Skip main navigation

Epigenetic Modifications in Cardiovascular Aging and Diseases

Originally publishedhttps://doi.org/10.1161/CIRCRESAHA.118.312497Circulation Research. 2018;123:773–786

    Abstract

    Aging is associated with a progressive decline in cardiovascular structure and function. Accumulating evidence links cardiovascular aging to epigenetic alterations encompassing a complex interplay of DNA methylation, histone posttranslational modifications, and dynamic nucleosome occupancy governed by numerous epigenetic factors. Advances in genomics technology have led to a profound understanding of chromatin reorganization in both cardiovascular aging and diseases. This review summarizes recent discoveries in epigenetic mechanisms involved in cardiovascular aging and diseases and discusses potential therapeutic strategies to retard cardiovascular aging and conquer related diseases through the rejuvenation of epigenetic signatures to a young state.

    Aging is broadly defined as a time-dependent functional decline. Aging is an inexorable movement of life and unfortunately poses the greatest threat for several cardiovascular diseases (CVDs), including atherosclerosis, coronary artery diseases, hypertension, heart failure, atrial fibrillation, myocardial infarction, and cardiac hypertrophy.

    With age, the heart and vasculature gradually show homeostatic imbalance, vascular stiffening, and fibrosis, as well as increased left ventricular (LV) wall thickness leading to accentuated tissue adaptations and decreased stress tolerance (Figure 1).1,2 Continuous efforts have been made to delineate the crucial mechanisms behind the deterioration of cardiovascular functions during aging. From these studies, epigenetic alterations have emerged as a crucial link between the intrinsic genetic landscape and extrinsic environmental influences. Various risk factors for CVDs, such as diabetes mellitus, nutrition, smoking, stress, hypertension, and circadian rhythm, often manifest as modifications of epigenetic marks.3

    Figure 1.

    Figure 1. General schematics of chromatin structure underpinning cardiovascular aging. The heart and vessels constitute the cardiovascular system. Cardiac aging is featured by chamber enlargement with ventricular wall thickening, whereas vessel aging is characterized by arterial thickening, fibrosis, and deposition of atherosclerotic plaques. The 3-dimensional (3D) architecture of chromatin is regulated by DNA methylation, histone post-transcriptional modifications (PTMs), and chromatin remodelers. These chromatin modifications act coordinately to control RNA transcription. On transcription, RNA processing and modifications add another layer of control on protein synthesis. The RNA products, including long noncoding RNAs (lncRNAs), microRNAs, and circular RNAs (circRNAs), in turn regulate chromatin remodeling, gene transcription, and mRNA processing and modifications. Notably, epigenetic modifications also regulate the expression of epigenetic players, such as epigenetic-related enzymes, which in turn modulate the 3D architecture of chromatin.

    Epigenetics represents a wide range of changes in gene expression independent of changes in DNA sequence. These changes can occur via DNA methylation or hydroxymethylation, histone modifications, and chromatin remodeling. Recent studies have identified histone variants, microRNAs (miRNAs), and long noncoding RNAs (lncRNAs) as additional epigenetic markers (Figure 1).4–7 Epigenetic dysregulation is associated with CVD pathophysiologies during aging. For example, at multiple stages, atherosclerosis is closely associated with epigenetic changes in key players, such as RELA, NOS3, KLF4, and APOE, which are regulated through various layers of epigenetic modifications.8,9

    In this article, we review the mechanisms underlying how epigenetic changes contribute to cardiovascular aging and CVDs. We focus on a subset of CVDs, including atherosclerosis and cardiomyopathies, to elucidate the central mechanisms of diverse epigenetic modifications and related enzymes in the pathogenesis of these symptoms. New therapeutic approaches that target the epigenetic machineries for prevention or treatment of CVDs are also discussed.

    Interplay Between Cardiovascular Aging and Diseases

    Cardiac aging is a complex process characterized by decreased heart functions and ventricular and atrial remodeling. This process includes LV wall thickening because of cardiomyocyte hypertrophy, increased left atrial size, and vascular intimal thickening and stiffening because of collagen and calcium deposition (Figure 1).10 Among the well-defined hallmarks in aging, including genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, dysregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication,11 many features are often observed in CVDs as well. For example, telomere shortening is related to CVDs. Telomere attrition reflects the cumulative burden of inflammatory, oxidative, and mechanical stress on the cardiovascular system,12 thereby linking aging with CVDs.

    Arteriosclerosis is referred to as the degenerative stiffness of the artery walls. Notably, a specific form of arteriosclerosis, atherosclerosis, is the cumulative result of endovascular inflammation, lipid oxidation, and plaque formation. Arteriosclerosis is a progressive, diffuse, age-related process.13 With aging and atherosclerosis regulating similar biochemical pathways in vascular alterations, vessel aging may be considered a prodromal stage of atherosclerotic diseases, or, conversely, atherosclerosis may be considered a form of accelerated arterial aging. In addition to atherosclerosis, aortic stiffening is among the earliest age-related manifestations that affect blood vessel structure and function13; increased aortic wall stiffness may emerge in childhood and progress with age.13 Additionally, the prevalence of atrial fibrillation increases with age-related progressive cardiac fibrosis as profibrotic signals are upregulated by aging.10

    DNA Methylation and Demethylation

    The most well-known epigenetic DNA modification is methylation at 5-cytosine, which is essential for proper gene expression, transposon silencing, alternative splicing, and genome stability.14,15 DNA methylation patterns are routinely maintained by DNMT (DNA methyltransferase) 1, while de novo DNA methylation is typically mediated by DNMT3A and DNMT3B. DNA methylation can be reversed via the oxidation of 5-methylcytosine to 5-hydroxymethylcytosine (5-hmC) by the TET (ten-eleven translocation) proteins.16 There are a variety of strategies for profiling genome-wide DNA methylation, such as whole genome bisulfite sequencing, reduced representation bisulfite sequencing, methylated-DNA immunoprecipitation, and methyl CpG (C-phosphate-G) immunoprecipitation with different depths. Tet-assisted bisulfite sequencing technology enables single-base-resolution detection of 5-hmC17; furthermore, a new method called oxidative bisulfite sequencing can discriminate between 5-methylcytosine and 5-hmC markers in genomic DNA at single-base resolution.18 These modifications were initially considered to be stable markers for gene silencing; however, based on these technologies, DNA methylation is now known to change with chronological age.

    Several studies have associated DNA methylation with cardiovascular aging and diseases. Cardiovascular risk factors, such as smoking, low dietary folate, and increased plasma homocysteine, induce dysregulated DNA methylation. The methylation status of hundreds of CpG sites has been used as an epigenetic clock to measure human aging rates.19–21 DNA methylation-based estimation is well correlated with the incidence of CVDs in a 10-year follow-up study of 832 participants, all aged 70 years at the time of examination.22

    In addition, global DNA hypomethylation is observed in atherosclerotic lesions in humans and animal models,23–25 whereas the promoter regions of atheroprotective genes, such as ESR1/2, ABCA1, and KLF4, are often hypermethylated in atherosclerosis.3,26–28 To date, the mechanisms underlying the changes in DNA methylation in cardiovascular aging and CVDs remain obscure and controversial. For instance, increased DNMT1 in macrophage is correlated with decreased PPAR-γ (peroxisome proliferator-activated receptor gamma) and increased proinflammatory cytokines in ApoE-knockout mice fed an atherogenic diet, as well as in patients with atherosclerosis.29 Consistently, the treatment of human umbilical vein endothelial cells with LDL (low-density lipoprotein) cholesterol—a major risk factor for coronary heart diseases—results in DNMT1 activation and aberrant gene expression. Conversely, DNMT1 expression is diminished in human atherosclerotic plaques.30

    Interestingly, the DNA methylation patterns in CVDs share some common features with developmental alterations. The methylation patterns of cardiomyocytes isolated from failing mouse hearts partially resemble those of neonatal mice.31 The 5-hmC landscape also shifts toward a neonatal-like distribution pattern in pathologically hypertrophic hearts. It has been suggested that TET2 may be a master epigenetic regulator of key cardiovascular genes, including MYH7, MYOCD, SRF, and KLF4.32 Additionally, the DNA hydroxymethylation level decreases on the induction of cardiac hypertrophy. Notably, the enrichment of 5-hmC at retrotransposon-LINEs is specific to hypertrophic conditions, indicating genomic instability.33 Yet, how TETs are implicated in cardiovascular aging and many other CVDs remains elusive.16,33

    Histone Modifications and Chromatin Remodeling

    Chromatin remodeling refers to the process by which the eukaryotic genomes wrap into nucleosome-based chromatin. Nucleosome remodeling changes the architecture of chromatin and affects the accessibility of DNA to elements facilitating transcription initiation and elongation, replication, recombination, and repair. One form of chromatin remodeling is the covalent modification of histones by histone acetyltransferases, deacetylases, methyltransferases, and kinases.7 Another form of chromatin remodeling is the noncovalent modification mediated by ATP-dependent chromatin remodeling complexes that physically exchange, move, or remove nucleosomes along DNA. Post-translational modifications (PTMs) of core histone components play vital roles in the activation and repression of gene transcription.34 Histone H2A, histone H2B, histone H3, and histone H4 can be modified by methylation, acetylation, ubiquitination, phosphorylation, SUMOylation, GlcNAcylation, carbonylation, and ADP-ribosylation, collectively constituting the histone code.7 The protein complexes that add, remove, and recognize various PTMs are referred to as histone writers, erasers, and readers, respectively.7,35 Growing evidence shows that the dysregulation of epigenetic regulators in histone modifications is a predisposing factor for cardiac aging and diseases.

    For monitoring histone modifications and binding of chromatin remodeling factors, chromatin immunoprecipitation combined with high-throughput sequencing is used to infer the distributions of proteins (such as core histone components with specific PTMs, transcriptional factors, and epigenetic enzymes) bound to or associated with DNA.36 This technique is a powerful tool to profile the genome-wide patterns of various histone modifications and to determine chromatin states, improving the current understanding of DNA-protein interactions at a genome-wide scale. In addition, the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat–associated 9) system serves as a platform for designing artificial transcriptional activators or repressors targeting almost any designated sequence and as an artificial bait for chromatin immunoprecipitation combined with high-throughput sequencing.

    Using new multiomics technologies, cardiomyocyte-specific global histone modification signature profiling in human failing hearts reveals that pathological gene expression is often accompanied by the alterations of active histone marks (H3K9ac, H3K27ac, H3K4me3, and H3K36me3) and repressive marks (H3K27me3).37 Chromatin immunoprecipitation combined with high-throughput sequencing against 7 types of histone marks in hypertrophic mouse cardiomyocytes shows that 3 marks are associated with active regulatory regions (H3K9ac, H3K27ac, and H3K4me3), 3 are associated with repressed regulatory regions (H3K9me2, H3K9me3, and H3K27me3), and 1 is preferentially distributed within transcribed genes (H3K79me2).37 Notably, 9.1% of the chromatin of these cardiomyocytes shows changes in the enrichment of at least 1 type of histone mark. These chromatin remodeling changes are responsible for the gene regulation contributing to cardiac hypertrophy. For example, the downregulation of genes clustered in oxidative stress and gene transcription regulation pathways are associated with epigenetic changes, including decreased H3K9ac, increased H3K9me3, and increased H3K27me3.37

    Histone Methylation and Demethylation

    H3K27me3—one of the most well-studied repressive marks—is governed by the histone methyltransferase of the PRC (polycomb repressive complex) 2, which contains the catalytic subunit EZH2 (enhancer of zeste homolog 2) and the H3K27-specific JmjC-domain demethylases UTX (lysine-specific demethylase 6A) and JMJD3 (lysine-specific demethylase 6B; Figure 2). EZH2 prevents cardiac pathology by repressing the expression of Six1—a homeodomain transcription factor functioning in cardiac progenitor cells that is silenced on cardiac differentiation (Figure 2).38 In contrast, the cardiomyocyte-specific overexpression of Rae28—a component of PRC1—causes cardiomyocyte apoptosis, abnormal myofibrils, and dilated cardiomyopathy.39 By comparison, an active histone mark, H3K79me3, is catalyzed by the histone-lysine N-methyltransferase DOT1L (histone H3-K79 methyltransferase) in mammals. DOT1L is downregulated in dilated cardiomyopathy; its cardiac-specific deletion results in the global loss of H3K79me2/3, including the loss of H3K79me2/3 enrichment on the DMD (dystrophin) gene, which encodes a membrane-associated protein whose mutation causes not only dilated cardiomyopathy but also muscular dystrophy. Consistently, DMD protein level is decreased 75% in DOT1L-deficient hearts that recapitulate the phenotypes in patients with dilated cardiomyopathy.40 Similarly, in patients with dilated cardiomyopathy, H3K9me2/3 decreases, whereas H3K4me2 increases with increased MLL3 (myeloid/lymphoid or mixed-lineage leukemia protein 3) in the LV. Implantation of an LV assist device reverses cardiomyocyte remodeling, along with the upregulation of the H3K9me3 methyltransferase SUV39H1 (suppressor of variegation 3-9 homolog 1) and the downregulation of the H3K9me3 JMJD demethylases (Figure 2).41,42

    Figure 2.

