Skip to main content

Epigenome alterations in aortic valve stenosis and its related left ventricular hypertrophy

Abstract

Aortic valve stenosis is the most common cardiac valve disease, and with current trends in the population demographics, its prevalence is likely to rise, thus posing a major health and economic burden facing the worldwide societies. Over the past decade, it has become more than clear that our traditional genetic views do not sufficiently explain the well-known link between AS, proatherogenic risk factors, flow-induced mechanical forces, and disease-prone environmental influences. Recent breakthroughs in the field of epigenetics offer us a new perspective on gene regulation, which has broadened our perspective on etiology of aortic stenosis and other aortic valve diseases. Since all known epigenetic marks are potentially reversible this perspective is especially exciting given the potential for development of successful and non-invasive therapeutic intervention and reprogramming of cells at the epigenetic level even in the early stages of disease progression. This review will examine the known relationships between four major epigenetic mechanisms: DNA methylation, posttranslational histone modification, ATP-dependent chromatin remodeling, and non-coding regulatory RNAs, and initiation and progression of AS. Numerous profiling and functional studies indicate that they could contribute to endothelial dysfunctions, disease-prone activation of monocyte-macrophage and circulatory osteoprogenitor cells and activation and osteogenic transdifferentiation of aortic valve interstitial cells, thus leading to valvular inflammation, fibrosis, and calcification, and to pressure overload-induced maladaptive myocardial remodeling and left ventricular hypertrophy. This is especcialy the case for small non-coding microRNAs but was also, although in a smaller scale, convincingly demonstrated for other members of cellular epigenome landscape. Equally important, and clinically most relevant, the reported data indicate that epigenetic marks, particularly certain microRNA signatures, could represent useful non-invasive biomarkers that reflect the disease progression and patients prognosis for recovery after the valve replacement surgery.

Background

Aortic valve stenosis (AS) is the most frequent heart valve disease among adults in the Western societies with ever increasing prevalence due to the rapidly aging population [1,2,3]. According to the recent population-based studies performed in Europe and North America the pooled prevalence of total and symptomatic severe AS cases in the general elderly population (≥ 75 years of age) is estimated to 12.4, and 3.4%, respectively [4]. Moreover, with current trends in the population demographics by the year 2050 there will be an estimated 2.1 million European and 1.4 million North American patients with symptomatic severe AS [4]. Furthermore, the even more pronounced demographic changes in Africa, Asia, and South America will further increase the absolute number of AS patients [2, 4, 5]. Therefore, in a very recent future, AS is likely to become a major health and economic burden facing the worldwide societies [2, 4, 5].

The major cause of AS is thickening, fibrosis, and calcification of a previously normal tricuspid valve (TAV) or a congenital bicuspid valve (BAV), while a historically prevailing rheumatic heart valve disease, and still the most common cause of AS in developing countries accounts for only 10% of diagnosed cases [2, 6, 7].

No matter the cause, development of AS starts with the risk of leaflet changes and progresses over many years from early lesions to subsequent narrowing (stenosis) of the aortic valve orifice [8]. During that time, the genetic predisposition or otherwise induced faulty valve repair system in concordance with continuous blood born mechanical forces and proatherogenic risk factors (i.e., hyperhomocysteinemia, hyperlipidemia, abnormal calcium metabolism, smoking, metabolic syndrome, diabetes, hypertension, chronic renal failure, male gender, age) leads to endothelial dysfunctions followed by disruption of the subendothelial basement membrane, extracellular accumulation of plasma-derived atherogenic lipoproteins and infiltration/activation of monocyte-macrophage cells, mast cells, and T lymphocytes [3, 9,10,11,12,13,14]. That leads to intracellular lipid deposition, generation of oxidative stress with accumulation of oxidized lipids and apolipoproteins, foamy cells formation, and upregulation of various pro-fibrotic and pro-inflammatory factors with concomitant inhibition of plasma derived or locally presents anti-calcific proteins. Acting together, these factors promote extensive extracellular matrix remodeling, and activation of signaling pathways that promote neovascularization, inflammation and calcification [3, 9,10,11,12,13,14]. Concomitant transformation of normally quiescent valvular interstitial cells (qVICs) to active myofibroblastic (aVICs) phenotype in the valve interstitium and their subsequent differentiation to osteoblast-like cells (obVICs) with activation of pro-osteogenic signaling pathways is thought to be one of the central mechanisms contributing to the initiation and progression of AS [15]. In addition, a subset of aortic valvular endothelial cells (VECs) undergoing endothelial- to-mesenchymal transition (EMT) and/or circulating osteoprogenitor cells (COPCs) may also contribute to valvular calcification/ossification either by redifferentiating to an osteoblast-like phenotype or by promoting VICs activation through paracrine signaling [16,17,18].

Based on this timely dependent change of tissue organization, the disease has been divided in two successive functional categories: aortic valve sclerosis (ASc) and aortic valve stenosis. ASc represents the initial, clinically mostly silent stage of disease with calcification and mild fibrous thickening of the aortic leaflets without marked functional obstruction of left ventricular outflow [8, 19]. Contrary, aortic valve stenosis as the more advanced stage of the disease comprise serious impairment of leaflet motion with subsequent obstruction of blood flow resulting in maladaptive left ventricular hypertrophy (LVH), myocardial fibrosis (MF), and a propensity for systolic and diastolic dysfunction and heart failure (HF) [3, 8, 20].

Currently there are no effective pharmacological remedies to prevent or slow the progression of AS and aortic valve replacement (AVR) either surgical or less invasive transcatheter (TAVR) approach is still the only clinical therapy at hand for its successful treatment [21, 22, 23]. Thus, a better understanding of mechanisms involved in the pathogenesis and progression of AS could lead to novel diagnostic, prognostic and therapeutic targets and eventual development of noninvasive therapeutic options.

Recent observations suggest that the full pathological spectrum of AS lesions cannot be entirely accounted for by hereditary predisposition or growing list of differentially expressed genes. Moreover, traditional genetic views do not sufficiently explain the well-known link between AS, proatherogenic risk factors, and disease-prone environmental influences. Thus, it has become clear that other regulatory mechanisms are essential, and a compelling argument for an epigenetic contribution is rapidly emerging [11, 14].

Epigenetics refers to mitotically and meiotically stable (heritable), and functionally relevant DNA and chromatin modifications that are not caused by alterations (mutations) in the primary DNA sequence itself [24]. They can be either inherited or accumulated throughout a lifetime. Furthermore, given that each cell in our organism contains the identical genomic DNA the epigenetic mechanisms are fundamental to proper lineage commitment, cell fate determination, organogenesis, and ultimately, whole body homeostasis. More specifically, by altering the chromatin architecture and the accessibility of DNA coding and regulatory regions they orchestrate the spatiotemporal gene expression in a cell-type and even allele-specific (maternally or paternally imprinted loci) manner [25].

Four major epigenetic mechanisms (Fig. 1) have been characterized: DNA methylation and hydroxymethylation, covalent histone modifications and incorporation of histone variants, ATP-dependent chromatin remodeling, and chromatin and gene regulation by non-coding RNAs (ncRNAs) including small micro RNAs (microRNAs/miRs) and long non-coding RNAs (lncRNAs) [26,27,28,29,30,31]. These mutually interdependent epigenetic alterations, collectively named the epigenome, have profound effects on cellular repertoire of active genes [32, 33]. Furthermore, the well-known characteristics of epigenetic regulatory mechanisms: stability, adaptability, and reversibility, are all equally important in maintaining and changing cellular phenotype and function in both health and disease.

Fig. 1
figure 1

Major epigenetics mechanisams acting in mammalian cells. Presented are the four epigenetic mechanisams and their major impact on cellular gene regulation. Some writers (proteins that establish epigenetic marks) and riders (proteins that interpret epigenetic marks) are also illustrated. DNMT DNA methyl transferase, CBX3 Chromobox 3, CLOCK clock circadian regulator, DPF3A double PHD fingers 3a, 5hm 5 methyl cytosine, HAT1 histone acetyltransferase 1, HMT histone methyl transferase, ING2 inhibitor of growth family member 2, KMT2A lysine methyltransferase 2a, MeCP2 methyl CpG binding protein 2, MBD1 methyl-CpG-binding domain protein 1, MLL 1–5 family of lysine methyltransferases, MYSTs family of histone acetyltransferase, p300 histone acetyltransferase P300, PRMT1 protein arginine methyltransferase 1, p160 MYB binding protein 1a, SAM S-adenosyl methionine, SET1/ASH2 histone methyltransferase complex, SUV39H1 histone-lysine N-methyltransferase

First, through epigenetic modifications genes are switched on and off in a more durable fashion than by any other mechanisms of gene regulation. Secondly, epigenetic alterations undergo dynamic changes during development and in response to the various nutritional, behavioral and environmental stimuli [34]. Notably, early changes of epigenetic regulatory mechanisms caused by fetal environment may influence the adult phenotype, including an individual’s susceptibility to cardiovascular diseases (CVD) later in life, and the late onset of CVDs may well be linked to age-related alterations of epigenetic marks [35,36,37,38,39]. Finally, and clinically most relevant, all known epigenetic marks are reversible, thus opening the possibility for prophylactic or therapeutic intervention and reprogramming of cells even in the early stages of disease progression.

Herein, we provide a comprehensive overview of currently known epigenetic mechanisms involved in the control of gene expression in the native and infiltratory aortic valve cells, and discusses their roles in the pathogenesis and the progression of AS. Myocardial epigenome alterations due to pressure overload (PO) LVH induced by AS will also be covered.

DNA methylation

The data examining the role of DNA methylation changes in etiology of AS are only beginning to emerge.

For instance, Nwachukwu et al. reported dramatically increased levels of DNMT3B [DNA (Cytosine-5-)-methyltransferase 3 beta] expression in human AS compared to control valves, that was associated with an increase in global DNA methylation [40]. Furthermore, through site-specific methylation analysis, they identified more than 6000 differentially methylated sites between normal and stenotic valves [40]. Interestingly AS leaflets also showed four times higher expression of pro-osteogenic marker osterix (SP7/OSX) [40].

Furthermore, Sritharen et al. showed that genetic inactivation of DNMT3B protects against activation of osteogenic pathways and slows the progression of AS [41]. In their experiment, aortic valves from haploinsufficient mice (LDLR−/−/APOB100/100, DNMT3B+/−) showed increased expression of FABP4 (fatty acid binding protein 4; opposes osteogenesis) and SMAD6 (SMAD family member 6; opposes bone morphogenetic protein /BMP/ signaling) gene products while expression of the osteogenic genes MSX2 (MSH homeobox 2) and SPP1/OPN (secreted phosphoprotein 1; bone sialoprotein I, osteopontin) were substantially reduced [41].

Additional proof for the involvement DNA methylation changes in etiology of AS was reported by Radhakrishna et al. [42]. Their comparative DNA methylation analysis of neonatal dried blood spots obtained from newborns with AS disease and gestational-age matched controls revealed 59 significantly altered (hypomethylated or dimethylated) CpG methylation sites in the coding and/or promoter regions of 52 genes [42]. More specifically, they observed a significant methylation changes in APOA5 (apolipoprotein A5), PCSK9 (proprotein convertase and subtilisin/knexin-type 9), DUSP27 (dual-specificity phosphatase 27), RUNX1 (runt-related transcription factor 1), and TXNRD2 (thioredoxin reductase 2) gene thus concluding that their altered expression is likely responsible for congenital AS [42]. Importantly, many of these differentially methylated CpG sites demonstrated good to excellent diagnostic accuracy for the prediction of AS status, thus raising the possibility to be used as molecular screening markers for non-invasive risk estimation and disease detection [42].

Importantly, Gilsbach et al. reported that altered methylation pattern of CpG sites may also contribute to regulation of LVH as a response to chronic PO induced by AS [43]. They identified 1280 differentially methylated CpG sites in myocardial biopsies from AS patients with cardiac hypertrophy (CH) and 1365 CpG sites in patients with HF, with 523 of them significantly altered in both patient groups [43]. In addition, 496 of these differentially methylated CpG sites were concordantly altered both in hypertrophic and in HF tissue samples [43].

The first piece of evidence for the role of gene specific alteration of DNA methylation marks in AS was reported by Nagy and Back [44]. They showed that treatment of human AVICs with the DNA methyltransferase inhibitor 5-Aza-2′-deoxycytidine increases ALOX5/5LO (5-lipoxygenase) transcriptional levels and the production of the proinflammatory mediator LTB4 (leukotriene B4) [44]. These in vitro findings were confirmed in surgically explanted calcified aortic valves exhibiting reduced promoter methylation of ALOX5 accompanied with significantly higher transcriptional levels compared with non-calcified valve tissue [44]. The same group has also previously reported that the local upregulation of 5-lipoxygenase pathway (ALOX5, ALOXAP/FLAP/5-LO activating protein, LTA4H/Leukotriene A4 hydrolase and LTC4S/Leukotriene C4 synthase) in human aortic valves leads to leukotriene-induced effects on aortic VICs (enhanced leukocyte recruitment, inflammation, increased reactive oxygen species /ROS/ production, LTB4-induced matrix metalloproteinase /MMP/ secretion, matrix remodeling, and calcification) and correlates significantly with the expression of osteogenic marker genes (BMP2/6 and runt-related transcription factor/RUNX2) and severity of stenosis [45].

Gene-specific DNA-methylation changes in human AVICs were also reported in promoter region of H19 (imprinted maternally expressed non-protein coding transcript) by Hadji et al. [46]. They showed that promoter region of this lncRNA is heavily methylated in healthy aortic valves resulting with no expression of its transcripts. Contrary, promoter hypomethylation observed in stenotic valves leads to increased H19 expression which, in turn, decreases NOTCH1 transcription (prevents the recruitment of P53 to NOTCH1 promoter), and consequently increases the level of NOTCH1 downstream targets RUNX2, BMP2, and BGLAP/OCN (bone gamma-carboxyglutamate (Gla) protein; osteocalcin) thus promoting osteogenic reprograming of AVICs [46].

Finally, reduced expression of EGFR (epidermal growth factor receptor) associated with hypermethylation and dysregulation of its 5-hydroxymethylation pattern has also been linked with abnormal valve differentiation leading to the calcific AS and LVH in mice [47]. Since EGFR protein normally suppresses BMP pathway, attenuation of EGFR signaling may predispose differentiation of VICs to a calcifying cell phenotype [47].

Data accumulating from other tissues also suggest that DNA methylation may be involved in transcriptional regulation of genes implicated in AS, especially the ones involved in osteoblastic transdifferentiation of VICs.

For example, it is well known that obVICs are characterized by markers of osteoblastic differentiation, such as induction of ALP (alkaline phosphatase) [48]. Interestingly, DNA methylation plays important role in regulation of ALP expression in cells of the osteoblastic lineage, especially in its progressive transcriptional silencing during the osteoblasts to osteocyte transition [49, 50]. Notably, the degree of ALP promoter methylation is inversely associated with the transcriptional levels of ALP positive cells, with osteocytes, which do not express ALP showing high CpG island methylation [49].

