET structural domain with highly conserved nucleus and the pre-SET and post-SET structural domain with abundant cysteine sequence (31,78,79). The denomination of SET structural domain is constituted with 3 initial letters of 3 genes from expressing SET structural domain found at the earliest, which are Su(var)3–9, E(z) and Trx (80). Furthermore, histone demethylase can catalyze histone lysine to demethylation, and then affect the level of methylation. At present, the demethylase of histone mainly has two types: lysine specific demethylase (LSD1) and Jmjc domain-containing histone demethylase (JHDM) (81,82).

3. Histone lysine methylation and heart development

Epigenetic modification, including methylation and acetylization plays an important role in the regulation of gene expression. Studies have indicated that histone methylation can reduce or increase its affinity for charged DNA, loosen or tighten the chromatin structure to affect the accessibility and interactions between transcriptional factors and DNA templates, ultimately promote or inhibit gene expression (83,84). Recent evidence supports a prominent role for histone methylation in normal and aberrant heart development (8587). A recent study demonstrated that changes in histone methylation levels in histone H3 that binds with critical promoter parts of the ssTnI gene can cause the corresponding changes in ssTnI gene expression, which indicated that histone methylation was involved in the regulation of myofibril gene expression in the heart during development (88). Additionally, Hand2 and Irx4 transcription factors have been shown to be reduced in SMYD1-deficient mice, suggesting that SMYD1-mediated histone methylation is necessary for the expression of these essential cardiac transcription factors (89). Hence, these findings illustrate the pervasive roles of histone methylation in the process of heart development.

4. Related enzymes of histone lysine methylation and congenital heart disease

CHD is the most common type of birth defect, manifesting as obstacles in the process of embryonic heart or blood vessel development, which may result in the morphology, structure, function and metabolic abnormalities of heart and blood vessels (2,3,5). According to the statistics, CHD has become the first reason for birth defects and the main cause of perinatal death and death in children (2). The causes of CHD are not yet completely clear; however, most scholars consider that many types of CHD are caused by a single gene mutation and chromosome aberration, and most types of CHD belong to complex genetic diseases, which are caused by the interaction between genetic factors and environmental factors (90,91). Studies have shown that histone lysine methylation modification as part of the epigenetic regulation, is involved in the development of heart and blood vessels, which is also one of the causes of CHD (92,93). The level of histone lysine methylation is determined by the balance of histone methylation and demethylation, which is a process by which methyl groups are transferred onto or removed from the amino acids of histone proteins. Histone methyltransferase and histone demethylase catalyze histone methylation and demethylation, respectively. In most cases, under the action of the methylation and demethylation, the histone tails relax or surround, which can loosen or inhibit DNA of transcription factors so as to turn the genes in DNA 'off' and 'on', resulting in the normal or aberrant expression of related genes and leading to abnormal heart development. To understand the research progress of histone lysine methylation and CHD, we summarize the known histone lysine modifying enzymes which regulates CHD in Table I.

Table I

Known histone lysine modifying enzymes involved in congenital heart diseases.

Table I

Known histone lysine modifying enzymes involved in congenital heart diseases.

Abnormal patternSpeciesRelated cardiac phenotype
Modifiers of H3K4
 Trithotax group proteins
  MLL2DeficiencyMiceEmbryonic lethal
DeficiencyMouse embryonic stem cellsAbnormal proliferation and cardiac lineage differentiation
MutationHumansAtrial/ventricular septal defects, aortic coarctation
 SMYDs
  SMYD1DeficiencyZebrafishImmature myofibers, non-beating heart
DeficiencyMiceEmbryonic lethal, hypoplastic right ventricle
  SMYD3DeficiencyZebrafishPericardial edema, abnormal expression of heart-chamber markers
 T-box transcription factors
  TBX1MutationHumansDiGeorge syndrome, double outlet right ventricle, ventricular septal defect
  TBX2MutationHumansVentricular septal defects
  TBX3MutationMice, humansCardiac conduction dysplasia
  TBX5DeficiencyMiceDefective epicardial and coronary blood vessel formation
MutationHumansHolt-Oram syndrome
  TBX18DeficiencyMiceDefective epicardium and coronary vessels
  TBX20 Mutation/deficiencyMiceFailed heart looping, defective chamber differentiation, cardiomyopathy, arrhythmias
MutationHumansSeptal defects, valvulogenesis defects, cardiomyopathy
  DPF3Mutation or deletionZebrafishIncomplete cardiac looping, defective ventricular contractility and muscular fibers
  PTIPDeficiencyMiceAbnormal cardiac conduction system, ventricular arrhythmia
  SETD7DeficiencyZebrafishDevelopmental heart edema
  LSD1DeficiencyMiceVentricular septal defects, salt-sensitive hypertension
  BCORMutationHumans Oculo-facio-cardio-dental (OFCD) syndrome
 Modifiers of H3K9
  G9a and GLPDeficiencyMiceEmbryonic lethality, atrioventricular septal defects
  EHMT1 Mutation/deficiencyHumansChromosome 9q subtelomere deletion syndrome with atrial/ventriculap septal defect
  Blimp-1/PRDMDeficiencyMiceVentricular septal defect and persistent arterial trunk
  Jarid2 Mutation/deficiencyMiceVentricular/atrial septal defect, double-outlet right ventricle, dilated atria
 Modifiers of H3K27
  Polycomb group proteins
  PRC1 Mutation/deficiencyMiceBone dysplasia and heart development defects
  Ezh2 Mutation/deficiencyMiceDouble outlet right ventricle, persistent truncus arteriosus, ventricular septal defects, atrial septal defects, atrioventricular canal defects and enlarged aortic valves, postnatal myocardial pathology
  UTXDeficiencyMiceEmbryonic lethality, reduced somite counts, heart malformation
DeficiencyMouse embryonic stem cellsFailed to develop heart-like rhythmic contractions
  Jmjd3DeficiencyMiceEmbryonic lethality
DeficiencyMouse embryonic stem cellsImpaired mesoderm and subsequent endothelial and cardiac differentiation
 Modifiers of H3K36
  NSD1 Mutation/deficiencyHumansSotos syndrome
  WHSC1DeficiencyMicePerinatal lethal, atrial and ventricular septal defects
MutationHumansWolf-Hirschhorn sydrome
  Jmjd5DeficiencyMiceEmbryonic lethal
 Modifiers of H3K79
  DOT1LDeficiencyMiceEmbryonic lethal, yolk sac angiogenesis defects, cardiac dilatation, cardiomyocyte death, systolic dysfunction, conduction abnormalities
 Modifiers of H4K20
  NSD1 Mutation/deficiencyHumansSotos syndrome, ventricular/atrial septal defect, patent ductus arteriosus
H3K4 methylation and CHD
Trithotax group (TrxG) proteins

During heart development, several cardiac progenitor pools give rise to diverse cell lineages, such as cardiomyocytes, vascular smooth muscle cells, fibroblasts that form the connective tissues and endothelial cells of the endocardium. The heart expresses many epigenetic factors, including both histone modifying proteins and chromatin remodelers. Among the epigenetic factors, TrxG proteins are special family of chromatin factors that regulate developmental gene expression in the heart (94). TrxG proteins function in multi-subunit complexes, three TrxG complexes, the MLL complex, the BRM/BAF complex and a supercomplex, and have been purified in mammalian cells (95). TrxG proteins are evolutionarily conserved H3K4 methyltransferases that maintain the transactivation states of lineage-specific genes during embryonic development. Multiple TrxG genes are normally expressed in the mouse heart. Due to the essential function of TrxG genes, constitutive knockouts of key TrxG genes often result in lethality during early embryogenesis before cardiac phenotypes can be analyzed. The differentiation of mouse embryonic stem cells (ESCs) toward mesodermal and endodermal lineages is severely altered and, in particular, the cardiac lineage differentiation of ESCs is completely abolished in the absence of MLL2, a TrxG member. Moreover, the expression of core cardiac transcription factors and the levels of H3K4 trimethylation of these cardiac-specific promoters are significantly decreased by the loss of MLL2 (96). Taken together, these results reveal a critical role for MLL2 in the proliferation and cardiac lineage differentiation of mouse ESCs, and provide critical insight not only into the novel role of the TrxG protein in cardiac development, but also into their clinical significance in related CHD.

SMYDs

SET and myeloid, nervy and DEAF-1 (MYND) domain-containing proteins (SMYDs), including SMYD1–5, have two functional protein domains, SET (mediates histone lysine methylation activity) and MYND (mediates the protein-protein interaction and binds to DNA motifs) domains (80). SET-MYND-domain 1 (SMYD1/BOP) encodes an evolutionary conserved histone methyltransferase containing a split SET domain interrupted by a MYND domain, which includes two members SMYD1a and SMYD1b, can catalyze H3K4 methylation (97). The expression of SMYD1 is restricted to skeletal and cardiac muscles in humans, fish, chickens and mice. There is evidence to indicate that SMYD1 plays important roles in cardiac differentiation, development and function (98). The global knockdown of SMYD1a and SMYD1b in zebrafish has been shown to result in the disruption of myofibril formation and an absence of beating of the heart. Molecular and cellular experiments showed that myofibers in embryos in which SMYD1 was knocked down appeared as immature myofibers with centrally located nuclei and disorganized myofibrils, indicating that SMYD1 played a critical role in myofibers maturation and contraction (97). Conventional null SMYD1 mice die in utero around embryonic day 10.5 (E10.5) due to heart defects, including disrupted maturation of ventricular cardiomyocytes and malformation of the right ventricle (99). However, Diehl et al recently reported that SET and MYND domain containing 2 (SMYD2), is capable of H3K4 methylation when bound to Hsp90a and acts on non-histone targets by inhibiting the functional activity of p53 via methylation of p53, lysine 370, which was differentially expressed during cardiac development with highest expression in the neonatal heart (100). To elucidate the functional role of SMYD2 in the heart, they generated knockout mice harboring a cardiomyocyte-specific deletion of SMYD2 and performed histological, functional and molecular experiments. Unexpectedly, cardiac deletion of SMYD2 was dispensable for proper morphological and functional development of the murine heart (100). H3K4 methyltransferase SMYD3 is highly expressed within developing zebrafish heart and knockdown of it led to severe defects such as pericardial edema and abnormal expression of three heart-chamber markers in cardiac morphogenesis (101). These results indicate that SMYD3 plays an important role in heart development and its proper functioning is essential for normal heart morphogenesis during development.