    Figure 2. Chromatin remodeling at open regions by removal of repressive marks. Chromatin is conceptualized into 2 structural states: (right) an open and active state with active histone modifications loosening the nucleosome and allowing for the binding of transcriptional machineries or (left) a condensed and inactive state with repressive histone modifications closing the nucleosome and blocking the accessibility of the transcriptional machinery to genes. Top, H3K9me2/3 (green dots) is catalyzed by histone methyltransferases G9a/GLP (histone-lysine N-methyltransferase EHMT1/2) and removed by the histone demethylase JMJD2A (lysine-specific demethylase 4A). Pathological hypertrophy drives the expression of miR-217 and thus blocks G9a/GLP, resulting in a reduction of the H3K9 methylation that impairs heterochromatin stability and derepresses the fetal gene program. In contrast, JMJD2A decreases H3K9me3, activates expression of a pressure overload-induced gene, and leads to postnatal myocardial pathology. Bottom, H3K27me3 (yellow dots) is catalyzed by EZH2 (enhancer of zeste homolog 2) and removed by UTX (lysine-specific demethylase 6A) or JMJD3 (lysine-specific demethylase 6B). Downregulation of EZH2 in adult hearts destabilizes cardiac gene expression and leads to the expression of fetal genes and profibrosis factors, causing postnatal myocardial pathology.

    In the atherosclerotic plaques of patients with carotid artery stenosis, changes in histone modifications facilitate the development and progression of atherosclerosis. There are significantly reduced H3K9 and H3K27 methylation levels, particularly in smooth muscle cells (SMCs) and lymphocytes. With the progression of atherosclerosis, H3K4 methylation increases in SMCs; H3K9ac and H3K27ac also increase in atherosclerotic SMCs and macrophages, correlating with the increase in histone acetyltransferase activity of GCN5L (GCN5-like protein 1) and MYST1 (histone acetyltransferase KAT8). In addition, in endothelial cells, H3K9ac increases in atherosclerotic plaques.43 Mutations in genes involved in epigenetic regulation have been associated with congenital heart defects.44,45 In particular, mutations in human LMNA cause laminopathies. The best-characterized laminopathy is Hutchinson-Gilford progeria syndrome (HGPS)45—a rare premature aging disease in which most patients die from atherosclerotic CVDs at ≈13 years of age. There are striking commonalities in the epigenetic defects between HGPS and physiological aging. In human stem cell-based HGPS models, the mark for facultative heterochromatin H3K27me3 and its methyltransferase EZH2 is diminished; the mark for constitutive heterochromatin H3K9me3 also decreases with its binding partner HP1α (chromobox protein homolog 5; also known as CBX5).46–50 Progressive disorganization of heterochromatin also occurs in a Werner Syndrome (WS)–specific stem cell model.50 Future systemic studies will depict the epigenetic changes in isogenic HGPS and WS stem cell models to elucidate the mechanisms underlying premature and normal aging and to identify effective treatments for atherosclerosis.50

    The aforementioned studies suggest that in cardiac aging, the loss of repressive marks and the gain of active marks may result in the upregulation of aging-promoting genes, aberrant activation of repetitive sequences, and an increase in transcriptional noise. These findings are consistent with the observation that at the single-cell level, cardiomyocytes isolated from 27-month-old mice exhibit considerable variations in the transcription levels of a panel of heart-specific housekeeping genes compared with those of young mice.51 The reversible nature of epigenetic modifications makes it possible to regulate and reverse some phenotypes by physical activity and dietary restriction.52–56 In a North Texas Health study, participants with high vegetable and fruit intake have higher LINE-1 methylation levels than those on a diet of mostly meats and carbohydrates.57 Flavonoids exhibit beneficial effects against heart diseases because of their effects on DNMT and a decrease in gene-specific methylation, demonstrating the potential of nonpharmacological adjuncts as therapies for aging individuals and patients with CVDs.58

    Although important roles for histone PTMs and chromatin remodeling in cardiovascular aging and CVDs have been revealed, how the histone code is regulated remains unknown. Continuous efforts are devoted to understanding how histone writers, erasers, and readers function in aging. For example, in normal adult hearts, the H3K9 dimethyltransferases G9a/GLP (histone-lysine N-methyltransferase EHMT1/2) interact with EZH2 and the transcription factor MEF2C (myocyte-specific enhancer factor 2C) to maintain the heterochromatin needed for repressing fetal genes,59 thus protecting against pathological cardiac hypertrophy. However, pathological hypertrophy drives the expression of miR-217 and blocks G9a/GLP, resulting in a reduction of H3K9me2, which unlocks the terminally differentiated state of cardiomyocytes and derepresses the fetal gene program, leading to pathological remodeling of the cardiomyocyte transcriptome.60 In another case, the forced expression of a H3K9me3 demethylase, JMJD2A (lysine-specific demethylase 4A), exacerbates cardiac hypertrophy under stress, whereas the loss of JMJD2A attenuates the hypertrophic response.61

    HDACs

    Another focus of the pioneering studies on histone modifications in cardiovascular aging includes HDACs (histone deacetylases). There are 4 distinct subtypes of HDACs: class I HDACs (HDACs 1, 2, 3, and 8), class II HDACs (IIa: HDACs 4, 5, 7, and 9; IIb: HDACs 6 and 10), class III nicotinamide adenine dinucleotide-dependent SIRT (sirtuin) enzymes (Sirt1–7), and the sole class IV enzyme HDAC 11.35,62 The activities of these HDACs are directly modulated by metabolic intermediates, such as nicotinamide adenine dinucleotide, acetyl-CoA, and β-hydroxybutyrate, which in turn transduce the impact to epigenetic changes.35,63 How these HDACs and SIRTs trigger the transcriptional shift in heart or vascular failure is partially elucidated.64

    Various studies in mouse models with HDAC deletion or overexpression have elucidated the function of HDACs in the regulation of cardiac hypertrophy. Simultaneous cardiomyocyte-specific deletion of HDAC1 and HDAC2 results in dilated cardiomyopathy and neonatal lethality.65 Mice with a cardiac-specific loss of HDAC3 develop massive cardiac hypertrophy and survive only 3 to 4 months.66 Mice lacking either HDAC5 or HDAC9 are sensitive to hypertrophic stimuli, and simultaneous knockout of both genes leads to spontaneous cardiac enlargement. Loss of HDAC7 results in the thinning of myocardial walls and dilation of atria.35 Thus, HDACs are critical for heart homeostasis. Notably, most of the aforementioned phenotypes attribute to abnormalities in the growth and maturation of the cardiomyocytes. To date, little is known on how specific HDACs regulate epigenetic modifications and affect the gene expression involved in aging and aging-related diseases. Sporadic studies demonstrate that HDAC9 expression is increased 3-fold in carotid plaques compared with that in normal arteries; a similar upregulation occurs in aortic and femoral plaques.67 In addition, various polymorphisms of HDAC9 are associated with a risk of large-vessel stroke via promoting atherosclerosis.68–70

    The beneficial effects of SIRTs as deacetylases against a variety of cardiovascular pathologies, including endothelial dysfunction, atherothrombosis, myocardial infarction, and LV hypertrophy, have been well documented and extensively discussed elsewhere64,71–78 and are, therefore, not discussed in detail in this review. Interestingly, 8 types of nonacetyl short-chain Lys acylations on histones utilizing acyl-CoA metabolites as substrates have been identified: succinylation, crotonylation, malonylation, butyrylation, propionylation, 2-hydroxyisobutyrylation, glutarylation, and β-hydroxybutyrylation.79 In addition to acting as deacetylases, SIRTs can mediate short-chain Lys deacylations, such as depropionylation and debutyrylation. In particular, SIRT5 has little deacetylase activity but is a robust desuccinylase. SIRT5 knockout mice develop hypertrophic cardiomyopathy with protein lysine succinylation predominantly accumulating in the heart. Further investigations will help understand how different SIRTs orchestrate the proportional mixture of distinct histone acylations in response to different metabolic stimuli.

    Chromatin Remodeling

    There are at least 4 distinct families of chromatin remodelers: SWI/SNF (switch/sucrose nonfermentable), ISWI (imitation SWI of drosophila melanogaster), NuRD (nucleosome remodeling deacetylase), and INO80 (chromatin-remodeling ATPase INO80). These complexes can dissect the original histone-DNA contacts, thus leading to nucleosome repositioning. Emerging evidence has implicated chromatin remodelers in cardiovascular aging and CVDs. For example, a Ser818Cys mutation in INO80D—a subunit of INO80—accelerates arterial aging in humans.80 In addition, several NuRD complex components are downregulated during HGPS premature and normal aging.45

    The most well-studied chromatin remodeling complex SWI/SNF, also called the BAF complex, consists of 12 protein subunits with either BRG1 (ATP-dependent helicase SMARCA4) or BRM (ATP-dependent helicase SMARCA2) as the essential ATPase subunit. Accumulating data have shown that the SWI/SNF complex is highly associated with cardiovascular function and pathology, positioning this complex as a potential therapeutic target for pathological cardiac hypertrophy and heart failure. A 70% decrease in BRG1 expression is reported in the myocardium of patients with congenital heart diseases. In mouse and human hypertrophic hearts, BRG1 is activated and assembles with G9a/GLP and DNMT3 to repress the expression of MYH6 (myosin-6) by H3K9/CpG methylation in its promoter region (Figure 3).81 BRG1 also interacts with its embryonic partners, PARP (poly ADP ribose polymerase) and HDACs, to regulate gene expression during cardiac hypertrophy in mice (Figure 3).82 In addition to BRG1, in hypertrophic hearts resulting from salt-induced hypertension, several other SWI/SNF subunits, including Baf180 and Baf60c, that are essential for heart development are upregulated.83 Moreover, alterations in the association of the SWI/SNF complex with genomic DNA have been observed in injured adult mouse hearts.84 Therefore, on BRG1 activation, all cardiac stimuli ultimately converge on the BAF-mediated regulation of key cardiac function genes, such as MHC, MYH6, and ABCA1, and the cardiac-specific lncRNA MHRT (myosin heavy chain-associated RNA transcript) to induce CVDs.85,86 Conversely, the inhibition of BRG1 or other SWI/SNF components, such as Baf60c and Baf180, blunts cardiac regeneration.87 The controversy between these findings suggests that the precise mechanisms of how the SWI/SNF complex contributes to the aging process require further investigations.

    Figure 3.

