The Epigenetic Promise to Improve Prognosis of Heart Failure and Heart Transplantation Chiara Sabia, Antonietta Picascia, Vincenzo Grimaldi, Cristiano Amarelli, Ciro Maiello, Claudio Napoli PII: DOI: Reference:
S0955-470X(17)30017-4 doi: 10.1016/j.trre.2017.08.004 YTRRE 459
To appear in:
Transplantation Reviews
Please cite this article as: Sabia Chiara, Picascia Antonietta, Grimaldi Vincenzo, Amarelli Cristiano, Maiello Ciro, Napoli Claudio, The Epigenetic Promise to Improve Prognosis of Heart Failure and Heart Transplantation, Transplantation Reviews (2017), doi: 10.1016/j.trre.2017.08.004
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Transplantation Reviews, Review Article, Revised form THE EPIGENETIC PROMISE TO IMPROVE PROGNOSIS OF HEART FAILURE AND HEART
, Cristiano Amarelli 3, Ciro Maiello
3
&
MA NU
Claudio Napoli 1,4. 1
1,2
SC
Chiara Sabia 1*, Antonietta Picascia 1, Vincenzo Grimaldi
RI P
T
TRANSPLANTATION
U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant
Immunology, Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria, Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Università degli Studi della Campania “L. Vanvitelli”, Italy.
Department of Sciences and Technologies, University of Sannio, Benevento, Italy.
3
Department of Cardiovascular Surgery and Transplants, Monaldi Hospital, Azienda dei Colli, Naples, Italy.
4
SDN Foundation, Institute of Diagnostic and Nuclear Development, IRCCS, Via Gianturco 113, 80143
ED
2
CE
PT
Naples, Italy
AC
Chiara Sabia and Antonietta Picascia contributed equally to this work.
*Corresponding author Dr. Chiara Sabia U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria, Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Università degli Studi della Campania “L. Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy E-mail address:
[email protected]
ACCEPTED MANUSCRIPT Transplantation Reviews, Review Article, Revised form
T
INTRODUCTION
RI P
Heart transplantation (HT) remains the gold standard treatment for end-stage heart failure (HF) ensuring the highest long-term survival; however, due to the shortage of organ donors, the left ventricular assist device
SC
(LVAD) provides a backup strategy for patients as bridge to transplantation or when clinical contraindications are present [1,2]. HF represents the end-stage of several cardiac pathological conditions
MA NU
including atherosclerosis, coronary heart disease (CHD) and dilated, hypertrophic and restrictive cardiomyopathies [3-5].
Human genetic studies have identified mutations that are associated with the onset and progression of cardiovascular disease (CVD). A growing body of evidence indicates the involvement of epigenetics in the
ED
development of CVD. In some cases, epigenetic modifications, a set of heritable and acquired modifications of the genome that alter gene expression without changing the DNA sequence, are stable and inherited by
PT
future generations, but in other cases many epigenetic changes are dynamic and responsive to environmental
CE
stimuli. [4,6-8].
Typically, epigenetic modifications regulate gene expression and are grouped into three main classes: DNA
AC
methylation, histone modifications and RNA-based mechanisms. DNA methylation consists in the addition of a methyl group on the carbon 5 of the cytosine residues (5mC) located in CpG dinucleotides. CpG dinucleotides are distributed throughout the genome, but display a high frequency in particular regions, termed “CpG islands”, that are localized in different genomic sites including gene promoter and body, and intergenic regions with enhancers and repetitive sequences. DNA methylation is catalyzed by a group of enzymes, the DNA methyltrasferase (DNMTs) and S-adenosylmethionine is the donor of the methyl groups. DNA methylation promotes gene silencing either directly, by blocking the binding of transcription factors to DNA, or indirectly, by binding MBPs (methyl-binding proteins) that, in turn, induce gene repression by recruiting co-repressors [9]. Acetylation or deacetylation of lysine residues in selected nucleosomal histone tails promotes gene activation or silencing, respectively [10]. Acetylation is a dynamic process regulated by two groups of enzymes: histone acetyltransferases (HATs) and histone deacetylases (HDACs), whose expression and activity are
ACCEPTED MANUSCRIPT finely regulated. Also, histone methylation can be associated with either activation or repression of transcription, depending on the degree of methylation of Lys or Arg residues. Furthermore, microRNAs (miRNAs), a class of small endogenous non-coding ribonucleic acids, regulates
RI P
T
gene expression at post-transcriptional level, through either transcript degradation or translational repression by binding to the 3 'UTR of the target mRNA [11]. MiRNAs originate from a primary transcript that is
for the processing by the endonuclease Dicer [12].
SC
cleaved in the nucleus by the RNase Drosha, thus resulting in a pre-miRNA that is exported to the cytoplasm
MA NU
Epigenetic mechanisms have been involved in the onset and progression of CVD as well as hypertrophic and dilated cardiomyopathies leading to HF, and have been proposed as potential new strategies for treating these diseases.
Here we discuss about epigenetic changes associated with all the clinical conditions leading to HF, and we also report the epigenetic mechanisms involved both in graft outcome and in diagnostic workup during post-
PT
ED
transplantation period.
EPIGENETIC MODIFICATIONS IN CARDIOVASCULAR DISEASE LEADING TO HEART
CE
TRANSPLANTATION
The development of HF implies the complex regulation of gene networks including both re-activation of
AC
fetal genes and/or re-expression of postnatal genes and leading to cardiac remodeling [13,14]. Even in disorders that recognize important environmental causes, such as CHD and hypertension, the risk of HF depends to some extent on genetic predisposition (Fig. 1). Epigenetic modifications can contribute to exacerbate the various stage of HF, by regulating synthesis of protein structures and by increasing the activity of cardiac fibroblasts with consequences on heart contractility and hypertrophy (Table 1). It is still unclear whether DNA methylation, histone changes, and miRNA synthesis are the cause or the effect of the failing hearts, but they could also represent an ideal target for future therapeutic strategies in HF [38].
ACCEPTED MANUSCRIPT DNA Methylation The effect of DNA hyper and /or hypomethylation on the expression of genes involved in several pathways underlying CVD has been studied in various experimental models (Table 1).