Recent studies have also identified SOST (sclerostin; regulates osteoblast activity and serves as a Wnt /wingless-type MMTV integration site family member/pathway antagonist and a potent inhibitor of bone formation) gene expression as a novel marker of valvular calcification [51,52,53]. Notably, AS patients have significantly higher serum levels of SOST protein when compared to healthy subjects [51, 54]. Furthermore, tissues close to the calcified regions exhibit positive sclerostin staining, which is not observed in non-calcified control valves. In addition, SOST mRNA is significantly upregulated in calcified valves compared to non-calcified leaflets [51]. This increase in the SOST mRNA and protein levels is accompanied by the expression of prototypic markers (RUNX2 and BGLAP/OCN) of osteogenic transdifferentiation [51].

Importantly, SOST protein is produced by osteocytes and not osteoblasts and is regulated by DNA methylation during the final stage of osteoblast-to-osteocyte transition [55]. In addition, the SOST methylation marks are inversely correlated with its expression in osteoblastic cells but in this case with heavily methylated promoter in osteoblasts, which do not express SOST, while this same region is largely hypomethylated in human osteocytes [55,56,57].

Furthermore, the promoter regions of DLX5 and OSX genes that exhibit increase expression in human VICs were also found methylated in non-osteogenic and unmethylated in osteogenic cell lines expressing these genes [58]. In addition, Arnsdorf et al. demonstrated that mechanical stimulation, essential for maintaining bone homeostasis, may reduce DNA methylation at the SPP1/OPN (osteopontin) promoter accompanied with corresponding increase in gene expression [59]. Interestingly, significant upregulation of SPP1 expression was found in human AS valves in relation to preoperative transvalvular pressure gradient as well as in cultured VICs in relation to the mechanical strain applied [60, 61].

However, it should be emphasized that even though obVICs and cells of osteoblastic lineage share similar phenotypic markers, the applicability of the above findings to AS is currently unknown and remains largely to be established. A side by side comparison between VICs and several cell types in different stages of osteoblastic lineage commitment and differentiation performed by Monzack and Masters have shown stark differences in both the level and pattern of gene expression [62]. Whether they also include the noticeable dissimilarity in patterns of epigenetic gene regulation is currently quite unknown.

Major epigenetic markers in human aortic VICs are presented in Fig. 2.

Fig. 2
figure 2

Epigenetic mechanisms currently associated with aortic VICs. ALP alkaline phosphatase, liver/bone/kidney, ALOX5 arachidonate 5-lipoxygenase, AVICs aortic valve interstitial cells, BAV bicuspid aortic valve, BGLAP/OCN bone gamma-carboxyglutamate protein/osteocalcin, BMP2 bone morphogenetic protein 2, BMPR2 bone morphogenetic protein receptor type 2, β-catenin CTNNB1/catenin (cadherin-associated protein), beta 1, CASP3 Caspase 3, apoptosis-related cysteine peptidase, DLX5 distal-less homeobox 5, DNMT3b DNA (Cytosine-5-)-methyltransferase 3 beta, ERK1 extracellular signal-regulated kinase 1/MAPK3 mitogen-activated protein kinase 3, ERK2 extracellular signal-regulated kinase 2/MAPK1 mitogen-activated protein kinase 1, H19 imprinted maternally expressed non-protein coding transcript, IKKβ inhibitor of kappa light polypeptide gene enhancer in b-cells, kinase beta, NF-Kb nuclear factor kappa b signaling pathway, IGF-1 insulin-like growth factor 1, ILα interleukin alpha, ILβ interleukin beta, IL8 interleukin 8, NOTCH1 notch homolog 1, translocation-associated (drosophila), JAG2 jagged, MMP1 matrix metallopeptidase 1, MMP2 matrix metallopeptidase 2, MMP9 matrix metallopeptidase 9, MMP14 matrix metallopeptidase 14, MMP16 matrix metallopeptidase 16, OSX SP7 transcription factor/osterix, p65 RELA/RELA proto-oncogene, NF-KB subunit, RUNX2 runt related transcription factor 2, SMAD1 SMAD family member 1, SMAD3 SMAD family member 3, SMAD5 SMAD family member 5, SMAD7 SMAD family member 7, SPP1 secreted phosphoprotein 1/OPN osteopontin/BNSP bone sialoprotein I, TGFβ1 transforming growth factor beta 1, TGFBR2 transforming growth factor beta receptor 2, WNT5a Wnt family member 5a

DNA methylation changes may also regulate gene expression pattern in aortic valve endothelial cells. Thus for example, White et al. reported DNMT3B as one of the genes affected by shear stress and decreased NOTCH1 (Notch/drosophila/homolog 1/translocation-associated) signaling in primary human AVECs [63]. It is well known that NOTCH1 regulates calcification related gene networks in human vascular and valvular endothelium [63]. Notably, NOTCH1 signaling is higher on the ventricular side, and it is considered protective for calcification [64, 65]. Furthermore, human NOTCH1 mutations can cause both familial and sporadic AS disease [66].

Possible NOTCH1-dependent transcriptional and epigenetic mechanisms underlying this phenomenon are quite recently revealed by Theodoris et al. [67]. They used hiPSC (human-induced pluripotent stem cell)-derived ECs generated from two patients with nonsense mutations in NOTCH1 and related unaffected individual to investigate how NOTCH1 haploinsufficiency may cause aortic valve calcification [67].

Upon shear stress NOTCH1 (+/−) ECs exhibited dysregulated epigenetic state and aberrant upregulation of pro-osteogenic, pro-oxidative and pro-inflammatory signaling pathways, thus recapitulating observed AVECs phenotype in AS [67]. Contrary, wild-type (WT; NOTCH1+/+) ECs cells under the same conditions suppressed these and activated homeostatic anti-atherogenic gene networks [67]. This was accompanied with corresponding alterations of DNA methylation marks, with so called ‘CpG Island shores’ (sequences up to 2 kb distant to CpG islands in promoter regions) showing the largest enrichment of methylation changes [67]. Their findings agree with previous reports of CpG shore regions displaying the most methylation differences in the context of specific tissue types regardless of species of origin [68].

Concludingly, all the above data, although some of them mostly inferential (osteoblastic transdifferentiation of VICs) and yet to be experimentally validated, strongly suggests that DNA methylation might be an important regulatory mechanism involved in the initiation and progression of AS.

Histone and chromatin marks

Emerging reports suggest that various HAT/HDAC (histone acetyltransferase/histone deacetylase) enzyme complexes have also an important roles in the patohistogenesis of AS.

For instance, Carter et al. have reported reduced mRNA and protein levels of class III histone deacetylase SIRT1 (silent information regulator-two (1) in AS patients when compared to controls [69]. In addition, the levels of SIRT1 expression were inversely correlated with transcriptional activity of RETN (resistin) gene in infiltrated macrophages, thus possibly contributing to the development of AS-associated inflammation [69]. Interestingly, Mohty et al. reported that higher resistin plasma levels in the elderly AS patients (≥ 70 years) are associated with lower LDL cholesterol and increased valvular calcification and inflammation compared to younger middle-aged patients [70]. This may well be associated with corresponding age-related changes in SIRT1 expression.

Roos et al. reported a significant age-related decrease of SIRT1, −2, −3,-4, −6 and SIRT7 expression in aortic valves of both SOD2 (superoxide dismutase (2) haploinsufficient (SOD2−/+) and normal (SOD2+/+) mice and increased expression of SIRT5 in aged, compared to young SOD2+/− mice [71]. They also reported markedly reduced expression of SIRT6 in aortic valves from AS patients while knockout of SIRT6 expression in mice resulted in a dramatic progeroid phenotype, with a much greater propensity for development of cardiovascular calcification and AS [72]. The same group showed that SIRT6 inhibition promotes the formation of calcified nodules, induces amplification of pro-osteogenic signaling pathways and favors osteogenic differentiation of cultured mouse aortic VSMCs and VICs obtained from SIRT6-deficient mice, porcine, or human patients [73]. Also, SIRT6-deficient cells exhibited increased p-SMAD1/5/8 levels while the global SIRTs inhibition resulted with increased histone acetylation and protein levels of osteogenic markers SP7 and RUNX2. Furthermore, SIRTs inhibition has also enhanced the transcriptional responses of SP7 and RUNX2 gene in BMP2 treated VSMCs and VICs [73, 74].

Moreover, the data obtained in hypercholesterolemic (LA, LDLR −/− APOB 100/100) mice that were WT (LA-SIRT6+/+) or heterozygous for SIRT6 (LA-SIRT6+/−) suggested the context-dependent role of SIRT6 in hypercholesterolemia-induced valvular dysfunction, and modulation of hypertrophic responses to progressive increases in LV afterload [74,75,76]. Notably, 12-month-old LA-SIRT6+/− mice fed a western diet showed dramatically worsened aortic valve dysfunction and AS compared to age-matched WT or 3-month-old LA-SIRT6+/− mice that was also associated with reductions in LV ejection fraction [74]. In addition, reductions of SIRT6 in the presence of chronic LV pressure overload significantly accelerated the rate of AS development in female but not male mice thus highlighting the biological influence of gender related factors on disease progression and AS-induced LV dysfunction [76].

The class I (HDAC1 and HDAC3) and class II (HDAC5, HDAC7, and HDAC9) HDAC family members were also found differentially expressed in human and porcine aortic valve cells [63, 77, 78]. For instance, HDAC1 was reported as shear-responsive transcripts thus suggesting its possible involvement in flow mediated regulation of side-specific gene expression in aortic valve endothelium [77]. In addition, HDAC3, −7, and −9 transcripts levels have been found decreased in HOTAIR (HOX transcript antisense RNA) siRNA treated VICs cells while HDAC5 was detected as one of gene transcripts that were activated by flow and affected by decreased NOTCH1 activity in human AVECs [63, 78].

It is well known that reduced HDAC1 expression and its decreased recruitment to the promoters of osteoblast marker genes represent an important step for osteoblasts differentiation and vascular calcification [79, 80]. It has been also reported that HDAC3 physically interacts with RUNX2 thus suppressing its transcriptional activity [81] Furthermore, dysregulation of HDAC3 mediated epigenetic silencing of TGFβ1 (transforming growth factor beta 1) is required during valve development to maintain VICs quiescence and may represent a predisposing factor for development of BAV and other congenital heart diseases [82]. In addition, HDAC5 is involved in RUNX2 degradation while HDAC7 represses its activity during osteoblastogenesis in deacetylase independent manner [82,83,84,85]. Furthermore, since HDAC7 suppresses the expression of MMP10, its downregulation observed in VICs may play a role in biomechanical and HOTAIR lncRNA dependent upregulation of procalcific and inflammatory genes related to BAV induced AS [78, 86, 87]. Nevertheless, despite all of the above data, the exact role, if any, of these HDAC enzymes in development and progression of AS has yet to be experimentally validated.

The importance of histone code alterations in the development of AS was also reinforced by previously mentioned analysis of hiPSC-derived ECs reported by Theodoris et al. [67]. They showed a clear relationship between the expression of oxidative, inflammatory and calcification related genes in WT (NOTCH1+/+) and haploinsufficient (NOTH1+/−) ECs mediated by fluid-flow conditions and NOTCH1-dependent distribution of activated (H3K4me3, H3K27ac, and H3K4me1) and repressive (H3K27me3) histone marks [67]. More specifically, the shear stress response in WT ECs resulted in activation of anti-osteogenic and anti-oxidation genes and repression of proinflammatory loci that was correlated with the occupancy of their enhancers by repressive H3K27me3 or active H3K27ac histone marks [67]. Quite contrary, upon shear stress Notch1+/− ECs exhibited aberrant upregulation of pro-osteogenic genes associated with increased H3K4me3, H3K27ac, and H3K4me1 histone marks while H3K27ac marks were increased in active enhancers of pro-inflammatory genes under both static and shear flow conditions [67]. Moreover, anti-calcific and anti-atherogenic genes downregulated by NOTCH1 haploinsufficiency showed decreased H3K27ac active enhancer marks as well as increased repressive H3K27me3 marks [67]. Importantly, alterations of histone code were in most cases accompanied with corresponding changes of DNA methylation marks [67]. More specifically, the regions hypermethylated in Notch1 haploinsufficient ECs lost H3K4me3 or H3K27ac activating marks present in WT ECs while regions hypomethylated in N1+/− ECs gained H3K4me3 or H3K27ac marks [67].

Although it remains to be determined whether the used hiPSC-derived ECs accurately model molecular pathways induced in diseased human AVECs, these data presented by Theodoris et al. represent a significant step forward in deciphering the complexity and mutual interconnection of epigenome alterations underlying the pathogenesis of AS.

Other genes involved in regulation of epigenetic histone marks and/or chromatin remodeling that were found differentially expressed in stenotic aortic valves are presented in Table 1. Given the important role the mechanical forces, proatherogenic risk factors and intrinsic (epi)genetic variants, as well as, age and gender are playing in disease onset and progression, the known transcriptional profile of corresponding histone and chromatin remodeling factors induced by them in experimentally modified aortic valve endothelial and interstitial cells are presented as well. Specified genes are selected based on reanalysis of published data using DAVID (database for annotation visualization and integrated discovery), EpiFactors (database for epigenetic factors, corresponding genes and products) and Genecard (the human gene database) free online databases [88,89,90,91].

Table 1 Genes involved in regulation of epigenetic histone marks and/or chromatin remodeling

Histone/chromatin alteration in AS-induced LVH

Alteration of histone marks and chromatin remodeling might also contribute to the pathological response of LV myocardium to increased afterload in AS, thus leading to CH and HF [92]. Although the initial hypertrophic responses seem to be an adaptation to those stimuli, the sustained stress may lead to reactivation of fetal genes, which is possible due to the interplay of transcription factors, HAT/ADAC and ATP-dependent chromatin remodeling complexes [92].

Thus, for example, Bovil et al. reported the reduced expression of JARID2 (Jumonji and AT-rich interaction domain containing 2) by mechanical stress in LV biopsy samples from AS patients [93]. They showed that JARID2 regulates the transcription of ANF (atrial natriuretic factor), MYL7 (myosin light chain 7), and MYH2 (myosin heavy chain 2), and contributes to reexpression of the fetal gene program in cardiomyocytes subjected to increased afterload [93]. More specifically, the reduced JARID2 expression contributes to increased ANF and MYL7 and decreased MYH2 expression [93]. In addition, Sanulli et al. reported that JARID2 interacts with PRC2 (Polycomb repressive complex 2) responsible for methylation of histone H3 lysine 27 (H3K27me2/3) through its EZH (enhancer of zeste; EZH1/2) members and plays an essential role in regulating gene expression during embryonic development [94]. This JARID2 mediated recruitment of the PRC2 complex to target genes is also required for proper expression of NOTCH1 [95].

