Next Article in Journal
Nematicidal Volatiles from Bacillus atrophaeus GBSC56 Promote Growth and Stimulate Induced Systemic Resistance in Tomato against Meloidogyne incognita
Next Article in Special Issue
The Evolving Role of Fetuin-A in Nonalcoholic Fatty Liver Disease: An Overview from Liver to the Heart
Previous Article in Journal
ThHSFA1 Confers Salt Stress Tolerance through Modulation of Reactive Oxygen Species Scavenging by Directly Regulating ThWRKY4
Previous Article in Special Issue
Male and Female Animals Respond Differently to High-Fat Diet and Regular Exercise Training in a Mouse Model of Hyperlipidemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetics, microRNA and Metabolic Syndrome: A Comprehensive Review

Liggins Institute, The University of Auckland, Auckland 1023, New Zealand
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(9), 5047; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22095047
Submission received: 31 March 2021 / Revised: 6 May 2021 / Accepted: 7 May 2021 / Published: 10 May 2021
(This article belongs to the Special Issue Metabolic Syndrome: From Molecular Mechanisms to Novel Therapies)

Abstract

:
Epigenetics refers to the DNA chemistry changes that result in the modification of gene transcription and translation independently of the underlying DNA coding sequence. Epigenetic modifications are reported to involve various molecular mechanisms, including classical epigenetic changes affecting DNA methylation and histone modifications and small RNA-mediated processes, particularly that of microRNAs. Epigenetic changes are reversible and are closely interconnected. They are recognised to play a critical role as mediators of gene regulation, and any alteration in these mechanisms has been identified to mediate various pathophysiological conditions. Moreover, genetic predisposition and environmental factors, including dietary alterations, lifestyle or metabolic status, are identified to interact with the human epigenome, highlighting the importance of epigenetic factors as underlying processes in the aetiology of various diseases such as MetS. This review will reflect on how both the classical and microRNA-regulated epigenetic changes are associated with the pathophysiology of metabolic syndrome. We will then focus on the various aspects of epigenetic-based strategies used to modify MetS outcomes, including epigenetic diet, epigenetic drugs, epigenome editing tools and miRNA-based therapies.

1. Introduction

The burden of non-communicable diseases (NCDs) or chronic diseases is increasing globally at an alarming rate and is currently the leading cause of mortality worldwide [1], with NCDs presently accounting for more than 70% of all deaths, most of which are preventable [2]. These diseases include cardiovascular diseases (CVD), chronic respiratory disease, cancer and type 2 diabetes (T2DM) [3]. The main risk factors underpinning these diseases involve aberrant changes in an individual’s metabolic and physiological profile that eventually result in the development of metabolic syndrome (MetS). MetS is defined as a multifactorial condition consisting of several inter-related anthropometric and biochemical features, including increased visceral adiposity, high fasting blood glucose, high blood pressure, low high-density lipoprotein-cholesterol (HDL-C) and high triglycerides, with further immune and vascular alterations [4]. The risk profile of MetS underlies the progressive development of diseases, including CVD and T2DM [5]. Complex interactions between environmental factors, including dietary and lifestyle, with the genetic and epigenetic makeup of an individual, are reported to be responsible for the pathophysiology of MetS [3,6,7]. However, the exact aetiology underpinning the development of MetS remains to be fully established [5].
Recent advances in the rapidly evolving field of epigenetics have revealed a complex network of shared interconnections between epigenetic machinery and human diseases [8,9,10,11]. Epigenetic modifications are reversible, differ across cell types and potentially lead to increased disease susceptibility by producing long-term changes in gene transcription [12,13]. Due to their key roles as mediators of gene regulation, epigenetic modifications may play a crucial role in developing pathological conditions [13]. Understanding the epigenetic machinery underlying MetS and how such epigenetic processes can provide utility in the development of diagnostic tools (i.e., biomarkers) and therapies is fundamental in improving the quality of life in individuals affected by MetS. Epigenetic changes can either be inherited or accumulated throughout a lifetime but, unlike genetic changes, are reversible and can therefore provide potential targets for MetS prevention and intervention [11]. Epigenetic patterns can also be altered under different developmental processes, dietary alterations, lifestyles or metabolic status, thus highlighting the importance of epigenetic factors as underlying processes in the aetiology of metabolic diseases [13,14,15].
This review will reflect on the classical epigenetic changes frequently associated with the pathophysiology of MetS, including DNA methylation and histone modifications. In addition to these mechanisms, we will then describe how miRNAs contribute to the epigenetic machinery of MetS by considering, firstly, how epigenetic mechanisms regulate miRNA expression and function and, secondly, how miRNAs reciprocally regulate the classical epigenetic mechanisms of DNA methylation and histone modifications. Finally, we will focus on epigenetic-based strategies used to modify MetS outcomes, including epigenetic diet, epigenetic drugs, epigenome editing tools and miRNA-based therapies.

2. The Classical Epigenetic Mechanisms in MetS

Epigenetics refers to the DNA chemistry changes that result in gene transcription modification and translation independently of the underlying DNA coding sequence. Epigenetic changes are increasingly thought to be of importance both in normal physiological processes and disease conditions [13]. The “classic” epigenetic mechanisms encompass DNA methylation and histone modifications, both of which can reduce or prevent RNA transcription and may be heritable [13,16,17]. Growing evidence has reported the function of classic epigenetic mechanisms in the regulation of gene expression [11,18,19]. However, the role of epigenetic marks in the development and progression of MetS is not clearly understood. Thus, exploration of epigenetic changes in the setting of the MetS may provide a deeper understanding of the molecular mechanisms and pathways involved. In this section, we focus on these classic epigenetic processes associated with the pathophysiology of MetS.

2.1. DNA Methylation

DNA methylation refers to a covalent and reversible transfer of a methyl group from S-adenosyl methionine (SAM) to the pyrimidine C5 position of the cytosine residues on genomic 5′-C-phosphate-G-3′ (CpG) dinucleotides, resulting in the formation of 5-methylcytosine (5-mC) [20]. A group of specific enzymes known as DNA methyltransferases (DNMTs) function as catalysers to inscribe the methylation marks on the genomic DNA. DNMTs are categorized into either maintenance DNMTs (DNMT1, DNMT2) that are involved in restoring the existing pattern of DNA methylation during cell replication, or de novo DNMTs (DNMT3a, DNMT3b and DNMT3L) responsible for catalysing new methylation of genomic DNA during embryonic development [21]. These DNMTs work in coordination to maintain a methylation pattern that supports a balanced transcriptional control over the genome. Methylation patterns within the promoter sequences of the DNA results in the downregulation of gene expression, while it has been reported that DNA methylation within the gene upregulates its expression. In contrast, the ten-eleven translocation (TET) family of enzymes, through their methylcytosine dioxygenase activity, assist in the removal of existing methylation marks by causing the conversion of 5-mC to 5-hydroxymethylcytosine (5-hmC) [20].
The association between aberrant DNA methylation and disease state has been studied across various disorders, including CVDs, autoimmune disorders and various cancers [22,23]. In recent years, epigenome-wide association studies (EWASs) have identified global and locus-specific epigenetic changes potentially involved in the pathophysiological mechanisms responsible for the development of MetS (Table 1) [24,25]. For example, an EWAS study performed in individuals with MetS demonstrated decreased methylation of CPT1A, a gene with a key role in regulating mitochondrial fatty acid oxidation (FAO), to be correlated with the increased metabolic risk and overall MetS phenotype [25]. In a recent EWAS study in African American adults, MetS was consistently associated with increased methylation in the ABCG1, a gene that encodes a protein in the ATP-binding cassette transporter family and is involved in intra- and extra-cellular signalling and lipid transport [24,26]. Additionally, it has been shown that marked differences exist in the prevalence of MetS and that of the DNA methylation patterns among middle-aged African Americans and white individuals diagnosed with MetS [25].
Further, with the acknowledged role of adipose tissue in the maintenance of metabolic homeostasis and altered adipogenesis associated with the MetS, studies have been undertaken to understand better the role of epigenetic processes in regulating the adipocyte function in the setting of such metabolic disorders [23,27,28]. A recent study by Daniel et al. [23] identified global DNA methylation of LINE-1 to be associated with metabolic deterioration and glucose metabolism in visceral adipose tissue of individuals with and without MetS. Strong associations between methylation of specific genes involved in substrate metabolism (LPL and PPARA) and inflammation (TNF) with that of MetS status was also observed in these individuals [23]. Besides, it has been identified that alterations in dietary patterns could modulate the methylation patterns of key metabolic genes and hence their expression [29,30]. For example, it has been demonstrated that dietary total antioxidant capacity is positively correlated with the global DNA methylation of LINE-1 levels in individuals with MetS after an 8-week hypocaloric diet [31]. Taken together, it is evident that DNA methylation has an important role to play in MetS development and progression; however, most of these studies only report associative effects and not cause–effect relationships. Of note, a potential limitation of observations to date is that many studies utilise analysis on whole tissues with mixed cell populations, thus, changes in methylation patterns observed may be due to tissue or cell population heterogeneity [32]. Furthermore, only small changes in methylation (1–10%) have been associated with complex diseases, and these observed differences in methylation profiles could arise as a result of variations in cell populations within the same tissue [33]. However, the functional consequences of such small absolute changes in methylation patterns in the setting of MetS cannot be discounted.
Table 1. Human studies showing associations between aberrant DNA methylation and features of metabolic health.
Table 1. Human studies showing associations between aberrant DNA methylation and features of metabolic health.
DiseaseDifferentially Methylated GenesSample TypeRef
MetSCPT1ACD4+ T cells[34]
MetSABCG1Blood (buffy coat)[26]
MetSLINE-1Visceral adipose tissue[23]
T2DMPPARGC1APancreatic islets[35]
T2DMTXNIPLeucocytes[36]
HypercholesterolemiaTNNTIBlood[37]

2.2. Histone Modifications

Classical epigenetic mechanisms also involve post-translational modifications of histone proteins by specialized histone-modifying enzymes, resulting in changes to chromatin architecture and gene expression regulation [38]. There are several post-translational modifications of histone proteins, with acetylation, phosphorylation, methylation and ubiquitination of histones being the most commonly observed [39]. Methylation of histones could result in either an either an increase or decrease in gene expression. For example, methylation of histone H3 at lysines 9, 27, and 36 would reduce gene expression, while methylation of histone H3 at lysines 4 and 79 and methylation of histone H4 at lysine 20 would generally result in an increase in proximal gene expression [40]. The dynamics underlying these changes are mediated through histone methyl transferases that place methyl groups and histone demethylases that remove methylation [39]. Similarly, acetylation of lysines on histones H3 and H4 is associated with increased transcription of nearby genes. Acetyl groups are placed by histone acetyl transferases and removed by histone deacetylases (HDAC). Histone phosphorylation is less well studied but is suggested to increase gene expression in combination with H3K56ac [9]. Histone variants such as H2A.Z and H3.3 have been demonstrated to play specific roles in regulating chromatin structure and function by influencing transcription of nearby genes [15].
Alterations in histone modifications are identified as essential components of epigenetic networks, controlling energy homeostasis and altering adipocyte thermogenesis, thereby contributing to the pathogenesis of MetS (Table 2) [28,41]. For example, a study involving mice deficient in histone demethylase, HDM2a, an enzyme responsible for H3K9 demethylation, reported the development of adult-onset obesity, hypertriglyceridemia and hypercholesterolemia, as well as insulin resistance (IR), in the deficient mice as compared to the wild type [41]. Additionally, histone modifications are also reported to interact with pathways related to the development of IR and inflammation, important hallmarks of MetS [42]. Consistent with this notion, IR has been shown to positively correlate with HDAC3 activity and HDAC3 mRNA levels in peripheral blood mononuclear cells of T2DM patients [43]. Moreover, sirtuins (SIRT, a class of HDACs) are reported to act as metabolic regulators of glucose homeostasis and IR-associated inflammation [44,45]. A lack of SIRT1-, SIRT2- and SIRT6-dependent deacetylation and activation of specific adipose gene programs have been shown to contribute to the development of metabolic disorders, including obesity andT2DM [46,47], thus further suggesting a role for histone modifications in the aetiology of metabolic disorders.