T-box (TBX) transcription factors

TBX transcription factors share a highly conserved DNA-binding domain and play critical roles in embryonic development (102). Six members of TBX family (TBX1, TBX18 and TBX20 of the TBX1 subfamily, and TBX2, TBX3 and TBX5 of the TBX2 subfamily) are required for the cardiac morphogenesis in mammals (103). TBX1 interacts with H3K4 methyltransferase to enhance its H3K4 monomethylation status through T-box, regulates expression of related genes by epigenetic patterns (104). TBX1 mutation can lead to DiGeorge syndrome (DGS), which is the most common microdeletion syndrome, and is characterized by congenital cardiac, craniofacial and immune system abnormalities (105). Additionally, Pan et al reported that a novel heterozygous TBX1 mutation, p.Q277X, was identified in an index patient with double outlet right ventricle and ventricular septal defect (106). TBX2 gene is expressed in the myocardium of the atrioventricular canal, outflow tract and inflow tract and plays a critical role in heart chamber formation (107). The genomic deletion and duplication of TBX2 gene have been found to be associated with ventricular septal defects (108). The evolutionary conserved TBX3 gene encodes T-box transcription factors and locus forms a CTCF independent autonomous regulatory domain with multiple combinatorial regulatory elements, which plays crucial roles in the development and homeostasis of the cardiac conduction system in humans and mice (109). Previous studies have found that TBX5 is expressed in the proepicardial organ or septum transversum, which is required for the normal development of proepicardium/proepicardial organ cells, as well as proper epicardial formation and maturation (110). Additionally, TBX5 deficiency delays epicardiac cell attachment to the myocardium and impairs production of epicardial-derived cells and their migration into the myocardium, and results in abnormal coronary vasculogenesis and murine ischemic cardiomyopathy (111). Clinical studies have shown that Holt-Oram syndrome is caused by mutations in TBX5, which is a human inherited disorder and manifests as left pericardium agenesis and anomalous coronary arteries along with ventricular septal defects (112114). These findings all demonstrate that TBX5 is essential for epicardial development in hearts and establishment of the coronary vasculature. Similar to TBX5, TBX18 is also highly expressed in proepicardial cells and proepicardium, TBX18-deficient proepicardium produces an epicardium and coronary vasculature with structural and functional defects, and that remodeling of the disorganized subepicardial plexus in TBX18-deficient hearts produced a mature coronary artery network with fewer distributing conduit vessels and smaller lumen profiles, which indicates that TBX18 plays critical role in coronary development (115). However, TBX20 is necessary in heart development by regulating cardiomyocyte proliferation and regional specification and formation of cardiac chambers and valves; TBX20 mutations in mice can result in the failure of heart looping, developmental arrest, and the lack of chamber differentiation, and loss of TBX20 in mice leads to cardiomyopathy with associated arrhythmias and death (116,117). More seriously, mutations in human TBX20 result in cardiac malformations including septal defects, double outlet right ventricle and cardiomyopathy (118,119). These findings provide novel insight into the molecular mechanism underlying CHD and suggest potential implications for the development of novel preventive and therapeutic strategies for CHD.

DPF3

DPF3 is a member of the highly conserved d4 protein family, which is characterized by a double PHD finger in the C-terminal and has two splice variants DPF3a and DPF3b in human and mice (120). In the process of embryonic development, DPF3 is expressed both in heart and somites of mouse, chicken and zebrafish, which is important epigenetic regulation factor for heart and muscle development by associated with the BAF chromatin remodeling complex and binds methylated lysine residues of H3K4 (121). Previous studies have found that DPF3 mutation or deletion leads to incomplete cardiac looping, attenuated ventricular contractility and disassembled muscular fibers caused by the transcriptional deregulation of structural and regulatory proteins in the heart, which all demonstrate that DPF3 is responsible for cardiac development imbalance, ventricular septal defect and other cardiac disorders (121).

Pax transactivation domain interacting protein (PTIP)

PTIP is an essential cofactor for H3K4me by KMT2C/D, which is encoded by the Paxip1 gene and is essential for embryonic development in mice and flies (122124). As a critical component of the KMT2C/D complex, the loss of PTIP leads to reduced levels of H3K4me3 in whole embryos, ESCs and Drosophila larvae (125,126). Stein et al demonstrated that temporal and tissue-specific deletion of PTIP reduces H3K4 methylation level and alters the transcriptional program in nondividing cardiomyocytes. It is suggested that a role for KMT2 complexes not just in establishing active chromatin domains but also in the maintenance of the differentiated state over time. Furthermore, the loss of PTIP-mediated H3K4me results in significant changes in the physiology of the cardiomyocytes, suggesting that PTIP deletion is the direct cause of premature ventricular beats, a harbinger of lethal ventricular arrhythmias in nondividing cardiomyocytes (42,127).

SET domain containing protein 7 (SETD7)

SETD7 also termed as SET7/9, is another type of histone lysine methyltransferase and only has SET domain for methyltransferase activity, but not MYND domain, which is initially discovered as a specific methyltransferase for nonmethylated H3K4. Tao et al found that the knockdown of SETD7 showed the defects in skeletal muscle formation and myofibril structures in a zebrafish developmental model (128). To examine the function of SETD7 in heart development, Kim et al firstly demonstrated that SETD7 was highly expressed in developing zebrafish heart and knockdown of it led to severe defects in cardiac morphogenesis such as developmental heart edema. Furthermore, the double knockdown of SMYD3 and SETD7 caused synergistic defects in heart development. Similar to the knockdown effect, the overexpression of SETD7 also caused the heart morphogenesis defects in zebrafish (85). These results indicate that the histone modifying enzyme, SETD7, plays an important role during heart development and its proper functioning is essential for normal heart morphogenesis during development.

Lysine-specific demethylase 1 (LSD1)

LSD1 (also known as AOF2/KDM1A), is a member of a group of enzymes with lysine specific demethylase activity. LSD1 performs enzymatic activity toward di- and monomethyl H3K4 and H3K9 respectively; the specificity for H3K9 arises when LSD1 binds to the androgen receptor, resulting in a shift of its activity from H3K4 (129). LSD1 interacts with proteins mostly through the tower domain, an extended helical structure. Furthmore, there is evidence to indicate that LSD1-interacting proteins can regulate the activity and specificity of LSD1 in developmental processes (130,131). Nicholson et al found that mice homozygous for a hypomorphic LSD1 allele exhibit a failure to survive after birth perinatally due to heart defects, with the majority of animals suffering from ventricular septal defects (132). Therefore, the above-mentioned studies thereby illuminate a novel role for LSD1 in the development of the mammalian heart.

BCL-6 corepressor (BCOR)

It was found that BCOR inhibited gene transcription by interacting with BCL-6, and BCOR mutation resulted in abnormal activation of AP-2a, which was a key factor that mediated the differentiation of bone marrow mesenchymal stem cells (MSCs) (133). Fan et al also pointed out that BCOR recruited a histone demethylase JHDM1B to the target gene promoter, resulting in the demethylation of H3K4me3 and H3K36me2 and transcription repression of genes; however, BCL-6 mutation may impair the recruitment of JHDM1B to chromatin, resulted in increased methylation levels of H3K4 and H3K36 (133). Abnormal histone methylation due to BCOR mutation may affect BCL-6 binding to the AP-2a promoter, causing aberrant activation of gene and resulting in the in occurrence of oculo-facio-cardio-dental (OFCD) syndrome, which is a rare genetic disorder characterized by teeth with extremely long roots, and craniofacial, eye and congenital cardiac abnormalities include septal defect and mitral valve defect abnormalities (133135). On the whole, it was identified that BCOR mutation affected heart development and AP-2α played a role in congenial heart defects associated with OFCD patients, and indicated that BCOR may be a novel target for diagnostic and treatment strategies of OFCD syndrome.

H3K9 methylation and CHD
G9a and GLP

G9a and GLP are known as major H3K9 mono-and di-methyltransferases and contribute to transcriptional silencing, which play critical biological roles in various cells and tissues. For example, G9a and GLP are indispensable for mouse early development; G9a or GLP knockout mice exhibit embryonic lethality around E9.5 due to severe growth defects (38,136,137). In order to clarify the roles of G9a and GLP in cardiac development, Inagawa et al analyzed the phenotypes of cardiomyocyte specific GLP knockout and G9a knockdown mice, it was shown that the H3K9me2 level decreased markedly in the nuclei of the cardiomyocytes of these mice, and the mice exhibited neonatal lethality and severe cardiac defects characterized by atrioventricular septal defects (138). These data indicated that G9a and GLP were required for H3K9me2 in cardiomyocytes and performed an essential role in normal morphogenesis of the atrioventricular septum through regulation of the size of the atrioventricular cushion.