Very recently, Cui et al. reported a significant upregulation of DPF3/BAF45C [Double PHD Fingers 3/BRG1-Associated Factor 45C, a histone acetylation and methylation reader of the SWI/SNF (SWItch/Sucrose Non-Fermentable)-like ATP-dependent BAF (BRG/BRM-associated factor) chromatin remodeling complex] and its two individual splice variants DPF3a (BAF45c1) and DPF3b (BAF45c2) in hypertrophic hearts of patients with hypertrophic cardiomyopathy or AS [96]. Importantly, DPF3a is expressed as a fetal-like gene, whose activation by CSNK2 (casein kinase 2; phosphorylates DPF3a at serine 348) upon hypertrophic stimuli switches cardiac fetal gene expression from being silenced by HEY (the hairy and enhancer of split-related family of basic helix-loop-helix /bHLH/ transcription factors; HEY1/2 and HEYL) proteins to being re-activated by BRG1/SMARCA4 (brahma-related gene 1; SWI/SNF related, matrix associated, actin-dependent regulator of chromatin, subfamily a, member 4) [96]. Consequently, the transcription of downstream targets such as NPPA (natriuretic peptide A), FOXO1 (Forkhead box O1), GATA4 (growth arrest and DNA damage-inducible protein 45; mediates active DNA demethylation pathways), TBX3 (T-box 3) and SMAD7 (SMAD family member 7) were found significantly upregulated, thus promoting cardiac hypertrophy [96].

As already known, BRG1 is highly expressed in the developing heart while BRM/SMARCA2 (Brahma; SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2) represents the prominent catalytic unit of BAF chromatin remodeling complex in adult cardiomyocytes [96]. However, upon hypertrophic stimuli adult cardiomyocytes switch to a fetal-like state and BRG1 is reactivated and with its embryonic partners (HDACs and poly (ADP ribose) polymerases /PARPs/) forms BRG1-HDAC-PARP chromatin remodeling complex to induce a pathological gene expression [96].

The importance of BRG1 for aortic valve development was also previously reported by Akerberg et al. [97]. They showed that endocardial BRG1-deficient embryos develop thickened aortic valves that are frequently bicuspid and progress to a myxomatous, disease-like state but does not become overtly calcified [97].

Taken together, these data provide new insight into the complexity of epigenetic gene regulation in diseased aortic valve cells and affected myocardium. Without any doubt, we currently see only the very tip of histone and chromatin alterations lying under the surface of AS patohistogenesis with numerous new players expected to emerge.

LncRNAs

Long noncoding RNAs are also emerging as powerful epigenetic regulators of AS. The recent comparative transcriptome analysis of calcified aortic valves and 32 other human tissues reported by Wang et al. revealed a total of 725 aortic valve-specific lncRNAs [98]. However, the exact role of lncRNA transcripts in patohistogenesis of AS is still poorly known and limited to few examples.

We already mentioned the role of H19 in osteogenic transdifferentiation of AVICs reported by Hadji et al. [46]. They also detected three upregulated (AFAP1-AS1, CCND1 ncRNA, and PRINS) and five downregulated (AK082072, APO-AS1, OIP5-AS1, PTENP1-AS, and SOX2-OT) lncRNAs in calcific aortic valves when compared to control [46]. It is known that the exon 1 of H19 harbors a primary miRNA sequence, which generates miR-675-3p and miR-675-5p, and miR-675-5p was found to directly target H19 and counteracted osteoblast differentiation. However, Hadji et al. did not detect the expression of miR-675 neither in control or calcified aortic valves [46].

Previously, Carrion et al. have reported decreased transcript levels of HOTAIR (HOX transcript antisense RNA) in human BAVs as compared to the normal TAVs and in VICs exposed to cyclic stretch [78]. They also detected significant upregulation of several genes associated with calcification of VICs subjected to cyclic stretch (ALP and BMP2) and HOTAIR siRNA treatment (ALP, BMP 1/4/6, bone morphogenic protein receptor type 2/BMPR2, endothelin 1/EDN1, periostin/POSTN, SOST, matrix γ-carboxyglutamate (Gla) protein/MGP, and MMP2/10/12) [78]. Furthermore, the mechanoresponsive HOTAIR expression and epigenetic regulation (recruitment of PRC2 complexes which mediate trimethylation of lysine 27 on histone H3/H3K27me3 to ALP and BMP2 promoters) of calcification related genes were significantly repressed by WNT β-catenin signaling pathway [78]. Interestingly HOTAIR siRNA treatment of cultured VICs resulted with decreased expression of DNMT3B and altered expression of number of genes involved in histone and chromatin modification (Table 1) and genes encoding various miR (miR-1228, miR-1978 and miR-586) and lncRNA transcripts (Table 2).

Table 2 LncRNA transcripts differentially expressed in stenotic aortic valves and experimentally modified aortic valve cells

Implication of lncRNAs in AS development is also demonstrated by the role of intergenic lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) which is associated with osteogenic transdifferentiation of human VICs. Its upregulated expression in AS compared to control valves was first reported by Bosse et al. [99]. Contrary to them, Zhu et al. detected significantly lower levels of MALAT1, both in calcific valves and osteoblastic differentiating VICs accompanied by lower levels of TWIST1 (twist-related protein 1) protein [100]. Furthermore, they showed that MALAT1 directly interacts with TWIST1 and could enhance its stability while the TWIST1 overexpression prevents VICs calcification induced by inhibition of MALAT1 [100]. Possible mechanism was previously indicated by study of Zhang et al. demonstrating that TWIST1 negatively regulates osteoblastic transdifferentiation of human VICs through direct inhibition of RUNX2 [101].

LncRNAs associated with AS-induced LVH

Recent studies had shown that lncRNAs may also play an important role in regulation of AS-induced cardiac hypertrophy. Thus, Viereck et al. reported a significant upregulation of CHAST (cardiac hypertrophy-associated lncRNA transcript) lncRNA in hypertrophic heart tissue from AS patients and in human embryonic stem cell-derived cardiomyocytes upon hypertrophic stimuli [102]. Furthermore, Ounzain et al. reported the upregulation of CARMEN (cardiac mesoderm enhancer-associated noncoding RNA) in patients with idiopathic dilated cardiomyopathy (DCM) and AS [103]. The same group quite recently reported WISPER (WISP2 super-enhancer associated RNA) lncRNA as a novel cardiac fibroblast enriched super-enhancer related lncRNA, whose expression was significantly correlated with cardiac fibrosis both in a murine model of myocardial infarction and in AS patients [104]. Furthermore, previous study by Ounzain et al. reported differential expression of Novlnc6, Novlnc23, and Novlnc44 lncRNA in LV biopsy samples from dilated cardiomyopathy (DCM) and AS patients when compared to control LV tissue [105]. In contrast to DCM group, patients with AS were not associated with downregulation of enhancer derived lncRNA Novlnc6, or its predicted target gene NKX2–5 (NK2 Transcription Factor Related, Locus 5). However, they detected significant downregulation of Novlnc44 while the downregulation of Novlnc23 was not statistically significant [105].

Contrary to the above, Peters et al. indicate no role of MALAT1 in PO-induced CH, myocardial remodeling and HF in mouse model(TAC- transverse aortic constriction, and angiotensin II-induced CH) of AS despite its reported role in regulation (binding and inhibition) of antihypertrophic miR-133 [106].

Other lncRNAs transcripts differentially regulated in AS compared to control aortic valve tissue are presented in Table 2. They are selected based on bioinformatic reanalysis of published data using DAVID database for annotation visualization and integrated discovery; NCBI Gene website; Genecard, the human gene database; lncrnadb, the reference database for functional long noncoding RNAs, and HGNC web base for gene nomenclature [88, 89, 107,108,109,110]. However, functional roles for most of these lncRNA transcripts in the vasculature and other human tissues are largely unknown.

MicroRNAs

MicroRNA (miRNA/miR) are the best studied epigenetic regulatory mechanism in AS and other cardiovascular diseases [26, 111,112,113]. Up to now, several qRT-PCR and miRNome-wide microarray studies, (Table 3) reported differential expression of more than 200 microRNAs in stenotic aortic valve leaflets or experimentally modified aortic valve cells [77, 87, 99, 114,115,116,117,118,119,120,121,122,123,124].

Table 3 Dysregulated microRNA in stenotic valves and experimentally modified aortic valve cells

Numerous functional studies (Table 4 ) revealed that many of these miRNA transcripts are involved in activation and osteogenic differentiation of AVICs [92, 115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138]. In addition, these ossification-related miRNAs, so called osteomiRs, have also a prominent role in calcification of other cardiovascular tissue settings [139,140,141,142,143,144,145,146].

Table 4 Functional analysis of dysregulated microRNAs in stenotic aortic valves and experimentally modified aortic valve cells

For example, Balderman et al. have reported that BMP2 decreases microRNA-30b and microRNA-30c thus promoting calcification of VSMCs [140, 141]. In addition, Ding et al. reported that miR-30e represses the osteogenic program (reduction of the osteogenic panel: dermatopontin/DPT, decorin/DCN, RUNX2, BMP4, SPP1/OPN, IGF2/insulin-like growth factor 2, ALP, and BGLAP/OCN) in bone marrow-derived mesenchymal stem cells and SMCs by targeting IGF2 and drives their differentiation into adipogenic or SMC lineage [125]. They showed that injections of antimiR-30e increases IGF2 expression in the mouse aorta and significantly enhances OPN protein expression and calcium deposition in aortic valves [125]. They also showed that NFYC (nuclear transcription factor gamma subunit c) gene and its hosted miR-30e transcripts are down-regulated with age and atherosclerosis and inversely proportional to the expression of the osteogenic genes RUNX2, OPN, and IGF2 [125]. MiR-30b/c/d transcripts were also predicted to target RUNX2, while miR-29a and miR-29c were known to target collagen production and miR-2861 has been shown to affect RUNX2 activity (inhibitor of RUNX2 protein) in mouse osteoblasts by targeting HDAC5 [116, 142]. In addition, Hu et al. and Xia et al. have detected the regulatory feed-back loop between RUNX2, miR-3960 and miR-2861 acting during the osteoblastic differentiation and in osteogenic transdifferentiation of VSMCs, correspondingly [143, 144]. Their results suggested that RUNX2 could directly induce the expression of miR-3960/miR-2861 cluster (located at the same loci) by binding to the putative binding site of its promoter [143, 144]. Furthermore, Goettsch et al. reported the involvement of miR-125b in osteogenic transdifferentiation of VSMCs in in vitro and in vivo experimental settings by targeting the osteoblast transcription factor SP7/OSX while When et al. showed that miR-125b regulates transdifferentiation and calcification of VSMCs in a high-phosphate environment by targeting ETS1 (ETS proto-oncogene 1), a known transcription factor involved in osteoblastogenesis and extracellular matrix (ECM) mineralization [145, 146]. However, the applicability of these findings in the settings of AS largely remains to be established.

MiRNA transcripts dysregulated in AS have also multifactorial impact on endothelial dysfunction, inflammation, and endothelial-dependent myofibroblastic activation and osteoblastic transdifferentiation of AVICs (Table 4). For instance, Ohukainen et al. detected miR-125b as one of the most prominent dysregulated miRNAs in AS compared to control valve tissue. They also detected an increased expression of the inflammatory chemokines CCL3 (chemokine (C-C motif) ligand 3) and CCL4 (chemokine (C-C motif) ligand 4) in macrophages and αSMA-positive myofibroblasts and confirmed the CCL4 as a direct target of miR-125b in cultured human THP-1 macrophages, thus showing the connection between microRNA and inflammatory gene expression in AS [92].

Various flow-sensitive miRNAs, known as mechano-miRs, are also detected in stenotic valves. For instance, Patel et al. have identified 5 upregulated and 10 down regulated miRNAs (Table 3) in human BAV tissue and AVICs exposed to cyclic stretch [87, 119]. They also found that stretch-modulated repression of miR-148a-3p and miR-19b (Tables 3 and 4) may be sufficient to activate macrophages and to promote inflammatory and osteogenic signaling pathways in AVICs [87, 119]. Shear and side-dependent miRNAs that regulate key mechanosensitive genes were also detected in human and porcine AVECs (Tables 3 and 4) [77, 122, 123].

Altered expression of endothelial mechano-miRNAs is also detected by Theodoris et al. in hiPSC-based modeling of human NOTCH1 mutations in AS [67]. They reported a significant dysregulation of 181 small ncRNAs in NOTCH1+/− compared to WT ECs in static versus shear stress conditions. Among them miR-30d, miR-663, miR-1260b and miR-3960 were upregulated while miR-20a/b, miR-21, miR-26a, mir-29c, miR-30e, miR-106a and miR-126 were downregulated in NOTCH1+/− ECs [67].

Moreover, differential expression of miRNAs was also detected in circulatory osteoprogenitor cells that play a significant role in pathogenesis of AS disease. Thus, a recent study reported by Takahashi et al. detected a higher levels of pro-osteogenic miR-30c in the AS group compared to controls and lower levels of miR-31, miR-106a, miR-148a, miR-204, miR-211, and miR-424, previously reported as negative regulators of pro-osteogenic pathways in mesenchymal stem cells [147,148,149,150]. Also, the degree of aortic valve calcification in their samples was weakly positively correlated with the number of COPCs and miR30c levels. Furthermore, the number of COPCs and the level of BGLAP/OCN protein in these cells was positively correlated with miR-30c and negatively correlated with the levels of remaining miRNAs [147]. Importantly, after surgical procedure both the number of COPCs and the levels of miR30c were decreased while the levels of the other miRNAs remained the same [147]. Moreover, the observed changes in miRNAs levels were greater after AVR than TAVR procedure. That may be explained by the procedural differences between these two surgical procedures, with TAVR leaving the residual diseased tissue around the prosthetic valve, which may continue to activate osteogenic processes via dysregulation of ossification related miRNAs [147].

Finally, Li et al. recently reported the role of macrophage miRNAs in regulation of AS [134]. They found that M1 macrophage-derived microvesicles deliver miR-214 promote pro-osteogenic differentiation of VICs and subsequent aortic valve calcification through downregulation of TWIST1, a direct target of miR-214 [134]. The upregulation of miR-214 of aortic valve samples was accompanied with both valve calcification and M1 macrophage polarization. In addition, the functional involvement of miR-214/TWIST1 in the osteogenic differentiation of VICs was further confirmed by intravenous treatment of hypercholesterolemic APOE−/− mice with a miR-214 inhibitor, which significantly suppressed valve calcification and resulted in the upregulation of TWIST1 [134].

Circulatory microRNAs

The utility of circulating miRNAs, as potential diagnostic, and prognostic biomarkers of AS, has also attracted considerable attention over the recent years. They have been correlated both with LV structural and functional impairment and with the outcome of AS patients after surgery.

For instance, the role of circulatory miR-21 as an indicator of LV fibrosis in AS patients in response to PO was supported by findings of Villar et al. and Fabiani et al. while Coffey et al. reported the higher levels of circulatory miR-21-5p in AS patients without coronary artery disease (CAD), but no apparent difference was found between groups with CAD [151,152,153]. Moreover, they showed that plasma levels of miR-21 and miR-21-5p correlated directly with the mean transvalvular gradients thus underscoring the value of circulating miR-21 as a biomarker for MF [151,152,153].