3. Epigenetics and miRNAs in MetS

While growing evidence has demonstrated an important role of classical epigenetic mechanisms, including DNA methylation and histone modifications, it is important also to consider factors outside of the nucleotide sequence that could affect gene expression. These factors alter the gene expression by affecting underlying mechanisms responsible for how transcription factors and other proteins bind to DNA, thus changing gene expression, which affects metabolism. In the context of such factors, there is increasing recognition of the role of small RNA molecules, particularly microRNA (miRNA), in the epigenetic regulation of gene expression [13,19,51].
miRNAs are evolutionary conserved, short non-coding RNA molecules of size ~18–24 nucleotides and are known to be involved in post-transcriptional and transcriptional regulation of gene expression and protein synthesis [52]. More than 2500 mature miRNA species have been discovered in the human genome [53]. It has been reported that >60% of protein-coding genes in the human genome are targeted by miRNAs [54], with a single miRNA being able to target and regulate several thousand mRNAs [55]. miRNAs function by binding to complementary sequences on the 3′-untranslated region (3′UTR) of the target messenger RNA (mRNA), thereby decreasing its stability and translation efficiency [56]. Dysregulation in the expression of miRNAs has been shown to modulate pathological pathways involved in the development of various diseases [57], such as T2DM [58,59], cancer [60,61] and CVD [62,63], and therefore have utility as potential biomarkers or diagnostic tools for both normal physiological and disease states [64].
Interestingly, the interaction of miRNAs with their gene targets is dynamic and dependent on various factors, such as their subcellular location, including mitochondria, endoplasmic reticulum and multivesicular bodies, their abundance, target mRNAs and the affinity of the miRNA–mRNA interaction [65]. Emerging studies to date have not only established the role of miRNAs in key physiological processes, including cell proliferation, DNA repair, cellular differentiation, insulin secretion, aging, metabolism and apoptosis of the host cell [66,67], but have shown them to be secreted into extracellular fluids and transported to target cells via vesicular bodies, such as exosomes, microvesicles or by binding to Argonaute (Ago) proteins (25) acting as the cell to cell mediators, thereby regulating their gene expression in a paracrine or endocrine manner [68,69].
In addition to their post-transcriptional functions, miRNAs are also reported to be involved in transcriptional gene regulation by binding to specific sequences of epigenetic events on DNA [70]. For example, miRNAs control the expression of various epigenetic regulators, such as DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) [71]. Similarly, DNA methylation and histone modifications can regulate the expression of some miRNAs. Taken together, miRNAs and epigenetic regulators cooperate to modulate the expression of mutual targets by forming a feedback loop. Therefore, although not strictly considered epigenetic factors, miRNAs contribute to gene expression modulation through epigenetic mechanisms. Any disruption of this complex regulation may participate in the development of different diseases, including MetS.

3.1. Epigenetically-Regulated miRNAs in MetS

Previous studies investigating the pathophysiology of MetS have revealed a complex network of reciprocal interconnections between those of miRNAs and the classic epigenetic machinery of DNA methylation and histone modifications [19,51,72]. In addition to regulating structural genes by the classical mechanisms, they have an important role in regulating miRNA expression [51,73]. DNA methylation regulates miRNA transcription either by hyper- or hypo-methylation of the promoter regions of miRNA genes [74]. Indeed, around 50% of the miRNAs are associated with CpG islands [75], and methylation of these sites on miRNA promoters can result in their modified expression [73,76]. For instance, it has been observed that pancreatic-β cells of individuals with T2DM have a cluster of miRNAs that are epigenetically regulated by hypermethylation of the DLK1–MEG3 locus on chromosome 14q32 [77]. A study by Wang et al. [78] examining expression levels and DNA methylation of miR-375, one of the most abundant miRNAs in the human pancreatic β cells, identified hypermethylation of its promoter and downregulated expression in the plasma of individuals with impaired glucose tolerance compared to those with normal glucose tolerance. Further, methylation of CpG sites located in the coding regions of miR-1203, miR-412 and miR-216a is responsible for their differential expression in obese children’s peripheral blood compared to non-obese children [79].
Epigenetic regulation of miRNA expression cannot be represented by DNA methylation alone but also involves histone post-translational modifications [80,81]. While the impact of alterations in the histone modifications on miRNA expression has not been extensively investigated in MetS per se, limited studies to date have implicated a regulatory role of histone-modified epigenetic regulations on miRNA expression during various pathophysiological mechanisms responsible for MetS development. Among these modifications, chromatin structures have been associated with the biogenesis and post-transcriptional regulation of miRNAs [82]. Several miRNAs related to metabolism are shown to be regulated by a repressive chromatin structure involving H3K27me3 mediated by an epigenetic regulator enhancer of zester homolog 2 (EZH2) [83,84]. Expression of miR-101-3p, a pancreatic islet-enriched miRNA with a role in insulin secretion and β cell functioning [85], is demonstrated to be regulated by H3K27me3 modification in EZH2. Moreover, epigenetically regulated alterations in miRNAs induced by suboptimal maternal nutrition or endocrine factors are also reported to be responsible for altered gene expression and to promote offspring and adult MetS phenotypes [86,87]. Altogether, these studies provide evidence for a functional link between classical epigenetic regulations of miRNA underpinning pathophysiological processes characterised as causal factors in the development of MetS.

3.2. miRNA-Induced Epigenetic Regulation in MetS

miRNAs are reported to reciprocally regulate the classical epigenetic processes and play an essential role in the pathophysiological mechanisms of various diseases, such as cancer and T2DM [88]. Numerous studies provide insights into miRNA contributing to the pathophysiology of metabolic-related diseases through the regulation of DNA methylation [78,89]. miRNAs are shown to regulate the expression of key DNA methyltransferases (DNMTs), including DNMT3a and DNMT3b and methylation-related proteins involved in de novo methylation [90]. For instance, miR-148, an identified circulatory biomarker of MetS [91], targets DNMT3b at the penultimate exon of their coding regions [92]. Similarly, members of the miR-29 family are shown to target DNMT3a and DNMT3b [87] directly. Dysregulation in the expression levels of miR-29 has been linked to impaired metabolic function, including altered insulin sensitivity and glucose metabolism, thus contributing to the pathogenesis of MetS [93,94]. In addition, a study involving a model of obesity in the mouse identified hepatic miR-29b to regulate DNMT3a and the hormone-encoding gene energy homeostasis-associated (Enho) in modulating insulin sensitivity [93].
It is well known that miRNA-mediated epigenetic changes induce obesity-associated adipose tissue inflammation, a significant factor responsible for developing IR and T2DM [95]. For example, it has been shown that obese adipocyte-derived exosomes have an increased expression of miR-29a. When transferred into adipocytes, myocytes and hepatocytes, these exosomes could result in IR both in in vitro and in vivo models [96]. Furthermore, SIRT1, a class-III histone deacetylase with an essential role in inflammation and metabolic homeostasis, has been identified to be regulated by miRNAs in the adipose tissue of individuals with obesity [44,97]. For instance, miR-377, an important regulator of adipogenesis, has been shown to target the 3’-UTR of SIRT1 mRNA directly, and downregulate its protein abundance, thereby promoting obesity-induced inflammation and IR [98]. Decreased insulin sensitivity, an important hallmark of MetS, has also been reported to be affected by miRNA-induced epigenetic alterations, partly through suppression of SIRT1 [47]. For example, miR-221, an miRNA known to affect adipocyte differentiation, metabolic homeostasis and insulin signalling [99], has been shown to promote adipose tissue inflammation by negatively regulating SIRT1 and decreasing adipose tissue insulin sensitivity [100]. miRNAs are also reported to suppress the differentiation of preadipocytes in a mouse model of obesity by involving the interaction of miR-138-5p with the 3’UTR of EZH2 [101]. Additionally, miR-22-3p acts as an inhibitor of adipogenic differentiation by suppressing histone deacetylase-6 (HDAC6) in human adipose tissue-derived mesenchymal stem cells [102]. Considering the evidence to date, these findings provide a framework for miRNAs in regulating the epigenetic machinery underlying the pathophysiological mechanisms linked to MetS development and progression.

4. Epigenetic Strategies for MetS Prevention/Reversal

The epigenome is in constant feedback with both the genotype and phenotype of an organism, and it has a profound effect on the pathophysiology of MetS [42]. Moreover, it has been reported that environmental factors, including diet and nutrition, can trigger the change in epigenetic regulation responsible for the pathophysiology of MetS. Therefore, developing interventions to modify the epigenetic alterations would enable ameliorating MetS and related metabolic disorders. These epigenetic-based strategies would include pharmacological interventions, epigenetic-based diets, editing tools for modifying the epigenome and miRNA-based therapeutics and diagnostic tools.

4.1. Epigenetic–Pharmacological Interventions

Epigenetic modifications are potentially reversible, and therefore pharmacological interventions targeted at these modifications hold great potential for the prevention, diagnosis, treatment and prognosis of MetS [22]. Drugs targeting epigenetic changes are termed either “epigenetic drugs” or “epidrugs”. Possible epidrugs would involve therapies that can alter DNA methylation, histone/chromatin modifications or miRNA expression [103]. Epidrugs mainly act as inhibitors for most of their targets but can also potentially activate them. Currently, several drugs targeting the enzymes necessary for undergoing modifications and nucleoside and non-nucleoside analogues of epigenetic changes are undergoing preclinical and clinical trials [14].
Since aberrant methylation of metabolic gene promoters and overexpression of DNMTs has been established as one of the major key players in the pathophysiology of MetS, demethylating agents such as DNMT inhibitors appear to offer promise as potential therapeutic targets for MetS. DNMT inhibitors function by blocking the key methylation enzymes such as DNMT1, DNMT3a and DNMT3b, which subsequently blocks DNA methylation. For instance, 5-azacitidine, a structural analogue of cytosine, when incorporated into DNA, is methylated by covalently bound DNMT3a, eventually inhibiting the enzyme activity [104]. A study by You et al. [105] utilizing insulin-resistant 3T3-L1 adipocytes identified an increased percentage of insulin-stimulated glucose uptake upon treatment with 5-azacitidine [105]. Another study employing a rat model of obesity observed that DNA hypermethylation was associated with reduced insulin sensitivity [106]. Using insulin-resistant rat hepatocytes, the authors observed that treatment with 5-azacitidine could restore insulin sensitivity [106].
One other class of epidrugs would involve modulators of histone acetylation, i.e., inhibitors of histone acetyltransferase (HATi) and inhibitors or activators of histone deacetylase (HDAC) [107]. For instance, tannic acid (TA), a plant-derived hydrolysable tannin polyphenol and a novel HATi, is reported to potentially attenuate lipid accumulation and ameliorate the development of non-alcoholic fatty liver diseases via inhibition of HAT activity [108]. Evidence from several preclinical and clinical trials suggests that other HDACis, including valproic acid (VPA), sodium phenylbutyrate (PBA) and trichostatin A (TSA), could have beneficial roles in reducing fat accumulation, IR, inflammation and gluconeogenesis, thus exerting anti-diabetic effects [109,110,111,112,113]. A different category of epidrugs would include regulators of SIRTs [114]. For example, fluvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG CoA), is reported to act as the activator of SIRT6 in liver cell models via phosphorylation of the AMPKα and SREBP-1 pathway, thereby maintaining cholesterol regulation [115]. Apart from acting as an activator, small molecules, such as 2,4-dioxo-N-(4-(pyridin-3-yloxy) phenyl)-1,2,3,4-tetrahydroquinazoline-6-sulfonamide, are identified to act as Sirt6 inhibitors in mice models of T2DM [116]. This molecule was identified to improve glucose tolerance in the mice by increasing the glucose transporters GLUT1 and GLUT1 and reducing insulin, triglycerides and cholesterol levels.
Epidrugs have the potential to provide novel therapeutic agents for various diseases, including MetS. However, there are still challenges remaining for implementing epigenetic-based approaches in the clinic [14]. In particular, to achieve the promise of reversing epigenetic alterations by genome-wide-acting epidrugs, a comprehensive understanding of their mechanism of action and specificity in relation to a specific disease and tissue is required [117].