RI P
T
In vitro studies and animal models
In animal model, a significant global hypomethylation status was observed in aortic samples, before the clinical evidence of atherosclerosis, by supporting the hypothesis that in CVD, some aberrations in DNA
SC
methylation can be prelude to the manifestation of disease [39]. Several studies have focused on the effects
MA NU
in vivo and in vitro of demethylating agents as 5-aza-2′-deoxycytidine (5-AzaD). Recently, in LDL receptor knockout mice (Ldlr-/-), it has been shown that treatment with 5-AzaD reduces inflammation and downregulates genes such as tumor necrosis factor TNF, interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS), besides a lowering of macrophage migration that contributes to
ED
regulating chemotaxis genes [15,16]. The effects of 5-AzaD inhibitors have been evaluated in human primary fibroblast cell line and in a spontaneously hypertensive rat model, by showing an antifibrotic and
PT
antihypertrophic action due to the lowering of myocardial collagen levels and myocyte size in vivo and the reduction of collagen I, collagen III, and α-smooth muscle actin in vitro[23]. It has been established HF is
CE
associated with hypermethylation-induced downregulation of different genes, including sarcoplasmic reticular ATPase and Pitx2c [30,31], which are involved in calcium homeostasis and cardiac electrical
AC
activity [32]. Previously, it has been demonstrated that tissue inhibitor of matrix metalloprotease 4 (TIMP4) is downregulated in volume overload-induced HF [40]; besides, a fine regulation of TIMP4 and matrix metalloprotease 9 (MMP9) was found. TIPM4 is the endogenous inhibitor of MMP9 in the heart and its overexpression can confer cardioprotection [41]. Chaturvedi and colleagues found the presence of 7 methylated CpGi in the TIMP4 promoter during the progression of HF that along with the up-regulated levels of miR-122a led to its epigenetic silencing [33]. Human studies Interestingly, despite the inherent difficulty in obtaining heart tissue from living patients and easy deterioration of these tissues, Movassagh and colleagues demonstrated a differential DNA methylation profile in human cardiac tissue from end-stage cardiomyopathic hearts and normal control hearts [24]. Particularly, using both a methylated DNA immunoprecipitation-chip (MeDIP-chip) and a bisulfite-(BS)
ACCEPTED MANUSCRIPT PCR and high-throughput sequencing, they identified three angiogenesis-related genetic loci (AMOTL2, ARHGAP24 and PECAM1) differentially methylated in left ventricular (LV) tissue from male patients compared to controls [24]. By comparing LV tissues from failed and normal human hearts, differences of
RI P
T
methylation have been identified in promoter CpGi, in intragenic CpGi and in H3K36Me3 enriched regions [25]. It has been hypothesized that distinct epigenomic patterns control the expression of genes involved in the regulation of human end-stage cardiomyopathy [25]. The high density of gene mutations associated with
SC
the development of hypertrophic cardiomyopathy led to investigate the cardiac myosin binding protein C
MA NU
gene (MYBPC3), the gene most frequently associated with the onset of cardiomyopathies. In pooled samples of human placenta, the methylation level of the CpGi resulted to be gene-specific as demonstrated by comparison of the coding regions of MYBPC3 with those of the skeletal muscle isoform MYBPC2 [26]. Regarding dilated cardiomyopathy, Haas and colleagues observed distinct patterns of altered DNA methylation in four genes with still unclear function, such as Lymphocyte antigen 75 (LY75), Tyrosine
ED
kinase-type cell surface receptor HER3 (ERBB3), Homeobox B13 (HOXB13) and Adenosine receptor A2A
PT
(ADORA2A) in LV heart tissue in vitro [27]. Recently, a total of 1828 differentially methylated probes have been identified by comparing the methylomes between 18 left ventricles and 9 right ventricles, with
CE
hypermethylated regions in severely affected LV [28]. A global analysis of gene promoter DNA methylation, performed on failing and non-failing human LV, identified changes in promoters of genes associated with
AC
CVD and dilated cardiomyopathy [42]. Recently, genome-wide DNA methylation analysis confirmed a differential gene methylation pattern in HF patients compared to normal controls using blood leukocytes [43]. These results strengthen the importance of evaluating DNA methylation changes in CVD and support the therapeutic use of compounds that influence the activity of epigenetic modulators such as DNMTs. Indeed, epigenetic modifiers such as DNA methylation inhibitors could be used for treating cardiac hypertrophy-associated disorders [23].
Histone Modifications In vitro studies and animal models Several in vitro and animal studies suggested the involvement of histone modifications in the onset and progression of CVD. The function of HDAC1, HDAC2 and HDAC3 has been investigated in knockout mice
ACCEPTED MANUSCRIPT that showed cardiac arrhythmias, and dilated and hypertrophic cardiomyopathy [44,45]. Previously, rats with HF showed altered trimethylation on lysine 4 or 9 of histone H3 [46]. In an elegant in vivo experiment, Papait and colleagues analyzed cardiomyocytes isolated from the LV of mice subjected to transverse aortic
RI P
T
constriction (TAC) and showed different regions of the genome with changes of their histone profile. Indeed, H3K9ac, H3K4me9, H3K79me2 and H3K27me3 showed a differential distribution in hypertrophic heart [47].
SC
HDAC inhibitors (HDACi) possess a wide range of cytoprotective activities such as anti-inflammatory,
MA NU
antioxidant, anti-apoptotic, anti-fibrotic, and antihypertrophic properties, which are beneficial for the treatment of a variety of CVDs. In support of this hypothesis, preclinical studies demonstrated that HDACi, such as trichostatin A (TSA) and the novel MPT0E014 exert a beneficial effect on heart remodeling, thereby diminishing the emergence of HF [48, 49].
The inhibition of HDAC3, as well as the deletion of HDAC9, reduces atherogenic macrophage functions
ED
through an increased accumulation of total acetylated H3 and H3K9 at the promoters of ATP-binding
PT
cassette transporter (ABCA1), ABCG1, and peroxisome proliferator-activated receptor γ (PPARγ) [50]. However, clinical implications for the therapeutic use of histone modulating enzymes need to be further
CE
investigated [51].
In a model of cardiac hypertrophy, treatment with a selective HDACi, magnesium valproate, prevented
AC
cardiac hypertrophy, improved hemodynamic functions, and also increased survival rate [52]. Human studies
In humans, Lu et al. showed a downregulation of SIRT1, a member of a family of protein deacetylases, in hearts of patients with advanced HF [53]. In patients with abdominal aortic aneurysm, an increased expression of HDACs 1, 2, 4 and 7 was found in abdominal aorta samples compared with those from donors [54]. These finding are coherent with the recognition of the fundamental role of reversible protein acetylation in the pathological remodeling of the failed heart.
ACCEPTED MANUSCRIPT MiRNA In vitro studies and animal models Increasing evidence shows that the expression of specific mRNAs is strongly correlated with the regulation
RI P
T
of genes involved in onset and progression of CVD leading to HF.
The levels of miRNAs were found to vary along with the development and progression of CVD, modulate the inflammatory response and play also an anti-hypertrophic role (Table 1)
SC
Many miRNAs, such as miR-195 and miR-1, change their expression pattern during the development of
MA NU
cardiac hypertrophy consequent to pressure-overload and their deregulation is an underlying cause of cellular dysfunction and disease in mouse cardiac myocytes [55,56]. Moreover, in hearts of Smad4-deleted mice, TGF-β-regulated miR-27 plays a pivotal role in the development of cardiac hypertrophy and HF and miR27b resulted significantly over-expressed in hearts from transgenic mice with cardiac hypertrophy at 3 months of age [57]. MiR-214 was over-expressed in the presence of hypertrophic cardiomyopathy and
ED
regulated negatively EZH2, the major histone methyltransferase of Polycomb repressor complex [55,58].
PT
Kim and colleagues have lately shown that miR-185 was significantly down-regulated in myocardial cells of mice subjected to TAC, indicating its substantial anti-hypertrophic role in failed heart [59].