    Figure 3. BRG1 (ATP-dependent helicase SMARCA4)-mediated chromatin remodeling during cardiovascular aging. BRG1 (ATP-dependent helicase SMARCA2) is the central catalytic subunit of BAF (BRG1/Brm-associated factor) chromatin remodeling complexes. BRG1 forms complexes with G9a/GLP (histone-lysine N-methyltransferase EHMT1/2) and DNMT3 (DNA methyltransferase 3) to repress Myh6 (myosin-6) gene expression, inducing a fetal gene transition from Myh6 to Myh7. In addition, BRG1 interacts with the scaffolding lncRNA (long noncoding RNA) MANTIS to facilitate the transcription of angiogenesis genes, such as SOX18 (transcription factor SOX-18), SMAD6 (mothers against decapentaplegic homolog 6), and COUP-TFII (COUP transcription factor 2). On cardiovascular stress, BRG1 interacts with the chromatin-modifying enzymes HDAC (histone deacetylase) 2/9 and PARP (poly ADP ribose polymerase) to repress the transcription of a heart-specific lncRNA, MHRT (myosin heavy chain-associated RNA transcript), during cardiac hypertrophy. MHRT in turn binds to BRG1 and antagonizes BRG1 occupancy on its chromatinized DNA targets, thus preventing the pathological Myh6-to-Myh7 switch. MHRT also recruits HDAC5 to myocardin, resulting in deacetylation and inhibition of the transcription of smooth muscle genes, including Sm22α (transgelin), Acta2 (aortic smooth muscle), and Myh11 (myosin-11).

    In addition, BRG1 is upregulated in patients with aortic dissection and dilation. In vitro BRG1 overexpression upregulates genes clustered in RRAD (Ras-related associated with diabetes) and inhibits migration and proliferation of human SMCs.88 Another component of the SWI/SNF complex, Baf60a, is a diet-sensitive regulator of cholesterol homeostasis; liver-specific Baf60a inactivation protects mice from diet-induced atherosclerosis (Table 1).89

    Table 1. Key Epigenetic Regulators in Cardiovascular Aging and Diseases

    Epigenetic MarksFunctionEnzymeEnzyme ActivityChange in Epigenetic Mark With AgingSpeciesReference
    5mCRepressiveDNMT1DNA methyltransferaseIncreasedApoE-knockout mice and patients with atherosclerosis29
    DNMT1DNA methyltransferaseIncreasedHuman umbilical vein endothelial cells treated with LDL cholesterol26
    DNMT1DNA methyltransferaseDecreasedHuman atherosclerotic plaques30
    H3K9me3RepressiveG9a/GLPH3K9 dimethyltransferasesDecreasedMouse model of pathological hypertrophy81
    JMJD2AH3K9me3 demethylaseIncreasedMouse model of cardiac hypertrophy61
    HP1αH3K9me3-binding partnerDecreasedHuman stem cell-based HGPS models45,50
    H3K27me3RepressiveEZH2Histone H3K27 methyltransferaseDecreasedHuman stem cell-based HGPS models45
    H3K79me3RepressiveDOT1LHistone H3K79 methyltransferaseDecreasedPatients with dilated cardiomyopathy40
    H3K4me2ActiveMLL3Lysine N-methyltransferaseIncreasedPatients with dilated cardiomyopathy42
    H3K9ac and H3K27acActiveGCN5L and MYST1Histone acetyltransferaseIncreasedPatients with carotid artery stenosis43
    Chromatin remodelersNDBRG1Chromatin remodelersIncreasedPatients with aortic dissection and dilation88
    IncreasedMouse model of cardiac hypertrophy82

    5mC indicates 5-methylcytosine; BRG1, ATP-dependent helicase SMARCA4; DNMT1, DNA methyltransferase 1; DOT1L, histone H3-K79 methyltransferase; EZH2, enhancer of zeste homolog 2; G9a/GLP, histone-lysine N-methyltransferase EHMT1/2; GCN5l, GCN5-like protein 1; MLL3, myeloid/lymphoid or mixed-lineage leukemia protein 3; and MYST1, histone acetyltransferase KAT8.

    In summary, nucleosome dynamics, occupancy, and positioning within chromatin is influenced by chromatin remodelers. Enzymatic complexes, such as the BAF complex, reposition chromatin to facilitate access to condensed genomic DNA by gene-regulatory elements. The loosening state of chromatin regions may correlate with dysregulated gene transcription in cardiac aging and diseases. By using novel genome-wide chromatin conformation capture (Hi-C) and transposase-accessible chromatin sequencing technology, open chromatin landscapes during aging can be profiled, thereby enhancing current understanding of how dysregulation of a chromatin remodeling complex contributes to cardiovascular aging and diseases.

    miRNAs, lncRNAs, and Circular RNAs

    A large part of the human genome is transcribed into noncoding RNAs (ncRNAs). These ncRNAs play important roles in age-associated CVDs and have been viewed as potential biomarkers and therapeutic targets. There are several subfamilies of ncRNAs, such as linear small-size miRNAs (<200 nucleotides) and lncRNAs (>200 nucleotides), as well as circular RNAs (circRNAs) consisting of a covalently closed continuous loop. Deep RNA sequencing followed by analyses with different algorithms can be used to detect genome-wide mRNAs, miRNAs, lncRNAs, and circRNAs.90 Here, we mainly focus on lncRNAs and circRNAs in cardiac aging and diseases.

    LncRNAs

    lncRNAs regulate multiple biological pathways in cardiac development, aging, and diseases. Different from miRNAs, lncRNAs are poorly conserved across species. There are ≈100 000 of lncRNAs in the human genome, and many of these molecules are cardiac enriched.86 LncRNAs are classified into 6 categories based on their genomic locations, including the dominant intergenic lncRNAs between 2 protein-coding genes, intronic lncRNAs in the introns of protein-coding genes, sense and antisense lncRNAs in the sense or antisense strands of protein-coding genes, bidirectional promoter lncRNAs in promoter regions, and enhancer lncRNAs in the enhancer regions of the genome.

    In recent years, many studies have profiled the changes in lncRNAs in CVDs and aging by deep sequencing.91 lncRNA profiling of mouse hearts with myocardial infarction has identified hundreds of novel cardiac-specific lncRNAs proximal to cardiac-specific enhancers, contributing to maladaptive cardiac pathological remodeling.92 Some of these lncRNAs are conserved in humans and dynamically modulated in CVDs, including dilated cardiomyopathy and aortic stenosis. In nonischemic myocardial biopsies from patients with dilated ischemic cardiomyopathy and heart failure, 9 lncRNAs (CDKN2B-AS1/ANRIL, RMRP, RNY5, SOX2-OT, SRA1 EGOT, H19, HOTAIR, and LOC285194/TUSC7) are significantly modulated. RMRP, H19, and HOTAIR are also upregulated in hypertrophic hearts in a mouse model.93 H19 binds to miR-103/107 and targets the FADD (Fas-associated protein with death domain) pathway in a myocardial ischemia/reperfusion injury model,94 whereas HOTAIR is a competing endogenous RNA that regulates PTEN (phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase) by inhibiting miRNA-19 and miRNA 29b.93,95 Intriguingly, CDKN2B-AS1/ANRIL, HOTAIR, and LOC285194/TUSC7 are similarly upregulated in the peripheral blood mononuclear cells of these patients, suggesting their potential as novel blood-based predictive biomarkers for heart failure.96 Other candidate lncRNA markers include MIAT (myocardial infarction-associated transcript) and SENCR (smooth muscle and endothelial cell-enriched migration/differentiation-associated lncRNA), which is related to diastolic dysfunction and myocardial infarction.97,98 Single-cardiomyocyte nuclear transcriptomes of normal and failing hearts further reveal nodal lncRNAs as key regulators of cardiomyocyte cell cycle.99 Importantly, Wisper (Wisp2 superenhancer-associated RNA)—a cardiac fibroblast-enriched lncRNA—correlates with the severity of fibrosis in diseased human hearts. Wisper controls the expression of profibrotic lysyl hydroxylase 2, which in turn regulates extracellular matrix deposition and the proliferation and survival of heart fibroblasts. Depletion of Wisper attenuates cardiac fibrosis in mice,100 highlighting its therapeutic potential for cardiac fibrosis.100

    A majority of lncRNAs are present in the nucleus. Emerging evidence shows that lncRNAs recruit epigenetic factors and trigger chromatin remodeling, thereby leading to the activation or repression of genes in the nucleus.101 Expression of the Myheart or MHRT—a highly sequentially and structurally conserved lncRNA transcribed divergently to MYH6 and overlapping with MYH7 in the antisense direction—gradually increases from the fetus to adulthood and serves as a safeguard for cardiac health in mice and humans.102 MHRT binds to the helicase domain of BRG1, antagonizing BRG1 occupancy on its chromatin targets and suppressing the pathological Myh6-to-Myh7 switch, thus protecting mouse hearts from cardiomyopathies (Figure 3). Under pathological stress, activated BRG1 forms a chromatin repressor complex with HDAC2/9 and PARP to repress MHRT transcription, and thus the heart loses the protection from MHRT (Figure 3).102 In addition, MHRT ameliorates cardiac hypertrophy via myocardin—a strong smooth and cardiac muscle-specific transactivator that binds to promoters harboring CArG box motif and activates a panel of downstream muscle-specific genes, including TAGLN (transgelin), ACTA2 (aortic smooth muscle), and MYH11 (myosin-11). MHRT recruits HDAC5 to myocardin, resulting in the deacetylation or direct inhibition of MYOCD gene transcription (Figure 3). Conversely, as part of a positive feedback loop, myocardin can promote the expression of MHRT by binding to its promoter.103 In addition to MHRT, BRG1 interacts with MANTIS—a scaffolding lncRNA—to stabilize the SWI/SNF chromatin remodeling complex that mediates the transcription of important angiogenesis genes, such as SOX18 (transcription factor SOX-18), SMAD6 (mothers against decapentaplegic homolog 6), and COUP-TFII (COUP transcription factor 2), by ensuring efficient RNA polymerase II binding.104 Furthermore, a cardiac-enriched lncRNA, cardiac hypertrophy-associated epigenetic regulator, is implicated in maladaptive cardiac remodeling via direct interaction with the catalytic subunit of PRC2 and inhibition of histone H3 lysine 27 methylation, thus regulating genes involved in cardiac hypertrophy.105

    In summary, these paradigms for lncRNA-chromatin interactions show that lncRNAs play roles in shaping genome organization and controlling gene expression levels in the cardiovascular system.101,106 New technologies, such as CRISPR/Cas9-based gain/loss of function, RNA immunoprecipitation, and sequencing, will provide a comprehensive understanding of the lncRNA biology and identify therapeutic targets for the prevention and treatment of CVDs within reach.107 Given the different expression of lncRNAs among various species, it will be necessary to focus on the pathways regulated by lncRNAs or to directly study the lncRNAs in primates.

    circRNAs

    In recent years, circRNAs have added another layer of complexity to ncRNA biology. circRNAs are pervasively expressed in mammalian tissues and often act as transcriptional/translational regulators or miRNA sponges.90,108–111 Approximately 15 318 and 3017 cardiac circRNAs have been identified in the human and mouse, respectively.112 In human and mouse failing hearts, the abundance of circRNAs increases along with the re-expression of fetal genes, such as NPPA and MYH7.113 By microarray expression profiling, 63 differentially expressed circRNAs in diseased mouse hearts have been identified.114 Several circRNAs (such as circRNAs from CHD7 (chromodomain-helicase-DNA-binding protein 7) and DNAJC6 (putative tyrosine-protein phosphatase auxilin) are dynamically expressed in response to stress in human stem cell-derived cardiomyocytes.115 Although multiple studies have profiled the expression patterns of circRNAs in the cardiovascular system, their functions have yet to be defined.112

    The circAmotl1 is identified as a circRNA interacting with PDK1 (3-phosphoinositide-dependent protein kinase 1) and AKT1 (proline-rich AKT1 substrate 1), leading to phosphorylation and nuclear translocation of AKT1 and thus enhancing cardiomyocyte survival.116 This circRNA dramatically decreases in aged human hearts. Inversely, a circRNA generated from FOXO3 (Forkhead box protein O3), which forms a complex with p21 (cyclin-dependent kinase inhibitor 1) and CDK2 (cell division protein kinase 2) and abolishes the function of CDK2 to promote the cell cycle,117 is highly expressed in the heart tissues of aged mice and human. The ectopic expression of circFoxo3 induces cellular senescence in cultured mouse embryonic fibroblasts118 and aggravates cardiomyopathy, whereas silencing circFoxo3 alleviates these symptoms.