4.2. Epigenetic Diets

Dietary factors play a significant role in human health, with dietary bioactive compounds reported to act as key mediators of epigenetic reprogramming [15,118]. The diets or dietary compounds identified to mediate metabolic programming through epigenetic modifications are termed “epigenetic diets”. Epigenetic diets not only modify the classic epigenetic mechanisms of DNA methylation and histone modifications, but they have also been identified to interact with miRNAs and are therefore involved in the dynamic regulation of gene expression, controlling cellular phenotypes linked to both the prevention and progression of disease phenotypes [119].
Growing evidence involving human clinical and dietary intervention studies supports the importance of individual nutrients, whole foods and dietary patterns in preventing and managing metabolic disorders by altering the epigenetic modifications [118,119]. Among such diets, long-chain omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to have anti-obesity effects by modulating epigenetic mechanisms responsible for the underlying pathophysiology, including lipid metabolism adipokine regulation, adipose tissue inflammation and adipogenesis. Several animal and human studies have also reported consumption of n-3 PUFAs to modulate the expression of miRNAs involved in key metabolic pathways, such as lipid metabolism and inflammation [120]. Likewise, Mediterranean diets, due to their abundant source of phytochemicals, are reported to have positive health effects by modulating the epigenomic mechanisms regulating processes related to metabolic homeostasis and pathophysiological pathways involved in metabolic disorders [121,122]. An example of one of the essential components found in the Mediterranean diet would be that of sulforaphane, an isothiocyanate known to reduce hepatic glucose production and improve glucose control in individuals with obesity and T2DM [123,124]. Based on the molecular mechanism of action of sulforaphane, epigenetic control of histone deacetylase (HDAC) and DNA methylation activity could be a possible mechanism underlying the observed changes in glucose control [125]. Both acute and long-term Mediterranean dietary patterns have also been shown to modulate miRNA expression associated with the pathogenesis of MetS, such as inflammatory gene regulation, atherogenic mechanisms and adipogenesis [126,127,128,129]. For example, a study analysing the expression of miRNAs in response to 8 weeks of intake of a hypocaloric Mediterranean diet in individuals with MetS reported an altered expression of miRNAs (decreased miR-155 and increased let-7b) that are involved in the pathogenesis of CVD [127].
Another example of dietary-derived HATi would involve curcumin, a polyphenolic compound in turmeric. Curcumin is reported to decrease hyperglycaemia-induced cytokine production in monocytes via reducing HAT activity [15,130]. Among activators of histone deacetylase, resveratrol, a small polyphenolic compound, has been identified to improve hepatic gluconeogenesis under IR conditions by acting via translocation of HDAC4 from the nucleus to the cytoplasm, thereby modulating the energy metabolism pathway [131,132]. Subsequently, several human clinical trials have shown positive health effects of resveratrol supplementation in individuals with obesity, NAFLD or T2DM [133,134,135]. In addition, resveratrol supplementation is also identified to modulate the expression of inflammation-related miRNAs [136,137]. For instance, a study by Carneriro et al. reported that a daily intake of grape extract and resveratrol-containing supplement resulted in the upregulation of miR-21, miR-181b, miR-663 and miR-30c and the downregulation of miR-155 in peripheral blood mononuclear cells of T2DM and hypertensive patients with coronary artery disease [138]. Moreover, the authors also showed an inverse relationship between the upregulated miRNAs and inflammatory cytokine gene expression [138].
Additionally, natural phenolic compounds, including catechin, epicatechin and their oligomers proanthocyanidin and epigallocatechin-3-gallate (EGCG), are considered biological modulators of MetS [30,139]. In both human and animal models of obesity, catechins are shown to attenuate dyslipidaemia and IR and reduce concentrations of inflammatory cytokines [140,141,142]. It has been reported that catechins mediate these effects, in part, by modulating epigenetic mechanisms such as DNMT inhibition, increasing HDAC activity or by inhibition of HAT activity [143,144]. Further, polyphenols are also reported to modulate miRNA expression involved in regulating genes and pathways underlying MetS [145]. For example, using a high-fat diet-fed mice model, it was shown that following intake of green tea for 12 weeks, mice showed a decrease in adipose miR-335 with an increase in energy expenditure and a reduction in adipose tissue inflammation and IR-associated gene expression [146].
Another class of epigenetic diets would include diets that are involved in the modulation of one-carbon metabolism. One-carbon metabolism comprises a complex network of pathways involved in transferring and utilising one-carbon units necessary for nucleic acid biosynthesis, amino acid metabolism and methylation processes [147,148]. Growing evidence has suggested important links of one-carbon metabolism with insulin sensitivity pathways, fat deposition and energy homeostasis [149,150,151]. These diets would involve nutrients containing methyl donors such as choline, betaine and folate that serve functional roles across the body through their metabolic, epigenetic and immunomodulatory properties [152,153,154]. Based on the fact that one-carbon metabolism involves methyl donors for epigenetic reactions and that miRNA has an important role to play in the modulation of these mechanisms, this might suggest a bi-directional association between one-carbon metabolites and miRNA profiles [155,156].
Collectively, epigenetic diets may act as important adjuncts to the management of MetS. Supplementation of epigenetic diets would help enhance metabolic homeostasis by ameliorating processes involved in the pathophysiology of MetS, including inflammation, obesity, glucose intolerance and insulin insensitivity. Understanding the molecular targets of epigenetic diets in relation to maintaining metabolic homeostasis and the pathophysiology of metabolic disorders could help discover novel and effective therapeutic targets.

4.3. Epigenome Editing Tools

Changes in epigenetic patterns can ultimately alter an entire metabolic pathway, and these epigenetically disturbed pathways could serve as key targets for the treatment of various metabolic diseases, including MetS. Given the reversible nature of epigenetic modifications, developing tools to regulate gene expression by modifying these epigenetic states could be of significant importance [157,158]. The widely used epigenetic editing tools include the zinc finger proteins (ZFPs), transcription activator-like effectors (TALEs) and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-deactivated nuclease CRISPR-associated Cas9 protein system (dCas9) [159,160]. ZFPs and TALEs are modular DNA-binding proteins that form specific interactions between amino acid side chains of the DNA-binding domain and the nucleotides of target DNA sequences [157,161]. In contrast, the CRISPR-dCas9 targets DNA by enabling RNA:DNA base pair complementarity [160]. These tools, combined with epigenetic enzymes such as DNA methyltransferases, histone lysine/methyltransferases and histone deacetylases/demethylases, enable the reversibility of epigenetic modifications in a gene-specific manner.
The use of epigenetic editing tools for MetS is still in its infancy, although there is growing interest in using these tools for other related metabolic diseases, such as T2DM [162]. Since the loss of insulin-secreting β-cells is characteristic of the diabetes pathophysiology and because these cells do not proliferate readily, expanding the source of these cells remains a challenge. To enhance the rapid proliferation of human β-cells, a study by Ou et al. [163] utilized epigenetic editing tools to promote the proliferation of human β-cells in a diabetic, immune-compromised mice model. The authors used in situ human β-cells to target demethylation of imprinting control region 2 (ICR2) using a transcription activator-like effector protein fused to the catalytic domain of TET1 (ICR2-TET1) and repressed expression of p57 (cell cycle inhibitor). Further, transplantation of these epigenetically edited β-cells into diabetic immune-compromised mice reduced blood glucose levels. Another example includes epigenetically modified expression of Pcsk9, a gene responsible for regulating circulating cholesterol levels [157]. Systemic administration of a dual-vector adeno-associated viral 8 system (AAV8) system expressing dCas9 fused to the Krüppel-associated box epigenetic repressor motif (dCas9KRAB) and a Pcsk9-targeting guide RNA (gRNA) in the liver of adult mice resulted in reductions in the levels of both circulating Pcsk9 and cholesterol. Taken together, these studies establish the potential of epigenetic editing tools for dissecting gene regulation mechanisms, understanding the pathophysiological mechanism of MetS and modulating gene expression for therapeutic applications.

4.4. miRNA-Based Therapy and Diagnostics

Growing evidence has established dysregulated miRNA expression as a molecular signature in MetS. Like classical epigenetic mechanisms, targeting miRNAs is considered a potential method for developing novel therapeutic targets for MetS. miRNA-based therapeutics rely on inhibition of the upregulated miRNAs (antagomiRs) or the restoration of downregulated miRNAs (mimics). Moreover, because miRNAs are stably present in human bio-fluids and are disease- and tissue-specific, quantifying their abundance in blood samples has gained significant attention given their potential utility as biomarkers of MetS diagnosis, prognosis and response to treatment.

4.4.1. miRNA Therapeutics

Considering that miRNAs are involved in regulating multiple pathophysiological pathways with relevance to different components of MetS, there is a growing interest in establishing miRNA-based therapeutics [164,165]. miRNA therapeutics can be either in the form of miRNA mimics that amplify the impact of a specific depressed miRNA, or antagomiRs that suppress a specific overexpressed miRNA expression. To date, several miRNAs are in different phases of preclinical and clinical trials to be classified as novel therapeutics for several diseases [166,167]. An example of an miRNA-based therapeutic for cardio-metabolic diseases is MGN-9103, a locked nucleic acid (LNA)-modified antisense oligonucleotide (ASO) by Viridian Therapeutics, formerly known as miRagen Therapeutics [168]. MGN-9103 is an antagomiR designed against miR-208, a cardiac-specific miRNA with known benefits for cardiac function and has a therapeutic potential in improving systemic insulin sensitivity and glucose tolerance that contribute to MetS. Another antagomiR includes anti-miR-33, developed by Regulus Therapeutics, which is currently in preclinical stages of development. The use of anti-miR-33 on atherosclerosis regression in diabetic mice was shown to overcome the deleterious effects of T2DM [169].
Although the development of miRNA-based therapeutics could represent a novel treatment approach for various diseases, several challenges remain, including their stability and mode of delivery [170,171]. Commercially prepared miRNA molecules are quite unstable, therefore natural and chemically modified molecules such as 2′-O-methyl (2′-OMe) or LNA are used to stabilize and reduce their high reactivity [170]. Secondly, accurate delivery of these therapeutics to the desired tissue for treatment specificity remains a challenge. Approaches undertaken for the precise delivery of these miRNA therapeutics involve using designed nanoparticles or liposome-like particles incorporating miRNAs that can be targeted to different organs, or combining miRNA with a specific molecule that will bind to the cells of interest and enhance endocytosis [168].

4.4.2. miRNA Biomarkers

Emerging research into the identification of reliable and sensitive biomarkers for the progression and development of different diseases have supported the use of circulatory miRNAs [52,172]. Any deviation from normal miRNA-mediated regulatory networks appear to be a common characteristic of various disease pathogenesis, including cancer, CVDs and other metabolic diseases, thus suggesting circulatory miRNAs could be essential components in the disease pathobiology [173,174]. Circulatory miRNAs in clinical samples are reported to be highly stable, and they can withstand unfavourable physiological conditions, such as variations in pH and storage [175]. Moreover, the diagnosis of diseases with similar aetiologies remains a significant challenge [176]. The abundance of circulatory miRNAs is reproducible with disease and tissue specificity [177]. Since miRNAs meet most of the required criteria for being a biomarker, including accessibility, high specificity and sensitivity, there is an increasing interest in the utility of circulatory miRNAs as potential biomarkers for diagnosis, as well as markers of disease progression [178]. It has to be also highlighted that the therapeutical modulation of distinct miRNAs is gaining importance for the management of patients with MetS features, particularly for T2DM; for instance, it has been recently shown that novel anti-diabetic agents may exert a direct epigenetic effect in T2DM patients, regulating miRNAs involved in the maintenance of endothelial cell homeostasis, and that this effect is independent of the metabolic control [179].
Although several diagnostic measures, including body mass index (BMI), total body fat and blood levels of glucose, HbA1c, lipids and pro-inflammatory cytokines, are available as markers of metabolic health, diagnosis is usually missed in the early stages of these diseases as these markers are generally only detectable upon disease progression [180]. On the other hand, miRNAs have gained increasing attention as a possible means to provide insights into the complex gene regulatory mechanisms involved both in normal physiology and in pathophysiological processes by acting as mediators (and markers) of disease [181]. It has also been reported that variations in miRNA expression profiles can be seen as the body’s integrative response to genetic susceptibility and environmental effects, hence with potential as predictive, diagnostic and prognostic markers [182]. While studies performed both in in vitro and in vivo experimental models have highlighted the associations between altered circulatory miRNA abundances and the dysregulation of contributory factors related to cardio-metabolic diseases [183,184], little is known about the relationship between circulatory miRNAs and the early stages of MetS prior to the development of overt T2DM or CVD.
In relation to miRNAs as biomarkers of MetS, we have previously reported the circulatory miRNAs miR-15a-5p and miR-17-5p as predictive biomarkers of MetS [4]. Both of these miRNAs have an identified role in regulating molecular mechanisms underlying the pathophysiology of metabolic derangements, including β-cell apoptosis, IR and central obesity, which is well known [185,186]. We also observed miR-15a-5p and miR-17-5p abundance to significantly correlate with individual MetS components, including BMI, waist circumference, plasma HDL, plasma glucose and blood pressure. Since MetS represents a complex pathophysiology, correlations of these miRNAs with more than one component suggest that the identified miRNAs might be involved in regulating complex metabolic pathways responsible for the development and progression of MetS. Further, it has been identified that different miRNAs can regulate different components of MetS (Table 3) [187]. One such example would include an increased abundance of miR-221 and let-7g in the circulation of an Asian cohort with MetS [187]. The abundance of both miRNAs was observed to increase with an increasing number of MetS components presented, with let-7g and miR-221 abundance increasing in individuals with more than four components of MetS.
Nonetheless, research into the use of miRNAs as biomarkers for MetS is still in its infancy. To date, the findings generally lack reproducibility, with examples of inconsistent reporting across groups that have analysed the same disease pathology [204,205]. Some of this discordance can be overcome by the use of standardized protocols controlling for both pre-analytical conditions, including sample handling (collection, transport and storage), population under investigation (healthy or diseased) and analytical factors including abundance quantification platforms involving qPCR, high throughput sequencing (HTS), microarray and data normalization strategies (endogenous and exogenous normalizers).

5. Conclusions

Considering the metabolic diversity of MetS, together, both classical and miRNA-regulated epigenetic modifications likely act as determining factors for its underlying pathophysiology, and thus may constitute another level of regulation in mediating disease risk. While studies have implicated putative roles for DNA methylation, histone modifications and miRNA regulation on the pathophysiology of MetS, there remains a gap to determine the cause-and-effect relationship between these epigenetic mechanisms and MetS. Therefore, future studies on the dysregulation of the classical miRNA epigenetics machinery in MetS would enable a deeper understanding of MetS pathophysiology. From a therapeutic standpoint, both classical and miRNA-based epigenetic drugs and diets are, in this regard, a flourishing scope for future research directions in the setting of MetS.