CE
Regarding statin administration in case of hypertrophic hearts, atorvastatin downregulated miR-22 in mice [60]. The functional role of miR-22 in cardiac remodeling in response to stress was reported both in cardiac
AC
hypertrophy and dilated cardiomyopathy. In particular, miR-22 expression was related with reorganization of the sarcomeres, increased size of cardiomyocytes and LV in mouse models [61]. Human studies
MiR-214 over-expression has been observed in hypertrophic cardiac tissue of patients with valvular diseases as well as in HF mouse model, and its inhibition with antisense oligonucleotides (antagomiRNAs), restored the EZH2 expression, thus protecting heart function [62,63]. Barsanti and colleagues observed 13 upregulated and 10 downregulated miRNAs when comparing end-stage HF patients undergoing transplantation with or without a previous LVAD support [64]. The expression level of 7 of these miRNAs correlated with the improvement of cardiac index; several target genes of these miRNAs belonged to the integrin signaling network involved in the remodeling process [64]. In addition, the levels of 186 miRNAs were measured in sera of chronic systolic HF patients respect to healthy controls,
ACCEPTED MANUSCRIPT demonstrating that elevated serum levels of miR-423-5p, miR-320a, miR-22, miR-92b correlate with severe clinical prognostic parameters, being involved in remodeling process following myocardial injury [65]. In a cohort of pediatric patients with dilated cardiomyopathy, several miRNAs resulted differently expressed
RI P
T
between children who recovered ventricular function and children who died or required HT. Therefore, the circulating miRNA profile of children with severe HF secondary to dilated cardiomyopathy could be a biomarker for outcome prediction [66]. MiR-155, previously shown to be a biomarker of dilated
SC
cardiomyopathy also in adult population, is implicated in cardiac disease and in the development of
MA NU
hypertrophy in vivo. Indeed, reduced levels of inflammation-associated miR-155 and SMC-associated miR145 were found in whole blood and plasma of CHD patients suggesting that the alteration of miR-155 levels is not limited to endothelial or vascular cells. Accordingly, CHD patients exhibit a miRNA-linked alteration in the inflammatory capacity of blood mononuclear cells [67,68].
In some circumstances, the correlation between biomarkers and miRNAs not only depends on the time of
ED
measurement, but also on the disease state as recently reported in patients with HF at different stage. Several
PT
miRNAs, identified in HF patients with a worse prognosis, seem to be linked to targets and pathways leading to cardiac disease, including cardiac remodeling/fibrosis, inflammation and angiogenesis [48,69]. As a
CE
whole, miRNAs are the more effective players in the modulation of key signaling elements and enzymes in
AC
the pathogenesis and progression of HF [70].
EPIGENETIC INSIGHTS IN HEART-TRANSPLANTED PATIENTS Although HT still represents the last chance for patients with end-stage HF, the outcome of the transplantation and, first of all, patients’ survival are closely related to several post-transplantation immune and non-immune complications such as infections, acute and chronic rejection, hypertension, kidney failure and malignancies [71]. Particularly, cardiac allograft vasculopathy (CAV), a manifestation of chronic injury, is commonly associated with graft deterioration and failure and the invasive allograft biopsy remains the main approach used for rejection surveillance. Over the years, several prognostic markers have been associated with the development of CAV, including the donor specific anti-HLA antibodies that develop despite the immunosuppressive therapies currently used in the management of transplanted patients. Among the non-immunological factors the most relevant are the
ACCEPTED MANUSCRIPT age of the donor, ischemia-reperfusion injury and cytomegalovirus infection [72]. The adoption of epigenetic mechanisms as non-invasive biomarkers for prediction and monitoring of CAV is actively investigated and their involvement in transplantation medicine needs further insight. Until now, few studies have focused on
RI P
T
the role of DNA methylation, histone modification and miRNA activity in controlling and monitoring the adverse post-transplantation events as well as the immunosuppressive therapies. Furthermore, an open
MA NU
that contribute to the graft outcome [73] (Fig. 1).
SC
question regards the contribute of both donor and recipient epigenetic signatures in the biological processes
DNA methylation
DNA methylation plays a pivotal role in the balanced immune response versus graft, by regulating the immune cells, but a direct link between this epigenetic mechanism and rejection is partially unknown. During immune response, DNA-methylation regulates T cell differentiation as well as instability and
ED
plasticity which need to be considered if a therapy based on regulatory T cell is carried out [74,75]. In this
PT
regard, FOXP3+Treg cells play an important role in the suppression of immune response and in the induction of tolerance in organ transplantation, although their contribution in favoring the tolerance or immune
CE
activation in human is still unclear. Indeed, intragraft FOXP3+ T cells have been associated with graft acceptance and better graft survival in both mice and humans, but higher FOXP3 mRNA expression in
AC
cardiac biopsies has also been associated with rejection. Likely, the expression of FOXP3 in different cell types could explain these results [76,77]. The preservation of self-tolerance has been further demonstrated in mice models, where two methylation modifiers, 5-AzaD and TSA, modulated the expression of FOXP3 thus promoting the conversion of naïve T cells into regulatory T cells [78] (Table 2).
Histone modification In Table 2 are described some evidence of histone modification involvement in post-transplantation period (Table 2).Therapeutic manipulation of FOXP3 acetylation through HDACi could favor the development and suppressive functions of Treg cells also in experimental models of transplant rejection [79,80].
ACCEPTED MANUSCRIPT The impact of epigenetic modifications on B cell function and its involvement in antibody-mediated rejection is still to be explored. HDACi, including SAHA, are under investigation as potential anti-rejection drugs. They synergize with tacrolimus in the prevention of rejection by promoting T effector cell apoptosis
RI P
T
and by favoring Treg. Moreover, SAHA treatment attenuates B cell-mediated rejection, prolongs graft survival and reduces serum levels of alloantibodies and IgG deposition [81,82]. Probably, SAHA prevents allograft rejection by balancing the Treg and Th17 ratio as well as by driving the balance towards Treg
SC
deviation [81]. An additional study in a mouse model of cardiac transplantation evaluated the impact of the
MA NU
HDACi, valproic acid, on proliferation, apoptosis, class switch recombination, differentiation and secretion of immunoglobulins on isolated murine B cells. Indeed, valproic acid reduced antibody generation significantly in a dose-dependent manner without altering B cell proliferation and apoptosis, thus suggesting a promising therapeutic role of this agent after organ transplantation [83]. The graft survival time could be further prolonged thanks to HDACi, such as TSA and the newly developed FR276457. This latter compound
ED
has been administered to a rat transplantation model alone or in combination with tacrolimus as
PT
immunosuppressive drugs, to improve overall survival time and to prevent the activation of T cells [84]. In humans, curcumin (a HAT inhibitor) in combination with cyclosporine A suppressed Th1 cytokines in ex
AC
MicroRNA
CE
vivo peripheral blood lymphocytes from kidney transplantation patients [85].