Euchromatin histone methyl transferase 1 (EHMT1)

EHMT1 is located on chromosome 9 and encodes Eu-HMTase1, which is a type of methyltransferase found in the region of the chromatin region. EHMT1 could specifically modify H3K9 methylation and thus inhibit the activity of related genes. Some studies have found EHMT1 mutation or deletion is the main cause of chromosome 9q subtelomere deletion syndrome (9qSTDS), approximately half of affected individuals have congenital heart defects primarily characterized by atrial septal defect or ventricular septal defect (139,140).

Blimp-1/PRDM

The PR/SET domain zinc-finger transcriptional repressor Blimp-1/PRDM is initially cloned as a negative regulator of key transcription factors expression and encoded by PRDM1, which contains PR/SET domain in N-terminal and C2H2 zinc finger structure in C-terminal, and thus plays essential roles in primordial germ cell specification, placental, heart, and forelimb development, plasma cell differentiation, and T-cell homeostasis through regulating DNA binding, nuclear input and recruitment of histone modifying enzymes (141). Blimp-1/PRDM causes H3K4 methylation by recruiting of histone modifying enzymes, which inhibits relevant genes expression and thus regulates the development of embryos. In the development of embryos, Blimp-1/PRDM deficiency caused serious cardiac defects including ventricular septal defect and persistent arterial trunk (141,142).

Jarid2

Jarid2, also termed as Jumonji (jmj), the founding member of the Jumonji family, all of which contain the JmjC domain that generally confers histone demethylase activities, which can catalyze H3K9 methylation and function as a transcriptional repressor and to interact with other nuclear factors. Lee et al found that the Jarid2 homozygous mouse embryos show heart malformations, including ventricular septal defect, noncompaction of the ventricular wall, double-outlet right ventricle, and dilated atria; furthermore, expression of Jarid2 in the interventricular septum, ventricular wall and outflow tract, which is correlated well with the locations of defects observed in the hearts of mutant mice. These results indicate that Jarid2 plays an important role in embryonic heart development (143). At the molecular level, Kim et al demonstrated that Jarid2 can inhibit the proliferation of cardiomyocytes, and inhibit the expression of atrial natriuretic peptide (ANP) by repressing the interaction with transcription factor Nkx2.5 and GATA4 (144). Other studies have also found that mutations or deletions of Jarid2 could increase H3K9 and H3K36 methylation level, resulting in the abnormal expression of development-related genes and thus inducing atrial septal defect or ventricular septal defect and other cardiac defects (145,146).

H3K27 methylation and CHD
Polycomb group proteins

Polycomb group proteins are key regulators of gene expression during development and differentiation, silencing genes via regulation of the chromatin structure, which act in complexes that have specific catalytic functions important for transcriptional repression. In mammals, 2 major Polycomb group complexes exist: polycomb repressive complex 1 (PRC1) and PRC2. Whereas PRC1 ubiquitinates histone H2A on Lys119,1 PRC2 catalyzes dimethylation and trimethylation of H3K27, generating H3K27me2/3 (147). Weston et al pointed out that Rae28 protein, the core component of PRC1, which made PRC1 bind to H3K27me3 and then formed chromatin tight structure to prevent the occurrence of transcription. Rae28 mutation or deletion mice tend to perform bone dysplasia and heart development defects (148). However, Ezh2, the major histone methyltransferase of PRC2, trimethylates H3K27 and is essential for embryonic development (149). Delgado-Olguín et al have shown that Ezh2 stabilizes cardiac gene expression and prevents cardiac pathology, but Ezh2 deletion in cardiac progenitors causes postnatal myocardial pathology and destabilizes cardiac gene expression, which suggests that Ezh2 is essential for stable postnatal heart gene expression and homeostasis (150). Furthermore, Chen et al demonstrated that a variety of cardiovascular structural malformations were observed in the Ezh2 mutant mice, including double outlet right ventricle, persistent truncus arteriosus, membranous and muscular ventricular septal defects, atrial septal defects, atrioventricular canal defects and enlarged aortic valves, which defined an indispensible role of Ezh2 in normal cardiovascular development (151).

Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX)

Histone demethylase UTX, also known as KDM6A, that specifically targets the repressive H3K27me3 modification plays an important role in the activation of 'bivalent' genes in response to specific developmental cues. Welstead et al showed that UTX-deficient embryos had reduced somite counts, neural tube closure defects and heart malformation that presented between E9.5 and E13.5 (152). Other studies have also found that UTX-deficient ESCs failed to develop heart-like rhythmic contractions under a cardiac differentiation condition; UTX deficient mice exhibited severe defects in heart development and embryonic lethality; these data establish that UTX is required for heart development acts as a critical switch to activate the cardiac developmental program (153,154).

Jumonji domain-containing protein 3 (Jmjd3)

Jmjd3 (KDM6B), another H3K27 demethylase, functions redundantly with UTX. Jmjd3 is induced and participates in Hox gene expression during development, neuronal differentiation and inflammation, and recent data suggest that Jmjd3 inhibits reprogramming by inducing cellular senescence (155). Jmjd3 deficient mice showed embryonic lethality before E6.5, suggesting a crucial role of Jmjd3 in early embryonic development (156,157). The ablation of Jmjd3 in mouse ESCs impaired mesoderm and subsequent endothelial and cardiac differentiation. These results clarify that Jmjd3 is necessary for mesoderm differentiation and cardiovascular lineage commitment (158).

H3K36 methylation and CHD
Nuclear receptor SET domain containing gene 1 (NSD1)

NSD1 is a structure containing the SET domain proteins, with specific H3K36 and H4K20 methyltransferase activity, which is associated with Sotos syndrome by haploinsufficiency (159,160). A very frequent feature among Sotos syndrome patients with intragenic mutations was the presence of congenital heart defects or heart conduction defects, including isolated atrial septal defect, atrial septal defect in association with other structural abnormalities (patent ductus arteriosus; aortic valve dysplasia; ventricular septal defect and aortic coarctation) (161). This evidence indicates that NSD1 may be a cause of a higher prevalence of congenital heart defect in Sotos syndrome patients, which may be a novel target of diagnosis and treatment strategy for Sotos syndrome.

Wolf-Hirschhorn syndrome candidate 1 (WHSC1). WHSC1 contains AWS-SET-PostSET domain structure with methyltransferase activity, is encoded by Wolf-Hirschhorn syndrome related regional gene, is homologous with H3K36 specific methyltransferase Set2 in yeast, which can make H3K36 mono- di-, tri-methylated. WHSC1 regulates the expression of related genes through interaction with other transcription factors such as Nkx2.5, in embryonic heart, WHSC1 is found to interact with Nkx2.5 to repress Pdgfra expression (162). Loss of WHSC1 resulted in reduction of H3K36me3 at the Pdgfra locus and upregulation of Pdgfra. Loss-of-WHSC1 mice are perinatal lethal with significant growth retardation and die within 10 days after birth. Deletion of a critical region of human chromosome 4q16.3 containing WSHC1 gene is associated with craniofacial malformations, growth retardation, learning disability and congenital heart defects. WHSC1-null mice display a variety of atrial and ventricular septal defects that manifest those in Wolf-Hirschhorn sydrome patients (163).

Jumonji C domain-containing 5 (Jmjd5)

Jmjd5 is a histone demethylase that specifically removes methyl moieties from dimethylated H3K36 and exerts a pro-proliferative effect on a large of cells. Strong Jmjd5 expression was observed only in the yolk sac at E8.5, Jmjd5 was robustly expressed in E10.5 embryos at several sites, including the heart and eye, which indicated that Jmjd5 may play an important role in heart and eye development. Jmjd5 deficiency mice embryos showed delayed development already at E8.5, embryonic lethal around E10 and were actively resorbed at E10.5 (164,165). Collectively, these data indicate that Jmjd5 is essential during embryonal development including heart development.

H3K79 methylation and CHD

H3K79 methylation is related to gene activation and DNA damage repair. Histone methylation occurs at H3K79 is catalyzed by yeast disruptor of telomeric silencing (DOT1) and its mammalian homolog, DOT1L. DOT1 is a kind of evolutionary highly conservative histone methyltransferase, which does not contain the SET domain structure, can be specific to different methylation levels in the H3K79. Compared with other histone lysine methylation, in yeast, DOT1 activity is positively regulated during transcription elongation through Rad6-Bre1 mono-ubiquitination of H2B (166). Recently, loss-of-function experiments revealed a critical role of DOT1L during mouse embryogenesis, as germline Dot1L knockout caused lethality at E10.5 with growth impairment, yolk sac angiogenesis defects, and cardiac dilation (167). In addition, cardiac-specific knockout of DOT1L resulted in increased mortality rate with chamber dilation, increased cardiomyocyte cell death, systolic dysfunction and conduction abnormalities (168). These phenotypes mimic those exhibited in patients with dilated cardiomyopathy. Interestingly, Nguyen et al demonstrated that DOT1L is downregulated in idiopathic DCM patient samples compared with normal controls (168). Therefore, the above studies not only establish a critical role for DOT1L-mediated H3K79 methylation in cardiomyocyte function, but also open new avenues for the diagnosis and treatment of CHD.