Author Contributions

Conceptualization: F.R.; Original draft preparation: F.R., M.H.V. and R.F.M.; Critical review and editing: M.H.V. and R.F.M. All authors have read and agreed to the published version of the manuscript.

Funding

M.H.V. was supported by grants from the Health Research Council of New Zealand (19/297). M.H.V. is also the recipient of a James Cook Research Fellowship from the Royal Society of New Zealand.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Reddy, K.S. Global Burden of Disease Study 2015 provides GPS for global health 2030. Lancet 2016, 388, 1448–1449. [Google Scholar] [CrossRef] [Green Version]
  2. Roth, G.A.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef] [Green Version]
  3. Alwan, A. Global Status Report on Noncommunicable Diseases 2010; World Health Organization: Geneva, Switzerland, 2011; pp. 1–160. ISBN 9789241564229. [Google Scholar]
  4. Ramzan, F.; D’Souza, R.F.; Durainayagam, B.R.; Milan, A.M.; Markworth, J.F.; Miranda-Soberanis, V.; Sequeira, I.R.; Roy, N.C.; Poppitt, S.D.; Mitchell, C.J.; et al. Circulatory miRNA biomarkers of metabolic syndrome. Acta Diabetol. 2020, 57, 203–214. [Google Scholar] [CrossRef] [PubMed]
  5. Vassallo, P.; Driver, S.L.; Stone, N.J. Metabolic Syndrome: An Evolving Clinical Construct. Prog. Cardiovasc. Dis. 2016, 59, 172–177. [Google Scholar] [CrossRef] [PubMed]
  6. Robberecht, H.; De Bruyne, T.; Hermans, N. Effect of various diets on biomarkers of the metabolic syndrome. Int. J. Food Sci. Nutr. 2017, 68, 627–641. [Google Scholar] [CrossRef] [PubMed]
  7. Kurian, M.; Wolfe, B.M.; Ikramuddin, S. Metabolic Syndrome and Diabetes: Medical and Surgical Management; Springer: New York, NY, USA, 2016. [Google Scholar] [CrossRef]
  8. Panzeri, I.; Pospisilik, J.A. Epigenetic control of variation and stochasticity in metabolic disease. Mol. Metab. 2018, 14, 26–38. [Google Scholar] [CrossRef] [PubMed]
  9. Park, J.H.; Yoo, Y.; Park, Y.J. Epigenetics: Linking Nutrition to Molecular Mechanisms in Aging. Prev. Nutr. food Sci. 2017, 22, 81–89. [Google Scholar] [CrossRef]
  10. Quilichini, E.; Haumaitre, C. Implication of epigenetics in pancreas development and disease. Best Pract. Res. Clin. Endocrinol. Metab. 2015, 29, 883–898. [Google Scholar] [CrossRef] [Green Version]
  11. Costantino, S.; Mohammed, S.A.; Ambrosini, S.; Paneni, F. Epigenetic processing in cardiometabolic disease. Atherosclerosis 2019, 281, 150–158. [Google Scholar] [CrossRef] [Green Version]
  12. Kanherkar, R.R.; Bhatia-Dey, N.; Csoka, A.B. Epigenetics across the human lifespan. Front. Cell Dev. Biol. 2014, 2, 49. [Google Scholar] [CrossRef] [Green Version]
  13. Xu, W.; Wang, F.; Yu, Z.; Xin, F. Epigenetics and cellular metabolism. Genet. Epigenet. 2016, 1, 43–51. [Google Scholar] [CrossRef] [Green Version]
  14. Berdasco, M.; Esteller, M. Clinical epigenetics: Seizing opportunities for translation. Nat. Rev. Genet. 2019, 20, 109–127. [Google Scholar] [CrossRef] [PubMed]
  15. Ramos-Lopez, O.; Milagro, F.I.; Riezu-Boj, J.I.; Martinez, J.A. Epigenetic signatures underlying inflammation: An interplay of nutrition, physical activity, metabolic diseases, and environmental factors for personalized nutrition. Inflamm. Res. 2020, 1, 1. [Google Scholar]
  16. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic modifications: Basic mechanisms and role in cardiovascular disease. Circulation 2011, 123, 2145–2156. [Google Scholar] [CrossRef] [Green Version]
  17. Wei, J.W.; Huang, K.; Yang, C.; Kang, C.S. Non-coding RNAs as regulators in epigenetics (Review). Oncol. Rep. 2017, 37, 3–9. [Google Scholar] [CrossRef] [Green Version]
  18. Ouni, M.; Schürmann, A. Epigenetic contribution to obesity. Mamm. Genome 2020, 31, 134–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Muhonen, P.; Holthofer, H. Epigenetic and microRNA-mediated regulation in diabetes. Nephrol. Dial. Transplant. 2009, 24, 1088–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [Green Version]
  21. Edwards, J.R.; Yarychkivska, O.; Boulard, M.; Bestor, T.H. DNA methylation and DNA methyltransferases. Epigenetics and Chromatin 2017, 10, 23. [Google Scholar] [CrossRef] [Green Version]
  22. Ma, X.; Kang, S. Functional implications of DNA methylation in adipose biology. Diabetes 2019, 68, 871–878. [Google Scholar] [CrossRef] [Green Version]
  23. Castellano-Castillo, D.; Moreno-Indias, I.; Sanchez-Alcoholado, L.; Ramos-Molina, B.; Alcaide-Torres, J.; Morcillo, S.; Ocaña-Wilhelmi, L.; Tinahones, F.; Queipo-Ortuño, M.; Cardona, F. Altered Adipose Tissue DNA Methylation Status in Metabolic Syndrome: Relationships Between Global DNA Methylation and Specific Methylation at Adipogenic, Lipid Metabolism and Inflammatory Candidate Genes and Metabolic Variables. J. Clin. Med. 2019, 8, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Nuotio, M.L.; Pervjakova, N.; Joensuu, A.; Karhunen, V.; Hiekkalinna, T.; Milani, L.; Kettunen, J.; Järvelin, M.R.; Jousilahti, P.; Metspalu, A.; et al. An epigenome-wide association study of metabolic syndrome and its components. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef]
  25. Chitrala, K.N.; Hernandez, D.G.; Nalls, M.A.; Mode, N.A.; Zonderman, A.B.; Ezike, N.; Evans, M.K. Race-specific alterations in DNA methylation among middle-aged African Americans and Whites with metabolic syndrome. Epigenetics 2020, 15, 462–482. [Google Scholar] [CrossRef]
  26. Akinyemiju, T.; Do, A.N.; Patki, A.; Aslibekyan, S.; Zhi, D.; Hidalgo, B.; Tiwari, H.K.; Absher, D.; Geng, X.; Arnett, D.K.; et al. Epigenome-wide association study of metabolic syndrome in African-American adults. Clin. Epigenet. 2018, 10, 49. [Google Scholar] [CrossRef]
  27. Remely, M.; De La Garza, A.L.; Magnet, U.; Aumueller, E.; Haslberger, A.G. Obesity: Epigenetic regulation-Recent observations. Biomol. Concepts 2015, 6, 163–175. [Google Scholar] [CrossRef]
  28. Inagaki, T. Histone demethylases regulate adipocyte thermogenesis. Diabetol. Int. 2018, 9, 215–223. [Google Scholar] [CrossRef] [PubMed]
  29. McKay, J.A.; Mathers, J.C. Diet induced epigenetic changes and their implications for health. Acta Physiol. 2011, 202, 103–118. [Google Scholar] [CrossRef]
  30. Číž, M.; Dvořáková, A.; Skočková, V.; Kubala, L. The role of dietary phenolic compounds in epigenetic modulation involved in inflammatory processes. Antioxidants 2020, 9, 691. [Google Scholar] [CrossRef]
  31. Garcia-Lacarte, M.; Milagro, F.I.; Zulet, M.A.; Martinez, J.A.; Mansego, M.L. LINE-1 methylation levels, a biomarker of weight loss in obese subjects, are influenced by dietary antioxidant capacity. Redox Rep. 2016, 21, 67–74. [Google Scholar] [CrossRef] [Green Version]
  32. Marousez, L.; Lesage, J.; Eberlé, D. Epigenetics: Linking early postnatal nutrition to obesity programming? Nutrients 2019, 11, 2966. [Google Scholar] [CrossRef] [Green Version]
  33. Leenen, F.A.D.; Muller, C.P.; Turner, J.D. DNA methylation: Conducting the orchestra from exposure to phenotype? Clin. Epigenet. 2016, 8, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Das, M.; Sha, J.; Hidalgo, B.; Aslibekyan, S.; Do, A.N.; Zhi, D.; Sun, D.; Zhang, T.; Li, S.; Chen, W.; et al. Association of DNA methylation at CPT1A locus with metabolic syndrome in the genetics of lipid lowering drugs and diet network (GOLDN) study. PLoS ONE 2016, 11. [Google Scholar] [CrossRef]
  35. Ling, C.; Del Guerra, S.; Lupi, R.; Rönn, T.; Granhall, C.; Luthman, H.; Masiello, P.; Marchetti, P.; Groop, L.; Del Prato, S. Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. Diabetologia 2008, 51, 615–622. [Google Scholar] [CrossRef] [Green Version]
  36. Florath, I.; Butterbach, K.; Heiss, J.; Bewerunge-Hudler, M.; Zhang, Y.; Schöttker, B.; Brenner, H. Type 2 diabetes and leucocyte DNA methylation: An epigenome-wide association study in over 1500 older adults. Diabetologia 2016, 59, 130–138. [Google Scholar] [CrossRef]
  37. Guay, S.P.; Voisin, G.; Brisson, D.; Munger, J.; Lamarche, B.; Gaudet, D.; Bouchard, L. Epigenome-wide analysis in familial hypercholesterolemia identified new loci associated with high-density lipoprotein cholesterol concentration. Epigenomics 2012, 4, 623–639. [Google Scholar] [CrossRef] [PubMed]
  38. Karlić, R.; Chung, H.R.; Lasserre, J.; Vlahoviček, K.; Vingron, M. Histone modification levels are predictive for gene expression. Proc. Natl. Acad. Sci. USA 2010, 107, 2926–2931. [Google Scholar] [CrossRef] [Green Version]
  39. Marmorstein, R.; Trievel, R.C. Histone modifying enzymes: Structures, mechanisms, and specificities. Biochim. Biophys. Acta Gene Regul. Mech. 2009, 1789, 58–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Morgan, M.A.J.; Shilatifard, A. Reevaluating the roles of histone-modifying enzymes and their associated chromatin modifications in transcriptional regulation. Nat. Genet. 2020, 52, 1271–1281. [Google Scholar] [CrossRef]
  41. Inagaki, T.; Tachibana, M.; Magoori, K.; Kudo, H.; Tanaka, T.; Okamura, M.; Naito, M.; Kodama, T.; Shinkai, Y.; Sakai, J. Obesity and metabolic syndrome in histone demethylase JHDM2a-deficient mice. Genes to Cells 2009, 14, 991–1001. [Google Scholar] [CrossRef] [PubMed]
  42. Stols-Gonçalves, D.; Tristão, L.S.; Henneman, P.; Nieuwdorp, M. Epigenetic Markers and Microbiota/Metabolite-Induced Epigenetic Modifications in the Pathogenesis of Obesity, Metabolic Syndrome, Type 2 Diabetes, and Non-alcoholic Fatty Liver Disease. Curr. Diab. Rep. 2019, 19, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Sathishkumar, C.; Prabu, P.; Balakumar, M.; Lenin, R.; Prabhu, D.; Anjana, R.M.; Mohan, V.; Balasubramanyam, M. Augmentation of histone deacetylase 3 (HDAC3) epigenetic signature at the interface of proinflammation and insulin resistance in patients with type 2 diabetes. Clin. Epigenet. 2016, 8, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kotas, M.E.; Gorecki, M.C.; Gillum, M.P. Sirtuin-1 is a nutrient-dependent modulator of inflammation. Adipocyte 2013, 2, 113–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gillum, M.P.; Kotas, M.E.; Erion, D.M.; Kursawe, R.; Chatterjee, P.; Nead, K.T.; Muise, E.S.; Hsiao, J.J.; Frederick, D.W.; Yonemitsu, S.; et al. SirT1 regulates adipose tissue inflammation. Diabetes 2011, 60, 3235–3245. [Google Scholar] [CrossRef] [Green Version]
  46. Song, Y.S.; Lee, S.K.; Jang, Y.J.; Park, H.S.; Kim, J.H.; Lee, Y.J.; Heo, Y.S. Association between low SIRT1 expression in visceral and subcutaneous adipose tissues and metabolic abnormalities in women with obesity and type 2 diabetes. Diabetes Res. Clin. Pract. 2013, 101, 341–348. [Google Scholar] [CrossRef]