With the purpose of identifying new biomarkers, much attention has been focused on miRNAs due to their prominent role in the regulation of innate and adaptive immunity. MiRNAs regulate the expression of 30% of human genes, including those involved in cell proliferation, differentiation, DNA repair, metabolism, and apoptosis (Table 2). The expression profile of miRNAs is both cell/tissue/organ specific and disease-stage specific [86]. MiRNA levels are relatively stable unless cell injury or death occurs as after rejection and therefore miRNA profiling can be explored in different samples including biofluids. Several studies in animal models are still needed to translate the results into clinical setting. Recently, miR-223 was associated with rejection, differently from miR-146a, miR-15b, miR-23a, miR-27a, miR-451, miR-101 and miR-148a that were involved in tolerance process in a murine cardiac allograft. These miRNAs seem to be specific for
ACCEPTED MANUSCRIPT heart allograft because their expression pattern showed a different trend compared with hepatic allograft, perhaps for the different number of immune cells and for the tissue-specificity of miRNAs [87]. Most of the upregulated miRNAs are involved in inflammation, innate immune system and fibrosis, which
RI P
T
are all specific manifestations of rejection. MiRNA profiling of human endomyocardial biopsies of patients with acute cellular rejection and murine allograft, highlighted increased levels of miR-21, miR-142-3p, miR142-5p, miR-146a, miR-146b, miR-155, miR-222, miR-223 and miR-494, and a decreased level of miR-149-
SC
5p. Interestingly, miR-155, with a well-established role in inflammation and immunity by targeting IL-6 and
MA NU
SPl1, has been identified as the most upregulated miRNA after rejection [88]. Moreover, miR-155 can regulate Th1/Th17-related inflammation by ameliorating the tissue injury in murine chronic cardiac rejection [89]. Overall, these results suggest that miR-155 might be considered a responsive target for a novel therapeutic approach also in the post-transplantation period. The levels of certain circulating organ-specific miRNAs might be significantly upregulated during acute rejection, but significantly downregulated during
ED
chronic or late rejection [90].
PT
Increased levels of miR-21, together with cardiac fibrosis, were found in murine hearts after transplantation as well as in patients with cardiac rejection. In vitro, miR-21 was induced by IL-6 treatment and it activated a
CE
fibrotic gene programming by promoting monocyte-to-fibrocyte transition. In addition, inhibition of miR-21 in vivo successfully reduced fibrosis and fibrocyte accumulation in cardiac allografts [91]. MiRNA
AC
expression profiling in patients with acute biopsy-proven allograft rejection compared with control patients without rejection, both in heart allograft tissue and in serum, highlighted 7 miRNAs differentially expressed, namely miR-10a, miR-21, miR-31, miR-92a, miR-142-3p, miR-155, and miR-451. In particular, miR-10a, miR-31, miR-92a, and miR-155 strongly discriminated patients with allograft rejection from patients without rejection and showed a differential serological expression that correlated with their tissue expression. As reported, these four miRNAs are specific for inflammatory burdens in EC, inflammatory pathways, cardiomyocytes/interstitial cells and EC, respectively [92]. A cross-sectional study on 52 patients undergoing coronary angiography between 5 and 15 years after HT, showed that circulating levels of endotheliumenriched miR-21-5p, miR-92a-3p, miR-126-3p, and miR-126-5p were almost two-fold increased in patients with CAV than patients without CAV [93].
ACCEPTED MANUSCRIPT In the attempt to identify miRNAs as biomarkers for CAV monitoring, a microarray analysis reported miR34a, miR-98, miR-155, miR-204, miR-628-5p as differentially regulated in plasma of patients with CAV. In particular plasma levels of miR-628-5p and miR-155 were significantly increased and a miR628-5p value
RI P
T
above 1.336 predicted CAV with a sensitivity of 72% and a specificity of 83% [93].
These results support a differential expression of miRNAs not only at the tissue level, but also in serum and
SC
plasma, thus confirming their potential relevance as non-invasive biomarkers in HT rejection [92,94].
MA NU
CONCLUSIONS
Epigenetic mechanisms have a role in the control of the pathological processes occurring at cardiac level thus, their alteration often results in an impaired cardiac function. Several evidences point out the involvement of the different epigenetic modifications in the onset and progression of CVD leading to advanced HF. Concerning the post-transplantation period, few researches so far proved the direct
ED
involvement of epigenetics in the outcome of HT; accordingly, this field remains largely unexplored (Fig. 1).
PT
While waiting for a breakthrough in regenerative medicine to treat CVD [95], patients suitable for HT have a poor survival prognosis in the absence of any feasible alternative therapeutic treatment and the outcome of
CE
HT depends on several post-transplantation immune and non-immune complications. Major efforts are now directed toward the identification of new non-invasive and reliable biomarkers for the prediction of allograft
AC
rejection and for monitoring immunosuppressive therapy. Differently from other pathological conditions affecting the cardiovascular system [96], the current invasive endomyocardial biopsy represents still a diagnostic procedure not free from life-threatening complications for patients who develop heart insufficiency with reduced LV function. The mechanism of epigenetic regulation is not an independent actor, rather it is involved in a complex network of information exchange implicated at the transcriptional level. In this regard, much attention has been focused on miRNAs, because the levels of several circulating organ-specific miRNAs seem to be fluctuating over the time during acute or chronic rejection both in biological fluids and biopsies. The levels of circulating miRNAs are specific of organ/tissue transplanted, and influenced by organ/tissue mass as well; therefore, a panel of miRNAs rather than only one could be useful to cover the widest range of organ or tissue specificity for rejection diagnosis or for early prediction of the evolution of cardiomyopathy.
ACCEPTED MANUSCRIPT This approach is further confirmed by some ongoing clinical trials that analyze the expression of different miRNAs in heart transplanted patients; those miRNAs involved in the mechanisms of rejection might then be inhibited by the use of antagonists to stop or limit the damage (Table 3) [ https://clinicaltrials.gov ].
RI P
T
The multi-faceted field of transplantation medicine should be boosted to maximize the donor pool, enhance organ quality, overcome the life limiting complications of immunosuppression and infections, optimize organ allocation. However, a deeper understanding of the mechanisms of immunological tolerance and of the
SC
epigenetic tags implied in the balance of the immune response toward the graft, could allow the identification
MA NU
of patients at risk of rejection with respect to patients who develop tolerance. Epigenetic modifications have the undeniable advantage of being reversible by using pharmacological treatment. Indeed, in the clinical setting, epigenetic drugs are currently used in the treatment of several disease, mostly cancer, but preliminary data on animal models suggest that these drugs, in combination with immunosuppressive therapies, prolong graft survival in cardiac and renal recipients. It would be advisable to
ED
overcome the model of “one therapy/all patients” and further in-depth studies will evaluate if epigenetic
PT
mechanisms can be used for designing a more tailored therapy and as a valid strategy to modulate the balancing between tolerance and immunity in HT. Indeed, the opportunity to modulate pharmacologically
CE
epigenetic targets could allow to improve the immunosuppressive therapies towards the prevention and reduction of their side-effects.
AC
Although on one side it would be unwise to consider epigenetic promise as the panacea for all failed hearts, on the other side epigenetic mechanisms are emerging more and more convincingly as the right key for interpreting the still unexplained processes in the pre- and post-heart transplantation.
CONFLICT OF INTERESTS The authors declare that they have no conflict of interest concerning this article.
FUNDING This research did not receive any specific grant from funding agencies in the public, commercial, or not-forprofit sectors.