H4K20 methylation and CHD

H4K20 can be catalyzed to different forms of monomethylation, dimethylation and trimethylation, PR-SET7 can only single methylate H4K20, but double and triple methylation of H4K20 are catalyzed by two other methyltransferases SUV4-20h1 and SUV4-20h2 (169). It is shown that H4K20 methylation is related to transcription silence, H4K20me3 plays a vital role in the regulation of DNA damage but not directly regulates the expression of genes (170). In addition, Tatton-Brown and Rahman have found that NSD1 is a protein containing the SET domain structure, which with specific H4K20 and H3K36 methyltransferase activity (171). NSD1 mutation or deficiency is the main cause of Sotos syndrome which with a high incidence of CHD characterized by ventricular septal defect, atrial septal defects and patent ductus arteriosus (159,160).

5. Conclusion and prospection

Histone modification is the important content of epigenetics, which is not only showed as directly regulating gene expressions, but also influencing gene activity through DNA modification because of its intimate touching with DNA. However, single histone modification usually cannot come into effect individually, and it determines together the gene expression of genome through collaborative effect of multiple histone modifications; but, yet so far, about the mechanism of histone modification, especially the specific mechanism of regulation of histone modification is not quite clear. Therefore, although the histone lysine methylation modification is related to CHD, further intensive research is still needed to illuminate relationship at the level of molecule. At the same time, besides the genetic factors of CHD pathogenesis, there still exist outer environmental factors, so that the complexity of diagnosing and treatment of diseases has been increased. However, it is clear that the comprehensive and meticulous investigation of histone lysine methylation modifications may provide new insight and understaning into the exploration of CHD pathogenesis and targeted prevention.

Acknowledgments

We apologize to the colleagues whose studies could not be cited in this review due to space limitations. This study was supported by grants from the Fundamental Research Funds for the Central Universities (no. 2042016kf0074), the National Natural Science Foundation of China (no. 81170307) and Specialized Research Fund for the Doctoral Program of Higher Education (no. 20120141110013).

References

1 

Campos CM, Zanardo EA, Dutra RL, Kulikowski LD and Kim CA: Investigation of copy number variation in children with conotruncal heart defects. Arq Bras Cardiol. 104:24–31. 2015.

2 

Barros TL, Dias MdeJ and Nina RV: Congenital cardiac disease in childhood x socioeconomic conditions: A relationship to be considered in public health? Rev Bras Cir Cardiovasc. 29:448–454. 2014.PubMed/NCBI

3 

Steffensen TS and Spicer DE: Congenital coronary artery anomalies for the pathologist. Fetal Pediatr Pathol. 33:268–288. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Sanchez-Castro M, Pichon O, Briand A, Poulain D, Gournay V, David A and Le Caignec C: Disruption of the SEMA3D gene in a patient with congenital heart defects. Hum Mutat. 36:30–33. 2015. View Article : Google Scholar

5 

Vecoli C, Pulignani S, Foffa I and Andreassi MG: Congenital heart disease: The crossroads of genetics, epigenetics and environment. Curr Genomics. 15:390–399. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Gittenberger-de Groot AC, Calkoen EE, Poelmann RE, Bartelings MM and Jongbloed MR: Morphogenesis and molecular considerations on congenital cardiac septal defects. Ann Med. 46:640–652. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wang X, Li P, Chen S, Xi L, Guo Y, Guo A and Sun K: Influence of genes and the environment in familial congenital heart defects. Mol Med Rep. 9:695–700. 2014. View Article : Google Scholar

8 

Lalani SR and Belmont JW: Genetic basis of congenital cardiovascular malformations. Eur J Med Genet. 57:402–413. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Kathiriya IS, Nora EP and Bruneau BG: Investigating the transcriptional control of cardiovascular development. Circ Res. 116:700–714. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Meganathan K, Sotiriadou I, Natarajan K, Hescheler J and Sachinidis A: Signaling molecules, transcription growth factors and other regulators revealed from in-vivo and in-vitro models for the regulation of cardiac development. Int J Cardiol. 183:117–128. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Li Y, Klena NT, Gabriel GC, Liu X, Kim AJ, Lemke K, Chen Y, Chatterjee B, Devine W, Damerla RR, et al: Global genetic analysis in mice unveils central role for cilia in congenital heart disease. Nature. 521:520–524. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Andersen TA, Troelsen KL and Larsen LA: Of mice and men: Molecular genetics of congenital heart disease. Cell Mol Life Sci. 71:1327–1352. 2014. View Article : Google Scholar :

13 

Yuan S, Zaidi S and Brueckner M: Congenital heart disease: Emerging themes linking genetics and development. Curr Opin Genet Dev. 23:352–359. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Serra-Juhé C, Cuscó I, Homs A, Flores R, Torán N and Pérez-Jurado LA: DNA methylation abnormalities in congenital heart disease. Epigenetics. 10:167–177. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Zhang QJ and Liu ZP: Histone methylations in heart development, congenital and adult heart diseases. Epigenomics. 7:321–330. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Chang CP and Bruneau BG: Epigenetics and cardiovascular development. Annu Rev Physiol. 74:41–68. 2012. View Article : Google Scholar

17 

D'Urso A and Brickner JH: Mechanisms of epigenetic memory. Trends Genet. 30:230–236. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Becker PB and Workman JL: Nucleosome remodeling and epigenetics. Cold Spring Harb Perspect Biol. 5:a0179052013. View Article : Google Scholar : PubMed/NCBI

19 

Krishnakumar R and Blelloch RH: Epigenetics of cellular reprogramming. Curr Opin Genet Dev. 23:548–555. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Singmann P, Shem-Tov D, Wahl S, Grallert H, Fiorito G, Shin SY, Schramm K, Wolf P, Kunze S, Baran Y, et al: Characterization of whole-genome autosomal differences of DNA methylation between men and women. Epigenetics Chromatin. 8:432015. View Article : Google Scholar : PubMed/NCBI

21 

Ziegler-Birling C, Daujat S, Schneider R and Torres-Padilla ME: Dynamics of histone H3 acetylation in the nucleosome core during mouse pre-implantation development. Epigenetics. 11:553–562. 2016. View Article : Google Scholar :

22 

Dudakovic A, Camilleri ET, Xu F, Riester SM, McGee-Lawrence ME, Bradley EW, Paradise CR, Lewallen EA, Thaler R, Deyle DR, et al: Epigenetic control of skeletal development by the histone methyltransferase Ezh2. J Biol Chem. 290:27604–27617. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Huang F, Ramakrishnan S, Pokhrel S, Pflueger C, Parnell TJ, Kasten MM, Currie SL, Bhachech N, Horikoshi M, Graves BJ, et al: Interaction of the Jhd2 H3K4 demethylase with chromatin is controlled by histone H2A surfaces and restricted by H2B ubiquitination. J Biol Chem. 290:28760–28777. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Wang Z, Casas-Mollano JA, Xu J, Riethoven JJ, Zhang C and Cerutti H: Osmotic stress induces phosphorylation of histone H3 at threonine 3 in pericentromeric regions of Arabidopsis thaliana. Proc Natl Acad Sci USA. 112:8487–8492. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Yao Y, Des Marais TL and Costa M: Chromatin memory in the development of human cancers. Gene Technol. 3:1142014.

26 

Heo JB and Sung S: Encoding memory of winter by noncoding RNAs. Epigenetics. 6:544–547. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Torres IO and Fujimori DG: Functional coupling between writers, erasers and readers of histone and DNA methylation. Curr Opin Struct Biol. 35:68–75. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Jaworska J, Ziemka-Nalecz M and Zalewska T: Histone deacetylases 1 and 2 are required for brain development. Int J Dev Biol. 59:171–177. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Carr SM, Poppy Roworth A, Chan C and La Thangue NB: Post-translational control of transcription factors: Methylation ranks highly. FEBS J. 282:4450–4465. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Gupta N, Madapura MP, Bhat UA and Rao MR: Mapping of post-translational modifications of transition proteins, TP1 and TP2, and identification of protein arginine methyltransferase 4 and lysine methyltransferase 7 as methyltransferase for TP2. J Biol Chem. 290:12101–12122. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Binda O: On your histone mark, SET, methylate! Epigenetics. 8:457–463. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Lesne A, Foray N, Cathala G, Forné T, Wong H and Victor JM: Chromatin fiber allostery and the epigenetic code. J Phys Condens Matter. 27:0641142015. View Article : Google Scholar : PubMed/NCBI

33 

Zuchegna C, Aceto F, Bertoni A, Romano A, Perillo B, Laccetti P, Gottesman ME, Avvedimento EV and Porcellini A: Mechanism of retinoic acid-induced transcription: Histone code, DNA oxidation and formation of chromatin loops. Nucleic Acids Res. 42:11040–11055. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Gayatri S and Bedford MT: Readers of histone methylarginine marks. Biochim Biophys Acta. 1839:702–710. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Casciello F, Windloch K, Gannon F and Lee JS: Functional role of G9a histone methyltransferase in cancer. Front Immunol. 6:4872015. View Article : Google Scholar : PubMed/NCBI

36 

Ortega-Molina A, Boss IW, Canela A, Pan H, Jiang Y, Zhao C, Jiang M, Hu D, Agirre X, Niesvizky I, et al: The histone lysine methyltransferase KMT2D sustains a gene expression program that represses B cell lymphoma development. Nat Med. 21:1199–1208. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Cho MH, Park JH, Choi HJ, Park MK, Won HY, Park YJ, Lee CH, Oh SH, Song YS, Kim HS, et al: DOT1L cooperates with the c-Myc-p300 complex to epigenetically derepress CDH1 transcription factors in breast cancer progression. Nat Commun. 6:78212015. View Article : Google Scholar : PubMed/NCBI