Coffey et al. detected higher circulatory levels of miR-451a and miR-22-3p, and lover levels of miR-24-3p and miR-382–5p in AS patients compared to controls that also remained significantly after adjusting for age [153]. However, after qRT-PCR validation only miR-22-3p and miR-382–5p had levels expected from microarray analysis. In addition, miR-22-3p and miR-24-3p were increased, whereas miR-382-3p was reduced in AS participants with CAD [153]. Furthermore, similar to miR-21-5p, the circulatory levels of miR-382–5p in AS patients also showed a significant correlation with maximum transvalvular velocity and mean gradient, but not with LV mass index [153].

Significantly lower circulating levels in AS patients compared to healthy controls were also reported for miR-1, miR-133, and miR-378 by Chen et al. [154]. In addition, their results indicated that the lower levels of miR-378 may serve as predictor for LVH independent of the pressure gradient [154]. Similarily, Garcia et al. reported that the higher preoperative plasma levels of miR-133a can predict the reversibility of LV hypertrophy after AVR, while Røsjø et al. detected positive association between higher circulating levels of miR-210 and increased mortality during follow-up of AS patients independently of established risk indices, including NT-proBNP (N-terminal proBrain Natriuremic Peptide) levels [155, 156].

Quite recently Martínez-Micaelo et al. reported that plasma circulating miRNA expression profile in AS patient may also reflect the morphology of the aortic valve (bicuspid/tricuspid) [124]. Their miRNA-wide microarray and qRT-PCR comparison between the groups (healthy TAV subjects without aortic dilation, BAV patients without aortic dilation and BAV patients with aortic dilation) revealed that the expression levels of circulating miR-122, miR-130a and miR-486 are significantly influenced by the morphology of the aortic valve (BAV vs. TAV), while the expression pattern of miR-718 was inversely correlated with the aortic diameter and thus may possibly be used an independent predictor of aortic dilation [124]. In addition, the targeted gene prediction and putative function analysis of selected miRNAs revealed that the bicuspid valve morphology-associated miRNAs (miR-122, miR-130a and miR-486) most probably affects the TGFβ1 signaling pathway (a total of 32 targeted genes) while the dilatation related miR-718 may be associated with focal adhesion and blood vessel remodeling processes [124].

Another, blood based microarray profiling reported by Derda et al. detected that among 8 known cardiovascular miRNAs (miR-1, miR-21, miR-29a, miR-29b, miR-29c, miR-133a, miR-155 and miR-499) only miR-29a, and miR-29c have potential to distinguish between patients with AS and hypertrophic non-obstructive (HNCM) and obstructive (HOCM) cardiomyopathies [157]. More specifically, they found increased levels of miR-29a in HOCM patients that correlated markers of cardiac hypertrophy while miR-29c was upregulated in AS but not in the other patient groups [157].

The possibility that circulating miRNAs are differentially expressed in the blood of hypertrophic cardiomyopathy patients and those with LVH induced by AS has been further investigated by Roncarati et al. [158]. Among the miRNAs significantly increased in the plasma of their patients (miR-27a, miR-199a-5p, miR-26a, miR-145, miR-133a, miR-143, miR-199a-3p, miR-126-3p, miR-29a, miR-155, miR-30a, and miR-21) they found that correlation with LVH holds true for only miR-199a-5p, miR-27a, and miR-29a, whereas only miR-29a was significantly associated with both hypertrophy and fibrosis, thus identifying it as a potential biomarker for myocardial remodeling assessment in CH [158]. The similar trend was detected for only five miRNAs (miR-21, miR-26a, miR-27a, miR-30a, and miR-133a) in AS patients, whereas mir-29a levels showed no increased in AS relative to control, thus suggesting specific miR signatures for these two pathological conditions [158].

Interestingly, Varrone et al. reported that plasma levels of mRNA protein targets in AS patients may also be used for indirect measurement of myocardial miRNAs expression. They detected inverse relationship between myocardial expressions of miR-1 and circulating levels of (FABP3, heart-type fatty acid-binding protein-3) in AS patients with LVH [159]. Specifically, myocardial miR-1 expression was decreased while FABP3 levels were increased compared to controls [159]. Furthermore, the decrease of myocardial wall stress following TAVR procedure have led to downregulation of FABP3 levels to values comparable to ones for control subjects [159]. Moreover, the increased level of circulating IGF1 (insulin-like growth factor 1) in AS patients were also significantly blunted by the TAVR procedure, thus suggesting that this reciprocal relationship between miR-1 and FABP3 protein may be tightly controlled by the IGF1/miR-1/FABP3 signaling axis [159]. It seems that the hypertrophic response in AS patients is followed by the increase in IGF1 plasma levels and downregulation of miR-1 [159]. Eventually, the reduction of cellular miR-1 levels relieves its negative control over the expression of FABP3, thus leading to a prompt protein release into the circulation where it might be used for indirect measurement of myocardial miR-1 activity [159].

Pericardial fluid miRNAs

Several vascular-enriched miRNAs have also been detected in pericardial fluid (PF) of AS patients undergoing surgery. For instance, Miyamoto et al. reported significantly higher levels of miR-423-5p in PF compared to its plasma values [160]. Contrary, the levels of muscle-enriched (miR-133a) and vascular-enriched (miR-126 and miR-92a) miRNAs were found unaltered [160].

In another experiment, Kuosmanen et al. detected more than 70 miRNAs in PF collected from AS and other HF patients (coronary artery disease, mitral valve insufficiency, aortic valve insufficiency, and other cardiovascular disease) during open-heart surgery, with let-7b-5p, miR-16-5p, miR-21-5p, miR-125b-5p, and miR-451a being the most abundant [161]. However, despite the differences in disease etiologies (ischemic vs. nonischemic) or the stage of the HF the overall miRNA profiles were quite similar between the groups [161].

The existence of functional miRNAs in PF samples of AS patients was recently reported by Beltrami et al. [162]. Among the 359 detected miRNAs in PF and patient matched peripheral plasma samples they confirmed PF exosome enrichment for 15 of them (let-7b-5p, miR-15a-5p, miR-16-5p, miR-19b-3p, miR-21-5p, miR-21-5p, miR-23a-3p, miR-24-3p, miR-27a-3p, miR-27b-3p, miR-29a-3p, miR-29b-3p, miR-29c-3p, miR-126-3p, miR-451a) [162]. These miRNAs were also found co-expressed in patient’s myocardium and ascending aorta [162]. Furthermore, at functional level these PF derived exosomes were able to improve survival, proliferation, and networking of cultured endothelial cells, restore their angiogenic capacity and promote post-ischemic blood flow recovery and angiogenesis in mice models, all of which was partially mediated by exosomal transfer of let-7b-5p miRNA and decreased transcription of its targeted gene TGFBR1 [162]. Thus, it seems that PF enriched cardiovascular miRNAs may act as endocrine and paracrine signaling factors responsible for local crosstalk between cardiac cells, and some of them may be utilized to reflect the patient clinical status.

Myocardial miRNAs

Expressional profiling of myocardial biopsies from AS patients further confirms the regulatory role of miRNAs in development of LVH and fibrosis induced by AS [163].

The role of myocardial miR-21 in these processes was supported by several investigators. Villar et al. reported that AS patients featured higher myocardial expression levels of both primary (pri-miR-21) and mature miR-21 transcripts. They were restricted to the interstitial cells (cardiac fibroblasts) within the ECM, with no or very weak miR-21 signals detected in the cardiomyocytes of AS patients or control samples, correspondingly [151]. Moreover, both the myocardial and circulating levels of miR-21 were positively correlated with the myocardial expression levels of genes encoding collagen I, collagen III, fibronectin, TGFβ1 and effectors of TGFß signaling (SMAD2 and MAP3K7/TAK1 mitogen-activated protein kinase kinase kinase 7) together with negative correlation with the miR-21 targets (PTEN, phosphatase and tensin homolog; TIMP3, tissue Inhibitor of metalloproteinases; RECK, reversion inducing cysteine rich protein with kazal motifs, and PDCD4 programmed cell death 4/neoplastic transformation inhibitor), thus suggesting a link between the severity of the maladaptive remodeling and miR-21 upregulation in the myocardium [151]. Another study reported by Lorenzen et al. detected the link between increased OPN expression and activation of the transcription factor AP-1, with subsequent miR-21 induction and regulation of downstream antifibrotic targets (PTEN and SMAD7) in angiotensin 2 (ANGII/AGT)-induced cardiac cells and in LV biopsies from AS patients with myocardial fibrosis [164]. In addition, Garcia et al. reported a new TGFβ-dependent regulatory mechanism involved in the miR-21 transcription and posttranscriptional maturation, through the interaction of p-SMAD2/3 effectors with the ribonuclease DICER1 processor machinery [165]. They showed that this TGFβ-dependent facilitator mechanism could contribute to the pathogenesis of PO-induced myocardial remodeling both in the experimental mouse model with transverse aortic constriction (TAC), and in patients with AS [165].

The association of miRNAs with PO-induced cardiac hypertrophy and heart failure in mouse model (TAC) of AS has also been reported by Eskildsen et al. [166]. Together with several well-described cardiac disease related miRNAs (miR-21, miR-29, miR-133a, and miR-208b), they identified altered expression of three novel miRNAs; miR-24, miR-301a, and miR-335 in the left ventricle of TAC affected mice compared to controls [166]. Interestingly, the increased expression of miR-24, miR-301a, and miR-335 was not found in an animal model of myocardial infarction thus suggesting that their regulation is specific for AS and is not part of a general cardiac disease response [166]. Furthermore, the importance of miR-133a in regulation of AS induced LVH was also reported by Duisters et al. [167]. They found reduced miR-133 and miR-30c expression levels in several forms of PO–induced LVH including the hearts of AS patients [167]. In addition, the expression of both miRNAs was inversely related to the protein level of CTGF (connective tissue growth factor), a key molecule in the process of fibrosis and a powerful inducer of extracellular matrix (ECM) synthesis [167]. Moreover, several lines of evidence provided by this study confirmed a negative regulatory role of miR-133 and miR-30c on the levels of CTGF gene expression, not only by repressing CTGF translation but also by degrading its mRNA [167]. Interestingly, 34 of the 42 mammalian collagen genes are also predicted targets of miR-133, thus suggesting a major role for this miRNA in preventing cardiomyocyte collagen synthesis, and the quality of their surrounding ECM [167].

Recently, another study reported by Jiang et al. detected the negative correlation between miR-133 and lncRNA-ROR (regulator of reprogramming, also named lncRNA-ST8SIA3) in mouse (TAC) model of pressure overload induced cardiac hypertrophy [168]. Thus, it seems that lncRNA-ROR serves as the miR-133 sponge while on the other hand overexpression of miR-133 successfully attenuates the expression of lncRNA-ROR reversing its pro-hypertrophic influence leading to markedly decreased expression of fetal NPPA/ANP (natriuretic peptide A) and NPPB/BNP (natriuretic peptide B) genes [168]. In addition, Renaud et al. reported that members of class I and IIb HDACs may also play a role in the regulation of PO-induced miR-133a downregulation in mouse (TAC) model of cardiac hypertrophy [169]. More specifically, the treatment with the class I and IIb HDAC inhibitor Vorinostat (also known as SAHA- suberoylanilide hydroxamic acid) significantly attenuated TAC-Induced downregulation of miR-133a and diminished the upregulation of CTGF protein abundance and collagen deposition, thus suggesting that the effect of HDAC inhibition on miR-133a expression is reflected on its downstream fibrotic targets [169].

These two reports clearly indicate the importance of understanding the complex crosstalk between miRs and other epigenetic regulators such as lncRNAs and HDACs, thus providing the ground for innovative therapies to reset the epigenome alterations in AS and other heart diseases.

MiR-133a also emerged as a key element of the postoperative reverse remodeling process of LVH in the report of Villar et al. [170]. They identified the microRNA-133a levels in intraoperative biopsies as a significant positive predictor of left ventricular mass normalization in AS patients while β-myosin heavy chain expression and BMI constituted negative predictors [170].

Furthermore, Beaumont et al. have reported that myocardial down-regulation of miR-122 might also be involved in the development of myocardial fibrosis in AS patients, most probably through the up-regulation of TGFβ1 [171]. They also reported differential expression of 118 miRNAs (99 down-regulated and 18 up-regulated) in AS patients with severe myocardial fibrosis (SF) compared with the non-SF AS group [171]. The role of miRs in regulation of TGFβ pathway in the cardiomyocyte hypertrophy and interstitial fibrosis in the settings of AS was also reported by Tijsen et al. [172]. They showed that the members of the miR-15 family (miR-15a/b, miR-16, miR-195, miR-497, and miR-322), that are abundantly expressed in the heart and up-regulated in the diseased myocardium, directly or indirectly inhibit TGFβ-pathway by targeting TGFBR1 and several other genes (p38, SMAD2, SMAD3, SMAD7, and Endoglin/ENG) within this pathway [172].

Specific contribution of miRNAs to LV cardiomyopathy induced by AS was further confirmed by Ikeda et al. [173, 174]. They reported 87 differentially expressed miRNAs (among 428 examined) in LV samples of AS patients compared to samples from diseased hearts with DCM and ischemic cardiomyopathy/ICM and the non-failing control group [173, 174]. Among the miRNAs with known cardiac-enriched expression (miR-1, miR-133, and miR-208), miR-1 was downregulated in DCM and AS and tended to be downregulated in ICM patients while the expression of miR-133 and miR-208 were not significantly changed [173, 174]. In addition, the miR-214 exhibited the up-regulation in all three disease groups while the miR-19a and miR-19b were the most down-regulated miRs in DCM and AS, but not in ICM patients [173, 174]. MiR-24 has also been identified as significantly upregulated in AS patient group [173, 174]. Importantly, differential expression of 13 of these miRNAs was specific to AS, while eight miRNAs exhibited differential expression in cardiomyopathy groups (ICM and DCM) that did not overlap with the expression of AS specific miRs thus suggesting that altered expression of these miRNAs reflects distinct disease mechanisms or disease stage in AS compared with cardiomyopathy samples [173, 174].

Another miRNA-wide microarray study reported by Gallego et al. detected differential expression of 70 miRNAs (64 downregulated and 6 upregulated) between control subjects and two clusters of AS patient identified according to cardiomyocyte apoptotic index [175]. Among them miR-10b, miR-125b-2* and miR-338-3p were inversely correlated with cardiomyocyte apoptotic index [175]. They concluded that myocardial downregulation of miR-10b may be involved in increased cardiomyocyte apoptosis in AS patients, probably through the upregulation of apoptotic peptidase activating factor 1 (APAF1) gene expression, thus contributing to cardiomyocyte damage and to the development of heart failure [175].

Finally, Beaumont et al. reported reduced expression of miR-133 and miR-19b in the myocardial and serum samples of AS patients when compared to controls [176]. Moreover, both myocardial and serum miR-19b levels were found inversely correlated with expression levels of lysyl oxidase (LOX), collagen cross-linking and left ventricular stiffness in AS patients, particularly in patients with heart failure [176]. In addition, the expression levels of miR-29b, miR-1, miR-208a and miR-499-5p was under the limit while no differences were found for miR-21 levels between serum samples from AS patients and control subjects [176].