ACCEPTED MANUSCRIPT REFERENCES
[7] [8] [9] [10] [11] [12] [13]
[14] [15]
[16]
[17]
[18]
[19]
[20]
T
RI P
SC
MA NU
[6]
ED
[5]
PT
[3] [4]
CE
[2]
McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Böhm M, Dickstein K et al. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2012. Eur J Heart Fail 2012;14:803-69. Erratum in: Eur J Heart Fail 2013;15:361-2. Picascia A, Grimaldi V, Zullo A, Infante T, Maiello C, Crudele V, et al. Current concepts in histocompatibility during heart transplant. Exp Clin Transplant 2012;10:209-18. Papait R, Condorelli G. Epigenetics in heart failure. Ann NY Acad Sci 2010;1188:159-64. Napoli C, Crudele V, Soricelli A, Al-Omran M, Vitale N, Infante T, et al. Primary prevention of atherosclerosis: a clinical challenge for the reversal of epigenetic mechanisms? Circulation 2012;125:2363-73. Papait R, Greco C, Kunderfranco P, Latronico MV, Condorelli G. Epigenetics: a new mechanism of regulation of heart failure? Basic Res Cardiol 2013;108:361 Alvarez RM, Margulies KB. Epigenetic memory and cardiac cell therapy. J Am Coll Cardiol 2014;64:449-50. Raftopoulos L, Katsi V, Makris T, Tousoulis D, Stefanadis C, Kallikazaros I. Epigenetics, the missing link in hypertension. Life Sci 2015;129:22-6. Napoli C, Infante T, Casamassimi A. Maternal-foetal epigenetic interactions in the beginning of cardiovascular damage. Cardiovasc Res 2011;92:367-74. Cedar H, Bergman Y. Programming of DNA methylation patterns. Annu Rev Biochem 2012;81:97-117. Peserico A, Simone C. Physical and functional HAT/HDAC interplay regulates protein acetylation balance. J Biomed Biotechnol 2011;371832. Nishiguchi T, Imanishi T, Akasaka T. MicroRNAs and Cardiovascular Diseases. Biomed Res Int 2015;682857. Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003;425:415-9. Greco CM, Kunderfranco P, Rubino M, Larcher V, Carullo P, Anselmo A, et al. DNA hydroxymethylation controls cardiomyocyte gene expression in development and hypertrophy. Nat Commun 2016;7:12418. Napoli C, Grimaldi V, De Pascale MR, Sommese L, Infante T, Soricelli A. Novel epigenetic-based therapies useful in cardiovascular medicine. World J Cardiol 2016;8:211-9. Cao Q, Wang X, Jia L, Mondal AK, Diallo A, Hawkins GA, et al. Inhibiting DNA Methylation by 5-Aza-2'-deoxycytidine ameliorates atherosclerosis through suppressing macrophage inflammation. Endocrinology 2014;155:4925-38. Dunn J, Qiu H, Kim S, Jjingo D, Hoffman R, Kim CW, et al. Flow-dependent epigenetic DNA methylation regulates endothelial gene expression and atherosclerosis. J Clin Invest 2014;124:3187-99. Kumar A, Kumar S, Vikram A, Hoffman TA, Naqvi A, Lewarchik CM, et al. Histone and DNA methylation-mediated epigenetic downregulation of endothelial Kruppel-like factor 2 by lowdensity lipoprotein cholesterol. Arterioscler Thromb Vasc Biol 2013;33:1936-42. Kim YR, Kim CS, Naqvi A, Kumar A, Kumar S, Hoffman TA, et al. Epigenetic upregulation of p66shc mediates low-density lipoprotein cholesterol-induced endothelial cell dysfunction. Am J Physiol Heart Circ Physiol 2012;303:H189-96. Tikoo K, Patel G, Kumar S, Karpe PA, Sanghavi M, Malek V, et al. Tissue specific up regulation of ACE2 in rabbit model of atherosclerosis by atorvastatin: Role of epigenetic histone modifications, Biochem Pharmacol 2015;93:343-51. Wierda RJ, Rietveld IM, van Eggermond MC. Global histone H3 lysine 27 triple methylation levels are reduced in vessels with advanced atherosclerotic plaques. Life Sci 2015;129:3-9.
AC
[1]
ACCEPTED MANUSCRIPT
AC
CE
PT
ED
MA NU
SC
RI P
T
[21] Du F, Yu F, Wang Y, Hui Y, Carnevale K, Fu M, et al. MicroRNA-155 deficiency results in decreased macrophage inflammation and attenuated atherogenesis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2014;34:759-67. [22] Chen T, Huang Z, Wang L, Y. Wang, F. Wu, S. Meng et al. MicroRNA-125a-5p partly regulates the inflammatory response, lipid uptake, and ORP9 expression in oxLDL-stimulated monocyte/macrophages. Cardiovasc Res 2009;83:131-9. [23] Watson CJ, Horgan S, Neary R, Glezeva N, Tea I, Corrigan N, et al. Epigenetic Therapy for the Treatment of Hypertension-Induced Cardiac Hypertrophy and Fibrosis. J Cardiovasc Pharmacol Ther 2016;21:127-37. [24] Movassagh M, Choy MK, Goddard M, Bennett MR, Down TA, Foo RS. Differential DNA methylation correlates with differential expression of angiogenic factors in human heart failure. PLoS One 2010;5:e8564. [25] Movassagh M, Choy MK, Knowles DA, Cordeddu L, Haider S, Down T, et al. Distinct epigenomic features in end-stage failing human hearts. Circulation 2011;124:2411-22. [26] Meurs KM, Kuan M. Differential methylation of CpG sites in two isoforms of myosin binding protein C, an important hypertrophic cardiomyopathy gene. Environ Mol Mutagen 2011;52:161-4. [27] Haas J, Frese KS, Park YJ, Keller A, Vogel B, Lindroth AM, et al. Alterations in cardiac DNA methylation in human dilated cardiomyopathy. EMBO Mol Med 2013;5:413-29. [28] Jo BS, Koh IU, Bae JB, Yu HY, Jeon ES, Lee HY, et al. Methylome analysis reveals alterations in DNA methylation in the regulatory regions of left ventricle development genes in human dilated cardiomyopathy. Genomics 2016;108:84-92. [29] Zhang QJ, Chen HZ, Wang L, Liu DP, Hill JA, Liu ZP. The histone trimethyllysine demethylase JMJD2A promotes cardiac hypertrophy in response to hypertrophic stimuli in mice. J Clin Invest 2011;121:2447-56. [30] Kao YH, Cheng CC, Chen YC, Chung CC, Lee TI, Chen SA, et al. Hydralazine induced promoter demethylation enhances sarcoplasmic reticulum Ca2+ -ATPase and calcium homeostasis in cardiac myocytes. Lab Invest 2011;91:1291-7. [31] Kao YH, Chen YC, Chung CC. Heart failure and angiotensin II modulate atrial Pitx2c promotor methylation. Clin Exp Pharmacol Physiol 2013;40:379-84. [32] Kao YH, Lien GS, Chao TF, Chen YJ. DNA methylation inhibition: a novel therapeutic strategy for heart failure. Int J Cardiol 2014;176:232-3. [33] Chaturvedi P, Tyagi SC. Epigenetic silencing of TIMP4 in heart failure. J Cell Mol Med 2016;20:2089-2101. [34] Lkhagva B, Lin YK, Kao YH, Chazo TF, Chung CC, Chen SA, et al. Novel Histone Deacetylase Inhibitor Modulates Cardiac Peroxisome Proliferator-Activated Receptors and Inflammatory Cytokines in Heart Failure. Pharmacology 2015;96:184-91. [35] Nural-Guvener HF, Zakharova L, Nimlos J, Popovic S, Mastroeni D, Gaballa MA. HDAC class I inhibitor, Mocetinostat, reverses cardiac fibrosis in heart failure and diminishes CD90+ cardiac myofibroblast activation. Fibrogenesis Tissue Repair 2014;7:10. [36] Su M, Chen Z, Wang C, Song L, Zou Y, Zhang L, et al. Wang, Cardiac-Specific Overexpression of miR-222 Induces Heart Failure and Inhibits Autophagy in Mice. Cell Physiol Biochem 2016;39:1503-11. [37] Seok HY, Chen J, Kataoka M, Huang ZP, Ding J, Yan J, et al. Loss of microRNA-155 protects the heart from pathological cardiac hypertrophy. Circ Res 2014;114:1585-95. [38] Grimaldi V, Mancini FP, Casamassimi A, Al-Omran M, Zullo A, Infante T, et al. Potential benefits of cell therapy in coronary heart disease. J Cardiol 2013;62:267-76. [39] Lund G, Andersson L, Lauria M, Lindholm M, Fraga MF, Villar-Garea A, et al. DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein E. J Biol Chem 2004 9;279:29147-54.