38 

Liu N, Zhang Z, Wu H, Jiang Y, Meng L, Xiong J, Zhao Z, Zhou X, Li J, Li H, et al: Recognition of H3K9 methylation by GLP is required for efficient establishment of H3K9 methylation, rapid target gene repression, and mouse viability. Genes Dev. 29:379–393. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Nicetto D, Hahn M, Jung J, Schneider TD, Straub T, David R, Schotta G and Rupp RA: Suv4-20h histone methyltransferases promote neuroectodermal differentiation by silencing the pluripotency-associated Oct-25 gene. PLoS Genet. 9:e10031882013. View Article : Google Scholar : PubMed/NCBI

40 

Maes T, Mascaró C, Ortega A, Lunardi S, Ciceri F, Somervaille TC and Buesa C: KDM1 histone lysine demethylases as targets for treatments of oncological and neurodegenerative disease. Epigenomics. 7:609–626. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Belakavadi M, Dell J, Grover GJ and Fondell JD: Thyroid hormone suppression of β-amyloid precursor protein gene expression in the brain involves multiple epigenetic regulatory events. Mol Cell Endocrinol. 339:72–80. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Stein AB, Goonewardena SN, Jones TA, Prusick PJ, Bazzi AA, Belyavskaya JM, McCoskey MM and Dandar RA: The PTIP-associated histone methyltransferase complex prevents stress-induced maladaptive cardiac remodeling. PLoS One. 10:e01278392015. View Article : Google Scholar : PubMed/NCBI

43 

Mathiyalagan P, Keating ST, Du XJ and El-Osta A: Chromatin modifications remodel cardiac gene expression. Cardiovasc Res. 103:7–16. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Hohl M, Wagner M, Reil JC, Müller SA, Tauchnitz M, Zimmer AM, Lehmann LH, Thiel G, Böhm M, Backs J, et al: HDAC4 controls histone methylation in response to elevated cardiac load. J Clin Invest. 123:1359–1370. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Bingham AJ, Ooi L, Kozera L, White E and Wood IC: The repressor element 1-silencing transcription factor regulates heart-specific gene expression using multiple chromatin-modifying complexes. Mol Cell Biol. 27:4082–4092. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Cabrera JR, Olcese U and Horabin JI: A balancing act: Heterochromatin protein 1a and the polycomb group coordinate their levels to silence chromatin in Drosophila. Epigenetics Chromatin. 8:172015. View Article : Google Scholar : PubMed/NCBI

47 

Sautel CF, Cannella D, Bastien O, Kieffer S, Aldebert D, Garin J, Tardieux I, Belrhali H and Hakimi MA: SET8-mediated methylations of histone H4 lysine 20 mark silent heterochromatic domains in apicomplexan genomes. Mol Cell Biol. 27:5711–5724. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Guo X, Wang L, Li J, Ding Z, Xiao J, Yin X, He S, Shi P, Dong L, Li G, et al: Structural insight into autoinhibition and histone H3-induced activation of DNMT3A. Nature. 517:640–644. 2015. View Article : Google Scholar

49 

Minkovsky A, Sahakyan A, Rankin-Gee E, Bonora G, Patel S and Plath K: The Mbd1-Atf7ip-Setdb1 pathway contributes to the maintenance of X chromosome inactivation. Epigenetics Chromatin. 7:122014. View Article : Google Scholar : PubMed/NCBI

50 

Simon MD, Pinter SF, Fang R, Sarma K, Rutenberg-Schoenberg M, Bowman SK, Kesner BA, Maier VK, Kingston RE and Lee JT: High-resolution Xist binding maps reveal two-step spreading during X-chromosome inactivation. Nature. 504:465–469. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Wang J, Telese F, Tan Y, Li W, Jin C, He X, Basnet H, Ma Q, Merkurjev D, Zhu X, et al: LSD1n is an H4K20 demethylase regulating memory formation via transcriptional elongation control. Nat Neurosci. 18:1256–1264. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Sen P, Dang W, Donahue G, Dai J, Dorsey J, Cao X, Liu W, Cao K, Perry R, Lee JY, et al: H3K36 methylation promotes longevity by enhancing transcriptional fidelity. Genes Dev. 29:1362–1376. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Kang SC, Kim SK, Chai JC, Kim SH, Won KJ, Lee YS, Jung KH and Chai YG: Transcriptomic Profiling and H3K27me3 Distribution reveal both demethylase-dependent and independent regulation of developmental gene transcription in cell differentiation. PLoS One. 10:e01352762015. View Article : Google Scholar : PubMed/NCBI

54 

Copur Ö and Müller J: The histone H3-K27 demethylase Utx regulates HOX gene expression in Drosophila in a temporally restricted manner. Development. 140:3478–3485. 2013. View Article : Google Scholar : PubMed/NCBI

55 

You L, Nie J, Sun WJ, Zheng ZQ and Yang XJ: Lysine acetylation: Enzymes, bromodomains and links to different diseases. Essays Biochem. 52:1–12. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Migliori V, Phalke S, Bezzi M and Guccione E: Arginine/lysinemethyl/methyl switches: Biochemical role of histone arginine methylation in transcriptional regulation. Epigenomics. 2:119–137. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Davie JK and Dent SY: Transcriptional control: An activating role for arginine methylation. Curr Biol. 12:R59–R61. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Tessarz P and Kouzarides T: Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol. 15:703–708. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Strahl BD, Briggs SD, Brame CJ, Caldwell JA, Koh SS, Ma H, Cook RG, Shabanowitz J, Hunt DF, Stallcup MR, et al: Methylation of histone H4 at arginine 3 occurs in vivo and is mediated by the nuclear receptor coactivator PRMT1. Curr Biol. 11:996–1000. 2001. View Article : Google Scholar : PubMed/NCBI

60 

Wang H, Huang ZQ, Xia L, Feng Q, Erdjument-Bromage H, Strahl BD, Briggs SD, Allis CD, Wong J, Tempst P, et al: Methylation of histone H4 at arginine 3 facilitating transcriptional activation by nuclear hormone receptor. Science. 293:853–857. 2001. View Article : Google Scholar : PubMed/NCBI

61 

Schurter BT, Koh SS, Chen D, Bunick GJ, Harp JM, Hanson BL, Henschen-Edman A, Mackay DR, Stallcup MR and Aswad DW: Methylation of histone H3 by coactivator-associated arginine methyltransferase 1. Biochemistry. 40:5747–5756. 2001. View Article : Google Scholar : PubMed/NCBI

62 

Aoshima K, Inoue E, Sawa H and Okada Y: Paternal H3K4 methylation is required for minor zygotic gene activation and early mouse embryonic development. EMBO Rep. 16:803–812. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Shinsky SA, Monteith KE, Viggiano S and Cosgrove MS: Biochemical reconstitution and phylogenetic comparison of human SET1 family core complexes involved in histone methylation. J Biol Chem. 290:6361–6375. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Chen CW, Koche RP, Sinha AU, Deshpande AJ, Zhu N, Eng R, Doench JG, Xu H, Chu SH, Qi J, et al: DOT1L inhibits SIRT1-mediated epigenetic silencing to maintain leukemic gene expression in MLL-rearranged leukemia. Nat Med. 21:335–343. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Jiao L and Liu X: Structural basis of histone H3K27 trimethylation by an active polycomb repressive complex 2. Science. 350:aac43832015. View Article : Google Scholar : PubMed/NCBI

66 

Foda BM and Singh U: Dimethylated H3K27 is a repressive epigenetic histone mark in the protist Entamoeba histolytica and is significantly enriched in genes silenced via the RNAi pathway. J Biol Chem. 290:21114–21130. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Vieira FQ, Costa-Pinheiro P, Almeida-Rios D, Graça I, Monteiro-Reis S, Simões-Sousa S, Carneiro I, Sousa EJ, Godinho MI, Baltazar F, et al: SMYD3 contributes to a more aggressive phenotype of prostate cancer and targets Cyclin D2 through H4K20me3. Oncotarget. 6:13644–13657. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Bierhoff H, Dammert MA, Brocks D, Dambacher S, Schotta G and Grummt I: Quiescence-induced LncRNAs trigger H4K20 trimethylation and transcriptional silencing. Mol Cell. 54:675–682. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Zhang X, Tanaka K, Yan J, Li J, Peng D, Jiang Y, Yang Z, Barton MC, Wen H and Shi X: Regulation of estrogen receptor α by histone methyltransferase SMYD2-mediated protein methylation. Proc Natl Acad Sci USA. 110:17284–17289. 2013. View Article : Google Scholar

70 

Hossain MA, Chung C, Pradhan SK and Johnson TL: The yeast cap binding complex modulates transcription factor recruitment and establishes proper histone H3K36 trimethylation during active transcription. Mol Cell Biol. 33:785–799. 2013. View Article : Google Scholar :

71 

Ontoso D, Acosta I, van Leeuwen F, Freire R and San-Segundo PA: Dot1-dependent histone H3K79 methylation promotes activation of the Mek1 meiotic checkpoint effector kinase by regulating the Hop1 adaptor. PLoS Genet. 9:e10032622013. View Article : Google Scholar : PubMed/NCBI