Taken together the above data provide clear evidence that aberrantly expressed miRNAs are implicated in a diverse spectrum of pathophysiological pathways leading to AS development and progression, and consequently to pressure overload-induced myocardial remodeling and hypertrophy. This is obviously just a beginning of story with the list of novel miRNA players and the previously unknown roles for the already existent ones growing at a steady pace.

Conclusion

Herein we presented a growing body of experimental evidences to support the key role of epigenetic alterations in the etiology and progression of AS. Evidently they participate in crucial disease-prone phenotype changes of aortic valve cells, and regulate key processes underlying AS-induced valvular tissue remodeling and maladaptive myocardial hypertrophy, i.e., fibrosis, calcification, LV remodeling, and inflammation. As can be seen, alteration of DNA methylation marks may contribute to production of proinflammatory mediators (ALOX5 induced LTB4 production), and have role in osteogenic transformation of VICs (H19 induced NOTCH1 downregulation and subsequent upregulation of RUNX2, BMP2, and OCN) while some of them demonstrate promising biomarker potentials for the prediction of AS status (analysis of dried blood spots of neonatal AS patients). Proinflammatory and osteogenic role was also demonstrated for histon code marks (mediated by alterations of SIRTs gene expression), and their involvement in AS-induced pathological remodeling of left ventricular myocardium was also clearly established (e.g., reexpression of the fetal gene programs induced by altered expression of JARID2 and DPF3 genes). Apparently most studies were focused on the role of small non-coding miRNAs demonstrating their essential role in some of the key processes responsible for disease progression such as the phenotypic alterations of valvular endothelial and interstitial cells under pathologically altered blood born mechanical forces, induction of proinflammatory pathways, and osteogenic transdifferentiation of VICs (Table 4). Moreover, they seem to be crucial for the disease-prone role of monocyte-macrophage and COPC cells and have established role in regulation of myocardial fibrosis and LVH. Most importantly, specific miRNA signatures (e.g., plasma levels of miR-21, miR-210, and miR-378), in combination with clinical and functional imaging parameters, could represent useful non-invasive biomarkers of disease progression or recovery after aortic valve replacement. Less is known about the ATP-dependent chromatin remodeling processes and long noncoding RNAs and their extent and potential role in subclinical and clinical manifestation of AS have yet to be examined and experimentally validated in both small and large scale human and animal studies. Also, we currently see only the very tip of histone and DNA methylation marks lying under the surface of AS patohistogenesis. Since all epigenetic mechanisam in a given cell and tissue structure are mutually interdependent as clearly represented by Theodoris et al. A better understanding of epigenome landscape in native and infiltrating aortic valve cells and affected myocardium will certainly shed new insights into all aspects of AS pathology and add important incremental diagnostic and prognostic informations useful for risk stratification and patient management. Even more, since all known epigenetic marks are potentially reversible this perspective is especially exciting given the potential for development of successful and non-invasive therapeutic intervention and reprogramming of cells at the epigenetic level even in the early stages of disease progression.

Abbreviations

ACTA2/ αSMA:

alpha-smooth muscle actin

AFAP1:

Actin filament associated protein 1

AFAP1-AS1:

AFAP1 Antisense RNA 1

ALOX5/5LO:

5-lipoxygenase

ALOXAP/FLAP:

5-LO activating protein

ALP:

Alkaline phosphatase

ANF:

Atrial natriuretic factor

ANGII/AGT:

Angiotensin 2

APAF1:

Apoptotic peptidase activating factor 1

APOA1:

Apolipoprotein A1

APOA5:

Apolipoprotein A5

APO-AS1:

APOA1-antisense transcript 1

APOB:

Apolipoprotein B

APOE:

Apolipoprotein E

AS:

Aortic valve stenosis

ASc:

Aortic valve sclerosis

AVECs:

Aortic valve endothelial cells

aVICs:

Activated aortic valve interstitial cells

AVICs:

Aortic valve interstitial cells

AVR:

Aortic valve replacement

BAF:

BRG/BRM-associated factor

BAV:

Bicuspid aortic valve

BGLAP/OCN:

Bone gamma-carboxyglutamate (Gla) protein; osteocalcin

BMP:

Bone morphogenetic protein

BMPR2:

Bone morphogenic protein receptor type 2

BRG1/SMARCA4:

Brahma-related gene 1; SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4

BRM/SMARCA2:

Brahma; SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2

CAD:

Coronary artery disease

CARMEN:

Cardiac mesoderm enhancer-associated noncoding RNA

CCL:

Chemokine (C-C motif) ligand

CH:

Cardiac hypertrophy

CHAST:

Cardiac hypertrophy-associated lncRNA transcript

CMAI:

Cardiomyocyte apoptotic index

COPCs:

Circulatory osteoprogenitor cells

CSNK2:

Casein kinase 2

CTGF:

Connective tissue growth factor

CVD:

Cardiovascular diseases

DCM:

Dilated cardiomyopathy

DICER1:

Dicer 1, Ribonuclease III

DLX5:

Distal-less homeobox 5

DNMT:

DNA (Cytosine-5-)-methyltransferase

DPF3/BAF45C:

Double PHD fingers 3/BRG1-associated factor 45C

DUSP27:

Dual-specificity phosphatase 27

ECM:

Extracellular matrix

ECs:

Endothelial cells

EDN1:

Endothelin 1

EGFR:

Epidermal growth factor receptor

ENG:

Endoglin

ETS1:

ETS proto-oncogene 1

EZH:

Enhancer of zeste

FABP:

Fatty acid-binding protein

FOXO1:

Forkhead box O1

GATA4:

Growth arrest and DNA damage-inducible protein 45

H3K27ac:

Acetylation of histone H3 at lysine 27

H3K27me2/3:

Methylation of histone H3 lysine 27

H3K27me3:

Trimethylation of histone H3 at lysine 27

H3K4me1:

Monomethylation of histone H3 at lysine 4

H3K4me3:

Trimethylation of histone H3 at lysine 4

HAT:

Histone acetyltransferase

HDAC:

Histone deacetylase

HEY1/2:

Hes related family BHLH transcription factor with YRPW motif 1/2

HF:

Heart failure

hiPSC:

Human induced pluripotent stem cell

HNCM:

Hypertrophic non-obstructive cardiomyopathy

HOCM:

Hypertrophic obstructive cardiomyopathy

HOTAIR:

HOX transcript antisense RNA

ICM:

Ischemic cardiomyopathy

IGF:

Insulin-like growth factor

JARID2:

Jumonji and AT-rich interaction domain containing 2

LA:

LDLR-/-APOB100/100 mice

LDLR:

Low density lipoprotein

lncRNAs:

Long non-coding RNAs

LOX:

Lysyl oxidase

LTA4H:

Leukotriene A4 hydrolase

LTB4:

Leukotriene B4

LTC4S:

Leukotriene C4 synthase

LV:

Left ventricle

LVH:

Left ventricular hypertrophy

MALAT1:

Metastasis-associated lung adenocarcinoma transcript 1

MAP3K7/TAK1:

Mitogen-activated protein kinase kinase kinase 7

MF:

Myocardial fibrosis

MGP/MGLAP:

Matrix γ-carboxyglutamate (Gla) protein

microRNAs/miRNAs/miRs:

Small non-coding RNAs

MMP:

Matrix metalloproteinase

MSCs:

Mesenchymal stem cells

MSX2:

MSH homeobox 2

MYH2:

Myosin heavy chain 2

MYL7:

Myosin light chain 7

ncRNAs:

Non-coding RNAs

NFYC:

Nuclear transcription factor gamma subunit c

NOTCH1:

Notch/drosophila/homolog 1/translocation-associated

NPPA/ANP:

Natriuretic peptide A

NT-proBNP:

N-terminal probrain natriuremic peptide

obVICs:

Osteoblast-like aortic valve interstitial cells

PARP:

Poly (ADP ribose) polymerase

PCSK9:

Proprotein convertase and subtilisin/knexin-type 9

PDCD4:

Programmed cell death 4 (neoplastic transformation inhibitor)

PF:

Pericardial fluid

PO:

Pressure overload

POSTN:

Periostin

PPARγ:

Peroxisome proliferator-activated receptor γ

PRC2:

Polycomb repressive complex 2

PRINS:

Psoriasis associated non-protein coding RNA induced by stress

PTEN:

Phosphatase and tensin homologue

PTENP1:

Phosphatase and tensin homolog pseudogene 1

PTENP1-AS:

PTENP1-antisense RNA

qVICs:

Quiescent aortic valve interstitial cells

RECK:

Reversion inducing cysteine rich protein with kazal motifs

RETN:

Resistin

ROR:

Regulator of reprogramming, also named lncRNA

ROS:

Reactive oxygen species

RUNX:

Runt-related transcription factor 1

SAHA:

Suberoylanilide hydroxamic acid

SIRT:

Silent information regulator-two

SM22α:

Smooth muscle protein 22-alpha

SMAD:

SMAD family member

SMCs:

Smooth muscle cells

SOD:

Superoxide dismutase

SOST:

Sclerostin

SOX2:

SRY-Box 2

SOX2-OT:

SOX2 overlapping transcript

SP7/OSX:

Osterix

SPP1/OPN:

Osteopontin, secreted phosphoprotein 1; bone sialoprotein 1

SWI/SNF:

SWItch/sucrose non-fermentable)-like ATP-dependent BAF chromatin remodeling complex chromatin remodeling complex

TAC:

Transverse aortic constriction

TAV:

Tricuspid aortic valve

TAVR:

Transcatheter aortic valve replacement

TBX3:

T-box 3

TGFBR1:

Transforming growth factor beta receptor 1

TGFβ:

Transforming growth factor beta

TIMP3:

Tissue inhibitor of metalloproteinases

TWIST1:

Twist-related protein 1

TXNRD2:

Thioredoxin reductase 2

VICs:

Aortic valve interstitial cells

VSMCs:

Vascular smooth muscle cells

WISP2:

WNT1 inducible signaling pathway protein 2

WISPER:

WISP2 super-enhancer associated RNA

WT:

Wild type

References

  1. Iung B, Baron G, Butchart EG, Delahaye F, Gohlke-Bärwolf C, Levang OW, et al. A prospective survey of patients with valvular heart disease in Europe: The Euro Heart Survey on Valvular Heart Disease. Eur Heart J. 2003;24(13):1231–43.

    Article  PubMed  Google Scholar 

  2. Thaden JJ, Nkomo VT, Enriquez-Sarano M. The global burden of aortic stenosis. Prog Cardiovasc Dis. 2014;56(6):565–71.

    Article  PubMed  Google Scholar 

  3. Yutzey KE, Demer LL, Body SC, Huggins GS, Towler DA, Giachelli CM, et al. Calcific aortic valve disease: a consensus summary from the Alliance of Investigators on Calcific Aortic Valve Disease. Arterioscler Thromb Vasc Biol. 2014;34(11):2387–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Osnabrugge RL, Mylotte D, Head SJ, Van Mieghem NM, Nkomo VT, LeReun CM, et al. Aortic stenosis in the elderly: disease prevalence and number of candidates for transcatheter aortic valve replacement: a meta-analysis and modeling study. J Am Coll Cardiol. 2013;62(11):1002–12.

    Article  PubMed  Google Scholar 

  5. Goldbarg SH, Elmariah S, Miller MA, Fuster V. Insights into degenerative aortic valve disease. J Am Coll Cardiol. 2007;2550(13):1205–13.

    Article  Google Scholar 

  6. Iung B, Vahanian A. Epidemiology of valvular heart disease in the adult. Nat Rev Cardiol. 2011;8(3):162–72.

    Article  PubMed  Google Scholar 

  7. Gohlke-Bärwolf C, Minners J, Jander N, Gerdts E, Wachtell K, Ray S, et al. Natural history of mild and of moderate aortic stenosis-new insights from a large prospective European study. Curr Probl Cardiol. 2013;38(9):365–409.

    Article  PubMed  Google Scholar 

  8. Otto CM, Prendergast B. Aortic-valve stenosis- from patients at risk to severe valve obstruction. N Engl J Med. 2014;371(8):744–56.

    Article  CAS  PubMed  Google Scholar 

  9. Sathyamurthy I, Alex S. Calcific aortic valve disease: is it another face of atherosclerosis? Indian Heart J. 2015;67(5):503–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pawade TA, Newby DE, Dweck MR. Calcification in Aortic Stenosis: The Skeleton Key. J Am Coll Cardiol. 2015;66(5):561–77.

    Article  PubMed  Google Scholar 

  11. Miller JD, Weiss RM, Heistad DD. Calcific aortic valve stenosis: methods, models, and mechanisms. Circ Res. 2011;108(11):1392–412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Towler DA. Molecular and cellular aspects of calcific aortic valve disease. Circ Res. 2013;113(2):198–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Li C, Xu S, Gotlieb AI. The progression of calcific aortic valve disease through injury, cell dysfunction, and disruptive biologic and physical force feedback loops. Cardiovasc Pathol. 2013;22(1):1–8.

    Article  PubMed  Google Scholar 

  14. Butcher JT, Mahler GJ, Hockaday LA. Aortic valve disease and treatment: the need for naturally engineered solutions. Adv Drug Deliv Rev. 2011;63(4–5):242–68.

    Article  CAS  PubMed  Google Scholar 

  15. Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial cell phenotypes in regulating heart valve pathobiology. Am J Pathol. 2007;171(5):1407–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rattazzi M, Pauletto P. Valvular endothelial cells: guardians or destroyers of aortic valve integrity? Atherosclerosis. 2015;242(2):396–8.

    Article  CAS  PubMed  Google Scholar 

  17. Gössl M, Khosla S, Zhang X, Higano N, Jordan KL, Loeffler D, et al. Role of circulating osteogenic progenitor cells in calcific aortic stenosis. J Am Coll Cardiol. 2012;60(19):1945–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Chen JH, Yip CY, Sone ED, Simmons CA. Identification and characterization of aortic valve mesenchymal progenitor cells with robust osteogenic calcification potential. Am J Pathol. 2009;174(3):1109–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Coffey S, Cox B, Williams MJ. The prevalence, incidence, progression, and risks of aortic valve sclerosis: a systematic review and meta-analysis. J Am Coll Cardiol. 2014;63(25 Pt A):2852–61.

    Article  PubMed  Google Scholar 

  20. Pasipoularides A. Calcific Aortic Valve Disease: Part 1-Molecular Pathogenetic Aspects, Hemodynamics, and Adaptive Feedbacks. J Cardiovasc Transl Res. 2016;9(2):102–18.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP 3rd, Guyton RA, et al. 2014 AHA/ACC guideline for the management of patients with valvular heart disease: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63(22):2438–88.

    Article  PubMed  Google Scholar 

  22. Lindman BR, Clavel MA, Mathieu P, Iung B, Lancellotti P, Otto CM, Pibarot P. Calcific aortic stenosis. Nat Rev Dis Primers. 2016;2:16006.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Yurek LA, Jakub KE, Menacho MM. Severe Symptomatic Aortic Stenosis in Older Adults: Pathophysiology, Clinical Manifestations, Treatment Guidelines, and Transcatheter Aortic Valve Replacement (TAVR). J Gerontol Nurs. 2015;41(6):8–13.