ACCEPTED MANUSCRIPT
AC
CE
PT
ED
MA NU
SC
RI P
T
[40] Cox MJ, Hawkins UA, Hoit BD, Tyagi SC. Attenuation of oxidative stress and remodeling by cardiac inhibitor of metalloproteinase protein transfer. Circulation 2004;109:2123-8. [41] Zavadzkas JA, Stroud RE, Bouges S, Mukherjee R, Jones JR, Patel RK, et al. Targeted overexpression of tissue inhibitor of matrix metalloproteinase-4 modifies postmyocardial infarction remodeling in mice. Circ Res 2014;114:1435-45. [42] Koczor CA, Lee EK, Torres RA, Boyd A, Vega JD, Uppal K, et al. Detection of differentially methylated gene promoters in failing and nonfailing human left ventricle myocardium using computation analysis. Physiol Genomics 2013; 45:597-605. [43] Li B, Feng ZH, Sun H, Zhao ZH, Yang SB, Yang P. The blood genome-wide DNA methylation analysis reveals novel epigenetic changes in human heart failure. Eur Rev Med Pharmacol Sci 2017;21:1828-6. [44] Montgomery RL, Davis CA, Potthoff MJ, Haberland M, Fielitz J, Qi X, et al. Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility. Genes Dev 2007;21:1790-1802. [45] Montgomery RL, Potthoff MJ, Haberland M, Qi X, Matsuzaki S, Humphries KM, et al. Maintenance of cardiac energy metabolism by histone deacetylase 3 in mice. J Clin Invest 2008;118:3588-97. [46] Kaneda R, Takada S, Yamashita Y, Y.L. Choi, M. Nonaka-Sarukawa, M. Soda et al. Genomewide histone methylation profile for heart failure, Genes Cells 2009;14:69-77. [47] Papait R, Cattaneo P, Kunderfranco P, Greco C, Carullo P, Guffanti A, et al. Genome-wide analysis of histone marks identifying an epigenetic signature of promoters and enhancers underlying cardiac hypertrophy. Proc Natl Acad Sci U S A 2013;110:20164-9. [48] Grimaldi V, De Pascale MR, Zullo A, Soricelli A, Infante T, Mancini FP, et al. Evidence of epigenetic tags in cardiac fibrosis. J Cardiol 2017;69:401-8. [49] Kao YH, Liou JP, Chung CC, Lien GS, Kuo CC, Chen SA, et al. Histone deacetylase inhibition improved cardiac functions with direct antifibrotic activity in heart failure. Int J Cardiol 2013;168:4178-83. [50] Cao Q, Rong S, Repa JJ, St Clair R, Parks JS, Mishra N. Histone deacetylase 9 represses cholesterol efflux and alternatively activated macrophages in atherosclerosis development. Arterioscler Thromb Vasc Biol 2014;34:1871-9. [51] Chistiakov DA, Orekhov AN, Bobryshev YV. Treatment of cardiovascular pathology with epigenetically active agents: Focus on natural and synthetic inhibitors of DNA methylation and histone deacetylation. Int J Cardiol 2017;227:66-82. [52] Raghunathan S, Goyal RK, Patel BM. Selective inhibition of HDAC2 by magnesium valproate attenuates cardiac hypertrophy. Can J Physiol Pharmacol 2017;95:260-7. [53] Lu TM, Tsai JY, Chen YC, Huang CY, Hsu HL, Weng CF, et al. Downregulation of Sirt1 as aging change in advanced heart failure. J Biomed Sci 2014;21:57 [54] Galán M, Varona S, Orriols M, Rodríguez JA, Aguiló S, Dilmé J, et al. Induction of histone deacetylases (HDACs) in human abdominal aortic aneurysm: therapeutic potential of HDAC inhibitors. Dis Model Mech 2016 1;9:541-52. [55] van Rooij E, Sutherland LB, Liu N, Williams AH, McAnally J, Gerard RA, et al. A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci U S A 2006;103:18255-60. [56] Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res 2007;100:416-24. [57] Wang J, Song Y, Zhang Y, Xiao H, Sun Q, Hou N, et al. Cardiomyocyte overexpression of miR27b induces cardiac hypertrophy and dysfunction in mice. Cell Res 2012;22:516-27. [58] Yang T, Zhang GF, Chen XF, Gu HH, Fu SZ, Xu HF, et al. MicroRNA-214 provokes cardiac hypertrophy via repression of EZH2. Biochem Biophys Res Commun 2013;436:578-84.