    A recent pioneering study has linked circRNAs to the expression of aging-associated genes. circANRIL is the first circRNA found in the p16INK4A locus.119–121 The expression of circANRIL is correlated with p16INK4A/p14ARF transcription, as well as the risk of atherosclerotic vascular diseases. Initially identified as a marker of aberrant RNA splicing with no specific functions, circANRIL binds to a core subunit of the 60S-preribosomal assembly factor and impairs ribosome biogenesis in vascular SMCs. Consequently, circANRIL induces nucleolar stress and p53 activation, thus slowing down the overproliferation of SMCs in atherosclerosis. In addition, the expression of circANRIL increases in patients with coronary artery diseases, indicative of an atheroprotective role.119 In addition to circANRIL, circRNA hsa_circ_0010729 targets the HIF-1α (hypoxia-inducible factor 1-alpha) pathway by sponging miR-186,122 and circRNA WDR77 (methylosome protein 50) targets FGF-2 (fibroblast growth factor 2) by antagonizing miR-124; both of these molecules regulate vascular endothelial cell proliferation (Table 2).123

    Table 2. Key lncRNAs and circRNAs in Cardiovascular Aging and Diseases

    lncRNAs or circRNAsChange With AgingTargetsSpeciesReference
    CDKN2B-AS1/ANRIL, RMRP, RNY5, SOX2-OT, SRA1 EGOT, H19, HOTAIR, and LOC285194/TUSC7UpregulatedNDLV biopsies of patients with dilated ischemic cardiomyopathy and heart failure93,95
    H19UpregulatedmiR-103/107Myocardial ischemia/reperfusion injury mouse model94
    HOTAIRUpregulatedmiRNA-19 and miRNA 29bHypertrophic hearts of a mouse model95
    HOTAIR and LOC285194/TUSC7UpregulatedNDPeripheral blood mononuclear cells of dilated ischemic cardiomyopathy patients95
    MIATUpregulatedNDPatients with well-controlled type 2 diabetes mellitus98
    SENCRUpregulatedNDPatients with well-controlled type 2 diabetes mellitus97
    WisperUpregulatedLysyl hydroxylase 2Murine model of myocardial infarction and heart tissue from human patients experiencing aortic stenosis100
    MHRTDownregulatedBRG1 and myocardinMouse and human cardiomyopathies102
    MANTISDownregulatedBRG1Patients with idiopathic pulmonary arterial hypertension104
    ChaerDownregulatedCatalytic subunit of PRC2Hearts from individuals or mouse models with dilated cardiomyopathy105
    circAmotl1DownregulatedPDK1 and AKT1Aged human hearts116
    circFoxo3Upregulatedp21 and CDK2Heart tissues of aged mice and humans117,118
    circANRILUpregulatedINK4/ARF transcription, 60S-preribosomal assembly factorPatients with coronary artery diseases119,120

    AKT1 indicates proline-rich AKT1 substrate 1; BRG1, ATP-dependent helicase SMARCA4; CDK2, cell division protein kinase 2; Chaer, cardiac hypertrophy-associated epigenetic regulator; circRNA, circular RNA; lncRNA, long noncoding RNA; LV, left ventricle; MHRT, myosin heavy chain-associated RNA transcript; MIAT, myocardial infarction-associated transcript; miRNA, microRNA; p21, cyclin-dependent kinase inhibitor 1; PDK1, 3-phosphoinositide-dependent protein kinase 1; PRC2, polycomb repressive complex 2; SENCR, smooth muscle and endothelial cell-enriched migration/differentiation-associated long noncoding RNA; and Wisper, Wisp2 superenhancer-associated RNA.

    circRNA is an extracellular, stable, and detectable molecule in the bloodstream, suggesting its potential as a candidate noninvasive biomarker of CVDs.124 In fact, some lncRNAs and circRNAs have not only been identified as vital biomarkers but also as promising therapeutic targets in heart diseases.124,125 Based on the global transcriptomic analyses of plasma RNAs from patients with myocardial infarction, circulating long noncoding RNA LIPCAR has been identified as a novel biomarker for cardiac remodeling and for predicting survival in patients with heart failure.126 In peripheral blood mononuclear cells, Hsa_circ_0005836, hsa_circRNA_025016, and hsa_circ_0124644 were identified as diagnostic biomarkers for various CVDs.127,128 With increasingly more functions of circRNAs discovered, the use of biochemical enrichment strategies and deep sequencing coupled with novel bioinformatics approaches will further provide a comprehensive understanding of circRNAs in mammalian cardiac aging and diseases.

    mRNA Modifications—a New Era of the Epigenome

    In recent years, the discovery of mRNA modifications has opened a new realm of posttranslational regulations, also called the RNA code or epitranscriptome. N6-methyladenosine (m6A), referring to the methylation of the adenosine base at the nitrogen-6 position, is the most prevalent internal mRNA modification linked to increased RNA stability and translational efficiency and accuracy.52,53,129–135 The relationship between m6A and CVDs is just emerging. It is particularly intriguing that polymorphisms of the FTO (fat mass and obesity associated) gene—an m6A demethylase—are risk factors not only for obesity and metabolic abnormalities but also for CVDs.136,137 FTO protein level decreases with increased global m6A in ischemic mouse and human hearts; by comparison, AAV-mediated FTO overexpression significantly improves cardiac function in mice.138 Recent evidence shows that the NSun2 (tRNA [cytosine(34)-C(5))-methyltransferase]) methylates ICAM-1 (intercellular adhesion molecule 1) mRNA in the 5' untranslated region and 3' untranslated region, as well as the coding sequence, promoting ICAM-1 protein expression—a feature of the inflammatory response in endothelial cells.139

    Another conserved and pervasive form of RNA modification, adenosine-to-inosine RNA editing,140,141 also contributes to atherosclerosis. In endothelial cells, adenosine deaminase ADAR1 (double-stranded RNA-specific adenosine deaminase) acts on CTSS (cathepsin S) mRNA,142 promoting the recruitment of HuR (human antigen R) and thereby stabilizing CTSS mRNA. ADAR1 knockdown downregulates CTSS transcripts, whereas ADAR1 overexpression increases the adenosine-to-inosine RNA modification of CTSS mRNA and the expression of CTSS protein. Increased adenosine-to-inosine RNA editing of CTSS is observed in patients with atherosclerotic diseases.142 Our knowledge about the biological implications of epitranscriptomic regulations and especially their spatiotemporal changes under various physiological and pathological conditions is still limited.

    To profile the changes in epitranscriptomic marks during cardiovascular aging, DNA–RNA immunoprecipitation followed by sequencing has identified thousands of ncRNAs in the human genome.143 Chromatin isolation by RNA purification may also be used to intersect interactomes of RNA and chromatin at genome scale. In fact, it has already been applied to genomic mapping of long ncRNA occupancy in the genome.144 In addition, a variety of novel sequencing technologies, such as m6A-seq, MeRIP-seq, PA-m6A-seq, miCLIP, and m6A-LAIC-seq can be used to further depict the m6A modification landscape of cardiovascular tissues in a transcriptome-wide manner and reveal the fundamental roles of RNA modifications in cardiac aging and diseases.133

    Omics Technologies and Systemic Genomics Approaches

    With the aforementioned similarities in chromatin epigenetics between cardiovascular aging and CVDs, it would be interesting to use novel 3-dimensional genome techniques to investigate whether aged cardiomyocytes or vascular cells are associated with a plastic chromatin architecture. Advances in multiomics technologies, encompassing genomics, methylomics, epigenomics, transcriptomics, proteomics, and metabolomics will facilitate the revolutionization of the current understanding of epigenetics by probing chromatin remodeling at unprecedented spatial and temporal resolutions.145–147

    Long-range chromatin interactions and chromatin accessibility are robustly associated with gene expression regulation. By using new genomic tools, condensed or relaxed chromatin compartments can be mapped genome wide. For example, active regulatory regions, such as promoters and enhancers, located in open regions can be systemically identified across the human genome.36 Hi-C and chromatin interaction analysis with paired-end-tag sequencing are widely used to analyze chromatin interactions. Hi-C illustrates an accurate 3-dimensional architecture on a genome-wide scale, whereas chromatin interaction analysis with paired-end-tag sequencing reveals information about the 3-dimensional genome organization governed by a particular transcriptional factor or epigenetic regulator. Additionally, DNA adenine methyltransferase identification, sequencing of micrococcal nuclease sensitive sites, DNase I hypersensitive site sequencing, and transposase-accessible chromatin sequencing assays are all powerful and sensitive methods to probe genome-wide chromatin accessibility.36 High-resolution mapping of the 3-dimensional epigenome by Hi-C has suggested a plastic epigenome in failing hearts. A global structural remodeling of chromatin, including a significant decrease in surrounding local chromatin interactions and remodeling of long-range interactions of cardiac enhancers, underpins heart failure.148 Notably, cardiac-specific knockout of CTCF (CCCTC-binding factor)—an insulator protein in vertebrates—is sufficient to induce heart failure.148

    Every step in epigenetic regulation is governed by corresponding enzymes. Enzymes that alter nucleosome structure or position are at the center of gene and genome regulation. Cutting-edge proteomic technologies are used to detect global changes in the expression levels, PTMs (methylation, acetylation, succinylation, crotonylation, malonylation, and so forth), and interacting factors of these key epigenetic enzymes during cardiac aging and diseases. In particular, high-resolution, mass spectrometry-based proteomics further enables proteome-wide characterization and quantification of acetylation, ushering in the age of acetylomics.

    Bulk cells have primarily been used in omics analyses during the last decades. However, different types of cells in the cardiovascular system may be highly heterogeneous during aging and disease progression. Single-cell genomics, such as massively parallel single-cell RNA-seq, facilitate detailed transcriptome analysis to identify variants of key epigenetic enzymes/pathways in specific diseased cohorts or cell types.54,57,58,146 Altogether, new sequencing technologies have generated vast amounts of data. Systemic genomics approaches with metadimensional and multistaged algorithms will help depict a comprehensive picture of how multilayers of hierarchal chromatin organization govern gene regulation.

    Conclusions and Perspectives

    The current knowledge of the role of epigenetics in cardiovascular aging and diseases has substantially increased in the last decade. It has become clear that alterations in chromatin remodeling, DNA occupancy, and ncRNA expression all contribute to the progress of cardiovascular aging and diseases. However, our understanding of epigenetic regulation during aging in cardiovascular system remains fragmentary; it is debatable whether the overall changes in the epigenetic patterns return to an embryonic state or only shift to open chromatin. In illustrating the epigenetic landscape during pathological and physiological aging, the current studies have focused on the pathogenesis of CVDs but not merely the aging process itself. How aging-related epigenetic changes directly contribute to the transformation in CVDs remains to be unraveled. In addition, epigenetic factors co-occur and are closely interconnected during aging, but which are the primary driving forces and which are just consequences? Furthermore, how environmental stimuli, such as smoking and drinking, influence chromatin architecture in cardiac aging and diseases remains obscure. Environmental stimuli may influence the ROS (reactive oxygen species) level or DNA damage to affect the epigenetic stage or change the level of metabolites, such as nicotinamide adenine dinucleotide, acetyl-coenzyme A, or S-adenosylmethionine, to regulate the activity of chromatin-modifying enzymes.55 Novel multiomics technologies combined with integrative approaches make it feasible to profile different layers of epigenetic landscapes in a spatiotemporal manner, which will be invaluable in understanding the molecular mechanisms involved in cardiac aging and diseases. Time-resolved analyses of epigenetic responses to external stimuli illustrate the environment-epigenome interactions that modulate aging. Moreover, CRISPR/Cas9 editing technology makes it feasible to target specific epigenetic enzymes and manipulate epigenetic pathways in a locus-specific manner, facilitating the elucidation of the roles of particular epigenetic modifications. Isogeneic human stem cells can be created and differentiated into heart or vessel cells carrying mutations of interest from patients with CVDs.46,48–50,56,149,150 New findings can be validated in vivo in human cardiac tissues or at least in nonhuman primates. For future clinical applications, profiling studies of large cohorts is needed to investigate the feasibility of using epigenetic markers for the prognosis and diagnosis of human CVDs. Hopefully, these combined approaches will provide a deeper understanding of the mechanisms underlying cardiovascular aging and diseases and eventually facilitate the development of effective interventions for human cardiovascular disorders.