    Article  PubMed  Google Scholar 

  24. Waddington CH. The epigenotype. Endeavor 1942; 1:18–20. Reprinted in Int J Epidemiol. 2012; 41:10–13.

  25. Bonasio R, Tu S, Reinberg D. Molecular signals of epigenetic states. Science. 2010;330(6004):612–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kim JK, Samaranayake M, Pradhan S. Epigenetic mechanisms in mammals. Cell Mol Life Sci. 2009;66(4):596–612.

    Article  CAS  PubMed  Google Scholar 

  27. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12(12):861–74.

    Article  CAS  PubMed  Google Scholar 

  28. Zink LM, Hake SB. Histone variants: nuclear function and disease. Curr Opin Genet Dev. 2016;37:82–9.

    Article  CAS  PubMed  Google Scholar 

  29. Rizki G, Boyer LA. Lncing epigenetic control of transcription to cardiovascular development and disease. Circ Res. 2015;117(2):192–206.

    Article  CAS  PubMed  Google Scholar 

  30. Narlikar GJ, Sundaramoorthy R, Owen-Hughes T. Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes. Cell. 2013;154(3):490–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov. 2012;11(5):384–400.

    Article  CAS  PubMed  Google Scholar 

  32. Murr R. Interplay between different epigenetic modifications and mechanisms. Adv Genet. 2010;70:101–41.

    CAS  PubMed  Google Scholar 

  33. Winter S, Fischle W. Epigenetic markers and their cross-talk. Essays Biochem. 2010;48(1):45–61.

    Article  CAS  PubMed  Google Scholar 

  34. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33(Suppl):245–54.

    Article  CAS  PubMed  Google Scholar 

  35. Sun C, Burgner DP, Ponsonby AL, Saffery R, Huang RC, Vuillermin PJ, et al. Effects of early-life environment and epigenetics on cardiovascular disease risk in children: highlighting the role of twin studies. Pediatr Res. 2013;73(4 Pt 2):523–30.

    Article  CAS  PubMed  Google Scholar 

  36. Napoli C, Infante T, Casamassimi A. Maternal-foetal epigenetic interactions in the beginning of cardiovascular damage. Cardiovasc Res. 2011;92(3):367–74.

    Article  CAS  PubMed  Google Scholar 

  37. Brunet A, Berger SL. Epigenetics of aging and aging-related disease. J Gerontol A Biol Sci Med Sci. 2014;69(Suppl 1):S17–20.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Handy DE, Castro R, Loscalzo J. Epigenetic modifications: basic mechanisms and role in cardiovascular disease. Circulation. 2011;123(19):2145–56.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Whayne TF. Epigenetics in the development, modification, and prevention of cardiovascular disease. Mol Biol Rep. 2015;42(4):765–76.

    Article  CAS  PubMed  Google Scholar 

  40. Nwachukwu N, Hagler M, Kafa R, Roos C, Miller J. Evidence for altered DNA methylation as a major regulator of gene expression in calcific aortic valve disease. FASEB J. 2014;28(1):Suppl 671.15. http://www.fasebj.org/content/28/1_Supplement/671.15.short

    Google Scholar 

  41. Sritharen Y, Roos CM, Nwachukwu N, Kafa R, Hagler MA, Verzosa GC, et al. Genetic Inactivation of DNMT3b Protects Against Activation of Osteogenic Signaling and Slows Progression of Aortic Valve Stenosis in Mice. FASEB J. 2016;30(1):Suppl 1178.12.

    Google Scholar 

  42. Radhakrishna U, Albayrak S, Alpay-Savasan Z, Zeb A, Turkoglu O, Sobolewski P, et al. Genome-Wide DNA Methylation Analysis and Epigenetic Variations Associated with Congenital Aortic Valve Stenosis (AVS). PLoS One. 2016;11(5):e0154010.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Gilsbach R, Preißl S, Rühle F, Weiß S, Mühle A, Doenst T, et al. Genome-wide DNA methylation in chronic heart disease. Clin Res Cardiol. 2012;101(Suppl 1).V843. Available at http://www.abstractserver.de/dgk2012/ft/abstracts/V843.HTM.

  44. Nagy E, Bäck M. Epigenetic regulation of 5-lipoxygenase in the phenotypic plasticity of valvular interstitial cells associated with aortic valve stenosis. FEBS Lett. 2012;586(9):1325–9.

    Article  CAS  PubMed  Google Scholar 

  45. Nagy E, Andersson DC, Caidahl K, Eriksson MJ, Eriksson P, Franco-Cereceda A, et al. Upregulation of the 5-lipoxygenase pathway in human aortic valves correlates with severity of stenosis and leads to leukotriene-induced effects on valvular myofibroblasts. Circulation. 2011;123(12):1316–25. 14

    Article  CAS  PubMed  Google Scholar 

  46. Hadji F, Boulanger MC, Guay SP, Gaudreault N, Amellah S, Mkannez G, Bouchareb R, Marchand JT, Nsaibia MJ, Guauque-Olarte S, Pibarot P, Bouchard L, Bossé Y, Mathieu P. Altered DNA Methylation of Long Noncoding RNA H19 in Calcific Aortic Valve Disease Promotes Mineralization by Silencing NOTCH1. Circulation. 2016;134(23):1848–62.

    Article  CAS  PubMed  Google Scholar 

  47. Barrick CJ, Roberts RB, Rojas M, Rajamannan NM, Suitt CB, O’Brien KD, et al. Reduced EGFR causes abnormal valvular differentiation leading to calcific aortic stenosis and left ventricular hypertrophy in C57BL/6J but not 129S1/SvImJ mice. Am J Physiol Heart Circ Physiol. 2009;297(1):H65–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Mathieu P, Voisine P, Pépin A, Shetty R, Savard N, Dagenais F. Calcification of human valve interstitial cells is dependent on alkaline phosphatase activity. J Heart Valve Dis. 2005;14(3):353–7.

    PubMed  Google Scholar 

  49. Delgado-Calle J, Sañudo C, Sánchez-Verde L, García-Renedo RJ, Arozamena J, Riancho JA. Epigenetic regulation of alkaline phosphatase in human cells of the osteoblastic lineage. Bone. 2011;49(4):830–8.

    Article  CAS  PubMed  Google Scholar 

  50. Zhou GS, Zhang XL, Wu JP, Zhang RP, Xiang LX, Dai LC, et al. 5-Azacytidine facilitates osteogenic gene expression and differentiation of mesenchymal stem cells by alteration in DNA methylation. Cytotechnology. 2009;60(1–3):11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Koos R, Brandenburg V, Mahnken AH, Schneider R, Dohmen G, Autschbach R, et al. Sclerostin as a potential novel biomarker for aortic valve calcification: an in-vivo and ex-vivo study. J Heart Valve Dis. 2013;22(3):317–25.

    PubMed  Google Scholar 

  52. Brandenburg VM, Kramann R, Koos R, Krüger T, Schurgers L, Mühlenbruch G, et al. Relationship between sclerostin and cardiovascular calcification in hemodialysis patients: a cross-sectional study. BMC Nephrol. 2013;14:219.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Claes KJ, Viaene L, Heye S, Meijers B, d’Haese P, Evenepoel P. Sclerostin: Another vascular calcification inhibitor? J Clin Endocrinol Metab. 2013;98(8):3221–8.

    Article  CAS  PubMed  Google Scholar 

  54. Kapelouzou A, Tsourelis L, Kaklamanis L, Degiannis D, Kogerakis N, Cokkinos DV. Serum and tissue biomarkers in aortic stenosis. Glob Cardiol Sci Pract. 2015;2015(4):49.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Delgado-Calle J, Sañudo C, Bolado A, Fernández AF, Arozamena J, Pascual-Carra MA, et al. 56 DNA methylation contributes to the regulation of sclerostin expression in human osteocytes. J Bone Miner Res. 2012;27(4):926–37.

    Article  CAS  PubMed  Google Scholar 

  56. Delgado-Calle J, Arozamena J, Pérez-López J, Bolado-Carrancio A, Sañudo C, Agudo G, et al. Role of BMPs in the regulation of sclerostin as revealed by an epigenetic modifier of human bone cells. Mol Cell Endocrinol. 2013;369(1–2):27–34.

    Article  CAS  PubMed  Google Scholar 

  57. Delgado-Calle J, Riancho JA, Klein-Nulend J. Nitric oxide is involved in the down-regulation of SOST expression induced by mechanical loading. Calcif Tissue Int. 2014;94(4):414–22.

    Article  CAS  PubMed  Google Scholar 

  58. Lee JY, Lee YM, Kim MJ, Choi JY, Park EK, Kim SY, et al. Methylation of the mouse DIx5 and Osx gene promoters regulates cell type-specific gene expression. Mol Cells. 2006;22(2):182–8.

    CAS  PubMed  Google Scholar 

  59. Arnsdorf EJ, Tummala P, Castillo AB, Zhang F, Jacobs CR. The epigenetic mechanism of mechanically induced osteogenic differentiation. J Biomech. 2010;43(15):2881–6.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Chen JC, Chua M, Bellon RB, Jacobs CR. Epigenetic changes during mechanically induced osteogenic lineage commitment. J Biomech Eng. 2015;137(2):020902.

    Article  PubMed  Google Scholar 

  61. Lehmann S, Walther T, Kempfert J, Rastan A, Garbade J, Dhein S, et al. Mechanical strain and the aortic valve: influence on fibroblasts, extracellular matrix, and potential stenosis. Ann Thorac Surg. 2009;88(5):1476–83.

    Article  PubMed  Google Scholar 

  62. Monzack EL, Masters KS. Can valvular interstitial cells become true osteoblasts? A side-by-side comparison. J Heart Valve Dis. 2011;20(4):449–63.

    PubMed  PubMed Central  Google Scholar 

  63. White MP, Theodoris CV, Liu L, Collins WJ, Blue KW, Lee JH, et al. NOTCH1 regulates matrix gla protein and calcification gene networks in human valve endothelium. J Mol Cell Cardiol. 2015;84:13–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Fernández Esmerats J, Heath J, Jo H. Shear-Sensitive Genes in Aortic Valve Endothelium. Antioxid Redox Signal. 2016;25(7):401–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Acharya A, Hans CP, Koenig SN, Nichols HA, Galindo CL, Garner HR, et al. Inhibitory role of Notch1 in calcific aortic valve disease. PLoS One. 2011;6(11):e27743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, et al. Mutations in NOTCH1 cause aortic valve disease. Nature. 2005;437(7056):270–4.

    Article  CAS  PubMed  Google Scholar 

  67. Theodoris CV, Li M, White MP, Liu L, He D, Pollard KS, et al. Human disease modeling reveals integrated transcriptional and epigenetic mechanisms of NOTCH1 haploinsufficiency. Cell. 2015;160(6):1072–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41(2):178–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Carter S, Miard S, Roy-Bellavance C, Boivin L, Li Z, Pibarot P, et al. Sirt1 inhibits resistin expression in aortic stenosis. PLoS One. 2012;7(4):e35110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Mohty D, Pibarot P, Després JP, Cartier A, Arsenault B, Picard F, et al. Age-related differences in the pathogenesis of calcific aortic stenosis: the potential role of resistin. Int J Cardiol. 2010;142(2):126–32.

    Article  PubMed  Google Scholar 

  71. Roos CM, Hagler M, Zhang B, Oehler EA, Arghami A, Miller JD. Transcriptional and phenotypic changes in aorta and aortic valve with aging and MnSOD deficiency in mice. Am J Physiol Heart Circ Physiol. 2013;305(10):H1428–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Miller JD. Role of SIRT6 in calcific aortic valve disease. https://mayoclinic.pure.elsevier.com/en/projects/role-of-sirt6-in-calcific-aortic-valve-disease.

  73. Zhang H, Greiten LE, Zhang B, Miller JD. Sirtuin 6 Reduces Osteogenic Signaling in Aortic Valve Interstitial Cells and Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol. 2012;32:A366.

    Google Scholar 

  74. Roos CM, Zhang B, Verzosa G, Oehler EA, Hagler MA, Zhang H, et al. Role of Sirtuin 6 in the Initiation and Progression of Calcific Aortic Valve Disease. Arterioscler Thromb Vasc Biol. 2014;34:A27.

    Google Scholar 

  75. Verzosa G, Hagler M, Roos CM, Zhang B, Miller JD. Left ventricular overload is required for manifestation of overt cardiac phenotypes in SIRT6-deficient mice. http://heartvalvesociety.org/meeting/abstracts/2016/P20.cgi.

  76. Casaclang-Verzosa G, Hagler MA, Roos CM, Zhang B, Miller JD. SIRT6 alters left ventricular responses to increased afterload due to progressive valvular stenosis in mice. http://www.epostersonline.com/hvs2015/node/323.

  77. Holliday CJ, Ankeny RF, Jo H, Nerem RM. Discovery of shear- and side-specific mRNAs and miRNAs in human aortic valvular endothelial cells. Am J Physiol Heart Circ Physiol. 2011;301(3):H856–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Carrion K, Dyo J, Patel V, Sasik R, Mohamed SA, Hardiman G, et al. The long non-coding HOTAIR is modulated by cyclic stretch and WNT/β-CATENIN in human aortic valve cells and is a novel repressor of calcification genes. PLoS One. 2014;9(5):e96577.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Lee HW, Suh JH, Kim AY, Lee YS, Park SY, Kim JB. Histone deacetylase 1-mediated histone modification regulates osteoblast differentiation. Mol Endocrinol. 2006;20(10):2432–43.

    Article  CAS  PubMed  Google Scholar 

  80. Kwon DH, Eom GH, Ko JH, Shin S, Joung H, Choe N, et al. MDM2 E3 ligase-mediated ubiquitination and degradation of HDAC1 in vascular calcification. Nat Commun. 2016;7:10492.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Schroeder TM, Kahler RA, Li X, Westendorf JJ. Histone deacetylase 3 interacts with runx2 to repress the osteocalcin promoter and regulate osteoblast differentiation. J Biol Chem. 2004;279(40):41998–2007.

    Article  CAS  PubMed  Google Scholar 

  82. Lewandowski SL, Janardhan HP, Trivedi CM. Histone Deacetylase 3 Coordinates Deacetylase-independent Epigenetic Silencing of Transforming Growth Factor-β1 (TGF-β1) to Orchestrate Second Heart Field Development. J Biol Chem. 2015;290(45):27067–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kang JS, Alliston T, Delston R, Derynck R. Repression of Runx2 function by TGF-beta through recruitment of class II histone deacetylases by Smad3. EMBO J. 2005;24(14):2543–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Jeon EJ, Lee KY, Choi NS, Lee MH, Kim HN, et al. Bone morphogenetic protein-2 stimulates Runx2 acetylation. J Biol Chem. 2006;281(24):16502–11.