ACCEPTED MANUSCRIPT
AC
CE
PT
ED
MA NU
SC
RI P
T
[59] Kim JO, Song DW, Kwon EJ, Hong SE, Song HK, Min CK, et al. MiR-185 plays an antihypertrophic role in the heart via multiple targets in the calcium-signaling pathways. PLoS One 2015;10:e0122509. [60] Tu Y, Wan L, Bu L, Zhao D, Dong D, Huang T, et al. MicroRNA-22 downregulation by atorvastatin in a mouse model of cardiac hypertrophy: a new mechanism for antihypertrophic intervention. Cell Physiol Biochem 2013;31:997-1008. [61] Huang ZP, Chen J, Seok HY, Zhang Z, Kataoka M, Hu X, et al. MicroRNA-22 regulates cardiac hypertrophy and remodeling in response to stress. Circ Res 2013;112:1234-43. [62] Thum T, Condorelli G. Long noncoding RNAs and microRNAs in cardiovascular pathophysiology. Circ Res 2015;116:751-62. [63] Yang T, Gu H, Chen X, Fu S, Wang C, Xu H, et al. Cardiac hypertrophy and dysfunction induced by overexpression of miR-214 in vivo. J Surg Res 2014;192:317-25. [64] Barsanti C, Trivella MG, D'Aurizio R, El Baroudi M, Baumgart M, Groth M, et al. Differential regulation of microRNAs in end-stage failing hearts is associated with left ventricular assist device unloading. Biomed Res Int 2015;2015:592512. [65] Goren Y, Kushnir M, Zafrir B, Tabak S, Lewis BS, Amir O. Serum levels of microRNAs in patients with heart failure. Eur J Heart Fail 2012;14:147-54. [66] Miyamoto SD, Karimpour-Fard A, Peterson V, Auerbach SR, Stenmark KR, Stauffer BL, et al. Circulating microRNA as a biomarker for recovery in pediatric dilated cardiomyopathy. J Heart Lung Transplant 2015;34:724-33. [67] Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, et al. Circulating microRNAs in patients with coronary artery disease. Circulation Research 2010;107:677-84. [68] Bronze-da-Rocha E. MicroRNAs expression profiles in cardiovascular diseases. Biomed Res Int 2014;2014:985408. [69] Vegter EL, Schmitter D, Hagemeijer Y, Ovchinnikova ES, van der Harst P, Teerlink JR, et al. Use of biomarkers to establish potential role and function of circulating microRNAs in acute heart failure. Int J Cardiol 2016;224:231-9. [70] Zhang X, Schulze PC. MicroRNAs in heart failure: Non-coding regulators of metabolic function. Biochim Biophys Acta 2016;1862:2276-87. [71] Zuckermann A, Barten MJ. Surgical wound complications after heart transplantation. Transpl Int 2011; 24: 627-36. [72] Picascia A, Grimaldi V, Casamassimi A, De Pascale MR, Schiano C, Napoli C. Human leukocyte antigens and alloimmunization in heart transplantation: an open debate. J Cardiovasc Transl Res 2014;7:664-75. [73] Picascia A, Grimaldi V, Napoli C. From HLA typing to anti-HLA antibody detection and beyond: The road ahead. Transplant Rev (Orlando) 2016;30:187-94. [74] Suarez-Alvarez B, Rodriguez RM, Fraga MF, López-Larrea C. DNA methylation: a promising landscape for immune system-related diseases. Trends Genet 2012;28:506-14. [75] Youngblood B, Hale JS, Ahmed R. T-cell memory differentiation: insights from transcriptional signatures and epigenetics. Immunology 2013;139:277-84. [76] Dijke IE, Velthuis JH, Caliskan K, Korevaar SS, Maat AP, Zondervan PE, et al. Intragraft FOXP3 mRNA expression reflects antidonor immune reactivity in cardiac allograft patients. Transplantation 2007;83:1477-84. [77] Waldmann H, Adams E, Fairchild P, Cobbold S. Regulation and privilege in transplantation tolerance. J Clin Immunol 2008;28:716-25. [78] Moon C, Kim SH, Park KS, B.K. Choi, H.S. Lee, J.B. Park, et al. Use of epigenetic modification to induce FOXP3 expression in naive T cells. Transplant Proc 2009;41:1848-54. [79] de Zoeten EF, Wang L, Butler K, U.H. Beier, T. Akimova, H. Sai, et al. Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp31 T-regulatory cells. Mol Cell Biol 2011;31:2066-78.
ACCEPTED MANUSCRIPT
AC
CE
PT
ED
MA NU
SC
RI P
T
[80] Beier UH, Wang L, Han R, Akimova T, Liu Y, Hancock WW. Histone deacetylases 6 and 9 and sirtuin-1 control Foxp3+ regulatory T cell function through shared and isoform-specific mechanisms. Sci Signal 2012;5: ra45. [81] Zhang X, Han S, Kang Y, Guo M, Hong S, Liu F, et al. SAHA, an HDAC inhibitor, synergizes with tacrolimus to prevent murine cardiac allograft rejection, Cell. Mol. Immunol 2012;9:390-8. [82] Zhang X, Guo M, Kang Y, Liu F, Zheng X, Han S, et al. SAHA, an HDAC inhibitor, attenuates antibody-mediated allograft rejection. Transplantation 2013;96:529-37. [83] Ye J, Li J, Zhou M, Xia R, Liu R, Yu L. Modulation of Donor-Specific Antibody Production After Organ Transplantation by Valproic Acid: A Histone Deacetylase Inhibitor. Transplantation 2016;100:2342-51. [84] Kinugasa F, Yamada T, Noto T, Matsuoka H, Mori H, Sudo Y, et al. Effect of a new immunosuppressant histon deacetylase (HDAC) inhibitor FR276457 in a rat cardiac transplant model. Biol Pharm Bull 2008;31:1723-6. [85] Bharti AC, Panigrahi A, Sharma PK, Gupta N, Kumar R, Shukla S, et al. Clinical relevance of curcumin-induced immunosuppression in living-related donor renal transplantation: an in vitro analysis. Exp Clin Transplant 2010;8:161-71. [86] Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004;116:281-97. [87] Fujino M, Zhu P, Cai S, Nishio Y, Zhuang J, Li XK. MicroRNAs Involved in Acute Rejection and Tolerance in Murine Cardiac Allografts. Exp Clin Transplant 2016;14:424-30. [88] Van Aelst LN, Summer G, Li S, Gupta SK, Heggermont W, De Vusser K, et al. RNA Profiling in Human and Murine Transplanted Hearts: Identification and Validation of Therapeutic Targets for Acute Cardiac and Renal Allograft Rejection. Am J Transplant 2016;16:99-110. [89] Zhang A, Wang K, Zhou C, Gan Z, Ma D, Ye P, et al. Knockout of microRNA-155 ameliorates the Th1/Th17 immune response and tissue injury in chronic rejection. J Heart Lung Transplant 2017;36:175-84. [90] Wilflingseder J, Regele H, Perco P, Kainz A, Soleiman A, Mühlbacher F, et al. miRNA profiling discriminates types of rejection and injury in human renal allografts. Transplantation 2013;95:83541. [91] Gupta SK, Itagaki R, Zheng X, Batkai S, Thum S, Ahmad F, et al. miR-21 promotes fibrosis in an acute cardiac allograft transplantation model. Cardiovasc Res 2016;110:215-26. [92] Duong Van Huyen JP, Tible M, Gay A, Guillemain R, Aubert O, Varnous S, et al. MicroRNAs as non-invasive biomarkers of heart transplant rejection. Eur Heart J 2014;35:3194-202. [93] Neumann A, Napp LC, Kleeberger JA, Benecke N, Pfanne A, Haverich A, et al. MicroRNA 6285p as a novel biomarker for cardiac allograft vasculopathy. Transplantation 2017;101:e26. [94] Singh N, Heggermont W, Fieuws S, Vanhaecke J, Van Cleemput J, De Geest B. Endotheliumenriched microRNAs as diagnostic biomarkers for cardiac allograft vasculopathy. J Heart Lung Transplant 2015;34:1376-84. [95] Sommese L, Zullo A, Schiano C, Mancini FP, Napoli C. Possible muscle repair in the human cardiovascular system. Stem Cell Rev 2017;13:170-91. [96] Grimaldi V, Schiano C, Casamassimi A, Zullo A, Soricelli A, Mancini FP et al. Imaging techniques to evaluate cell therapy in peripheral artery disease: state of the art and clinical trials. Clin Physiol Funct Imaging 2016;36:165-78.
ACCEPTED MANUSCRIPT Legend to Fig. 1 Epigenetic mechanisms could contribute to establishing innovative diagnostic and predictive biomarkers, as
T
well as ground-breaking therapies for heart failure and heart transplantation rejection. In patients with failed
RI P
heart, for example histone methylation and miRNAs could represent a useful tool for diagnostic purpose. Furthermore, in heart transplanted patients epigenetic tags might also contribute to the balance between
AC
CE
PT
ED
MA NU
SC
rejection and tolerance by allowing a tailored therapeutic approach.