72 

Kim SK, Jung I, Lee H, Kang K, Kim M, Jeong K, Kwon CS, Han YM, Kim YS, Kim D, et al: Human histone H3K79 methyltransferase DOT1L protein [corrected] binds actively transcribing RNA polymerase II to regulate gene expression. J Biol Chem. 287:39698–39709. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Zhao XX, Zhang YB, Ni PL, Wu ZL, Yan YC and Li YP: Protein arginine methyltransferase 6 (Prmt6) is essential for early zebrafish development through the direct suppression of Gadd45alphaa stress sensor gene. J Biol Chem. 291:402–412. 2016. View Article : Google Scholar

74 

Feng Y, Hadjikyriacou A and Clarke SG: Substrate specificity of human protein arginine methyltransferase 7 (PRMT7): The importance of acidic residues in the double E loop. J Biol Chem. 289:32604–32616. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Su X, Zhu G, Ding X, Lee SY, Dou Y, Zhu B, Wu W and Li H: Molecular basis underlying histone H3 lysine-arginine methylation pattern readout by Spin/Ssty repeats of Spindlin1. Genes Dev. 28:622–636. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Nguyen HC, Wang M, Salsburg A and Knuckley B: Development of a plate-based screening assay to investigate the dubstrate dpecificity of the PRMT gamily of rnzymes. ACS Comb Sci. 17:500–505. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Tini M, Naeem H and Torchia J: Biochemical analysis of arginine methylation in transcription. Methods Mol Biol. 523:235–247. 2009. View Article : Google Scholar : PubMed/NCBI

78 

Del Rizzo PA and Trievel RC: Substrate and product specificities of SET domain methyltransferases. Epigenetics. 6:1059–1067. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Couture JF and Trievel RC: Histone-modifying enzymes: Encrypting an enigmatic epigenetic code. Curr Opin Struct Biol. 16:753–760. 2006. View Article : Google Scholar : PubMed/NCBI

80 

Spellmon N, Holcomb J, Trescott L, Sirinupong N and Yang Z: Structure and function of SET and MYND domain-containing proteins. Int J Mol Sci. 16:1406–1428. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Anand R and Marmorstein R: Structure and mechanism of lysine-specific demethylase enzymes. J Biol Chem. 282:35425–35429. 2007. View Article : Google Scholar : PubMed/NCBI

82 

Tsukada Y and Zhang Y: Purification of histone demethylases from HeLa cells. Methods. 40:318–326. 2006. View Article : Google Scholar : PubMed/NCBI

83 

Liu L, Jin G and Zhou X: Modeling the relationship of epigenetic modifications to transcription factor binding. Nucleic Acids Res. 43:3873–3885. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Bai H, Li Y, Gao H, Dong Y, Han P and Yu H: Histone methyltransferase SMYD3 regulates the expression of transcriptional factors during bovine oocyte maturation and early embryonic development. Cytotechnology. 68:849–859. 2016. View Article : Google Scholar :

85 

Kim JD, Kim E, Koun S, Ham HJ, Rhee M, Kim MJ and Huh TL: Proper activity of histone H3 lysine 4 (H3K4) methyltransferase is required for morphogenesis during zebrafish cardiogenesis. Mol Cells. 38:580–586. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Martinez SR, Gay MS and Zhang L: Epigenetic mechanisms in heart development and disease. Drug Discov Today. 20:799–811. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Dorn GW II and Matkovich SJ: Epitranscriptional regulation of cardiovascular development and disease. J Physiol. 593:1799–1808. 2015. View Article : Google Scholar :

88 

Zhao W, Liu L, Pan B, Xu Y, Zhu J, Nan C, Huang X and Tian J: Epigenetic regulation of cardiac myofibril gene expression during heart development. Cardiovasc Toxicol. 15:203–209. 2015. View Article : Google Scholar

89 

Park CY, Pierce SA, von Drehle M, Ivey KN, Morgan JA, Blau HM and 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. View Article : Google Scholar : PubMed/NCBI

90 

Drake KM, Comhair SA, Erzurum SC, Tuder RM and Aldred MA: Endothelial chromosome 13 deletion in congenital heart disease-associated pulmonary arterial hypertension dysregulates SMAD9 signaling. Am J Respir Crit Care Med. 191:850–854. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Geng J, Picker J, Zheng Z, Zhang X, Wang J, Hisama F, Brown DW, Mullen MP, Harris D, Stoler J, et al: Chromosome microarray testing for patients with congenital heart defects reveals novel disease causing loci and high diagnostic yield. BMC Genomics. 15:11272014. View Article : Google Scholar : PubMed/NCBI

92 

Zaidi S, Choi M, Wakimoto H, Ma L, Jiang J, Overton JD, Romano-Adesman A, Bjornson RD, Breitbart RE, Brown KK, et al: De novo mutations in histone-modifying genes in congenital heart disease. Nature. 498:220–223. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Ozanne SE and Constância M: Mechanisms of disease: The developmental origins of disease and the role of the epigenotype. Nat Clin Pract Endocrinol Metab. 3:539–546. 2007. View Article : Google Scholar : PubMed/NCBI

94 

Wang QT: Epigenetic regulation of cardiac development and function by polycomb group and trithorax group proteins. Dev Dyn. 241:1021–1033. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Geisler SJ and Paro R: Trithorax and Polycomb group-dependent regulation: A tale of opposing activities. Development. 142:2876–2887. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Wan X, Liu L, Ding X, Zhou P, Yuan X, Zhou Z, Hu P, Zhou H, Li Q, Zhang S, et al: Mll2 controls cardiac lineage differentiation of mouse embryonic stem cells by promoting H3K4me3 deposition at cardiac-specific genes. Stem Cell Rev. 10:643–652. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Tan X, Rotllant J, Li H, De Deyne P and 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. View Article : Google Scholar : PubMed/NCBI

98 

Du SJ, Tan X and Zhang J: SMYD proteins: Key regulators in skeletal and cardiac muscle development and function. Anat Rec (Hoboken). 297:1650–1662. 2014. View Article : Google Scholar

99 

Rasmussen TL, Ma Y, Park CY, Harriss J, Pierce SA, Dekker JD, Valenzuela N, Srivastava D, Schwartz RJ, Stewart MD, et al: Smyd1 facilitates heart development by antagonizing oxidative and ER stress responses. PLoS One. 10:e01217652015. View Article : Google Scholar : PubMed/NCBI

100 

Diehl F, Brown MA, van Amerongen MJ, Novoyatleva T, Wietelmann A, Harriss J, Ferrazzi F, Böttger T, Harvey RP, Tucker PW, et al: Cardiac deletion of Smyd2 is dispensable for mouse heart development. PLoS One. 5:e97482010. View Article : Google Scholar : PubMed/NCBI

101 

Fujii T, Tsunesumi S, Yamaguchi K, Watanabe S and Furukawa Y: Smyd3 is required for the development of cardiac and skeletal muscle in zebrafish. PLoS One. 6:e234912011. View Article : Google Scholar : PubMed/NCBI

102 

Papaioannou VE: The T-box gene family: Emerging roles in development, stem cells and cancer. Development. 141:3819–3833. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Greulich F, Rudat C and Kispert A: Mechanisms of T-box gene function in the developing heart. Cardiovasc Res. 91:212–222. 2011. View Article : Google Scholar : PubMed/NCBI

104 

Chen L, Fulcoli FG, Ferrentino R, Martucciello S, Illingworth EA and Baldini A: Transcriptional control in cardiac progenitors: Tbx1 interacts with the BAF chromatin remodeling complex and regulates Wnt5a. PLoS Genet. 8:e10025712012. View Article : Google Scholar : PubMed/NCBI

105 

Caprio C and Baldini A: p53 Suppression partially rescues the mutant phenotype in mouse models of DiGeorge syndrome. Proc Natl Acad Sci USA. 111:13385–13390. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Pan Y, Wang ZG, Liu XY, Zhao H, Zhou N, Zheng GF, Qiu XB, Li RG, Yuan F, Shi HY, et al: A novel TBX1 loss-of-function mutation associated with congenital heart disease. Pediatr Cardiol. 36:1400–1410. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Sedletcaia A and Evans T: Heart chamber size in zebrafish is regulated redundantly by duplicated tbx2 genes. Dev Dyn. 240:1548–1557. 2011. View Article : Google Scholar : PubMed/NCBI

108 

Pang S, Liu Y, Zhao Z, Huang W, Chen D and Yan B: Novel and functional sequence variants within the TBX2 gene promoter in ventricular septal defects. Biochimie. 95:1807–1809. 2013. View Article : Google Scholar : PubMed/NCBI

109 

van Weerd JH, Badi I, van den Boogaard M, Stefanovic S, van de Werken HJ, Gomez-Velazquez M, Badia-Careaga C, Manzanares M, de Laat W, Barnett P, et al: A large permissive regulatory domain exclusively controls Tbx3 expression in the cardiac conduction system. Circ Res. 115:432–441. 2014. View Article : Google Scholar : PubMed/NCBI

110 

Hatcher CJ, Diman NY, Kim MS, Pennisi D, Song Y, Goldstein MM, Mikawa T and Basson CT: A role for Tbx5 in proepicardial cell migration during cardiogenesis. Physiol Genomics. 18:129–140. 2004. View Article : Google Scholar : PubMed/NCBI

111 

Diman NY, Brooks G, Kruithof BP, Elemento O, Seidman JG, Seidman CE, Basson CT and Hatcher CJ: Tbx5 is required for avian and Mammalian epicardial formation and coronary vasculogenesis. Circ Res. 115:834–844. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Zhou L, Liu J, Olson P, Zhang K, Wynne J and Xie L: Tbx5 and Osr1 interact to regulate posterior second heart field cell cycle progression for cardiac septation. J Mol Cell Cardiol. 85:1–12. 2015. View Article : Google Scholar : PubMed/NCBI