    Article  CAS  PubMed  Google Scholar 

  85. Jensen ED, Schroeder TM, Bailey J, Gopalakrishnan R, Westendorf JJ. Histone deacetylase 7 associates with Runx2 and represses its activity during osteoblast maturation in a deacetylation-independent manner. J Bone Miner Res. 2008;23(3):361–72.

    Article  CAS  PubMed  Google Scholar 

  86. Chang S, Young BD, Li S, Qi X, Richardson JA, Olson EN. Histone deacetylase 7 maintains vascular integrity by repressing matrix metalloproteinase 10. Cell. 2006;126(2):321–34.

    Article  CAS  PubMed  Google Scholar 

  87. Patel V, Carrion K, Hollands A, Hinton A, Gallegos T, Dyo J, et al. The stretch responsive microRNA miR-148a-3p is a novel repressor of IKBKB, NF-κB signaling, and inflammatory gene expression in human aortic valve cells. FASEB J. 2015;29(5):1859–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID Bioinformatics Resources. Nature Protoc. 2009;4(1):44–57. Available at https://david.ncifcrf.gov/.

    Article  CAS  Google Scholar 

  89. Huang DW, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13. Available at https://david.ncifcrf.gov/.

    Article  CAS  Google Scholar 

  90. Medvedeva YA, Lennartsson A, Ehsani R, Kulakovskiy IV, Vorontsov IE, Panahandeh P, et al. EpiFactors: a comprehensive database of human epigenetic factors and complexes. Database (Oxford). 2015;2015:bav067. Available at http://epifactors.autosome.ru/.

    Article  CAS  Google Scholar 

  91. Rebhan M, Chalifa-Caspi V, Prilusky J, Lancet D. GeneCards: integrating information about genes, proteins and diseases. Trends Genet. 1997;13(4):163. Available at http://www.genecards.org/.

    Article  CAS  PubMed  Google Scholar 

  92. Kee HJ, Kook H. Roles and Targets of Class I and IIa Histone Deacetylases in Cardiac Hypertrophy. J Biomed Biotechnol. 2011;2011:928326.

    Article  PubMed  CAS  Google Scholar 

  93. Bovill E, Westaby S, Reji S, Sayeed R, Crisp A, Shaw T. Induction by left ventricular overload and left ventricular failure of the human Jumonji gene (JARID2) encoding a protein that regulates transcription and reexpression of a protective fetal program. J Thorac Cardiovasc Surg. 2008;136(3):709–16.

    Article  CAS  PubMed  Google Scholar 

  94. Sanulli S, Justin N, Teissandier A, Ancelin K, Portoso M, Caron M, et al. Jarid2 methylation via the PRC2 complex regulates H3K27me3 deposition during cell differentiation. Mol Cell. 2015;57(5):769–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Mysliwiec MR, Carlson CD, Tietjen J, Hung H, Ansari AZ, Lee Y. Jarid2 (Jumonji, AT rich interactive domain 2) regulates NOTCH1 expression via histone modification in the developing heart. J Biol Chem. 2012;287(2):1235–41.

    Article  CAS  PubMed  Google Scholar 

  96. Cui H, Schlesinger J, Schoenhals S, Tönjes M, Dunkel I, Meierhofer D, et al. Phosphorylation of the chromatin remodeling factor DPF3a induces cardiac hypertrophy through releasing HEY repressors from DNA. Nucleic Acids Res. 2016;44(6):2538–53.

    Article  PubMed  Google Scholar 

  97. Akerberg BN, Sarangam ML, Stankunas K. Endocardial Brg1 disruption illustrates the developmental origins of semilunar valve disease. Dev Biol. 2015;407(1):158–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wang J, Wang Y, Gu W, Ni B, Sun H, Yu T, et al. Comparative Transcriptome Analysis Reveals Substantial Tissue Specificity in Human Aortic Valve. Evol Bioinformatics Online. 2016 Jul 31;12:175–84.

    Google Scholar 

  99. Bossé Y, Miqdad A, Fournier D, Pépin A, Pibarot P, Mathieu P. Refining molecular pathways leading to calcific aortic valve stenosis by studying gene expression profile of normal and calcified stenotic human aortic valves. Circ Cardiovasc Genet. 2009;2(5):489–98.

    Article  PubMed  CAS  Google Scholar 

  100. Zhu Y, Chen H, Zhang X, Zhang L, Liu Y, Ren C. Downregulation of MALAT1 Promotes Aortic Valve Calcification by Inhibiting TWIST1 Expression. Lipid Cardiovasc Res. 2018;2(2):31–8.

    Google Scholar 

  101. Zhang XW, Zhang BY, Wang SW, Gong DJ, Han L, Xu ZY, et al. Twist-related protein 1 negatively regulated osteoblastic transdifferentiation of human aortic valve interstitial cells by directly inhibiting runt-related transcription factor 2. J Thorac Cardiovasc Surg. 2014;148(4):1700–1708.e1.

    Article  CAS  PubMed  Google Scholar 

  102. Viereck J, Kumarswamy R, Foinquinos A, Xiao K, Avramopoulos P, Kunz M, et al. Long noncoding RNA Chast promotes cardiac remodeling. Sci Transl Med. 2016;8(326):326ra22.

    Article  PubMed  CAS  Google Scholar 

  103. Ounzain S, Micheletti R, Arnan C, Plaisance I, Cecchi D, Schroen B, et al. CARMEN, a human super enhancer-associated long noncoding RNA controlling cardiac specification, differentiation and homeostasis. J Mol Cell Cardiol. 2015;89(Pt A):98–112.

    Article  CAS  PubMed  Google Scholar 

  104. Micheletti R, Plaisance I, Abraham BJ, Sarre A, Ting CC, Alexanian M, et al. The long noncoding RNA Wisper controls cardiac fibrosis and remodeling. Sci Transl Med. 2017;9(395).

  105. Ounzain S, Micheletti R, Beckmann T, Schroen B, Alexanian M, Pezzuto I, 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(6):353–68a.

    Article  PubMed  Google Scholar 

  106. Peters T, Hermans-Beijnsberger S, Beqqali A, Bitsch N, Nakagawa S, Prasanth KV, et al. Long Non-Coding RNA Malat-1 Is Dispensable during Pressure Overload-Induced Cardiac Remodeling and Failure in Mice. PLoS One. 2016;11(2):e0150236.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Quek XC, Thomson DW, Maag JL, Bartonicek N, Signal B, Clark MB, Gloss BS, Dinger ME. lncRNAdb v2.0: expanding the reference database for functional long noncoding RNAs. Nucleic Acid Res. 2014;43:D168–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Amaral PP, Clark MB, Gascoigne DK, Dinger ME, Mattick JS. lncRNAdb: a reference database for long noncoding RNAs. Nucleic Acids Res. 2011;39:D146–51.

    Article  CAS  PubMed  Google Scholar 

  109. Gray KA, Yates B, Seal RL, Wright MW, Bruford EA. genenames.org: the HGNC resources in 2015. Nucleic Acids Res. 2015;43(Database issue):D1079–85.

    Article  CAS  PubMed  Google Scholar 

  110. Clark K, Karsch-Mizrachi I, Lipman DJ, Ostell J, Sayers EW. GenBank. Nucleic Acids Res. 2016;44(D1):D67–72. Available at https://www.ncbi.nlm.nih.gov/gene/.

    Article  CAS  PubMed  Google Scholar 

  111. Romaine SP, Tomaszewski M, Condorelli G, Samani NJ. MicroRNAs in cardiovascular disease: an introduction for clinicians. Heart. 2015;101(12):921–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Vavuranakis M, Kariori M, Vrachatis D, Aznaouridis K, Siasos G, Kokkou E, et al. MicroRNAs in aortic disease. Curr Top Med Chem. 2013;13(13):1559–72.

    Article  CAS  PubMed  Google Scholar 

  113. Oury C, Servais L, Bouznad N, Hego A, Nchimi A, Lancellotti P. MicroRNAs in Valvular Heart Diseases: Potential Role as Markers and Actors of Valvular and Cardiac Remodeling. Int J Mol Sci. 2016;17(7).

  114. Ohukainen P, Syväranta S, Näpänkangas J, Rajamäki K, Taskinen P, Peltonen T, et al. MicroRNA-125b and chemokine CCL4 expression are associated with calcific aortic valve disease. Ann Med. 2015;47(5):423–9.

    Article  PubMed  CAS  Google Scholar 

  115. Nigam V, Sievers HH, Jensen BC, Sier HA, Simpson PC, Srivastava D, et al. Altered microRNAs in bicuspid aortic valve: a comparison between stenotic and insufficient valves. J Heart Valve Dis. 2010;19(4):459–65.

    PubMed  PubMed Central  Google Scholar 

  116. Riem Vis PW, Joziassen IC, Kluin J, Vink A, van Herwerden LA, Doevendans PAFM, et al. Differential expression of microRNAs in human calcific aortic valve disease. In: Riem Vis PW. Towards new therapies for calcific aortic valve disease, PhD thesis, Utrecht University 2011. ISBN: 978–90–393-55631-9. Available at http://dspace.library.uu.nl/handle/1874/211988.

  117. Yanagawa B, Lovren F, Pan Y, Garg V, Quan A, Tang G, et al. miRNA-141 is a novel regulator of BMP-2-mediated calcification in aortic stenosis. J Thorac Cardiovasc Surg. 2012;144(1):256–62.

    Article  CAS  PubMed  Google Scholar 

  118. Zhang M, Liu X, Zhang X, Song Z, Han L, He Y, et al. MicroRNA-30b is a multifunctional regulator of aortic valve interstitial cells. J Thorac Cardiovasc Surg. 2014;147(3):1073–1080.e2.

    Article  CAS  PubMed  Google Scholar 

  119. Dyo j. The Role of Stretch Responsive microRNAs in Aortic Valve Disease: miR-148a Repression of NF-kappaB Mediated Inflammation and miR-19b Modulation of Osteogenic Pathways. Available at http://escholarship.org/uc/item/9pp8n26j.

  120. Shi J, Liu H, Wang H, Kong X. MicroRNA Expression Signature in Degenerative Aortic Stenosis. Biomed Res Int. 2016;2016:4682172.

    PubMed  PubMed Central  Google Scholar 

  121. Coffey S, Williams MJ, Phillips LV, Galvin IF, Bunton RW, Jones GT. Integrated microRNA and messenger RNA analysis in aortic stenosis. Sci Rep. 2016;6:36904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Holliday JC. Discovery of shear- and side-dependent microRNAs and messenger RNAs in aortic valvular endothelium. PhD thesis, Georgia Institute of Technology, 2012. Available at http://hdl.handle.net/1853/47517.

  123. Rathan S, Ankeny CJ, Arjunon S, Ferdous Z, Kumar S, Fernandez Esmerats J, et al. Identification of side- and shear-dependent microRNAs regulating porcine aortic valve pathogenesis. Sci Rep. 2016;6:25397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Martínez-Micaelo N, Beltrán-Debón R, Baiges I, Faiges M, Alegret JM. Specific circulating microRNA signature of bicuspid aortic valve disease. J Transl Med. 2017;15(1):76.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Ding W, Li J, Singh J, Alif R, Vazquez-Padron RI, Gomes SA, et al. miR-30e targets IGF2-regulated osteogenesis in bone marrow-derived mesenchymal stem cells, aortic smooth muscle cells, and ApoE−/− mice. Cardiovasc Res. 2015;106(1):131–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Anselmo W, Branchetti E, Ayoub S, Grau JB, Sacks M, Levy RJ, et al. miR-143 and SOD mimetics regulate matricellular proteins and ameliorate valve functions in aortic valve sclerosis. 7th Biennial Heart Valve Biology & Tissue Engineering Meeting. 2016, AC22 Available at https://www.emedevents.com/c/medical-conferences-2016/7th-biennial-heart-valve-biology-and-tissue-engineering-meeting.

  127. Branchetti E, Lee S, Grau JB, Hazen SL, Levy RJ, Ferrari G. Locked Nucleic Acid miR-143 Ameliorates Aortic Valve Functions in a ROS-Mediated Murine Model of Aortic Valve Sclerosis. Arterioscler Thromb Vasc Biol. 2015;35(Suppl 1):A646.

    Google Scholar 

  128. Salim T, Arjunon S, Rathan S, Jo H, Yoganathan A. Role of Elevated Cyclic Stretch in Modulating MiR-181a and MiR-214 Expres-sions in Aortic Valve. 7th Biennial Heart Valve Biology & Tissue Engineering Meeting. 2016, AP10 Available at https://www.emedevents.com/c/medical-conferences-2016/7th-biennial-heart-valve-biology-and-tissue-engineering-meeting.

  129. Heath J, Esmerats JF, Simmons R, Kumar S, Jo H. Shear-Sensitive miRNA-181b Impairs Anti-Inflammatory Signaling in the Aortic Valve Endothelium. http://heartvalvesociety.org/meeting/abstracts/2016/A53.cgi.

  130. Wang Y, Chen S, Deng C, Li F, Wang Y, Hu X, et al. MicroRNA-204 Targets Runx2 to Attenuate BMP-2-induced Osteoblast Differentiation of Human Aortic Valve Interstitial Cells. J Cardiovasc Pharmacol. 2015;66(1):63–71.

    Article  CAS  PubMed  Google Scholar 

  131. Song R, Fullerton DA, Ao L, Zhao K, Meng X. Microrna-204 and -486 Regulate the Expression of Osteogenic Transcription Factors in Human Aortic Valve Cells: Evidence for Epigenetic Modulation of Valvular Cell Osteogenic Reprogramming. Circulation. 2013;128(Suppl 3):A18239.

    Google Scholar 

  132. Song R, Fullerton DA, Ao L, Zhao K, Reece TB, Cleveland JCJ, et al. The Pro-osteogenic Phenotype of Interstitial Cells of Calcified Human Aortic Valves is Due to Altered Expression of Mir-486 and -204. Circulation. 2015;132(Suppl 3):A18717.

    Google Scholar 

  133. Song R, Fullerton DA, Ao L, Yao O, Zhao K, Meng X. Microrna-486 Regulates Fibroblast-to-myofibroblast Transition in Human Aortic Valve Interstitial Cells. Arterioscler Thromb Vasc Biol. 2015;35:A165.

    Google Scholar 

  134. Li XF, Wang Y, Zheng DD, Xu HX, Wang T, Pan M, et al. M1 macrophages promote aortic valve calcification mediated by microRNA-214/TWIST1 pathway in valvular interstitial cells. Am J Transl Res. 2016;8(12):5773–83.

    PubMed  PubMed Central  Google Scholar 

  135. Esmerats JF, Heath J, Kumar S, Jo H. Role of miRNA-483-3p in Valvular Endothelial Dysfunction. Cardiology. 2016;134:136–310.

    Article  Google Scholar 

  136. Holliday-Ankeny CJ, Ankeny RF, Ferdous Z, Nerem RM, Jo H. The function of shear-responsive and side-dependent microRNA-486-5p in aortic valve endothelium. Cardiovasc Pathol. 2013;22(3):e50.