ACCEPTED MANUSCRIPT Table 1. Some evidence of epigenetic involvement in cardiovascular disease
Epigenetic Mechanism
Study Mo dels
Target/Effects
Ref
T
Cardiac Condition
15 AM
SC
RI P
5-aza-dC treatment downregulated expression of genes involved in inflammation and chemotaxis by attenuating macrophage migration and adhesion to endothelial cells and reduced macrophage infiltration into atherosclerotic plaques
DNA and histone methylation of KLF2dependent genes by LDL such as thrombomodulin, eNOS and PAI by leading to endothelial activation and dysfunction
17
MA NU
16
hypomethylation of two CpG dinucleotides and acetylation of histone 3 by LDL in the human increase in expression of endothelial intercellular adhesion molecule-1 (ICAM1) and decrease in expression of thrombomodulin (TM) by promoting proadhesive and procoagulant features
HC
ED PT CE
histone modification
AC
Atherosclerosis and Coronary Heart Disease
methylation
miRNAs
Cardiomyopathies
hypermethylation of 11 mechanosensitive genes such as HoxA5 and Klf3
methylation
18
Atorvastatin up-regulate ACE2 via epigenetic histone modifications
AM
19
Reduced global trimethylation levels of lysine 27 on histone H3 (H3K27me3) in vessels of advanced atherosclerotic plaques that could reflect a phenotype switching of SMC vessels
HT
20
miR-155 reduce the atherogenesis through a decrease of macrophage inflammation and an enhanced macrophage cholesterol efflux
AM
21
miR-125a mediates lipid uptake and decreases the inflammatory response through the lowering of several cytokines including IL-2, IL-6, TNF- and providing a posttranscriptional regulation of the proinflammatory response
HC
22
Antifibrotic and antihypertrofic action of methylation inhibitor due to the reduction of collagen I, collagen III, and α-smooth muscle
HC/AM
23
Differential DNA methylation profiles in promoter CpGi, in intragenic
HT
24,25
ACCEPTED MANUSCRIPT CpGi and in H3K36Me3 enriched regions, especially in three angiogenesis-related genetic loci (AMOTL2, ARHGAP24 and PECAM1) Differential methylation of CpG sites in two isoforms of myosin binding protein C
RI P
T
26
Distinct patterns of altered DNA methylation in four genes whit unclear function like Lymphocyte antigen 75 (LY75), Tyrosine kinase-type cell surface receptor HER3 (ERBB3), Homeobox B13 (HOXB13) and Adenosine receptor A2A (ADORA2A)
MA NU
SC
27
Alterations in DNA methylation the cisregulatory regions of cardiac development genes involved in ventricular development (e.g., TBX5 and HAND1)
AC
HF
CE
PT
methylation
Histone trimethyl-lysine demethylase JMJD2A promotes cardiac hypertrophy
AM
ED
histone modification
28
histone modification
miRNAs
29
Hypermethylation of genes including sarcoplasmic reticular ATPase and Pitx2c, involved in calcium homeostasis and cardiac electrical activity
30,31,3 2
Different methylated 7 CpG islands in the TIMP4 promoter
33
Higher levels of HDAC1, HDAC2, HDAC3, HDAC4 and HDAC6 whose inhibition improves cardiac function by modulating the cardiac peroxisome proliferatoractivated receptors and inflammatory cytokines
34 AM/cells
Differential expression patterns of HDAC1 and 2 whose inhibition reverses interstitial fibrosis
35
Cardiac-specific overexpression of miR222 induces heart failure and inhibits autophagy in mice
36
miR-155 as an inducer of pathological cardiomyocyte hypertrophy and suggest that inhibition of endogenous miR-155 might have clinical potential to suppress cardiac hypertrophy and heart failure
37
AM: animal model; HC: human cells; HT: human tissue
ACCEPTED MANUSCRIPT
Table 2. In vivo and in vitro epigenetic modifications involved in transplantation tolerance and rejection.
SOURCE
EFFECT/ OUTCOME
REFERENCES
DNA-methylation inhibition (5AzaD)
Mouse splenic naïve T cells
Increased FOXP3 expression that promote the conversion of naïve T cells to regulatory T cells
[78]
SC
RI P
T
EPIGENETIC SIGNATURE
MA NU
HDAC inhibition (TSA)
Mouse cardiac transplantation model
Modulation of Treg suppressive activity and HSP90 acetylation
[79]
HDAC6 inhibition (Tubastatin and ACY-738)
Mouse spleens and somatic lymph nodes
Deacethylation of FOXP3 and improvement of Treg suppressive function
[80]
Mouse cardiac transplantation model
Modulation of inflammatory cytokines and enhancement of Treg function
[81]
Isolated mouse B cells
Immunosuppression of splenic B cells
[82]
HDAC inhibition (VPA)
Mouse cardiac transplantation model
Inhibition of B cells and reduced antibody generation
[83]
HDAC inhibition (FR276457)
Rat
heterotopic cardiac transplantation model
Immunosuppressive effect on T cells proliferation
[84]
MiRNAs
Mouse cardiac transplantation
up-regulated miR-223 associated with rejection and down-regulated
[87]
ED
HDAC6 inhibition
PT
HDAC9 inhibition
AC
HDAC inhibition (SAHA)
CE
Sirt1 inhibition (EX-527)
ACCEPTED MANUSCRIPT model
miR-146a, -15b, -23a, -27a, -34a, -451, -101a, -101b, and -148a associated with tolerance
up-regulated miR-21, -142-3p, -1425p, -146a, -146b, -155, -222, 223, -494 and down-regulated miR-149-5p in rejection
[88]
Mouse cardiac transplantation model
miR-155-/- resistance to rejection and weakened Th17 cells mediated inflammation
[89]
ED
PT
RI P
SC
MA NU
Human EMB and serum
Plasma from cardiac transplanted patients
T
Human and mouse EMB
differential expression of miR-10a, 31, -92a, and -155 in rejection
[92]
miR-34a, miR-98, miR-155, miR-204, miR-628-5p differentially regulated in patients with CAV
[93]
Increased levels of miR-210-5p, -92a3p, -126-3p, and -126-5p in patients with CAV
[94]
CE
5AzaD, demethylating agent 5-aza-2′-deoxycytidine; CAV, cardiac allograft vasculopathy; EMB, endomyocardial biopsies; FOXP3, forkhead box P3; HDAC, histone deacetilase; MiRNAs, micro-RNA; SAHA,
AC
Suberoylanilide hydroxamic acid; Sirt1, sirtuin-1; TSA, trichostatin A; VPA, Valproic acid.
ACCEPTED MANUSCRIPT Table 3. Clinical ongoing trials on miRNA expression in heart-transplanted patients.
Status
Study Type
Outcome Measures
NCT02672683
Not
Observational
Circulating miRNAs measurement in patients with rejection
No
SC
yet recrui ting
RI P
T
Number
of enroll ed patien ts
400 (estim ated)
Completed
Randomized
Plasma miR-133b and miR-208a evaluation
60
NCT01848301
Active, not recrui ting
Interventional
Expression of miRNAs and relationship with endothelial function and intimal thickness
12
NCT02602834
Completed
Changes in miRNA levels with interval training compared to moderate training (1 week)
19
NCT03076580
Recruiting
microRNA-seq to determine the correlation of echocardiographic parameters of systolic and diastolic functional entry with circulating molecules (plasma specimen)
2000 (estim ated)
ED
MA NU
NCT02149316
AC
CE
PT
Interventional
Observational
PT
ED
MA NU
SC
RI P
T
ACCEPTED MANUSCRIPT
AC
CE
Figure 1