113 

Al-Qattan MM and Abou Al-Shaar H: Molecular basis of the clinical features of Holt-Oram syndrome resulting from missense and extended protein mutations of the TBX5 gene as well as TBX5 intragenic duplications. Gene. 560:129–136. 2015. View Article : Google Scholar : PubMed/NCBI

114 

Kimura M, Kikuchi A, Ichinoi N and Kure S: Novel TBX5 duplication in a Japanese family with Holt-Oram syndrome. Pediatr Cardiol. 36:244–247. 2015. View Article : Google Scholar

115 

Wu SP, Dong XR, Regan JN, Su C and Majesky MW: Tbx18 regulates development of the epicardium and coronary vessels. Dev Biol. 383:307–320. 2013. View Article : Google Scholar : PubMed/NCBI

116 

Cai X, Zhang W, Hu J, Zhang L, Sultana N, Wu B, Cai W, Zhou B and Cai CL: Tbx20 acts upstream of Wnt signaling to regulate endocardial cushion formation and valve remodeling during mouse cardiogenesis. Development. 140:3176–3187. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Shen T, Aneas I, Sakabe N, Dirschinger RJ, Wang G, Smemo S, Westlund JM, Cheng H, Dalton N, Gu Y, et al: Tbx20 regulates a genetic program essential to adult mouse cardiomyocyte function. J Clin Invest. 121:4640–4654. 2011. View Article : Google Scholar : PubMed/NCBI

118 

Zhang W, Chen H, Wang Y, Yong W, Zhu W, Liu Y, Wagner GR, Payne RM, Field LJ, Xin H, et al: Tbx20 transcription factor is a downstream mediator for bone morphogenetic protein-10 in regulating cardiac ventricular wall development and function. J Biol Chem. 286:36820–36829. 2011. View Article : Google Scholar : PubMed/NCBI

119 

Kirk EP, Sunde M, Costa MW, Rankin SA, Wolstein O, Castro ML, Butler TL, Hyun C, Guo G, Otway R, et al: Mutations in cardiac T-box factor gene TBX20 are associated with diverse cardiac pathologies, including defects of septation and valvulogenesis and cardiomyopathy. Am J Hum Genet. 81:280–291. 2007. View Article : Google Scholar : PubMed/NCBI

120 

Ishizaka A, Mizutani T, Kobayashi K, Tando T, Sakurai K, Fujiwara T and Iba H: Double plant homeodomain (PHD) finger proteins DPF3a and -3b are required as transcriptional co-activators in SWI/SNF complex-dependent activation of NF-κB RelA/p50 heterodimer. J Biol Chem. 287:11924–11933. 2012. View Article : Google Scholar : PubMed/NCBI

121 

Lange M, Kaynak B, Forster UB, Tönjes M, Fischer JJ, Grimm C, Schlesinger J, Just S, Dunkel I, Krueger T, et al: Regulation of muscle development by DPF3, a novel histone acetylation and methylation reader of the BAF chromatin remodeling complex. Genes Dev. 22:2370–2384. 2008. View Article : Google Scholar : PubMed/NCBI

122 

Liu Y, Huang Y, Fan J and Zhu GZ: PITX2 associates with PTIP-containing histone H3 lysine 4 methyltransferase complex. Biochem Biophys Res Commun. 444:634–637. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Kim D, Patel SR, Xiao H and Dressler GR: The role of PTIP in maintaining embryonic stem cell pluripotency. Stem Cells. 27:1516–1523. 2009. View Article : Google Scholar : PubMed/NCBI

124 

Fang M, Ren H, Liu J, Cadigan KM, Patel SR and Dressler GR: Drosophila ptip is essential for anterior/posterior patterning in development and interacts with the PcG and trxG pathways. Development. 136:1929–1938. 2009. View Article : Google Scholar : PubMed/NCBI

125 

Callen E, Faryabi RB, Luckey M, Hao B, Daniel JA, Yang W, Sun HW, Dressler G, Peng W, Chi H, et al: The DNA damage- and transcription-associated protein paxip1 controls thymocyte development and emigration. Immunity. 37:971–985. 2012. View Article : Google Scholar : PubMed/NCBI

126 

Daniel JA, Santos MA, Wang Z, Zang C, Schwab KR, Jankovic M, Filsuf D, Chen HT, Gazumyan A, Yamane A, et al: PTIP promotes chromatin changes critical for immunoglobulin class switch recombination. Science. 329:917–923. 2010. View Article : Google Scholar : PubMed/NCBI

127 

Stein AB, Jones TA, Herron TJ, Patel SR, Day SM, Noujaim SF, Milstein ML, Klos M, Furspan PB, Jalife J, et al: Loss of H3K4 methylation destabilizes gene expression patterns and physiological functions in adult murine cardiomyocytes. J Clin Invest. 121:2641–2650. 2011. View Article : Google Scholar : PubMed/NCBI

128 

Tao Y, Neppl RL, Huang ZP, Chen J, Tang RH, Cao R, Zhang Y, Jin SW and Wang DZ: The histone methyltransferase Set7/9 promotes myoblast differentiation and myofibril assembly. J Cell Biol. 194:551–565. 2011. View Article : Google Scholar : PubMed/NCBI

129 

Nicholson TB and Chen T: LSD1 demethylates histone and non-histone proteins. Epigenetics. 4:129–132. 2009. View Article : Google Scholar : PubMed/NCBI

130 

He Y, Tang D, Cai C, Chai R and Li H: LSD1 is required for hair cell regeneration in zebrafish. Mol Neurobiol. 53:2421–2434. 2016. View Article : Google Scholar

131 

van Riel B, Pakozdi T, Brouwer R, Monteiro R, Tuladhar K, Franke V, Bryne JC, Jorna R, Rijkers EJ, van Ijcken W, et al: A novel complex, RUNX1-MYEF2, represses hematopoietic genes in erythroid cells. Mol Cell Biol. 32:3814–3822. 2012. View Article : Google Scholar : PubMed/NCBI

132 

Nicholson TB, Singh AK, Su H, Hevi S, Wang J, Bajko J, Li M, Valdez R, Goetschkes M, Capodieci P, et al: A hypomorphic lsd1 allele results in heart development defects in mice. PLoS One. 8:e609132013. View Article : Google Scholar : PubMed/NCBI

133 

Fan Z, Yamaza T, Lee JS, Yu J, Wang S, Fan G, Shi S and Wang CY: BCOR regulates mesenchymal stem cell function by epigenetic mechanisms. Nat Cell Biol. 11:1002–1009. 2009. View Article : Google Scholar : PubMed/NCBI

134 

Hilton E, Johnston J, Whalen S, Okamoto N, Hatsukawa Y, Nishio J, Kohara H, Hirano Y, Mizuno S, Torii C, et al: BCOR analysis in patients with OFCD and Lenz microphthalmia syndromes, mental retardation with ocular anomalies, and cardiac laterality defects. Eur J Hum Genet. 17:1325–1335. 2009. View Article : Google Scholar : PubMed/NCBI

135 

Di Stefano C, Lombardo B, Fabbricatore C, Munno C, Caliendo I, Gallo F and Pastore L: Oculo-facio-cardio-dental (OFCD) syndrome: The first Italian case of BCOR and co-occurring OTC gene deletion. Gene. 559:203–206. 2015. View Article : Google Scholar : PubMed/NCBI

136 

Huang XJ, Ma X, Wang X, Zhou X, Li J, Sun SC and Liu H: Involvement of G9A-like protein (GLP) in the development of mouse preimplantation embryos in vitro. Reprod Fertil Dev. May 18–2015.Epub ahead of print. View Article : Google Scholar

137 

Tachibana M, Ueda J, Fukuda M, Takeda N, Ohta T, Iwanari H, Sakihama T, Kodama T, Hamakubo T and Shinkai Y: Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9. Genes Dev. 19:815–826. 2005. View Article : Google Scholar : PubMed/NCBI

138 

Inagawa M, Nakajima K, Makino T, Ogawa S, Kojima M, Ito S, Ikenishi A, Hayashi T, Schwartz RJ, Nakamura K, et al: Histone H3 lysine 9 methyltransferases, G9a and GLP are essential for cardiac morphogenesis. Mech Dev. 130:519–531. 2013. View Article : Google Scholar : PubMed/NCBI

139 

Kleefstra T, van Zelst-Stams WA, Nillesen WM, Cormier-Daire V, Houge G, Foulds N, van Dooren M, Willemsen MH, Pfundt R, Turner A, et al: Further clinical and molecular delineation of the 9q subtelomeric deletion syndrome supports a major contribution of EHMT1 haploinsufficiency to the core phenotype. J Med Genet. 46:598–606. 2009. View Article : Google Scholar : PubMed/NCBI

140 

Stewart DR and Kleefstra T: The chromosome 9q subtelomere deletion syndrome. Am J Med Genet C Semin Med Genet. 145C:383–392. 2007. View Article : Google Scholar : PubMed/NCBI

141 

Bikoff EK, Morgan MA and Robertson EJ: An expanding job description for Blimp-1/PRDM1. Curr Opin Genet Dev. 19:379–385. 2009. View Article : Google Scholar : PubMed/NCBI