    Article  Google Scholar 

  137. Esmerats JF, Heath J, Sandeep Kumar S, Jo H. Shear-Stress Regulates Aortic Valve Inflammation Via MiRNA-1237-3p. 7th Biennial Heart Valve Biology & Tissue Engineering Meeting. 2016, AC15 Available at https://www.emedevents.com/c/medical-conferences-2016/7th-biennial-heart-valve-biology-and-tissue-engineering-meeting.

  138. Esmerats JF, Heath J, Ankeny C, Kumar S, Jo H. miRNA-1237-3p modulates shear-stress dependent aortic valve endothelial inflammation. http://epostersonline.s3.amazonaws.com/hvs2016/hvs2016.0c000ac.NORMAL.pdf.

  139. Goettsch C, Hutcheson JD, Aikawa E. MicroRNA in cardiovascular calcification: focus on targets and extracellular vesicle delivery mechanisms. Circ Res. 2013;112(7):1073–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Balderman JA, Lee HY, Mahoney CE, Handy DE, White K, Annis S, et al. Bone morphogenetic protein-2 decreases microRNA-30b and microRNA-30c to promote vascular smooth muscle cell calcification. J Am Heart Assoc. 2012;1(6):e003905.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Leopold JA. MicroRNAs Regulate Vascular Medial Calcification. Cells. 2014;3(4):963-80. 

  142. Li H, Xie H, Liu W, Hu R, Huang B, Tan YF, et al. A novel microRNA targeting HDAC5 regulates osteoblast differentiation in mice and contributes to primary osteoporosis in humans. J Clin Invest. 2009;119(12):3666–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Hu R, Liu W, Li H, Yang L, Chen C, Xia ZY, et al. A Runx2/miR-3960/miR-2861 regulatory feedback loop during mouse osteoblast differentiation. J Biol Chem. 2011;286(14):12328–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Xia ZY, Hu Y, Xie PL, Tang SY, Luo XH, Liao EY, et al. Runx2/miR-3960/miR-2861 Positive Feedback Loop Is Responsible for Osteogenic Transdifferentiation of Vascular Smooth Muscle Cells. Biomed Res Int. 2015;2015:624037.

    PubMed  PubMed Central  Google Scholar 

  145. Goettsch C, Rauner M, Pacyna N, Hempel U, Bornstein SR, Hofbauer LC. MiR-125b regulates calcification of vascular smooth muscle cells. Am J Pathol. 2011;179(4):1594–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Wen P, Cao H, Fang L, Ye H, Zhou Y, Jiang L, et al. MiR-125b/Ets1 axis regulates transdifferentiation and calcification of vascular smooth muscle cells in a high-phosphate environment. Exp Cell Res. 2014;322(2):302–12.

    Article  CAS  PubMed  Google Scholar 

  147. Takahashi K, Satoh M, Takahashi Y, Osaki T, Nasu T, Tamada M, et al. Dysregulation of ossification-related miRNAs in circulating osteogenic progenitor cells obtained from patients with aortic stenosis. Clin Sci (Lond). 2016;130(13):1115–24.

    Article  CAS  Google Scholar 

  148. Huang J, Zhao L, Xing L, Chen D. MicroRNA-204 regulates Runx2 protein expression and mesenchymal progenitor cell differentiation. Stem Cells. 2010;28:357–64.

    PubMed  PubMed Central  Google Scholar 

  149. Vimalraj S, Selvamurugan N. MicroRNAs expression and their regulatory networks during mesenchymal stem cells differentiation toward osteoblasts. Int J Biol Macromol. 2014;66:194–202.

    Article  CAS  PubMed  Google Scholar 

  150. Baglìo SR, Devescovi V, Granchi D, Baldini N. MicroRNA expression profiling of human bone marrow mesenchymal stem cells during osteogenic differentiation reveals Osterix regulation by miR-31. Gene. 2013;527:321–31.

    Article  PubMed  CAS  Google Scholar 

  151. Villar AV, García R, Merino D, Llano M, Cobo M, Montalvo C, et al. Myocardial and circulating levels of microRNA-21 reflect left ventricular fibrosis in aortic stenosis patients. Int J Cardiol. 2013;167(6):2875–81.

    Article  PubMed  Google Scholar 

  152. Fabiani I, Scatena C, Mazzanti CM, Conte L, Pugliese NR, Franceschi S, et al. Micro-RNA-21 (biomarker) and global longitudinal strain (functional marker) in detection of myocardial fibrotic burden in severe aortic valve stenosis: a pilot study. J Transl Med. 2016;14(1):248.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Coffey S, Williams MJ, Phillips LV, Jones GT. Circulating microRNA Profiling Needs Further Refinement Before Clinical Use in Patients With Aortic Stenosis. J Am Heart Assoc. 2015;4(8):e002150.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Chen Z, Li C, Xu Y, Li Y, Yang H, Rao L. Circulating level of miR-378 predicts left ventricular hypertrophy in patients with aortic stenosis. PLoS One. 2014;9(8):e105702.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. García R, Villar AV, Cobo M, Llano M, Martín-Durán R, Hurlé MA, et al. Circulating levels of miR-133a predict the regression potential of left ventricular hypertrophy after valve replacement surgery in patients with aortic stenosis. J Am Heart Assoc. 2013;2(4):e000211.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Røsjø H, Dahl MB, Bye A, Andreassen J, Jørgensen M, Wisløff U, et al. Prognostic value of circulating microRNA-210 levels in patients with moderate to severe aortic stenosis. PLoS One. 2014 Mar 13;9(3):e91812.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Derda AA, Thum S, Lorenzen JM, Bavendiek U, Heineke J, Keyser B, et al. Blood-based microRNA signatures differentiate various forms of cardiac hypertrophy. Int J Cardiol. 2015;196:115–22.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Roncarati R, Viviani Anselmi C, Losi MA, Papa L, Cavarretta E, Da Costa MP, et al. Circulating miR-29a, among other up-regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol. 2014;63(9):920–7.

    Article  CAS  PubMed  Google Scholar 

  159. Varrone F, Gargano B, Carullo P, Di Silvestre D, De Palma A, Grasso L, et al. The circulating level of FABP3 is an indirect biomarker of microRNA-1. J Am Coll Cardiol. 2013;61(1):88–95.

    Article  CAS  PubMed  Google Scholar 

  160. Miyamoto S, Usami S, Kuwabara Y, Horie T, Baba O, Hakuno D, et al. Expression Patterns of miRNA-423-5p in the Serum and Pericardial Fluid in Patients Undergoing Cardiac Surgery. PLoS One. 2015;10(11):e0142904.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Kuosmanen SM, Hartikainen J, Hippeläinen M, Kokki H, Levonen AL, Tavi P. MicroRNA profiling of pericardial fluid samples from patients with heart failure. PLoS One. 2015;10(3):e0119646.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Beltrami C, Besnier M, Shantikumar S, Shearn AI, Rajakaruna C, Laftah A, et al. Human Pericardial Fluid Contains Exosomes Enriched with Cardiovascular-Expressed MicroRNAs and Promotes Therapeutic Angiogenesis. Mol Ther. 2017;25(3):679–93.

    Article  CAS  PubMed  Google Scholar 

  163. Iacopo F, Lorenzo C, Calogero E, Matteo P, Riccardo PN, Veronica S, et al. Review in Translational Cardiology: MicroRNAs and Myocardial Fibrosis in Aortic Valve Stenosis, a Deep Insight on Left Ventricular Remodeling. J Cardiovasc Echogr. 2016; 26(4):109-14.

  164. Lorenzen JM, Schauerte C, Hübner A, Kölling M, Martino F, Scherf K, et al. Osteopontin is indispensible for AP1-mediated angiotensin II-related miR-21 transcription during cardiac fibrosis. Eur Heart J. 2015;36(32):2184–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. García R, Nistal JF, Merino D, Price NL, Fernández-Hernando C, Beaumont J, et al. p-SMAD2/3 and DICER promote pre-miR-21 processing during pressure overload-associated myocardial remodeling. Biochim Biophys Acta. 2015;1852(7):1520–30.

    Article  PubMed  CAS  Google Scholar 

  166. Eskildsen TV, Schneider M, Zhai P, et al. Comprehensive microarray analysis identify dysregulated microRNAs in pressure overload affected hearts. Translational Biomedicine 2015: ISSN. 2172–0479. Available at http://www.transbiomedicine.com/translational-biomedicine/comprehensive-microarray-analysis-identify-dysregulated-micrornas-in-pressure-overload-affected-hearts.pdf.

  167. Duisters RF, Tijsen AJ, Schroen B, Leenders JJ, Lentink V, van der Made I, et al. miR-133 and miR-30 regulate connective tissue growth factor: implications for a role of microRNAs in myocardial matrix remodeling. Circ Res. 2009;104(2):170–8. 6p following 178

    Article  CAS  PubMed  Google Scholar 

  168. Jiang F, Zhou X, Huang J. Long Non-Coding RNA-ROR Mediates the Reprogramming in Cardiac Hypertrophy. PLoS One. 2016;11(4):e0152767.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Renaud L, Harris LG, Mani SK, Kasiganesan H, Chou JC, Baicu CF, et al. HDACs Regulate miR-133a Expression in Pressure Overload-Induced Cardiac Fibrosis. Circ Heart Fail. 2015;8(6):1094–104.

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Villar AV, Merino D, Wenner M, Llano M, Cobo M, Montalvo C, et al. Myocardial gene expression of microRNA-133a and myosin heavy and light chains, in conjunction with clinical parameters, predict regression of left ventricular hypertrophy after valve replacement in patients with aortic stenosis. Heart. 2011;97(14):1132–7.

    Article  PubMed  Google Scholar 

  171. Beaumont J, López B, Hermida N, Schroen B, San José G, Heymans S, et al. microRNA-122 down-regulation may play a role in severe myocardial fibrosis in human aortic stenosis through TGF-β1 up-regulation. Clin Sci (Lond). 2014;126(7):497–506.

    Article  CAS  Google Scholar 

  172. Tijsen AJ, van der Made I, van den Hoogenhof MM, Wijnen WJ, van Deel ED, de Groot NE, et al. The microRNA-15 family inhibits the TGFβ-pathway in the heart. Cardiovasc Res. 2014;104(1):61–71.

    Article  CAS  PubMed  Google Scholar 

  173. Ikeda S, Kong SW, Lu J, Bisping E, Zhang H, Allen PD, et al. Altered microRNA expression in human heart disease. Physiol Genomics. 2007;31(3):367–73.

    Article  CAS  PubMed  Google Scholar 

  174. Ikeda S, Pu WT. Expression and function of microRNAs in heart disease. Curr Drug Targets. 2010;11(8):913–25.

    Article  CAS  PubMed  Google Scholar 

  175. Gallego I, Beaumont J, López B, Ravassa S, José Gómez-Doblas JJ, Moreno MU, et al. Potential role of microRNA-10b down-regulation in cardiomyocyte apoptosis in aortic stenosis patients. Clin Sci (Lond). 2016;130(23):2139–49.

    Article  Google Scholar 

  176. Beaumont J, López B, Ravassa S, Hermida N, José GS, Gallego I, et al. MicroRNA-19b is a potential biomarker of increased myocardial collagen cross-linking in patients with aortic stenosis and heart failure. Sci Rep. 2017;7:40696.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Butcher JT. The effects of steady laminar shear stress on aortic valve cell biology. Georgia Institute of Technology, 2004. PhD Thesis, Georgia Institute of Technology 2004. Retrieved from http://hdl.handle.net/1853/4824.

  178. Porras AM, Shanmuganayagam D, Meudt JJ, Krueger CG, Hacker TA, Rahko PS, et al. Development of Aortic Valve Disease in Familial Hypercholesterolemic Swine: Implications for Elucidating Disease Etiology. J Am Heart Assoc. 2015;4(10):e002254.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  179. Esmerats JF, Heath J, Kumar S, Jo H. Role of miRNA-483-3p in Valvular Endothelial Dysfunction. Cardiology. 2016;134:178. Retrieved from https://www.karger.com/Article/Abstract/444511.

    Google Scholar 

  180. McCoy CM, Nicholas DQ, Masters KS. Sex-related differences in gene expression by porcine aortic valvular interstitial cells. PLoS One. 2012;7(7):e39980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Rusconi F, Ceriotti P, Miragoli M, Carullo P, Salvarani N, Rocchetti M, et al. Peptidomimetic Targeting of Cavβ2 Overcomes Dysregulation of the L-Type Calcium Channel Density and Recovers Cardiac Function. Circulation. 2016;134(7):534–46.

    Article  CAS  PubMed  Google Scholar 

  182. Guauque-Olarte S, Droit A, Tremblay-Marchand J, Gaudreault N, Kalavrouziotis D, Dagenais F, et al. RNA expression profile of calcified bicuspid, tricuspid, and normal human aortic valves by RNA sequencing. Physiol Genomics. 2016;48(10):749–61.

    Article  PubMed  Google Scholar 

  183. Padang R, Bagnall RD, Tsoutsman T, Bannon PG, Semsarian C. Comparative transcriptome profiling in human bicuspid aortic valve disease using RNA sequencing. Physiol Genomics. 2015;47(3):75–87.

    Article  CAS  PubMed  Google Scholar 

  184. Latif N, Sarathchandra P, Chester AH, Yacoub MH. Expression of smooth muscle cell markers and co-activators in calcified aortic valves. Eur Heart J. 2015;36(21):1335–45.

    Article  PubMed  Google Scholar 

  185. Lunde IG, Aronsen JM, Kvaløy H, Qvigstad E, Sjaastad I, Tønnessen T, et al. Cardiac O-GlcNAc signaling is increased in hypertrophy and heart failure. Physiol Genomics. 2012;44(2):162–72.

    Article  CAS  PubMed  Google Scholar 

  186. Heath J, Esmerats JF, Simmons R, Kumar S, Jo H. Shear-Sensitive miRNA-181b Impairs Anti-Inflammatory Signaling in the Aortic Valve Endothelium. Cardiology. 2016;134:167. Retrieved from https://www.karger.com/Article/Abstract/444511.

    Google Scholar 

  187. Nagy E, Caidahl K, Franco-Cereceda A, Bäck M. Increased transcript level of poly(ADP-ribose) polymerase (PARP-1) in human tricuspid compared with bicuspid aortic valves correlates with the stenosis severity. Biochem Biophys Res Commun. 2012;420(3):671–5.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work is supported by the Scientific Center of Excellence for Regenerative and Reproductive Medicine (CERRM) established in 2013 at the School of Medicine, University of Zagreb, by the decision of Ministry of Science, Education and Sport, Republic of Croatia.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Author information

Authors and Affiliations

Authors

Contributions

GI, ZD, MZ, and FP were major contributors in writing the manuscript. FP designed the study and extracted the data, conducted the bioinformatic analysis, and drew the tables with the help of GI, NI, GM, BD, LZ, and SI. All authors contributed to the content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Frane Paić.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gošev, I., Zeljko, M., Đurić, Ž. et al. Epigenome alterations in aortic valve stenosis and its related left ventricular hypertrophy. Clin Epigenet 9, 106 (2017). https://doi.org/10.1186/s13148-017-0406-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13148-017-0406-7

Keywords