  47. Li, F.; Li, H.; Jin, X.; Zhang, Y.; Kang, X.; Zhang, Z.; Xu, M.; Qian, Z.; Ma, Z.; Gao, X.; et al. Adipose-specific knockdown of Sirt1 results in obesity and insulin resistance by promoting exosomes release. Cell Cycle 2019, 18, 2067–2082. [Google Scholar] [CrossRef]
  48. Chen, J.; Wang, N.; Dong, M.; Guo, M.; Zhao, Y.; Zhuo, Z.; Zhang, C.; Chi, X.; Pan, Y.; Jiang, J.; et al. The metabolic regulator histone deacetylase 9 contributes to glucose homeostasis abnormality induced by hepatitis C virus infection. Diabetes 2015, 64, 4088–4098. [Google Scholar] [CrossRef] [Green Version]
  49. Paneni, F.; Costantino, S.; Battista, R.; Castello, L.; Capretti, G.; Chiandotto, S.; Scavone, G.; Villano, A.; Pitocco, D.; Lanza, G.; et al. Adverse epigenetic signatures by histone methyltransferase set7 contribute to vascular dysfunction in patients with type 2 diabetes mellitus. Circ. Cardiovasc. Genet. 2015, 8, 150–158. [Google Scholar] [CrossRef]
  50. McKenna, B.; Guo, M.; Reynolds, A.; Hara, M.; Stein, R. Dynamic recruitment of functionally distinct Swi/Snf chromatin remodeling complexes modulates Pdx1 activity in Islet β cells. Cell Rep. 2015, 10, 2032–2042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Piletič, K.; Kunej, T. MicroRNA epigenetic signatures in human disease. Arch. Toxicol. 2016, 90, 2405–2419. [Google Scholar] [CrossRef]
  52. Weiland, M.; Gao, X.-H.; Zhou, L.; Mi, Q.-S. Small RNAs have a large impact. RNA Biol. 2012, 9, 850–859. [Google Scholar] [CrossRef] [Green Version]
  53. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef] [PubMed]
  54. Akhtar, M.M.; Micolucci, L.; Islam, M.S.; Olivieri, F.; Procopio, A.D. Bioinformatic tools for microRNA dissection. Nucleic Acids Res. 2016, 44, 24–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ghorai, A.; Ghosh, U. miRNA gene counts in chromosomes vary widely in a species and biogenesis of miRNA largely depends on transcription or post-transcriptional processing of coding genes. Front. Genet. 2014, 5, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Sluijter, J.P.G.; Pasterkamp, G. MicroRNAs. Circ. Res. 2017, 120, 5–7. [Google Scholar] [CrossRef]
  57. Vishnoi, A.; Rani, S. (Eds.) MiRNA Biogenesis and Regulation of Diseases: An Overview. In MicroRNA Profiling. Methods in Molecular Biology; Humana Press: New York, NY, USA, 2017; Volume 1509, pp. 1–10. [Google Scholar] [CrossRef]
  58. Wu, L.; Dai, X.; Zhan, J.; Zhang, Y.; Zhang, H.; Zhang, H.; Zeng, S.; Xi, W. Profiling peripheral microRNAs in obesity and type 2 diabetes mellitus. APMIS 2015, 123, 580–585. [Google Scholar] [CrossRef]
  59. Martinez-Sanchez, A.; Rutter, G.A.; Latreille, M. MiRNAs in β-cell development, identity, and disease. Front. Genet. 2017, 7, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Qu, K.; Zhang, X.; Lin, T.; Liu, T.; Wang, Z.; Liu, S.; Zhou, L.; Wei, J.; Chang, H.; Li, K.; et al. Circulating miRNA-21-5p as a diagnostic biomarker for pancreatic cancer: Evidence from comprehensive miRNA expression profiling analysis and clinical validation. Sci. Rep. 2017, 7, 1692. [Google Scholar] [CrossRef] [Green Version]
  61. Nakata, K.; Ohuchida, K.; Mizumoto, K.; Kayashima, T.; Ikenaga, N.; Sakai, H.; Lin, C.; Fujita, H.; Otsuka, T.; Aishima, S.; et al. MicroRNA-10b is overexpressed in pancreatic cancer, promotes its invasiveness, and correlates with a poor prognosis. Surgery 2011, 150, 916–922. [Google Scholar] [CrossRef] [PubMed]
  62. Maegdefessel, L. The emerging role of microRNAs in cardiovascular disease. J. Intern. Med. 2014, 276, 633–644. [Google Scholar] [CrossRef] [Green Version]
  63. Ding, Y.; Sun, X.; Shan, P.-F. MicroRNAs and Cardiovascular Disease in Diabetes Mellitus. BioMed Res. Int. 2017, 2017, 1–8. [Google Scholar] [CrossRef] [Green Version]
  64. Goodwin, B.C. Biology without Darwinian spectacles. Biologist 1982, 21, 108–112. [Google Scholar]
  65. Molotski, N.; Soen, Y. Differential association of microRNAs with polysomes reflects distinct strengths of interactions with their mRNA targets. RNA 2012, 18, 1612–1623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wienholds, E.; Plasterk, R.H.A.; Lawitz, E.; Samani, N.J. MicroRNA function in animal development. FEBS Lett. 2005, 579, 5911–5922. [Google Scholar] [CrossRef] [Green Version]
  67. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Turchinovich, A.; Samatov, T.R.; Tonevitsky, A.G.; Burwinkel, B. Circulating miRNAs: Cell-cell communication function? Front. Genet. 2013, 4, 119. [Google Scholar] [CrossRef] [Green Version]
  69. Bär, C.; Thum, T.; De Gonzalo-Calvo, D. Circulating miRNAs as mediators in cell-to-cell communication. Epigenomics 2019, 11, 111–113. [Google Scholar] [CrossRef] [PubMed]
  70. Turchinovich, A.; Tonevitsky, A.G.; Burwinkel, B. Extracellular miRNA: A Collision of Two Paradigms. Trends Biochem. Sci. 2016, 41, 883–892. [Google Scholar] [CrossRef]
  71. Sætrom, P.; Snøve, O.; Rossi, J.J. Epigenetics and microRNAs. Pediatr. Res. 2007, 61, 17–23. [Google Scholar] [CrossRef]
  72. Tian, Y.; Mok, M.T.S.; Yang, P.; Cheng, A.S.L. Epigenetic activation of Wnt/β-catenin signaling in NAFLD-associated hepatocarcinogenesis. Cancers 2016, 8, 76. [Google Scholar] [CrossRef]
  73. Oltra, S.S.; Peña-Chilet, M.; Vidal-Tomas, V.; Flower, K.; Martinez, M.T.; Alonso, E.; Burgues, O.; Lluch, A.; Flanagan, J.M.; Ribas, G. Methylation deregulation of miRNA promoters identifies miR124-2 as a survival biomarker in Breast Cancer in very young women. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef]
  74. Glaich, O.; Parikh, S.; Bell, R.E.; Mekahel, K.; Donyo, M.; Leader, Y.; Shayevitch, R.; Sheinboim, D.; Yannai, S.; Hollander, D.; et al. DNA methylation directs microRNA biogenesis in mammalian cells. Nat. Commun. 2019, 10, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Weber, B.; Stresemann, C.; Brueckner, B.; Lyko, F. Methylation of human MicroRNA genes in normal and neoplastic cells. Cell Cycle 2007, 6, 1001–1005. [Google Scholar] [CrossRef]
  76. Yang, Y.; Wang, L.L.; Li, Y.H.; Gao, X.N.; Liu, Y.; Yu, L. Effect of CpG island methylation on MicroRNA expression in the k-562 cell line. Biochem. Genet. 2012, 50, 122–134. [Google Scholar] [CrossRef] [PubMed]
  77. Kameswaran, V.; Bramswig, N.C.; McKenna, L.B.; Penn, M.; Schug, J.; Hand, N.J.; Chen, Y.; Choi, I.; Vourekas, A.; Won, K.J.; et al. Epigenetic regulation of the DLK1-MEG3 MicroRNA cluster in human type 2 diabetic islets. Cell Metab. 2014, 19, 135–145. [Google Scholar] [CrossRef] [Green Version]
  78. Wang, X.; Chang, X.; Li, J.; Yin, L.; Sun, K. DNA methylation of microRNA-375 in impaired glucose tolerance. Exp. Ther. Med. 2014, 8, 775–780. [Google Scholar] [CrossRef] [Green Version]
  79. Mansego, M.L.; Garcia-Lacarte, M.; Milagro, F.I.; Marti, A.; Martinez, J.A. DNA methylation of miRNA coding sequences putatively associated with childhood obesity. Pediatr. Obes. 2017, 12, 19–27. [Google Scholar] [CrossRef]
  80. Lin, Y.C.D.; Huang, H.Y.; Shrestha, S.; Chou, C.H.; Chen, Y.H.; Chen, C.R.; Hong, H.C.; Li, J.; Chang, Y.A.; Chiew, M.Y.; et al. Multi-omics profiling reveals microRNA-mediated insulin signaling networks. BMC Bioinform. 2020, 21, 389. [Google Scholar] [CrossRef] [PubMed]
  81. Gosline, S.J.C.; Gurtan, A.M.; JnBaptiste, C.K.; Bosson, A.; Milani, P.; Dalin, S.; Matthews, B.J.; Yap, Y.S.; Sharp, P.A.; Fraenkel, E. Elucidating MicroRNA Regulatory Networks Using Transcriptional, Post-transcriptional, and Histone Modification Measurements. Cell Rep. 2016, 14, 310–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Tao, B.B.; Liu, X.Q.; Zhang, W.; Li, S.; Dong, D.; Xiao, M.; Zhong, J. Evidence for the association of chromatin and microRNA regulation in the human genome. Oncotarget 2017, 8, 70958–70966. [Google Scholar] [CrossRef] [Green Version]
  83. Wang, Y.; Wang, M.; Wei, W.; Han, D.; Chen, X.; Hu, Q.; Yu, T.; Liu, N.; You, Y.; Zhang, J. Disruption of the EZH2/miRNA/β-catenin signaling suppresses aerobic glycolysis in glioma. Oncotarget 2016, 7, 49450–49458. [Google Scholar] [CrossRef] [Green Version]
  84. Vanderkruk, B.; Hoffman, B.G. Metabolism as a central regulator of β-cell chromatin state. FEBS J. 2020, febs.15562. [Google Scholar] [CrossRef]
  85. Zheng, Y.; Wang, Z.; Tu, Y.; Shen, H.; Dai, Z.; Lin, J.; Zhou, Z. MiR-101a and miR-30b contribute to inflammatory cytokine-mediated β-cell dysfunction. Lab. Investig. 2015, 95, 1387–1397. [Google Scholar] [CrossRef] [Green Version]
  86. Floris, I.; Descamps, B.; Vardeu, A.; Mitić, T.; Posadino, A.M.; Shantikumar, S.; Sala-Newby, G.; Capobianco, G.; Mangialardi, G.; Howard, L.; et al. Gestational Diabetes Mellitus Impairs Fetal Endothelial Cell Functions Through a Mechanism Involving MicroRNA-101 and Histone Methyltransferase Enhancer of Zester Homolog-2. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 664–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Liao, X.; Zhou, Z.; Zhang, X. Effects of miR-195-5p on cell proliferation and apoptosis in gestational diabetes mellitus via targeting EZH2. Mol. Med. Rep. 2020, 22, 803–809. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, S.; Wu, W.; Claret, F.X. Mutual regulation of microRNAs and DNA methylation in human cancers. Epigenetics 2017, 12, 187–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Sun, K.; Chang, X.; Yin, L.; Li, J.; Zhou, T.; Zhang, C.; Chen, X. Expression and DNA methylation status of microRNA-375 in patients with type 2 diabetes mellitus. Mol. Med. Rep. 2013, 9, 967–972. [Google Scholar] [CrossRef] [Green Version]
  90. Chhabra, R. miRNA and methylation: A multifaceted liaison. ChemBioChem 2015, 16, 195–203. [Google Scholar] [CrossRef] [PubMed]
  91. Doghish, A.S.; Elsisi, A.M.; Amin, A.I.; Abulsoud, A.I. Circulating miR-148a-5p and miR-21-5p as novel diagnostic biomarkers in Egyptian male patients with metabolic syndrome. Can. J. Diabetes 2021. [Google Scholar] [CrossRef] [PubMed]
  92. Duursma, A.M.; Kedde, M.; Schrier, M.; Le Sage, C.; Agami, R. miR-148 targets human DNMT3b protein coding region. RNA 2008, 14, 872–877. [Google Scholar] [CrossRef] [Green Version]
  93. Yu-Han, H.; Kanke, M.; Kurtz, C.L.; Cubitt, R.; Bunaciu, R.P.; Miao, J.; Zhou, L.; Graham, J.L.; Hussain, M.M.; Havel, P.; et al. Acute suppression of insulin resistance-associated hepatic miR-29 in vivo improves glycemic control in adult mice. Physiol. Genom. 2019, 51, 379–389. [Google Scholar] [CrossRef]