    Nonstandard Abbreviations and Acronyms

    5-hmC

    5-hydroxymethylcytosine

    BAF

    BRG1/Brm-associated factor

    circRNA

    circular RNA

    CRISPR/Cas9

    clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat–associated 9

    CTCF

    CCCTC-binding factor

    CTSS

    cathepsin S

    CVD

    cardiovascular disease

    DNMT

    DNA methyltransferase

    eNOS

    endothelial NO synthase

    FADD

    Fas-associated protein with death domain

    FTO

    fat mass and obesity associated

    HDAC

    histone deacetylase

    HGPS

    Hutchinson-Gilford progeria syndrome

    HuR

    human antigen R

    LDL

    low-density lipoprotein

    lncRNA

    long noncoding RNA

    LV

    left ventricle

    m6A

    N6-methyladenosine

    MHRT

    myosin heavy chain-associated RNA transcript

    MIAT

    myocardial infarction-associated transcript

    miRNA

    microRNA

    ncRNA

    noncoding RNA

    NSun2

    NOP2/Sun domain family, member 2

    NuRD

    nucleosome remodeling deacetylase

    PARP

    poly ADP ribose polymerase

    PRC

    polycomb repressive complex

    PTM

    modification

    RRAD

    Ras-related associated with diabetes

    SENCR

    smooth muscle and endothelial cell-enriched migration/differentiation-associated long noncoding RNA

    SIRT

    sirtuin

    SMC

    smooth muscle cell

    SWI/SNF

    switch/sucrose nonfermentable

    TET

    ten-eleven translocation

    Wisper

    Wisp2 superenhancer-associated RNA

    Acknowledgments

    Because of limitations of space, we apologized for not being able to cite all important studies in this review. This work was supported by the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA16010100), the National Key Research and Development Program of China (2015CB964800, 2017YFA0103304, 2017YFA0102802, 2014CB910503, and 2018YFA0107203), the National Natural Science Foundation of China (grant No. 81625009, 81330008, 91749202, 91749123, 81371342, 81471414, 81422017, 31671429, 81601233, 81671377, 31601109, 31601158, 81771515, 81822018, 81870228, 81801399, 81801370, 31801010, and 81701388), the National High Technology Research and Development Program of China (2015AA020307), CAS (KJZDEW-TZ-L05 and CXJJ-16M271), Program of Beijing Municipal Science and Technology Commission (Z151100003915072), the Thousand Young Talents Program of China, Advanced Innovation Center for Human Brain Protection (117212), and Beijing Municipal Commission of Health and Family Planning (PXM2018_026283_000002).

    Footnotes

    *These authors contributed equally to this work.

    Correspondence to Guang-Hui Liu, PhD, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China, Email
    Jing Qu, PhD, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China, Email