142 

Morgan MA, Mould AW, Li L, Robertson EJ and Bikoff EK: Alternative splicing regulates Prdm1/Blimp-1 DNA binding activities and corepressor interactions. Mol Cell Biol. 32:3403–3413. 2012. View Article : Google Scholar : PubMed/NCBI

143 

Lee Y, Song AJ, Baker R, Micales B, Conway SJ and Lyons GE: Jumonji, a nuclear protein that is necessary for normal heart development. Circ Res. 86:932–938. 2000. View Article : Google Scholar : PubMed/NCBI

144 

Kim TG, Chen J, Sadoshima J and Lee Y: Jumonji represses atrial natriuretic factor gene expression by inhibiting transcriptional activities of cardiac transcription factors. Mol Cell Biol. 24:10151–10160. 2004. View Article : Google Scholar : PubMed/NCBI

145 

Mysliwiec MR, Carlson CD, Tietjen J, Hung H, Ansari AZ and Lee Y: Jarid2 (Jumonji, AT rich interactive domain 2) regulates NOTCH1 expression via histone modification in the developing heart. J Biol Chem. 287:1235–1241. 2012. View Article : Google Scholar :

146 

Mysliwiec MR, Bresnick EH and Lee Y: Endothelial Jarid2/Jumonji is required for normal cardiac development and proper Notch1 expression. J Biol Chem. 286:17193–17204. 2011. View Article : Google Scholar : PubMed/NCBI

147 

Dobreva G and Braun T: When silence is broken: Polycomb group proteins in heart development. Circ Res. 110:372–374. 2012. View Article : Google Scholar : PubMed/NCBI

148 

Weston AD, Ozolins TR and Brown NA: Thoracic skeletal defects and cardiac malformations: A common epigenetic link? Birth Defects Res C Embryo Today. 78:354–370. 2006. View Article : Google Scholar

149 

Sanulli S, Justin N, Teissandier A, Ancelin K, Portoso M, Caron M, Michaud A, Lombard B, da Rocha ST, Offer J, et al: Jarid2 methylation via the PRC2 complex regulates H3K27me3 deposition during cell differentiation. Mol Cell. 57:769–783. 2015. View Article : Google Scholar : PubMed/NCBI

150 

Delgado-Olguín P, Huang Y, Li X, Christodoulou D, Seidman CE, Seidman JG, Tarakhovsky A and Bruneau BG: Epigenetic repression of cardiac progenitor gene expression by Ezh2 is required for postnatal cardiac homeostasis. Nat Genet. 44:343–347. 2012. View Article : Google Scholar : PubMed/NCBI

151 

Chen L, Ma Y, Kim EY, Yu W, Schwartz RJ, Qian L and Wang J: Conditional ablation of Ezh2 in murine hearts reveals its essential roles in endocardial cushion formation, cardiomyocyte proliferation and survival. PLoS One. 7:e310052012. View Article : Google Scholar : PubMed/NCBI

152 

Welstead GG, Creyghton MP, Bilodeau S, Cheng AW, Markoulaki S, Young RA and Jaenisch R: X-linked H3K27me3 demethylase Utx is required for embryonic development in a sex-specific manner. Proc Natl Acad Sci USA. 109:13004–13009. 2012. View Article : Google Scholar : PubMed/NCBI

153 

Morales Torres C, Laugesen A and Helin K: Utx is required for proper induction of ectoderm and mesoderm during differentiation of embryonic stem cells. PLoS One. 8:e600202013. View Article : Google Scholar : PubMed/NCBI

154 

Lee S, Lee JW and Lee SK: UTX, a histone H3-lysine 27 demethylase, acts as a critical switch to activate the cardiac developmental program. Dev Cell. 22:25–37. 2012. View Article : Google Scholar

155 

Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rappsilber J, Issaeva I, Canaani E, Salcini AE and Helin K: UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature. 449:731–734. 2007. View Article : Google Scholar : PubMed/NCBI

156 

Li Q, Wang HY, Chepelev I, Zhu Q, Wei G, Zhao K and Wang RF: Stage-dependent and locus-specific role of histone demethylase Jumonji D3 (JMJD3) in the embryonic stages of lung development. PLoS Genet. 10:e10045242014. View Article : Google Scholar : PubMed/NCBI

157 

Burgold T, Voituron N, Caganova M, Tripathi PP, Menuet C, Tusi BK, Spreafico F, Bévengut M, Gestreau C, Buontempo S, et al: The H3K27 demethylase JMJD3 is required for maintenance of the embryonic respiratory neuronal network, neonatal breathing, and survival. Cell Rep. 2:1244–1258. 2012. View Article : Google Scholar : PubMed/NCBI

158 

Ohtani K, Zhao C, Dobreva G, Manavski Y, Kluge B, Braun T, Rieger MA, Zeiher AM and Dimmeler S: Jmjd3 controls mesodermal and cardiovascular differentiation of embryonic stem cells. Circ Res. 113:856–862. 2013. View Article : Google Scholar : PubMed/NCBI

159 

Chen CP, Lin CJ, Chern SR, Liu YP, Kuo YL, Chen YN, Wu PS, Town DD, Chen LF, Yang CW, et al: Prenatal diagnosis and molecular cytogenetic characterization of a 1.07-Mb microdeletion at 5q35.2-q35.3 associated with NSD1 haploinsufficiency and Sotos syndrome. Taiwan J Obstet Gynecol. 53:583–587. 2014. View Article : Google Scholar : PubMed/NCBI

160 

Park SH, Lee JE, Sohn YB and Ko JM: First identified Korean family with Sotos syndrome caused by a novel intragenic mutation in NSD1. Ann Clin Lab Sci. 44:228–231. 2014.PubMed/NCBI

161 

Sheth K, Moss J, Hyland S, Stinton C, Cole T and Oliver C: The behavioral characteristics of Sotos syndrome. Am J Med Genet A. 167A:2945–2956. 2015. View Article : Google Scholar : PubMed/NCBI

162 

Vallaster M, Vallaster CD and Wu SM: Epigenetic mechanisms in cardiac development and disease. Acta Biochim Biophys Sin (Shanghai). 44:92–102. 2012. View Article : Google Scholar

163 

Nimura K, Ura K, Shiratori H, Ikawa M, Okabe M, Schwartz RJ and Kaneda Y: A histone H3 lysine 36 trimethyltransferase links Nkx2-5 to Wolf-Hirschhorn syndrome. Nature. 460:287–291. 2009. View Article : Google Scholar : PubMed/NCBI

164 

Oh S and Janknecht R: Histone demethylase JMJD5 is essential for embryonic development. Biochem Biophys Res Commun. 420:61–65. 2012. View Article : Google Scholar : PubMed/NCBI

165 

Kawakami E, Tokunaga A, Ozawa M, Sakamoto R and Yoshida N: The histone demethylase Fbxl11/Kdm2a plays an essential role in embryonic development by repressing cell-cycle regulators. Mech Dev. 135:31–42. 2015. View Article : Google Scholar

166 

Mohan M, Herz HM, Takahashi YH, Lin C, Lai KC, Zhang Y, Washburn MP, Florens L and Shilatifard A: Linking H3K79 trimethylation to Wnt signaling through a novel Dot1-containing complex (DotCom). Genes Dev. 24:574–589. 2010. View Article : Google Scholar : PubMed/NCBI

167 

Jones B, Su H, Bhat A, Lei H, Bajko J, Hevi S, Baltus GA, Kadam S, Zhai H, Valdez R, et al: The histone H3K79 methyltransferase Dot1L is essential for mammalian development and heterochromatin structure. PLoS Genet. 4:e10001902008. View Article : Google Scholar : PubMed/NCBI

168 

Nguyen AT, Xiao B, Neppl RL, Kallin EM, Li J, Chen T, Wang DZ, Xiao X and Zhang Y: DOT1L regulates dystrophin expression and is critical for cardiac function. Genes Dev. 25:263–274. 2011. View Article : Google Scholar : PubMed/NCBI

169 

Beck DB, Burton A, Oda H, Ziegler-Birling C, Torres-Padilla ME and Reinberg D: The role of PR-Set7 in replication licensing depends on Suv4-20h. Genes Dev. 26:2580–2589. 2012. View Article : Google Scholar : PubMed/NCBI

170 

Lennartsson A and Ekwall K: Histone modification patterns and epigenetic codes. Biochim Biophys Acta. 1790:863–868. 2009. View Article : Google Scholar : PubMed/NCBI

171 

Tatton-Brown K and Rahman N: Clinical features of NSD1-positive Sotos syndrome. Clin Dysmorphol. 13:199–204. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2017
Volume 40 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yi X, Jiang X, Li X and Jiang D: Histone lysine methylation and congenital heart disease: From bench to bedside (Review). Int J Mol Med 40: 953-964, 2017
APA
Yi, X., Jiang, X., Li, X., & Jiang, D. (2017). Histone lysine methylation and congenital heart disease: From bench to bedside (Review). International Journal of Molecular Medicine, 40, 953-964. https://doi.org/10.3892/ijmm.2017.3115
MLA
Yi, X., Jiang, X., Li, X., Jiang, D."Histone lysine methylation and congenital heart disease: From bench to bedside (Review)". International Journal of Molecular Medicine 40.4 (2017): 953-964.
Chicago
Yi, X., Jiang, X., Li, X., Jiang, D."Histone lysine methylation and congenital heart disease: From bench to bedside (Review)". International Journal of Molecular Medicine 40, no. 4 (2017): 953-964. https://doi.org/10.3892/ijmm.2017.3115