  94. Kurtz, C.L.; Peck, B.C.E.; Fannin, E.E.; Beysen, C.; Miao, J.; Landstreet, S.R.; Ding, S.; Turaga, V.; Lund, P.K.; Turner, S.; et al. MicroRNA-29 fine-tunes the expression of key foxa2- Activated lipid metabolism genes and is dysregulated in animal models of insulin resistance and diabetes. Diabetes 2014, 63, 3141–3148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384. [Google Scholar] [CrossRef] [Green Version]
  96. Liu, T.; Sun, Y.C.; Cheng, P.; Shao, H.G. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochem. Biophys. Res. Commun. 2019, 515, 352–358. [Google Scholar] [CrossRef]
  97. Kurylowicz, A.; Owczarz, M.; Polosak, J.; Jonas, M.I.; Lisik, W.; Jonas, M.; Chmura, A.; Puzianowska-Kuznicka, M. SIRT1 and SIRT7 expression in adipose tissues of obese and normal-weight individuals is regulated by microRNAs but not by methylation status. Int. J. Obes. 2016, 40, 1635–1642. [Google Scholar] [CrossRef] [PubMed]
  98. Peng, J.; Wu, Y.; Deng, Z.; Zhou, Y.; Song, T.; Yang, Y.; Zhang, X.; Xu, T.; Xia, M.; Cai, A.; et al. MiR-377 promotes white adipose tissue inflammation and decreases insulin sensitivity in obesity via suppression of sirtuin-1 (SIRT1). Oncotarget 2017, 8, 70550–70563. [Google Scholar] [CrossRef] [PubMed]
  99. Ahonen, M.A.; Asghar, M.Y.; Parviainen, S.J.; Liebisch, G.; Höring, M.; Leidenius, M.; Fischer-Posovszky, P.; Wabitsch, M.; Mikkola, T.S.; Törnquist, K.; et al. Human adipocyte differentiation and composition of disease-relevant lipids are regulated by miR-221-3p. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158841. [Google Scholar] [CrossRef]
  100. Peng, J.; Zhou, Y.; Deng, Z.; Zhang, H.; Wu, Y.; Song, T.; Yang, Y.; Wei, H.; Peng, J. miR-221 negatively regulates inflammation and insulin sensitivity in white adipose tissue by repression of sirtuin-1 (SIRT1). J. Cell. Biochem. 2018, 119, 6418–6428. [Google Scholar] [CrossRef]
  101. Liu, Y.; Liu, H.; Li, Y.; Mao, R.; Yang, H.; Zhang, Y.; Zhang, Y.; Guo, P.; Zhan, D.; Zhang, T. Circular RNA SAMD4A controls adipogenesis in obesity through the miR-138-5p/EZH2 axis. Theranostics 2020, 10, 4705–4719. [Google Scholar] [CrossRef]
  102. Huang, S.; Wang, S.; Bian, C.; Yang, Z.; Zhou, H.; Zeng, Y.; Li, H.; Han, Q.; Zhao, R.C. Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression. Stem Cells Dev. 2012, 21, 2531–2540. [Google Scholar] [CrossRef] [Green Version]
  103. Altucci, L.; Rots, M.G. Epigenetic drugs: From chemistry via biology to medicine and back. Clin. Epigenet. 2016, 8, 1–3. [Google Scholar] [CrossRef] [Green Version]
  104. Cai, W.J.; Liang, X.F.; Yuan, X.C.; Li, A.X.; He, S. Changes of DNA Methylation Pattern in Metabolic Pathways Induced by High-Carbohydrate Diet Contribute to Hyperglycemia and Fat Deposition in Grass Carp (Ctenopharyngodon idellus). Front. Endocrinol. 2020, 11, 398. [Google Scholar] [CrossRef] [PubMed]
  105. You, D.; Nilsson, E.; Tenen, D.E.; Lyubetskaya, A.; Lo, J.C.; Jiang, R.; Deng, J.; Dawes, B.A.; Vaag, A.; Ling, C.; et al. Dnmt3a is an epigenetic mediator of adipose insulin resistance. eLife 2017, 6. [Google Scholar] [CrossRef]
  106. Jiang, M.; Zhang, Y.; Liu, M.; Lan, M.S.; Fei, J.; Fan, W.; Gao, X.; Lu, D. Hypermethylation of hepatic glucokinase and L-type pyruvate kinase promoters in high-fat diet-induced obese rats. Endocrinology 2011, 152, 1284–1289. [Google Scholar] [CrossRef] [Green Version]
  107. Daneshpajooh, M.; Eliasson, L.; Bacos, K.; Ling, C. MC1568 improves insulin secretion in islets from type 2 diabetes patients and rescues β-cell dysfunction caused by Hdac7 upregulation. Acta Diabetol. 2018, 55, 1231–1235. [Google Scholar] [CrossRef] [Green Version]
  108. Chung, M.Y.; Song, J.H.; Lee, J.; Shin, E.J.; Park, J.H.; Lee, S.H.; Hwang, J.T.; Choi, H.K. Tannic acid, a novel histone acetyltransferase inhibitor, prevents non-alcoholic fatty liver disease both in vivo and in vitro model. Mol. Metab. 2019, 19, 34–48. [Google Scholar] [CrossRef]
  109. Rakitin, A. Does valproic acid have potential in the treatment of diabetes mellitus? Front. Endocrinol. 2017, 8, 147. [Google Scholar] [CrossRef] [Green Version]
  110. Khan, S.; Komarya, S.K.; Jena, G. Phenylbutyrate and β-cell function: Contribution of histone deacetylases and ER stress inhibition. Epigenomics 2017, 9, 711–720. [Google Scholar] [CrossRef]
  111. Xiao, C.; Giacca, A.; Lewis, G.F. Sodium phenylbutyrate, a drug with known capacity to reduce endoplasmic reticulum stress, partially alleviates lipid-induced insulin resistance and β-cell dysfunction in humans. Diabetes 2011, 60, 918–924. [Google Scholar] [CrossRef] [Green Version]
  112. Sharma, S.; Taliyan, R. Histone deacetylase inhibitors: Future therapeutics for insulin resistance and type 2 diabetes. Pharmacol. Res. 2016, 113, 320–326. [Google Scholar] [CrossRef] [PubMed]
  113. Lu, J.C.; Chang, Y.T.; Wang, C.T.; Lin, Y.C.; Lin, C.K.; Wu, Z.S. Trichostatin A Modulates Thiazolidinedione-Mediated Suppression of Tumor Necrosis Factor α-Induced Lipolysis in 3T3-L1 Adipocytes. PLoS ONE 2013, 8, e71517. [Google Scholar] [CrossRef] [PubMed]
  114. Schiedel, M.; Robaa, D.; Rumpf, T.; Sippl, W.; Jung, M. The Current State of NAD+ -Dependent Histone Deacetylases (Sirtuins) as Novel Therapeutic Targets. Med. Res. Rev. 2018, 38, 147–200. [Google Scholar] [CrossRef]
  115. Kim, J.H.; Lee, J.M.; Kim, J.H.; Kim, K.R. Fluvastatin activates sirtuin 6 to regulate sterol regulatory element-binding proteins and AMP-activated protein kinase in HepG2 cells. Biochem. Biophys. Res. Commun. 2018, 503, 1415–1421. [Google Scholar] [CrossRef] [PubMed]
  116. Sociali, G.; Magnone, M.; Ravera, S.; Damonte, P.; Vigliarolo, T.; Von Holtey, M.; Vellone, V.G.; Millo, E.; Caffa, I.; Cea, M.; et al. Pharmacological Sirt6 inhibition improves glucose tolerance in a type 2 diabetes mouse model. FASEB J. 2017, 31, 3138–3149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Ganesan, A.; Arimondo, P.B.; Rots, M.G.; Jeronimo, C.; Berdasco, M. The timeline of epigenetic drug discovery: From reality to dreams. Clin. Epigenet. 2019, 11, 1–17. [Google Scholar] [CrossRef] [Green Version]
  118. Park, J.H.; Kim, S.H.; Lee, M.S.; Kim, M.S. Epigenetic modification by dietary factors: Implications in metabolic syndrome. Mol. Asp. Med. 2017, 54, 58–70. [Google Scholar] [CrossRef]
  119. Sánchez, I.; Reynoso-Camacho, R.; Salgado, L.M. The diet-induced metabolic syndrome is accompanied by whole-genome epigenetic changes. Genes Nutr. 2015, 10, 21. [Google Scholar] [CrossRef]
  120. Ortega, F.J.; Cardona-Alvarado, M.I.; Mercader, J.M.; Moreno-Navarrete, J.M.; Moreno, M.; Sabater, M.; Fuentes-Batllevell, N.; Ramírez-Chávez, E.; Ricart, W.; Molina-Torres, J.; et al. Circulating profiling reveals the effect of a polyunsaturated fatty acid-enriched diet on common microRNAs. J. Nutr. Biochem. 2015, 26, 1095–1101. [Google Scholar] [CrossRef]
  121. Bullón-Vela, V.; Abete, I.; Tur, J.A.; Pintó, X.; Corbella, E.; Martínez-González, M.A.; Toledo, E.; Corella, D.; Macías, M.; Tinahones, F.; et al. Influence of lifestyle factors and staple foods from the Mediterranean diet on non-alcoholic fatty liver disease among older individuals with metabolic syndrome features. Nutrition 2020, 71. [Google Scholar] [CrossRef]
  122. Martín-Peláez, S.; Fito, M.; Castaner, O. Mediterranean diet effects on type 2 diabetes prevention, disease progression, and related mechanisms. A review. Nutrients 2020, 12, 2236. [Google Scholar] [CrossRef]
  123. Pham, T.X.; Lee, J. Dietary regulation of histone acetylases and deacetylases for the prevention of metabolic diseases. Nutrients 2012, 4, 1868–1886. [Google Scholar] [CrossRef] [Green Version]
  124. Axelsson, A.S.; Tubbs, E.; Mecham, B.; Chacko, S.; Nenonen, H.A.; Tang, Y.; Fahey, J.W.; Derry, J.M.J.; Wollheim, C.B.; Wierup, N.; et al. Sulforaphane reduces hepatic glucose production and improves glucose control in patients with type 2 diabetes. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Kaufman-Szymczyk, A.; Majewski, G.; Lubecka-Pietruszewska, K.; Fabianowska-Majewska, K. The role of sulforaphane in epigenetic mechanisms, including interdependence between histone modification and DNA methylation. Int. J. Mol. Sci. 2015, 16, 29732–29743. [Google Scholar] [CrossRef] [Green Version]
  126. D’Amore, S.; Vacca, M.; Cariello, M.; Graziano, G.; D’Orazio, A.; Salvia, R.; Sasso, R.C.; Sabbà, C.; Palasciano, G.; Moschetta, A. Genes and miRNA expression signatures in peripheral blood mononuclear cells in healthy subjects and patients with metabolic syndrome after acute intake of extra virgin olive oil. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2016, 1861, 1671–1680. [Google Scholar] [CrossRef] [PubMed]
  127. Marques-Rocha, J.L.; Milagro, F.I.; Mansego, M.L.; Zulet, M.A.; Bressan, J.; Martínez, J.A. Expression of inflammation-related miRNAs in white blood cells from subjects with metabolic syndrome after 8 wk of following a Mediterranean diet-based weight loss program. Nutrition 2016, 32, 48–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Daimiel, L.; Micó, V.; Valls, R.M.; Pedret, A.; Motilva, M.J.; Rubió, L.; Fitó, M.; Farrás, M.; Covas, M.I.; Solá, R.; et al. Impact of Phenol-Enriched Virgin Olive Oils on the Postprandial Levels of Circulating microRNAs Related to Cardiovascular Disease. Mol. Nutr. Food Res. 2020, 64. [Google Scholar] [CrossRef]
  129. Ramzan, F.; D’Souza, R.F.; Durainayagam, B.R.; Milan, A.M.; Roy, N.C.; Kruger, M.C.; Henry, C.J.; Mitchell, C.J.; Cameron-Smith, D. Inflexibility of the plasma miRNA response following a high-carbohydrate meal in overweight insulin-resistant women. Genes Nutr. 2020, 15, 1–12. [Google Scholar] [CrossRef]
  130. Yun, J.M.; Jialal, I.; Devaraj, S. Epigenetic regulation of high glucose-induced proinflammatory cytokine production in monocytes by curcumin. J. Nutr. Biochem. 2011, 22, 450–458. [Google Scholar] [CrossRef] [Green Version]
  131. Zhao, H.; Shu, L.; Huang, W.; Song, G.; Ma, H. Resveratrol affects hepatic gluconeogenesis via histone deacetylase 4. Diabetes Metab. Syndr. Obes. Targets Ther. 2019, 12, 401–411. [Google Scholar] [CrossRef] [Green Version]
  132. Poulsen, M.M.; Jørgensen, J.O.L.; Jessen, N.; Richelsen, B.; Pedersen, S.B. Resveratrol in metabolic health: An overview of the current evidence and perspectives. Ann. N. Y. Acad. Sci. 2013, 1290, 74–82. [Google Scholar] [CrossRef] [PubMed]