    References

    • 1. Stern S, Behar S, Gottlieb S. Cardiology patient pages. Aging and diseases of the heart.Circulation. 2003; 108:e99–e101. doi: 10.1161/01.CIR.0000086898.96021.B9LinkGoogle Scholar
    • 2. Humphrey JD, Milewicz DM. Aging, smooth muscle vitality, and aortic integrity.Circ Res. 2017; 120:1849–1851. doi: 10.1161/CIRCRESAHA.117.311075LinkGoogle Scholar
    • 3. Ordovás JM, Smith CE. Epigenetics and cardiovascular disease.Nat Rev Cardiol. 2010; 7:510–519. doi: 10.1038/nrcardio.2010.104CrossrefMedlineGoogle Scholar
    • 4. Greco CM, Condorelli G. Epigenetic modifications and noncoding RNAs in cardiac hypertrophy and failure.Nat Rev Cardiol. 2015; 12:488–497. doi: 10.1038/nrcardio.2015.71CrossrefMedlineGoogle Scholar
    • 5. Venkatesh S, Workman JL. Histone exchange, chromatin structure and the regulation of transcription.Nat Rev Mol Cell Biol. 2015; 16:178–189. doi: 10.1038/nrm3941CrossrefMedlineGoogle Scholar
    • 6. Verdin E, Ott M. 50 years of protein acetylation: from gene regulation to epigenetics, metabolism and beyond.Nat Rev Mol Cell Biol. 2015; 16:258–264. doi: 10.1038/nrm3931CrossrefMedlineGoogle Scholar
    • 7. Strahl BD, Allis CD. The language of covalent histone modifications.Nature. 2000; 403:41–45. doi: 10.1038/47412CrossrefMedlineGoogle Scholar
    • 8. Gillette TG, Hill JA. Readers, writers, and erasers.Circ Res. 2015; 116:1245.LinkGoogle Scholar
    • 9. Han P, Hang CT, Yang J, Chang CP. Chromatin remodeling in cardiovascular development and physiology.Circ Res. 2011; 108:378–396. doi: 10.1161/CIRCRESAHA.110.224287LinkGoogle Scholar
    • 10. Dzeshka MS, Lip GY, Snezhitskiy V, Shantsila E. Cardiac fibrosis in patients with atrial fibrillation: mechanisms and clinical implications.J Am Coll Cardiol. 2015; 66:943–959. doi: 10.1016/j.jacc.2015.06.1313CrossrefMedlineGoogle Scholar
    • 11. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging.Cell. 2013; 153:1194–1217. doi: 10.1016/j.cell.2013.05.039CrossrefMedlineGoogle Scholar
    • 12. Sack MN, Fyhrquist FY, Saijonmaa OJ, Fuster V, Kovacic JC. Basic biology of oxidative stress and the cardiovascular system: Part 1 of a 3-Part Series.J Am Coll Cardiol. 2017; 70:196–211. doi: 10.1016/j.jacc.2017.05.034CrossrefMedlineGoogle Scholar
    • 13. Cavalcante JL, Lima JA, Redheuil A, Al-Mallah MH. Aortic stiffness: current understanding and future directions.J Am Coll Cardiol. 2011; 57:1511–1522. doi: 10.1016/j.jacc.2010.12.017CrossrefMedlineGoogle Scholar
    • 14. Smith ZD, Meissner A. DNA methylation: roles in mammalian development.Nat Rev Genet. 2013; 14:204–220. doi: 10.1038/nrg3354CrossrefMedlineGoogle Scholar
    • 15. Lev Maor G, Yearim A, Ast G. The alternative role of DNA methylation in splicing regulation.Trends Genet. 2015; 31:274–280. doi: 10.1016/j.tig.2015.03.002CrossrefMedlineGoogle Scholar
    • 16. Liu R, Jin Y, Tang WH, Qin L, Zhang X, Tellides G, Hwa J, Yu J, Martin KA. Ten-eleven translocation-2 (TET2) is a master regulator of smooth muscle cell plasticity.Circulation. 2013; 128:2047–2057. doi: 10.1161/CIRCULATIONAHA.113.002887LinkGoogle Scholar
    • 17. Yu M, Hon GC, Szulwach KE, Song CX, Zhang L, Kim A, Li X, Dai Q, Shen Y, Park B, Min JH, Jin P, Ren B, He C. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome.Cell. 2012; 149:1368–1380. doi: 10.1016/j.cell.2012.04.027CrossrefMedlineGoogle Scholar
    • 18. Booth MJ, Ost TW, Beraldi D, Bell NM, Branco MR, Reik W, Balasubramanian S. Oxidative bisulfite sequencing of 5-methylcytosine and 5-hydroxymethylcytosine.Nat Protoc. 2013; 8:1841–1851. doi: 10.1038/nprot.2013.115CrossrefMedlineGoogle Scholar
    • 19. Horvath S. DNA methylation age of human tissues and cell types.Genome Biol. 2013; 14:R115. doi: 10.1186/gb-2013-14-10-r115CrossrefMedlineGoogle Scholar
    • 20. Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock.Cell. 2012; 148:46–57. doi: 10.1016/j.cell.2012.01.003CrossrefMedlineGoogle Scholar
    • 21. Wilson R, Wahl S, Pfeiffer L, Ward-Caviness CK, Kunze S, Kretschmer A, Reischl E, Peters A, Gieger C, Waldenberger M. The dynamics of smoking-related disturbed methylation: a two time-point study of methylation change in smokers, non-smokers and former smokers.BMC Genomics. 2017; 18:805. doi: 10.1186/s12864-017-4198-0CrossrefMedlineGoogle Scholar
    • 22. Lind L, Ingelsson E, Sundstrom J, Siegbahn A, Lampa E. Methylation-based estimated biological age and cardiovascular disease.Eur J Clin Invest. 2018; 48:eci.12872.CrossrefGoogle Scholar
    • 23. Jiang YZ, Manduchi E, Stoeckert CJ, Davies PF. Arterial endothelial methylome: differential DNA methylation in athero-susceptible disturbed flow regions in vivo.BMC Genomics. 2015; 16:506. doi: 10.1186/s12864-015-1656-4CrossrefMedlineGoogle Scholar
    • 24. Zaina S. Unraveling the DNA methylome of atherosclerosis.Curr Opin Lipidol. 2014; 25:148–153. doi: 10.1097/MOL.0000000000000059CrossrefMedlineGoogle Scholar
    • 25. Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, Condom E, Ramírez-Ruz J, Gomez A, Gonçalves I, Moran S, Esteller M. DNA methylation map of human atherosclerosis.Circ Cardiovasc Genet. 2014; 7:692–700. doi: 10.1161/CIRCGENETICS.113.000441LinkGoogle Scholar
    • 26. Kumar A, Kumar S, Vikram A, Hoffman TA, Naqvi A, Lewarchik CM, Kim YR, Irani K. Histone and DNA methylation-mediated epigenetic downregulation of endothelial Kruppel-like factor 2 by low-density lipoprotein cholesterol.Arterioscler Thromb Vasc Biol. 2013; 33:1936–1942. doi: 10.1161/ATVBAHA.113.301765LinkGoogle Scholar
    • 27. Zhou J, Li YS, Wang KC, Chien S. Epigenetic mechanism in regulation of endothelial function by disturbed flow: induction of DNA hypermethylation by DNMT1.Cell Mol Bioeng. 2014; 7:218–224. doi: 10.1007/s12195-014-0325-zCrossrefMedlineGoogle Scholar
    • 28. Jiang YZ, Jiménez JM, Ou K, McCormick ME, Zhang LD, Davies PF. Hemodynamic disturbed flow induces differential DNA methylation of endothelial Kruppel-Like Factor 4 promoter in vitro and in vivo.Circ Res. 2014; 115:32–43. doi: 10.1161/CIRCRESAHA.115.303883LinkGoogle Scholar
    • 29. Yu J, Qiu Y, Yang J, Bian S, Chen G, Deng M, Kang H, Huang L. DNMT1-PPARγ pathway in macrophages regulates chronic inflammation and atherosclerosis development in mice.Sci Rep. 2016; 6:30053. doi: 10.1038/srep30053CrossrefMedlineGoogle Scholar
    • 30. Xie SA, Zhang T, Wang J, Zhao F, Zhang YP, Yao WJ, Hur SS, Yeh YT, Pang W, Zheng LS, Fan YB, Kong W, Wang X, Chiu JJ, Zhou J. Matrix stiffness determines the phenotype of vascular smooth muscle cell in vitro and in vivo: role of DNA methyltransferase 1.Biomaterials. 2018; 155:203–216. doi: 10.1016/j.biomaterials.2017.11.033CrossrefMedlineGoogle Scholar
    • 31. Gilsbach R, Preissl S, Grüning BA, Schnick T, Burger L, Benes V, Würch A, Bönisch U, Günther S, Backofen R, Fleischmann BK, Schübeler D, Hein L. Dynamic DNA methylation orchestrates cardiomyocyte development, maturation and disease.Nat Commun. 2014; 5:5288. doi: 10.1038/ncomms6288CrossrefMedlineGoogle Scholar
    • 32. Liu R, Jin Y, Tang WH, Qin L, Zhang X, Tellides G, Hwa J, Yu J, Martin KA. Ten-eleven translocation-2 (TET2) is a master regulator of smooth muscle cell plasticity.Circulation. 2013: 128:2047–2057. doi: 10.1161/CIRCULATIONAHA.113.002887LinkGoogle Scholar
    • 33. Greco CM, Kunderfranco P, Rubino M, Larcher V, Carullo P, Anselmo A, Kurz K, Carell T, Angius A, Latronico MV, Papait R, Condorelli G. DNA hydroxymethylation controls cardiomyocyte gene expression in development and hypertrophy.Nat Commun. 2016; 7:12418. doi: 10.1038/ncomms12418CrossrefMedlineGoogle Scholar
    • 34. Sen P, Shah PP, Nativio R, Berger SL. Epigenetic mechanisms of longevity and aging.Cell. 2016; 166:822–839. doi: 10.1016/j.cell.2016.07.050CrossrefMedlineGoogle Scholar
    • 35. Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy.Nat Rev Genet. 2009; 10:32–42. doi: 10.1038/nrg2485CrossrefMedlineGoogle Scholar
    • 36. Risca VI, Greenleaf WJ. Unraveling the 3D genome: genomics tools for multiscale exploration.Trends Genet. 2015; 31:357–372. doi: 10.1016/j.tig.2015.03.010CrossrefMedlineGoogle Scholar
    • 37. Papait R, Cattaneo P, Kunderfranco P, Greco C, Carullo P, Guffanti A, Viganò V, Stirparo GG, Latronico MV, Hasenfuss G, Chen J, Condorelli G. Genome-wide analysis of histone marks identifying an epigenetic signature of promoters and enhancers underlying cardiac hypertrophy.Proc Natl Acad Sci USA. 2013; 110:20164–20169. doi: 10.1073/pnas.1315155110CrossrefMedlineGoogle Scholar
    • 38. 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. 2012; 44:343–347. doi: 10.1038/ng.1068CrossrefMedlineGoogle Scholar
    • 39. Koga H, Kaji Y, Nishii K, Shirai M, Tomotsune D, Osugi T, Sawada A, Kim JY, Hara J, Miwa T, Yamauchi-Takihara K, Shibata Y, Takihara Y. Overexpression of Polycomb-group gene rae28 in cardiomyocytes does not complement abnormal cardiac morphogenesis in mice lacking rae28 but causes dilated cardiomyopathy.Lab Invest. 2002; 82:375–385.CrossrefMedlineGoogle Scholar
    • 40. 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. 2011; 25:263–274. doi: 10.1101/gad.2018511CrossrefMedlineGoogle Scholar
    • 41. Ito E, Miyagawa S, Fukushima S, Yoshikawa Y, Saito S, Saito T, Harada A, Takeda M, Kashiyama N, Nakamura Y, Shiozaki M, Toda K, Sawa Y. Histone modification is correlated with reverse left ventricular remodeling in nonischemic dilated cardiomyopathy.Ann Thorac Surg. 2017; 104:1531–1539. doi: 10.1016/j.athoracsur.2017.04.046CrossrefMedlineGoogle Scholar
    • 42. Jiang DS, Yi X, Li R, Su YS, Wang J, Chen ML, et al. The histone methyltransferase mixed lineage leukemia (MLL) 3 may play a potential role on clinical dilated cardiomyopathy.Mol Med. 2017; 23:196–203. doi: 10.2119/molmed.2017.00012CrossrefMedlineGoogle Scholar
    • 43. Greißel A, Culmes M, Burgkart R, Zimmermann A, Eckstein HH, Zernecke A, Pelisek J. Histone acetylation and methylation significantly change with severity of atherosclerosis in human carotid plaques.Cardiovasc Pathol. 2016; 25:79–86. doi: 10.1016/j.carpath.2015.11.001CrossrefMedlineGoogle Scholar
    • 44. Ashleigh AR, Vidu G. Genetics of congenital heart disease.Curr Cardiol Rev. 2010; 6:91–97.CrossrefMedlineGoogle Scholar
    • 45. Kubben N, Misteli T. Shared molecular and cellular mechanisms of premature ageing and ageing-associated diseases.Nat Rev Mol Cell Biol. 2017; 18:595–609. doi: 10.1038/nrm.2017.68CrossrefMedlineGoogle Scholar
    • 46. Kubben N, Zhang W, Wang L, Voss TC, Yang J, Qu J, Liu GH, Misteli T. Repression of the antioxidant NRF2 pathway in premature aging.Cell. 2016; 165:1361–1374. doi: 10.1016/j.cell.2016.05.017CrossrefMedlineGoogle Scholar
    • 47. Li Y, Zhang W, Chang L, et al. Vitamin C alleviates aging defects in a stem cell model for Werner syndrome.Protein Cell. 2016; 7:478–488. doi: 10.1007/s13238-016-0278-1CrossrefMedlineGoogle Scholar
    • 48. Liu GH, Barkho BZ, Ruiz S, et al. Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome.Nature. 2011; 472:221–225. doi: 10.1038/nature09879CrossrefMedlineGoogle Scholar
    • 49. Ren R, Ocampo A, Liu GH, Izpisua Belmonte JC. Regulation of stem cell aging by metabolism and epigenetics.Cell Metab. 2017; 26:460–474. doi: 10.1016/j.cmet.2017.07.019CrossrefMedlineGoogle Scholar
    • 50. Wu Z, Zhang W, Song M, Wang W, Wei G, Li W, Lei J, Huang Y, Sang Y, Chan P, Chen C, Qu J, Suzuki K, Belmonte JCI, Liu GH. Differential stem cell aging kinetics in hutchinson-gilford progeria syndrome and werner syndrome.Protein Cell. 2018: 9;333–350. doi: 10.1007/s13238-018-0517-8CrossrefMedlineGoogle Scholar
    • 51. Bahar R, Hartmann CH, Rodriguez KA, Denny AD, Busuttil RA, Dollé ME, Calder RB, Chisholm GB, Pollock BH, Klein CA, Vijg J. Increased cell-to-cell variation in gene expression in ageing mouse heart.Nature. 2006; 441:1011–1014. doi: 10.1038/nature04844CrossrefMedlineGoogle Scholar
    • 52. Lewis CJ, Pan T, Kalsotra A. RNA modifications and structures cooperate to guide RNA–protein interactions.Nat Rev Mol Cell Biol. 2017; 18:202–210. doi: 10.1038/nrm.2016.163CrossrefMedlineGoogle Scholar
    • 53. Chi KR. The RNA code comes into focus.Nature. 2017; 542:503.CrossrefMedlineGoogle Scholar
    • 54. Vanlandewijck M, He L, Mäe MA, et al. A molecular atlas of cell types and zonation in the brain vasculature.Nature. 2018; 554:475–480. doi: 10.1038/nature25739CrossrefMedlineGoogle Scholar
    • 55. Etchegaray JP, Mostoslavsky R. Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes.Mol Cell. 2016; 62:695–711. doi: 10.1016/j.molcel.2016.05.029CrossrefMedlineGoogle Scholar
    • 56. Wang S, Hu B, Ding Z, et al. ATF6 safeguards organelle homeostasis and cellular aging in human mesenchymal stem cells.Cell Discov. 2018; 4:2. doi: 10.1038/s41421-017-0003-0CrossrefMedlineGoogle Scholar
    • 57. Hupe M, Li MX, Kneitz S, Davydova D, Yokota C, Kele J, Hot B, Stenman JM, Gessler M. Gene expression profiles of brain endothelial cells during embryonic development at bulk and single-cell levels.Sci Signal. 2017; 10:eaag2476. doi: 10.1126/scisignal.aag2476CrossrefMedlineGoogle Scholar
    • 58. Khyzha N, Alizada A, Wilson MD, Fish JE. Epigenetics of atherosclerosis: emerging mechanisms and methods.Trends Mol Med. 2017; 23:332–347. doi: 10.1016/j.molmed.2017.02.004CrossrefMedlineGoogle Scholar
    • 59. 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. 2017; 136:1233–1246. doi: 10.1161/CIRCULATIONAHA.117.028561LinkGoogle Scholar
    • 60. 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. 2017; 127:335–348. doi: 10.1172/JCI88353CrossrefMedlineGoogle Scholar
    • 61. Zhang QJ, Chen HZ, Wang L, Liu DP, Hill JA, Liu ZP. The histone trimethyllysine demethylase JMJD2A promotes cardiac hypertrophy in response to hypertrophic stimuli in mice.J Clin Invest. 2011; 121:2447–2456. doi: 10.1172/JCI46277CrossrefMedlineGoogle Scholar
    • 62. McKinsey TA. Therapeutic potential for HDAC inhibitors in the heart.Annu Rev Pharmacol Toxicol. 2012; 52:303–319. doi: 10.1146/annurev-pharmtox-010611-134712CrossrefMedlineGoogle Scholar
    • 63. Choudhary C, Weinert BT, Nishida Y, Verdin E, Mann M. The growing landscape of lysine acetylation links metabolism and cell signalling.Nat Rev Mol Cell Biol. 2014; 15:536–550. doi: 10.1038/nrm3841CrossrefMedlineGoogle Scholar
    • 64. Hewitson R, Dargan J, Collis D, Green A, Moorjani N, Ohri S, Townsend PA. Heart failure: the pivotal role of histone deacetylases.Int J Biochem Cell Biol. 2013; 45:448–453. doi: 10.1016/j.biocel.2012.11.006CrossrefMedlineGoogle Scholar
    • 65. Montgomery RL, Davis CA, Potthoff MJ, Haberland M, Fielitz J, Qi X, Hill JA, Richardson JA, Olson EN. Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility.Genes Dev. 2007; 21:1790–1802. doi: 10.1101/gad.1563807CrossrefMedlineGoogle Scholar
    • 66. Montgomery RL, Potthoff MJ, Haberland M, Qi X, Matsuzaki S, Humphries KM, Richardson JA, Bassel-Duby R, Olson EN. Maintenance of cardiac energy metabolism by histone deacetylase 3 in mice.J Clin Invest. 2008; 118:3588–3597. doi: 10.1172/JCI35847CrossrefMedlineGoogle Scholar
    • 67. Markus HS, Mäkelä KM, Bevan S, Raitoharju E, Oksala N, Bis JC, O’Donnell C, Hainsworth A, Lehtimäki T. Evidence HDAC9 genetic variant associated with ischemic stroke increases risk via promoting carotid atherosclerosis.Stroke. 2013; 44:1220–1225. doi: 10.1161/STROKEAHA.111.000217LinkGoogle Scholar
    • 68. Zhou X, Guan T, Li S, Jiao Z, Lu X, Huang X, Ji Y, Ji Q. The association between HDAC9 gene polymorphisms and stroke risk in the Chinese population: a meta-analysis.Sci Rep. 2017; 7:41538. doi: 10.1038/srep41538CrossrefMedlineGoogle Scholar
    • 69. Qingxu G, Yan Z, Jiannan X, Yunlong L. Association between the gene polymorphisms of HDAC9 and the risk of atherosclerosis and ischemic stroke.Pathol Oncol Res. 2016; 22:103–107. doi: 10.1007/s12253-015-9978-8CrossrefMedlineGoogle Scholar
    • 70. Wang XB, Han YD, Sabina S, Cui NH, Zhang S, Liu ZJ, Li C, Zheng F. HDAC9 variant Rs2107595 modifies susceptibility to coronary artery disease and the severity of coronary atherosclerosis in a Chinese Han population.PLoS One. 2016; 11:e0160449. doi: 10.1371/journal.pone.0160449MedlineGoogle Scholar
    • 71. Winnik S, Auwerx J, Sinclair DA, Matter CM. Protective effects of sirtuins in cardiovascular diseases: from bench to bedside.Eur Heart J. 2015; 36:3404–3412. doi: 10.1093/eurheartj/ehv290CrossrefMedlineGoogle Scholar
    • 72. Favero G, Franceschetti L, Rodella LF, Rezzani R. Sirtuins, aging, and cardiovascular risks.AGE. 2015; 37:65.CrossrefGoogle Scholar
    • 73. Haigis MC, Sinclair DA. sirtuins in aging and age-related diseases.Masoro EJ, Austad SN, eds. In: Handbook of the Biology of Aging (7th edition). San Diego, CA: Academic Press; 2011:243–274.Google Scholar
    • 74. Wątroba M, Dudek I, Skoda M, Stangret A, Rzodkiewicz P, Szukiewicz D. Sirtuins, epigenetics and longevity.Ageing Res Rev. 2017; 40:11–19. doi: 10.1016/j.arr.2017.08.001CrossrefMedlineGoogle Scholar
    • 75. Cencioni C, Spallotta F, Mai A, Martelli F, Farsetti A, Zeiher AM, Gaetano C. Sirtuin function in aging heart and vessels.J Mol Cell Cardiol. 2015; 83:55–61. doi: 10.1016/j.yjmcc.2014.12.023CrossrefMedlineGoogle Scholar
    • 76. Bindu S, Pillai VB, Gupta MP. Role of sirtuins in regulating pathophysiology of the heart.Trends Endocrinol Metab. 2016; 27:563–573. doi: 10.1016/j.tem.2016.04.015CrossrefMedlineGoogle Scholar
    • 77. Hershberger K, Martin A, Hirschey M. Role of nad+ and mitochondrial sirtuins in cardiac and renal diseases.Nat Rev Neph. 2017; 13:4. doi: 10.1038/nrneph.2017.5Google Scholar
    • 78. van de Ven RAH, Santos D, Haigis MC. Mitochondrial sirtuins and molecular mechanisms of aging.Trends Mol Med. 2017; 23:320–331.CrossrefMedlineGoogle Scholar
    • 79. Sabari BR, Zhang D, Allis CD, Zhao Y. Metabolic regulation of gene expression through histone acylations.Nat Rev Mol Cell Biol. 2017; 18:90–101. doi: 10.1038/nrm.2016.140CrossrefMedlineGoogle Scholar
    • 80. Shameer K, Klee EW, Dalenberg AK, Kullo IJ. Whole exome sequencing implicates an INO80D mutation in a syndrome of aortic hypoplasia, premature atherosclerosis, and arterial stiffness.Circ Cardiovasc Genet. 2014; 7:607–614. doi: 10.1161/CIRCGENETICS.113.000233LinkGoogle Scholar
    • 81. Han P, Li W, Yang J, et al. Epigenetic response to environmental stress: assembly of BRG1-G9a/GLP-DNMT3 repressive chromatin complex on Myh6 promoter in pathologically stressed hearts.Biochim Biophys Acta. 2016; 1863:1772–1781. doi: 10.1016/j.bbamcr.2016.03.002CrossrefMedlineGoogle Scholar
    • 82. Hang CT, Yang J, Han P, Cheng HL, Shang C, Ashley E, Zhou B, Chang CP. Chromatin regulation by Brg1 underlies heart muscle development and disease.Nature. 2010; 466:62–67. doi: 10.1038/nature09130CrossrefMedlineGoogle Scholar
    • 83. Mehrotra A, Joe B, de la Serna IL. SWI/SNF chromatin remodeling enzymes are associated with cardiac hypertrophy in a genetic rat model of hypertension.J Cell Physiol. 2013; 228:2337–2342. doi: 10.1002/jcp.24404CrossrefMedlineGoogle Scholar
    • 84. Vieira JM, Howard S, Villa Del Campo C, et al. BRG1-SWI/SNF-dependent regulation of the Wt1 transcriptional landscape mediates epicardial activity during heart development and disease.Nat Commun. 2017; 8:16034. doi: 10.1038/ncomms16034CrossrefMedlineGoogle Scholar
    • 85. Huuskonen J, Vishnu M, Fielding PE, Fielding CJ. Activation of ATP-binding cassette transporter A1 transcription by chromatin remodeling complex.Arterioscler Thromb Vasc Biol. 2005; 25:1180–1185. doi: 10.1161/01.ATV.0000163186.58462.c5LinkGoogle Scholar
    • 86. Devaux Y, Zangrando J, Schroen B, Creemers EE, Pedrazzini T, Chang CP, Dorn GW, Thum T, Heymans S; Cardiolinc Network. Long noncoding RNAs in cardiac development and ageing.Nat Rev Cardiol. 2015; 12:415–425. doi: 10.1038/nrcardio.2015.55CrossrefMedlineGoogle Scholar
    • 87. Cahill TJ, Choudhury RP, Riley PR. Heart regeneration and repair after myocardial infarction: translational opportunities for novel therapeutics.Nat Rev Drug Discov. 2017; 16:699–717. doi: 10.1038/nrd.2017.106CrossrefMedlineGoogle Scholar
    • 88. Liao WL, Tan MW, Yuan Y, Wang GK, Wang C, Tang H, Xu ZY. Brahma-related gene 1 inhibits proliferation and migration of human aortic smooth muscle cells by directly up-regulating Ras-related associated with diabetes in the pathophysiologic processes of aortic dissection.J Thorac Cardiovasc Surg. 2015; 150:1292.e2–1301.e2. doi: 10.1016/j.jtcvs.2015.08.010CrossrefGoogle Scholar
    • 89. Meng ZX, Wang L, Chang L, Sun J, Bao J, Li Y, Chen YE, Lin JD. A diet-sensitive BAF60a-mediated pathway links hepatic bile acid metabolism to cholesterol absorption and atherosclerosis.Cell Rep. 2015; 13:1658–1669. doi: 10.1016/j.celrep.2015.10.033CrossrefMedlineGoogle Scholar
    • 90. Szabo L, Salzman J. Detecting circular RNAs: bioinformatic and experimental challenges.Nat Rev Genet. 2016; 17:679–692. doi: 10.1038/nrg.2016.114CrossrefMedlineGoogle Scholar
    • 91. Schiano C, Costa V, Aprile M, Grimaldi V, Maiello C, Esposito R, Soricelli A, Colantuoni V, Donatelli F, Ciccodicola A, Napoli C. Heart failure: pilot transcriptomic analysis of cardiac tissue by RNA-sequencing.Cardiol J. 2017; 24:539–553. doi: 10.5603/CJ.a2017.0052CrossrefMedlineGoogle Scholar
    • 92. Ounzain S, Micheletti R, Beckmann T, et al. Genome-wide profiling of the cardiac transcriptome after myocardial infarction identifies novel heart-specific long non-coding RNAs.Eur Heart J. 2015; 36:353a–368a. doi: 10.1093/eurheartj/ehu180CrossrefMedlineGoogle Scholar
    • 93. Lai Y, He S, Ma L, Lin H, Ren B, Ma J, Zhu X, Zhuang S. HOTAIR functions as a competing endogenous RNA to regulate PTEN expression by inhibiting miR-19 in cardiac hypertrophy.Mol Cell Biochem. 2017; 432:179–187. doi: 10.1007/s11010-017-3008-yCrossrefMedlineGoogle Scholar
    • 94. Wang JX, Zhang XJ, Li Q, Wang K, Wang Y, Jiao JQ, Feng C, Teng S, Zhou LY, Gong Y, Zhou ZX, Liu J, Wang JL, Li PF. MicroRNA-103/107 regulate programmed necrosis and myocardial ischemia/reperfusion injury through targeting FADD.Circ Res. 2015; 117:352–363. doi: 10.1161/CIRCRESAHA.117.305781LinkGoogle Scholar
    • 95. Yu F, Chen B, Dong P, Zheng J. HOTAIR epigenetically modulates PTEN expression via MicroRNA-29b: a novel mechanism in regulation of liver fibrosis.Mol Ther. 2017; 25:205–217. doi: 10.1016/j.ymthe.2016.10.015CrossrefMedlineGoogle Scholar
    • 96. Greco S, Zaccagnini G, Perfetti A, Fuschi P, et al. Long noncoding rna dysregulation in ischemic heart failure.J Transl Med. 2016; 14:183. doi: 10.1186/s12967-016-0926-5.CrossrefMedlineGoogle Scholar
    • 97. Gonzalo-Calvo D, Kenneweg F, Bang C, et al. Circulating long-non coding rnas as biomarkers of left ventricular diastolic function and remodelling in patients with well-controlled type 2 diabetes.Sci Rep. 2016; 6:37354. doi: 10.1038/srep37354.CrossrefMedlineGoogle Scholar
    • 98. Qu X, Du Y, Shu Y, Gao M, Sun F, Luo S, Yang T, Zhan L, Yuan Y, Chu W, Pan Z, Wang Z, Yang B, Lu Y. Miat is a pro-fibrotic long non-coding rna governing cardiac fibrosis in post-infarct myocardium.Sci Rep. 2017; 7:42657. doi: 10.1038/srep42657CrossrefMedlineGoogle Scholar
    • 99. See K, Tan WLW, Lim EH, Tiang Z, Lee LT, Li PYQ, Luu TDA, Ackers-Johnson M, Foo RS. Single cardiomyocyte nuclear transcriptomes reveal a lincrna-regulated de-differentiation and cell cycle stress-response in vivo.Nat Commun. 2017; 8:225. doi: 10.1038/s41467-017-00319-8CrossrefMedlineGoogle Scholar
    • 100. Micheletti R, Plaisance I, Abraham BJ, Sarre A, Ting CC, Alexanian M, Maric D, Maison D, Nemir M, Young RA, Schroen B, González A, Ounzain S, Pedrazzini T. The long noncoding RNA Wisper controls cardiac fibrosis and remodeling.Sci Transl Med. 2017; 9:eaai9118. doi: 10.1126/scitranslmed.aai9118CrossrefMedlineGoogle Scholar
    • 101. Melé M, Rinn JL. “Cat’s Cradling” the 3D genome by the act of LncRNA transcription.Mol Cell. 2016; 62:657–664. doi: 10.1016/j.molcel.2016.05.011CrossrefMedlineGoogle Scholar
    • 102. Han P, Li W, Lin CH, et al. A long noncoding rna protects the heart from pathological hypertrophy.Nature. 2014; 514:102–106. doi: 10.1038/nature13596CrossrefMedlineGoogle Scholar
    • 103. Luo Y, Xu Y, Liang C, Xing W, Zhang T. The mechanism of myocardial hypertrophy regulated by the interaction between mhrt and myocardin.Cell Signal. 2018; 43:11–20. doi: 10.1016/j.cellsig.2017.11.007CrossrefMedlineGoogle Scholar
    • 104. Leisegang MS, Fork C, Josipovic I, et al. Long noncoding rna mantis facilitates endothelial angiogenic function.Circulation. 2017; 136:65–79. doi: 10.1161/CIRCULATIONAHA.116.026991LinkGoogle Scholar
    • 105. Wang Z, Zhang XJ, Ji YX, et al. The long noncoding rna chaer defines an epigenetic checkpoint in cardiac hypertrophy.Nat Med. 2016; 22:1131–1139.CrossrefMedlineGoogle Scholar
    • 106. Han P, Yang J, Shang C, Chang C-P. Chromatin remodeling in heart failure.Backs J, McKinsey T, eds. In: Epigenetics in Cardiac Disease. Cardiac and Vascular Biology. Springer, Cham, 2016; 10.1007/978-3-319-41457-7_5Google Scholar
    • 107. Chatterjee S, Bär C, Thum T. Linc-ing the noncoding genome to heart function: beating hypertrophy.Trends Mol Med. 2017; 23:577–579. doi: 10.1016/j.molmed.2017.05.007CrossrefMedlineGoogle Scholar
    • 108. Memczak S, Jens M, Elefsinioti A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency.Nature. 2013; 495:333–338.CrossrefMedlineGoogle Scholar
    • 109. Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. Natural RNA circles function as efficient microrna sponges.Nature. 2013; 495:384–388.CrossrefMedlineGoogle Scholar
    • 110. Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L, Chen LL. Circular intronic long noncoding RNAs.Mol Cell. 2013; 51:792–806. doi: 10.1016/j.molcel.2013.08.017CrossrefMedlineGoogle Scholar
    • 111. Chen LL. The biogenesis and emerging roles of circular RNAs.Nat Rev Mol Cell Biol. 2016; 17:205.CrossrefMedlineGoogle Scholar
    • 112. Tan WL, Lim BT, Anene-Nzelu CG, Ackers-Johnson M, Dashi A, See K, Tiang Z, Lee DP, Chua WW, Luu TD, Li PY, Richards AM, Foo RS. A landscape of circular RNA expression in the human heart.Cardiovasc Res. 2017; 113:298–309. doi: 10.1093/cvr/cvw250MedlineGoogle Scholar
    • 113. Werfel S, Nothjunge S, Schwarzmayr T, Strom TM, Meitinger T, Engelhardt S. Characterization of circular RNAs in human, mouse and rat hearts.J Mol Cell Cardiol. 2016; 98:103–107. doi: 10.1016/j.yjmcc.2016.07.007CrossrefMedlineGoogle Scholar
    • 114. Wu HJ, Zhang CY, Zhang S, Chang M, Wang HY. Microarray expression profile of circular RNAs in heart tissue of mice with myocardial infarction-induced heart failure.Cell Physiol Biochem. 2016; 39:205–216.CrossrefMedlineGoogle Scholar
    • 115. Siede D, Rapti K, Gorska AA, Katus HA, Altmüller J, Boeckel JN, Meder B, Maack C, Völkers M, Müller OJ, Backs J, Dieterich C. Identification of circular RNAs with host gene-independent expression in human model systems for cardiac differentiation and disease.J Mol Cell Cardiol. 2017; 109:48–56. doi: 10.1016/j.yjmcc.2017.06.015CrossrefMedlineGoogle Scholar
    • 116. Zeng Y, Du WW, Wu Y, et al. A circular RNA binds to and activates akt phosphorylation and nuclear localization reducing apoptosis and enhancing cardiac repair.Theranostics. 2017; 7:3842–3855.CrossrefMedlineGoogle Scholar
    • 117. Du WW, Yang W, Chen Y, Wu ZK, Foster FS, Yang Z, Li X, Yang BB. Foxo3 circular rna promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses.Eur Heart J. 2017; 38:1402–1412. doi: 10.1093/eurheartj/ehw001MedlineGoogle Scholar
    • 118. Du WW, Yang W, Liu E, Yang Z, Dhaliwal P, Yang BB. Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2.Nucleic Acids Res. 2016; 44:2846–2858. doi: 10.1093/nar/gkw027CrossrefMedlineGoogle Scholar
    • 119. Holdt LM, Stahringer A, Sass K, et al. Circular non-coding RNA ANRIL modulates ribosomal rna maturation and atherosclerosis in humans.Nat Commun. 2016; 7:12429. doi: 10.1038/ncomms12429CrossrefMedlineGoogle Scholar
    • 120. Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z, Sharpless NE. Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk.PLoS Genet. 2010; 6:e1001233. doi: 10.1371/journal.pgen.1001233CrossrefMedlineGoogle Scholar
    • 121. Yap KL, Li S, Muñoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, Gil J, Walsh MJ, Zhou MM. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a.Mol Cell. 2010; 38:662–674. doi: 10.1016/j.molcel.2010.03.021CrossrefMedlineGoogle Scholar
    • 122. Dang R-Y, Liu F-L, Li Y. Circular rna hsa_circ_0010729 regulates vascular endothelial cell proliferation and apoptosis by targeting the mir-186/hif-1α axis.Biochem Biophys Res Commun. 2017; 490:104–110. doi: 10.1016/j.bbrc.2017.05.164CrossrefMedlineGoogle Scholar
    • 123. Chen J, Cui L, Yuan J, Zhang Y, Sang H. Circular RNA WDR77 target FGF-2 to regulate vascular smooth muscle cells proliferation and migration by sponging miR-124.Biochem Biophys Res Commun. 2017; 494:126–132. doi: 10.1016/j.bbrc.2017.10.068CrossrefMedlineGoogle Scholar
    • 124. Shi Q, Yang X. Circulating microrna and long noncoding rna as biomarkers of cardiovascular diseases.J Cell Physiol. 2016; 231:751–755. doi: 10.1002/jcp.25174CrossrefMedlineGoogle Scholar
    • 125. Li M, Ding W, Sun T, et al. Biogenesis of circular RNAs and their roles in cardiovascular development and pathology.FEBS J. 2018; 285:220–232. doi: 10.1111/febs.14191CrossrefMedlineGoogle Scholar
    • 126. Kumarswamy R, Bauters C, Volkmann I, Maury F, Fetisch J, Holzmann A, Lemesle G, de Groote P, Pinet F, Thum T. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure.Circ Res. 2014; 114:1569–1575. doi: 10.1161/CIRCRESAHA.114.303915LinkGoogle Scholar
    • 127. Zhao ZZ, Li XJ, Gao CY, Jian DD, Hao PY, Rao LX, Li M. Peripheral blood circular RNA hsa_circ_0124644 can be used as a diagnostic biomarker of coronary artery disease.Sci Rep. 2017; 7:39918. doi: 10.1038/srep39918CrossrefMedlineGoogle Scholar
    • 128. Reckman Y, Creemers E. Circulating circles predict postoperative atrial fibrillation.J Am Heart Assoc. 2018; 22:7. pii: e008261. doi: 10.1161/JAHA.117.008261Google Scholar
    • 129. Novoa EM, Mason CE, Mattick JS. Charting the unknown epitranscriptome.Nat Rev Mol Cell Biol. 2017; 18:339. doi: 10.1038/nrm.2017.49CrossrefMedlineGoogle Scholar
    • 130. Fu Y, Dominissini D, Rechavi G, He C. Gene expression regulation mediated through reversible m6a RNA methylation.Nature Rev Genet. 2014; 15:293.CrossrefMedlineGoogle Scholar
    • 131. Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications.Nat Rev Mol Cell Biol. 2016; 18:31–42. doi: 10.1038/nrm.2016.132CrossrefMedlineGoogle Scholar
    • 132. Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, Fu Y, Parisien M, Dai Q, Jia G, Ren B, Pan T, He C. N6-methyladenosine-dependent regulation of messenger RNA stability.Nature. 2014; 505:117–20. doi: 10.1038/nature12730CrossrefMedlineGoogle Scholar
    • 133. Li X, Xiong X, Yi C. Epitranscriptome sequencing technologies: decoding rna modifications.Nat Methods. 2016; 14:23.CrossrefMedlineGoogle Scholar
    • 134. Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation.Cell. 2017; 169:1187–1200. doi: 10.1016/j.cell.2017.05.045CrossrefMedlineGoogle Scholar
    • 135. Helm M, Motorin Y. Detecting rna modifications in the epitranscriptome: predict and validate.Nature Rev Genet. 2017; 18:275.CrossrefMedlineGoogle Scholar
    • 136. de Luis DA, Aller R, Izaola O, Primo D, Urdiales S, Romero E. Effects of a high-protein/low-carbohydrate diet versus a standard hypocaloric diet on weight and cardiovascular risk factors: role of a genetic variation in the rs9939609 FTO gene variant.J Nutrigenet Nutrigenomics. 2015; 8:128–136. doi: 10.1159/000441142CrossrefMedlineGoogle Scholar
    • 137. Frayling TM, Timpson NJ, Weedon MN, et al. A common variant in the fto gene is associated with body mass index and predisposes to childhood and adult obesity.Science. 2007; 316:889–894.CrossrefMedlineGoogle Scholar
    • 138. Mathiyalagan P, Martins A, Liang Y, et al. Abstract 12012: dysregulated adenosine methylation of rna in myocardial ischemia.Circulation. 2016; 134:A12012.LinkGoogle Scholar
    • 139. Luo Y, Feng J, Xu Q, Wang W, Wang X. NSun2 deficiency protects endothelium from inflammation via mRNA methylation of ICAM-1.Circ Res. 2016; 118:944–956. doi: 10.1161/CIRCRESAHA.115.307674LinkGoogle Scholar
    • 140. Zipeto MA, Jiang Q, Melese E, Jamieson CH. RNA rewriting, recoding, and rewiring in human disease.Trends Mol Med. 2015; 21:549–559. doi: 10.1016/j.molmed.2015.07.001CrossrefMedlineGoogle Scholar
    • 141. Nishikura K. A-to-I editing of coding and non-coding RNAs by ADARs.Nat Rev Mol Cell Biol. 2015; 17:83–96. doi: 10.1038/nrm.2015.4CrossrefMedlineGoogle Scholar
    • 142. Stellos K, Gatsiou A, Stamatelopoulos K, et al. Adenosine-to-inosine RNA editing controls cathepsin S expression in atherosclerosis by enabling hur-mediated post-transcriptional regulation.Nat Med. 2016; 22:1140–1150. doi: 10.1038/nm.4172CrossrefMedlineGoogle Scholar
    • 143. Santos-Pereira JM, Aguilera A. R loops: new modulators of genome dynamics and function.Nat Rev Genet. 2015; 16:583.CrossrefMedlineGoogle Scholar
    • 144. Chu C, Qu K, Zhong FL, Artandi SE, Chang HY. Genomic maps of long noncoding rna occupancy reveal principles of RNA-chromatin interactions.Mol Cell. 2011; 44:667–678. doi: 10.1016/j.molcel.2011.08.027CrossrefMedlineGoogle Scholar
    • 145. Lau E, Cao Q, Lam MPY, Wang J, Ng DCM, Bleakley BJ, Lee JM, Liem DA, Wang D, Hermjakob H, Ping P. Integrated omics dissection of proteome dynamics during cardiac remodeling.Nat Commun. 2018; 9:120. doi: 10.1038/s41467-017-02467-3CrossrefMedlineGoogle Scholar
    • 146. Raghow R. An ‘Omics’ perspective on cardiomyopathies and heart failure.Trends Mol Med. 2016; 22:813–827. doi: 10.1016/j.molmed.2016.07.007CrossrefMedlineGoogle Scholar
    • 147. Cuvier O, Fierz B. Dynamic chromatin technologies: from individual molecules to epigenomic regulation in cells.Nat Rev Genet. 2017; 18:457.CrossrefMedlineGoogle Scholar
    • 148. Rosa-Garrido M, Chapski DJ, Schmitt AD, et al. High-resolution mapping of chromatin conformation in cardiac myocytes reveals structural remodeling of the epigenome in heart failure.Circulation. 2017; 136:1613–1625. doi: 10.1161/CIRCULATIONAHA.117.029430LinkGoogle Scholar
    • 149. Wang P, Liu Z, Zhang X, Li J, Sun L, Ju Z, Li J, Chan P, Liu GH, Zhang W, Song M, Qu J. CRISPR/Cas9-mediated gene knockout reveals a guardian role of NF-κb/RelA in maintaining the homeostasis of human vascular cells [published online July 2, 2018].Protein Cell. doi: 10.1007/s13238-018-0560-5Google Scholar
    • 150. Geng L, Liu Z, Zhang W, et al. Chemical screen identifies a geroprotective role of quercetin in premature aging.Protein & Cell. 2018. doi: 10.1007/s13238-018-0567-yCrossrefGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.