  133. Hou, C.Y.; Tain, Y.L.; Yu, H.R.; Huang, L.T. The effects of resveratrol in the treatment of metabolic syndrome. Int. J. Mol. Sci. 2019, 20, 535. [Google Scholar] [CrossRef] [Green Version]
  134. Asgary, S.; Karimi, R.; Momtaz, S.; Naseri, R.; Farzaei, M.H. Effect of resveratrol on metabolic syndrome components: A systematic review and meta-analysis. Rev. Endocr. Metab. Disord. 2019, 20, 173–186. [Google Scholar] [CrossRef]
  135. Arzola-Paniagua, M.A.; García-Salgado López, E.R.; Calvo-Vargas, C.G.; Guevara-Cruz, M. Efficacy of an orlistat-resveratrol combination for weight loss in subjects with obesity: A randomized controlled trial. Obesity 2016, 24, 1454–1463. [Google Scholar] [CrossRef] [PubMed]
  136. Tili, E.; Michaille, J.J.; Adair, B.; Alder, H.; Limagne, E.; Taccioli, C.; Ferracin, M.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis 2010, 31, 1561–1566. [Google Scholar] [CrossRef]
  137. Gracia, A.; Fernández-Quintela, A.; Miranda, J.; Eseberri, I.; González, M.; Portillo, M.P. Are miRNA-103, miRNA-107 and miRNA-122 Involved in the Prevention of Liver Steatosis Induced by Resveratrol? Nutrients 2017, 9, 360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Tomé-Carneiro, J.; Larrosa, M.; Yáñez-Gascón, M.J.; Dávalos, A.; Gil-Zamorano, J.; Gonzálvez, M.; García-Almagro, F.J.; Ruiz Ros, J.A.; Tomás-Barberán, F.A.; Espín, J.C.; et al. One-year supplementation with a grape extract containing resveratrol modulates inflammatory-related microRNAs and cytokines expression in peripheral blood mononuclear cells of type 2 diabetes and hypertensive patients with coronary artery disease. Pharmacol. Res. 2013, 72, 69–82. [Google Scholar] [CrossRef]
  139. Casanova, E.; Salvadó, J.; Crescenti, A.; Gibert-Ramos, A. Epigallocatechin gallate modulates muscle homeostasis in type 2 diabetes and obesity by targeting energetic and redox pathways: A narrative review. Int. J. Mol. Sci. 2019, 20, 532. [Google Scholar] [CrossRef] [Green Version]
  140. Milagro, F.I.; Gómez-Abellán, P.; Campión, J.; Martínez, J.A.; Ordovás, J.M.; Garaulet, M. CLOCK, PER2 and BMAL1 DNA methylation: Association with obesity and metabolic syndrome characteristics and monounsaturated fat intake. Chronobiol. Int. 2012, 29, 1180–1194. [Google Scholar] [CrossRef] [PubMed]
  141. Yun, J.M.; Jialal, I.; Devaraj, S. Effects of epigallocatechin gallate on regulatory T cell number and function in obese v. lean volunteers. Br. J. Nutr. 2010, 103, 1771–1777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Bose, M.; Lambert, J.D.; Ju, J.; Reuhl, K.R.; Shapses, S.A.; Yang, C.S. The major green tea polyphenol, (-)-epigallocatechin-3-gallate, inhibits obesity, metabolic syndrome, and fatty liver disease in high-fat-fed mice. J. Nutr. 2008, 138, 1677–1683. [Google Scholar] [CrossRef]
  143. Yiannakopoulou, E.C. Targeting DNA methylation with green tea catechins. Pharmacology 2015, 95, 111–116. [Google Scholar] [CrossRef] [PubMed]
  144. Carlos-Reyes, Á.; López-González, J.S.; Meneses-Flores, M.; Gallardo-Rincón, D.; Ruíz-García, E.; Marchat, L.A.; Astudillo-De La Vega, H.; Hernández De La Cruz, O.N.; López-Camarillo, C. Dietary compounds as epigenetic modulating agents in cancer. Front. Genet. 2019, 10, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Corrêa, T.A.; Rogero, M.M. Polyphenols regulating microRNAs and inflammation biomarkers in obesity. Nutrition 2019, 59, 150–157. [Google Scholar] [CrossRef] [PubMed]
  146. Otton, R.; Bolin, A.P.; Ferreira, L.T.; Marinovic, M.P.; Rocha, A.L.S.; Mori, M.A. Polyphenol-rich green tea extract improves adipose tissue metabolism by down-regulating miR-335 expression and mitigating insulin resistance and inflammation. J. Nutr. Biochem. 2018, 57, 170–179. [Google Scholar] [CrossRef]
  147. Clare, C.E.; Brassington, A.H.; Kwong, W.Y.; Sinclair, K.D. One-Carbon Metabolism: Linking Nutritional Biochemistry to Epigenetic Programming of Long-Term Development. Annu. Rev. Anim. Biosci. 2019, 7, 263–287. [Google Scholar] [CrossRef]
  148. Ducker, G.S.; Rabinowitz, J.D. One-Carbon Metabolism in Health and Disease. Cell Metab. 2017, 25, 27–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Sherriff, J.L.; O’Sullivan, T.A.; Properzi, C.; Oddo, J.L.; Adams, L.A. Choline, its potential role in nonalcoholic fatty liver disease, and the case for human and bacterial genes. Adv. Nutr. 2016, 7, 5–13. [Google Scholar] [CrossRef] [Green Version]
  150. Radziejewska, A.; Muzsik, A.; Milagro, F.I.; Martínez, J.A.; Chmurzynska, A. One-Carbon Metabolism and Nonalcoholic Fatty Liver Disease: The Crosstalk between Nutrients, Microbiota, and Genetics. Lifestyle Genom. 2020, 13, 53–63. [Google Scholar] [CrossRef]
  151. Zeisel, S.H. Metabolic crosstalk between choline/1-carbon metabolism and energy homeostasis. Chem. Lab. Med. 2013, 51, 467–475. [Google Scholar] [CrossRef]
  152. Dibella, M.; Thomas, M.S.; Alyousef, H.; Millar, C.; Blesso, C.; Malysheva, O.; Caudill, M.A.; Fernandez, M.L. Choline intake as supplement or as a component of eggs increases plasma choline and reduces interleukin-6 without modifying plasma cholesterol in participants with metabolic syndrome. Nutrients 2020, 12, 3120. [Google Scholar] [CrossRef]
  153. Gao, X.; Wang, Y.; Randell, E.; Pedram, P.; Yi, Y.; Gulliver, W.; Sun, G. Higher Dietary Choline and Betaine Intakes are Associated with Better Body Composition in the Adult Population of Newfoundland, Canada. PLoS ONE 2016, 11, e0155403. [Google Scholar] [CrossRef] [Green Version]
  154. Dahlhoff, C.; Worsch, S.; Sailer, M.; Hummel, B.A.; Fiamoncini, J.; Uebel, K.; Obeid, R.; Scherling, C.; Geisel, J.; Bader, B.L.; et al. Methyl-donor supplementation in obese mice prevents the progression of NAFLD, activates AMPK and decreases acyl-carnitine levels. Mol. Metab. 2014, 3, 565–580. [Google Scholar] [CrossRef]
  155. Beckett, E.L.; Martin, C.; Choi, J.H.; King, K.; Niblett, S.; Boyd, L.; Duesing, K.; Yates, Z.; Veysey, M.; Lucock, M. Folate status, folate-related genes and serum miR-21 expression: Implications for miR-21 as a biomarker. BBA Clin. 2015, 4, 45–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Beckett, E.L.; Veysey, M.; Lucock, M. Folate and microRNA: Bidirectional interactions. Clin. Chim. Acta 2017, 474, 60–66. [Google Scholar] [CrossRef] [PubMed]
  157. Thakore, P.I.; Black, J.B.; Hilton, I.B.; Gersbach, C.A. Editing the epigenome: Technologies for programmable transcription and epigenetic modulation. Nat. Methods 2016, 13, 127–137. [Google Scholar] [CrossRef]
  158. Jin, H.J.; Jung, S.; DebRoy, A.R.; Davuluri, R.V. Identification and validation of regulatory SNPs that modulate transcription factor chromatin binding and gene expression in prostate cancer. Oncotarget 2016, 7, 54616–54626. [Google Scholar] [CrossRef] [Green Version]
  159. Waryah, C.B.; Moses, C.; Arooj, M.; Blancafort, P. Zinc fingers, TALEs, and CRISPR systems: A comparison of tools for epigenome editing. In Methods in Molecular Biology; Humana Press: New York, NY, USA, 2018; Volume 1767, pp. 19–63. [Google Scholar] [CrossRef]
  160. Syding, L.A.; Nickl, P.; Kasparek, P.; Sedlacek, R. CRISPR/Cas9 Epigenome Editing Potential for Rare Imprinting Diseases: A Review. Cells 2020, 9, 993. [Google Scholar] [CrossRef]
  161. Ceasar, S.A.; Rajan, V.; Prykhozhij, S.V.; Berman, J.N.; Ignacimuthu, S. Insert, remove or replace: A highly advanced genome editing system using CRISPR/Cas9. Biochim. Biophys. Acta Mol. Cell Res. 2016, 1863, 2333–2344. [Google Scholar] [CrossRef] [PubMed]
  162. Sgro, A.; Blancafort, P. Epigenome engineering: New technologies for precision medicine. Nucleic Acids Res. 2020, 48, 12453–12482. [Google Scholar] [CrossRef] [PubMed]
  163. Ou, K.; Yu, M.; Moss, N.G.; Wang, Y.J.; Wang, A.W.; Nguyen, S.C.; Jiang, C.; Feleke, E.; Kameswaran, V.; Joyce, E.F.; et al. Targeted demethylation at the CDKN1C/p57 locus induces human β cell replication. J. Clin. Investig. 2019, 129, 209–214. [Google Scholar] [CrossRef] [Green Version]
  164. Christopher, A.; Kaur, R.; Kaur, G.; Kaur, A.; Gupta, V.; Bansal, P. MicroRNA therapeutics: Discovering novel targets and developing specific therapy. Perspect. Clin. Res. 2016, 7, 68. [Google Scholar] [CrossRef] [PubMed]
  165. Rupaimoole, R.; Slack, F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–221. [Google Scholar] [CrossRef]
  166. Haussecker, D. Current issues of RNAi therapeutics delivery and development. J. Control. Release 2014, 195, 49–54. [Google Scholar] [CrossRef]
  167. Badalian-Very, G.; Hydbring, P. Clinical applications of microRNAs. F1000Research 2013, 2. [Google Scholar] [CrossRef]
  168. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.S. Therapeutic advances of miRNAs: A preclinical and clinical update. J. Adv. Res. 2021, 28, 127–138. [Google Scholar] [CrossRef]
  169. Distel, E.; Barrett, T.J.; Chung, K.; Girgis, N.M.; Parathath, S.; Essau, C.C.; Murphy, A.J.; Moore, K.J.; Fisher, E.A. MIR33 inhibition overcomes deleterious effects of diabetes mellitus on atherosclerosis plaque regression in mice. Circ. Res. 2014, 115, 759–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Bonneau, E.; Neveu, B.; Kostantin, E.; Tsongalis, G.J.; De Guire, V. How close are miRNAs from clinical practice? A perspective on the diagnostic and therapeutic market. Electron. J. Int. Fed. Clin. Chem. Lab. Med. 2019, 30, 114–127. [Google Scholar]
  171. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Doss, C.G.P.; Lee, S.S. Therapeutic miRNA and siRNA: Moving from Bench to Clinic as Next Generation Medicine. Mol. Ther. Nucleic Acids 2017, 8, 132–143. [Google Scholar] [CrossRef] [Green Version]
  172. Allegra, A.; Alonci, A.; Campo, S.; Penna, G.; Petrungaro, A.; Gerace, D.; Musolino, C. Circulating microRNAs: New biomarkers in diagnosis, prognosis and treatment of cancer (Review). Int. J. Oncol. 2012, 41, 1897–1912. [Google Scholar] [CrossRef] [Green Version]
  173. Vafaee, F.; Diakos, C.; Kirschner, M.B.; Reid, G.; Michael, M.Z.; Horvath, L.G.; Alinejad-Rokny, H.; Cheng, Z.J.; Kuncic, Z.; Clarke, S. A data-driven, knowledge-based approach to biomarker discovery: Application to circulating microRNA markers of colorectal cancer prognosis. NPJ Syst. Biol. Appl. 2018, 4, 20. [Google Scholar] [CrossRef] [PubMed]
  174. Pérez-Sánchez, C.; Arias-de la Rosa, I.; Aguirre, M.Á.; Luque-Tévar, M.; Ruiz-Limón, P.; Barbarroja, N.; Jiménez-Gómez, Y.; Ábalos-Aguilera, M.C.; Collantes-Estévez, E.; Segui, P.; et al. Circulating microRNAs as biomarkers of disease and typification of the atherothrombotic status in antiphospholipid syndrome. Haematologica 2018, 103, 908–918. [Google Scholar] [CrossRef] [Green Version]
  175. Liang, H.; Gong, F.; Zhang, S.; Zhang, C.Y.; Zen, K.; Chen, X. The origin, function, and diagnostic potential of extracellular microRNAs in human body fluids. Wiley Interdiscip. Rev. RNA 2014, 5, 285–300. [Google Scholar] [CrossRef]
  176. Lee, I.; Baxter, D.; Lee, M.Y.; Scherler, K.; Wang, K. The Importance of Standardization on Analyzing Circulating RNA. Mol. Diagnosis Ther. 2017, 21, 259–268. [Google Scholar] [CrossRef]
  177. Kondkar, A.A.; Abu-Amero, K.K. Utility of Circulating MicroRNAs as Clinical Biomarkers for Cardiovascular Diseases. BioMed Res. Int. 2015, 2015, 1–10. [Google Scholar] [CrossRef] [Green Version]
  178. Olivieri, F.; Capri, M.; Bonafè, M.; Morsiani, C.; Jung, H.J.; Spazzafumo, L.; Viña, J.; Suh, Y. Circulating miRNAs and miRNA shuttles as biomarkers: Perspective trajectories of healthy and unhealthy aging. Mech. Ageing Dev. 2017, 165, 162–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Giglio, R.V.; Nikolic, D.; Volti, G.L.; Stoian, A.P.; Banerjee, Y.; Magan-Fernandez, A.; Castellino, G.; Patti, A.M.; Chianetta, R.; Castracani, C.C.; et al. Liraglutide increases serum levels of microRNA27b,-130a and-210 in patients with type 2 diabetes mellitus: A novel epigenetic effect. Metabolites 2020, 10, 391. [Google Scholar] [CrossRef]
  180. Huang, Y.; Yan, Y.; Xv, W.; Qian, G.; Li, C.; Zou, H.; Li, Y. A New Insight into the Roles of MiRNAs in Metabolic Syndrome. BioMed Res. Int. 2018, 2018, 1–15. [Google Scholar] [CrossRef] [Green Version]
  181. Deiuliis, J.A. MicroRNAs as regulators of metabolic disease: Pathophysiologic significance and emerging role as biomarkers and therapeutics. Int. J. Obes. 2016, 40, 88–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Dumas, M.-E.; Emanueli, C. Circulating MicroRNAs to Predict the Risk for Metabolic Diseases in the General Population? Diabetes 2017, 66, 565–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Calderari, S.; Diawara, M.R.; Garaud, A.; Gauguier, D. Biological roles of microRNAs in the control of insulin secretion and action. Physiol. Genom. 2017, 49, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Párrizas, M.; Novials, A. Circulating microRNAs as biomarkers for metabolic disease. Best Pract. Res. Clin. Endocrinol. Metab. 2016, 30, 591–601. [Google Scholar] [CrossRef]
  185. Rawal, S.; Munasinghe, P.E.; Nagesh, P.T.; Lew, J.K.S.; Jones, G.T.; Williams, M.J.A.; Davis, P.; Bunton, D.; Galvin, I.F.; Manning, P.; et al. Down-regulation of miR-15a/b accelerates fibrotic remodelling in the Type 2 diabetic human and mouse heart. Clin. Sci. 2017, 131, 847–863. [Google Scholar] [CrossRef]
  186. Chen, Y.; Tian, L.; Wan, S.; Xie, Y.; Chen, X.; Ji, X.; Zhao, Q.; Wang, C.; Zhang, K.; Hock, J.M.; et al. MicroRNA-17-92 cluster regulates pancreatic beta-cell proliferation and adaptation. Mol. Cell. Endocrinol. 2016, 437, 213–223. [Google Scholar] [CrossRef]
  187. Wang, Y.-T.; Tsai, P.-C.; Liao, Y.-C.; Hsu, C.-Y.; Juo, S.-H. Circulating microRNAs have a sex-specific association with metabolic syndrome. J. Biomed. Sci. 2013, 20, 72. [Google Scholar] [CrossRef] [Green Version]
  188. Al‑Kafaji, G.; Al‑Mahroos, G.; Alsayed, N.; Hasan, Z.; Nawaz, S.; Bakhiet, M. Peripheral blood microRNA-15a is a potential biomarker for type 2 diabetes mellitus and pre-diabetes. Mol. Med. Rep. 2015, 12, 7485–7490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Heneghan, H.M.; Miller, N.; McAnena, O.J.; O’Brien, T.; Kerin, M.J. Differential miRNA expression in omental adipose tissue and in the circulation of obese patients identifies novel metabolic biomarkers. J. Clin. Endocrinol. Metab. 2011, 96, E846–E850. [Google Scholar] [CrossRef]
  190. He, Q.F.; Wang, L.X.; Zhong, J.M.; Hu, R.Y.; Fang, L.; Wang, H.; Gong, W.W.; Zhang, J.; Pan, J.; Yu, M. Circulating microRNA-21 is downregulated in patients with metabolic syndrome. Biomed. Environ. Sci. 2016, 29, 385–389. [Google Scholar] [CrossRef] [PubMed]
  191. Roncarati, R.; Viviani Anselmi, C.; Losi, M.A.; Papa, L.; Cavarretta, E.; Da Costa Martins, P.; Contaldi, C.; Saccani Jotti, G.; Franzone, A.; Galastri, L.; 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, 920–927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Liang, Y.-Z.; Dong, J.; Zhang, J.; Wang, S.; He, Y.; Yan, Y.-X. Identification of Neuroendocrine Stress Response-Related Circulating MicroRNAs as Biomarkers for Type 2 Diabetes Mellitus and Insulin Resistance. Front. Endocrinol. 2018, 9, 132. [Google Scholar] [CrossRef]
  193. Alrob, O.A.; Khatib, S.; Naser, S.A. MicroRNAs 33, 122, and 208: A potential novel targets in the treatment of obesity, diabetes, and heart-related diseases. J. Physiol. Biochem. 2017, 73, 307–314. [Google Scholar] [CrossRef] [PubMed]
  194. Al-Kafaji, G.; Al-Mahroos, G.; Abdulla Al-Muhtaresh, H.; Sabry, M.A.; Abdul Razzak, R.; Salem, A.H. Circulating endothelium-enriched microRNA-126 as a potential biomarker for coronary artery disease in type 2 diabetes mellitus patients. Biomarkers 2016, 1–11. [Google Scholar] [CrossRef]
  195. Kilic, I.D.; Dodurga, Y.; Uludag, B.; Alihanoglu, Y.I.; Yildiz, B.S.; Enli, Y.; Secme, M.; Bostanci, H.E. MicroRNA -143 and -223 in obesity. Gene 2015, 560, 140–142. [Google Scholar] [CrossRef]
  196. Karolina, D.S.; Armugam, A.; Tavintharan, S.; Wong, M.T.K.; Lim, S.C.; Sum, C.F.; Jeyaseelan, K. MicroRNA 144 impairs insulin signaling by inhibiting the expression of insulin receptor substrate 1 in type 2 diabetes mellitus. PLoS ONE 2011, 6, e22839. [Google Scholar] [CrossRef]
  197. Link, J.C.; Hasin-Brumshtein, Y.; Cantor, R.M.; Chen, X.; Arnold, A.P.; Lusis, A.J.; Reue, K. Diet, gonadal sex, and sex chromosome complement influence white adipose tissue miRNA expression. BMC Genom. 2017, 18, 89. [Google Scholar] [CrossRef] [Green Version]
  198. Al-Rawaf, H.A. Circulating microRNAs and adipokines as markers of metabolic syndrome in adolescents with obesity. Clin. Nutr. 2019, 38, 2231–2238. [Google Scholar] [CrossRef] [PubMed]
  199. Wang, X.; Sundquist, J.; Zöller, B.; Memon, A.A.; Palmér, K.; Sundquist, K.; Bennet, L. Determination of 14 circulating microRNAs in Swedes and Iraqis with and without diabetes mellitus type 2. PLoS ONE 2014, 9. [Google Scholar] [CrossRef]
  200. Motawae, T.M.; Ismail, M.F.; Shabayek, M.I.; Seleem, M.M. MicroRNAs 9 and 370 Association with Biochemical Markers in T2D and CAD Complication of T2D. PLoS ONE 2015, 10, e0126957. [Google Scholar] [CrossRef] [PubMed]
  201. Higuchi, C.; Nakatsuka, A.; Eguchi, J.; Teshigawara, S.; Kanzaki, M.; Katayama, A.; Yamaguchi, S.; Takahashi, N.; Murakami, K.; Ogawa, D.; et al. Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes. Metabolism 2015, 64, 489–497. [Google Scholar] [CrossRef] [Green Version]
  202. Wang, K.-J.; Zhao, X.; Liu, Y.-Z.; Zeng, Q.-T.; Mao, X.-B.; Li, S.-N.; Zhang, M.; Jiang, C.; Zhou, Y.; Qian, C.; et al. Circulating miR-19b-3p, miR-134-5p and miR-186-5p are promising novel biomarkers for early diagnosis of acute myocardial infarction. Cell. Physiol. Biochem. 2016, 38, 1015–1029. [Google Scholar] [CrossRef]
  203. Jiang, J.; Mo, H.; Liu, C.; Wu, B.; Wu, Z.; Li, X.; Li, T.; He, S.; Li, S.; You, Q.; et al. Inhibition of miR-186-5p contributes to high glucose-induced injury in AC16 cardiomyocytes. Exp. Ther. Med. 2018, 15, 627–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Zhang, J.; Zhao, H.; Gao, Y.; Zhang, W. Secretory miRNAs as novel cancer biomarkers. Biochim. Biophys. Acta Rev. Cancer 2012, 1826, 32–43. [Google Scholar] [CrossRef]
  205. Karolina, D.S.; Tavintharan, S.; Armugam, A.; Sepramaniam, S.; Pek, S.L.T.; Wong, M.T.K.; Lim, S.C.; Sum, C.F.; Jeyaseelan, K. Circulating miRNA Profiles in Patients with Metabolic Syndrome. J. Clin. Endocrinol. Metab. 2012, 97, E2271–E2276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 2. Human studies showing associations between histone modifications and features of metabolic health.
Table 2. Human studies showing associations between histone modifications and features of metabolic health.
ConditionHistone ModificationSample TypeRef
Insulin resistance and inflammationHDAC3Peripheral blood mononuclear cells[43]
Glucose metabolismHDAC9Liver[48]
Vascular dysfunction and T2DMSet7Peripheral blood mononuclear cells[49]
Obesity and T2DMSIRT1Visceral adipose tissue[46]
Glucose metabolismSwi/SnfPancreatic islet beta cells[50]
Table 3. Human studies reporting association between circulatory miRNAs and features of metabolic health.
Table 3. Human studies reporting association between circulatory miRNAs and features of metabolic health.
DiseasemiRNASample TypeRef
MetS/Atherosclerosis/ObesitymiR-15a-5p↓ Plasma[4,188]
MetS/ObesitymiR-17-5p↓ Serum/Plasma[4,189]
MetS
Chronic Heart Disease with T2DM
miR-21-3p↓ Plasma
↑ Plasma
[190]
Obesity/Chronic Heart DiseasemiR-29a-5p↑ Plasma[191]
T2DMlet-7 family↑ Plasma[192]
Obesity/T2DMmiR-122-5p↑ Plasma[193]
Atherosclerosis/T2DMmiR-126-5p↓ Plasma[194]
ObesitymiR-143-5p↓ Plasma[195]
T2DMmiR-144-5p↑ Plasma[196]
Obesity/T2DMmiR-221-3p↓ Plasma[197]
ObesitymiR-222-3p↑ Plasma[198]
T2DM/ObesitymiR-320a↑ Plasma[199]
T2DM/Diabetic CardiomyopathymiR-370-3p↑ Plasma[200]
T2DMmiR-375↑ Serum[201]
Diabetic CardiomyopathymiR-186-5p↑ Plasma[202,203]
↓ indicates decreased abundance compared to controls; ↑ indicates increased abundance compared to controls.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ramzan, F.; Vickers, M.H.; Mithen, R.F. Epigenetics, microRNA and Metabolic Syndrome: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 5047. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22095047

AMA Style

Ramzan F, Vickers MH, Mithen RF. Epigenetics, microRNA and Metabolic Syndrome: A Comprehensive Review. International Journal of Molecular Sciences. 2021; 22(9):5047. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22095047

Chicago/Turabian Style

Ramzan, Farha, Mark H. Vickers, and Richard F. Mithen. 2021. "Epigenetics, microRNA and Metabolic Syndrome: A Comprehensive Review" International Journal of Molecular Sciences 22, no. 9: 5047. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22095047

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop