Next Article in Journal
N-Glycoprofiling of SLC35A2-CDG: Patient with a Novel Hemizygous Variant
Next Article in Special Issue
Cholesterol-Related lncRNAs as Response Predictors of Atorvastatin Treatment in Chilean Hypercholesterolemic Patients: A Pilot Study
Previous Article in Journal
Association of Antihistamine Use with Increased Risk of Esophageal Squamous Cell Carcinoma: A Nationwide, Long-Term Follow-Up Study Using Propensity Score Matching
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure

College of Science, Technology, Engineering, Arts and Mathematics, Alvernia University, Reading, PA 19607, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(2), 579; https://doi.org/10.3390/biomedicines11020579
Submission received: 24 January 2023 / Revised: 10 February 2023 / Accepted: 13 February 2023 / Published: 16 February 2023

Abstract

:
Cardiovascular diseases are a major cause of death globally, with no cure to date. Many interventions have been studied and suggested, of which epigenetics and chromatin remodeling have been the most promising. Over the last decade, major advancements have been made in the field of chromatin remodeling, particularly for the treatment of heart failure, because of innovations in bioinformatics and gene therapy. Specifically, understanding changes to the chromatin architecture have been shown to alter cardiac disease progression via variations in genomic sequencing, targeting cardiac genes, using RNA molecules, and utilizing chromatin remodeler complexes. By understanding these chromatin remodeling mechanisms in an injured heart, treatments for heart failure have been suggested through individualized pharmaceutical interventions as well as biomarkers for major disease states. By understanding the current roles of chromatin remodeling in heart failure, a potential therapeutic approach may be discovered in the future.

Graphical Abstract

1. Introduction

Cardiovascular diseases (CVDs) have been the leading cause of death worldwide for many years [1,2]. It has been estimated that around 17.9 million lives are lost annually due to CVDs across the globe [2]. Currently, there are no cures for any type of CVDs; there are only preventative measures to alleviate risk factors and in extreme cases perform heart transplants [3,4]. CVDs include a multitude of pathological conditions, including atherosclerosis, ischemic heart disease, stroke, and heart failure, to name a few [5]. For this review, heart failure (HF) will be the focus. HF occurs due to structural or functional stress in the heart which no longer allows the heart to properly pump blood and oxygen to designated areas of the body [6,7,8]. This is often manifested in decreased cardiac output and increased internal cardiac pressure [7]. Annually, there are over 64 million cases of HF, accounting for over 346 billion US dollars, and these numbers are projected to rise [9]. It is predicted that the rate of HF will increase by 50% in low and middle sociodemographic regions by 2030 [9]. With no cure and increasing concern across the globe, there is a dire need to understand and treat this devastating disease.
Because of the lack of a cure for HF, many interventions have been utilized to attenuate risk factors. There is an assembly of drug-based interventions that have proven to be instrumental in treating the risk factors associated with HF, one of the most significant being chronic hypertension [10]. Heart transplants do occur, but not as frequently as they are needed [11]. To truly cure an injured heart, cardiac stem cells have been suggested and are currently being studied [12]. However, they have not been as successful as originally proposed due to engraftment issues and detrimental immune responses [13]. Alternatively, cardiac remodeling via epigenetic regulation has been studied and has had promising outcomes [14]. In recent research, the combination of epigenetic regulated chromatin remodeling methods with the techniques, advancements, and theories of stem cell regulation have been groundbreaking for the field of cardiac biology [15,16].
The role of chromatin remodeling in HF is not a new idea, partially due to the overwhelming application of epigenetics in cardiac biology [14,17,18,19]. Generally, it is believed that when epigenetic modifications are altered there is a progression or suppression of the cardiac disease state [20]. Normally, eukaryotic DNA is tightly condensed in chromatin, which is compacted around histones that form a nucleosome [21]. These histones have all been identified (H1, H2A, H2B, H3, H4) and are well understood in cardiac biology [22,23,24]. However, changes to these histones and the chromatin are less understood. Modifications such as a methylation and phosphorylation can be added to or removed from these structures, altering the chromatin architecture as well as the recruitment of complexes that can alter the overall chromatin structure [25]. These alterations dictate whether the DNA becomes more or less tightly condensed. The more compacted the DNA, the less available the genes, meaning less gene expression or transcription. If the chromatin is less condensed, the DNA is open and therefore the genes are more accessible and can allow for more gene expression and transcription [25]. Many of these mechanisms have been connected to the progression or suppression of genes involved in HF [17]. Specifically, chromatin remodeling has been the highlight of many investigations involving the development of the heart [23,24] as well as the disease state [25]. Chromatin remodeling is of specific importance because the architecture and rearrangement of the chromatin can dictate more than just one epigenetic site or change. Chromatin remodeling affects the accessibility of multiple transcription sites and the overall availability of the chromatin [26,27,28].
Targeting the changes in chromatin architecture or chromatin remodeling has been of interest over the past few years. With promising strategies, potential clinical trials, and the advancement of therapeutic approaches, chromatin remodeling in HF is a vital method for understanding this devasting disease to find a therapeutic approach to treating HF as depicted in Figure 1. The goal of this review is to summarize the most recent advancement in chromatin remodeling in the progression and treatment of HF.

2. Current Strategies for Detecting Chromatin Remodeling in Heart Failure

With advancements in bioinformatics as well as gene therapy technology, the ability to study chromatin as a clinical cardiac therapeutic has greatly increased over the last decade [29,30,31,32,33]. Because chromatin is often tightly compacted and densely organized in the nucleus of cells, it can be difficult to isolate [34]. Importantly, variations to the chromatin architecture are a three-dimensional, multifaceted challenge for researchers [18]. To truly find a therapeutic approach, it is important to know the current challenges and new advancements in studying chromatin remodeling in the heart. Currently, there are standard approaches to studying changes in chromatin remodeling and gene expression due to changes in epigenetic profiles [35,36,37]. Many of these approaches include a combination of genomics, RNA molecules, cardiac gene targets, and chromatin remodelers. Advancements in bioinformatics and gene therapy have proven to be pivotal in understanding pathological signaling in cardiac disease states; however, this can prove challenging and too broad for a true therapeutic approach. Additionally, targeting only a specific change in selective gene expression can be difficult and can cause other adverse downstream effects as well [38]. However, a combination of all of these advancements could provide information to build a comprehensive understanding of chromatin remodeling during HF.
A more recent and promising strategy is using small molecules to target chromatin remodeling in the heart. By using chromatin remodeling enzymes, histone modifiers, chromatin regulatory complexes, and even DNA modifiers, the changes in chromatin architecture can be directly studied. This has been shown with the Switch/Sucrose-Nonfermentable (SWI/SNF) chromatin remodeling complex in many oncological studies, but it could also prove valuable in the heart [39]. Similarly, small molecules such as long noncoding RNAs (lncRNAs) have been used to assess chromatin remodeling [40]. The lncRNAs have been directly associated with structural changes in the chromatin in many disease models, including during pathological challenges to the heart [28,41]. One of the biggest advancements for understanding chromatin remodeling during HF is through genetic profiling approaches via sequencing and bioinformatic-based analysis. Using ATAC sequencing, RNA sequencing, and ChIP sequencing has led to major advancements in understanding epigenetic changes during HF. Specifically, for chromatin remodeling, combining these approaches in a novel “multiomic” approach has led to major discoveries that will be addressed in this review.

3. Therapeutic Approaches Using Chromatin Remodeling in Heart Failure

3.1. Genomics and Bioinformatics

The use of genomics to understand chromatin alterations and all the downstream changes has become a steadfast approach to most clinical applications of HF research. Many genomics-based research techniques are incorporated into most current studies along with other techniques, as this review will highlight. It is important to emphasize some of the most influential sequencing techniques that have made an impact on the field of chromatin remodeling in cardiac biology. One of the main methods to study chromatin remodeling through genomics is through the Assay for Transposase Accessible Chromatin (ATAC) sequencing. This ATAC sequencing data allows researchers to understand chromatin remodeling, chromatin accessibility, and epigenetic profiles [37]. Through this sequencing method, studies have found pivotal transcriptional and chromatin accessibility differences in HF [42]. For example, one study used ATAC sequencing to understand trans-aortic constriction in mice and found predictors of chromatin structural changes that led to a pivotal understanding of gene expression changes during disease progression [43]. Another study used ATAC sequencing to analyze different genes found in hypoxia-induced stress associated with HF and found major changes in the chromatin’s accessibility during stress [44]. ATAC sequencing is often paired with other genomic techniques or gene expression data.
Many of the widely used techniques include ATAC sequencing, RNA sequencing, miRNA sequencing, and DNA sequencing. All of these are not new techniques; however, how the data is used and interpreted is constantly changing. For example, one study used the multiomic approach, meaning they used single-cell RNA sequencing (scRNA seq), single-cell ATAC (scATAC seq), bulk ATAC sequencing, and miRNA sequencing to study HF in murine models [45]. From these immense data sets, the authors found miRNA expression differences that aligned with chromatin accessibility profiles in mice with HF that were not seen in the control mice. All of this gave a new understanding of potential targets and mechanisms in the progression of HF [45]. Another study used the multiomic approach to map changes in cardiomyocytes during differentiation and found a specific connection to the ACTN2 gene that was not known previously [46]. Using DNA sequencing, a study performed genome-wide chromatin conformation capture (Hi-C) to study adult cardiomyocytes in HF [47]. This study focused on the loss of a chromatin structural protein, CTCF. The loss of CTCF led to increased HF in mice and structural differences in the epigenomic profile. Specifically, it was determined that the loss of CTCF led to a loss of chromatin interaction specifically around pathologically related cardiac genes [47]. Similarly, genomics has also been suggested and utilized as a biomarker for HF [48]. One study assessed 111 families with various types of heart disease and used data sets generated via whole genome sequencing. They found that sequencing could be used as an initial understanding of human based cardiac diseases [49]. Again, genomic-based research techniques are a necessary component of research used for clinical relevance; however, genomics only provides one piece of a very complex puzzle.

3.2. Targeting Cardiac Genes for Transcriptional Regulation

Chromatin remodeling in the heart is regulated by a multitude of genes. Some of these cardiac genes have been targeted during HF to assess changes in the cardiac disease state. For example, the transcription factor, Med1, was found to regulate chromatin remodeling in cardiomyocytes. Specifically, Med1 was found to synchronize the histone acetylation of lysine 27 (H3K27) which allowed for more open chromatin accessibility and therefore more gene expression [50]. One study found that changes in gene expression in GATA4 and NXK2.5 were associated with changes in the chromatin architecture [43]. Interestingly, the authors found a decrease in the protein CTCF, which is a chromatin structural protein, specifically after HF in murine cardiomyocytes [43]. Likewise, another study found that GATA4 plays a major role in the chromatin structure, specifically in cardiac disease progression as well as cardiac development [51]. He et al. describe these changes as GATA4 occupancy, which is significantly altered during fetal cardiac development. Similarly, they saw changes in gene expression and therefore chromatin occupancy during cardiac stress [51]. The transcription factor, AP-1, was found to play a pivotal role in cardiomyocyte regeneration and growth after injury in zebrafish [52]. Zebrafish are known to have an increased ability to regenerate and grow cardiomyocytes even into adulthood, unlike humans and mice, making them an interesting resource for studying HF [53]. Thus, this study connected AP-1 to the regulation of chromatin remodeling and accessibility in the heart, making it a viable target for cardiomyocyte regeneration after injury [52].
It has also been found that changes in chromatin architecture have a role in cardiac fibroblast phenotypic gene expression. After cardiac injury, there is often an influx of cardiac fibroblasts that form scar tissue in the heart. Too much fibrosis can lead to irreversible damage and decreased heart function [54]. It has been suggested that transcription factors such as GATA4, as well as MEF2C and TBX5, could be directly reprogrammed to alter their chromatin remodeling and therefore epigenetic signaling in fibrosis [52,53]. This has specifically been studied in cardiac fibroblasts in vitro and in vivo with high success [55,56,57,58,59]. Additionally, changes in chromatic structure were shown to affect the stiffness of cardiac fibroblasts after injury [60]. It was also found that topological change to cardiac fibroblasts significantly altered the chromatin remodeling and downstream cardiac gene expression associated with a decreased pathological response [61]. The chromatin remodeling protein, BRG1, was also found to be a vital regulator of cardiac fibrosis, specifically in regulating the endothelial-to-mesenchymal transition [62].
Chromatin remodeling has also been connected to metabolism during HF. Specifically, one study found that changes in the expression of beta hydroxybutyrate (BHB) were associated with alterations in chromatin remodeling and histone modifications that enhanced disease progression [63]. Chromatin remodeling and BHB expression were associated with cardiac protection via mitochondrial function and metabolism [63]. Overall, an important conclusion from assessing gene regulation is that changes in gene expression alone do not tell the entire story. There is a combination of regulation from gene expression, modifiers, and epigenetic marks that is necessary to fully understand chromatin remodeling in the heart, which is summarized in Table 1.

3.3. RNA Molecules

The use of small RNA molecules, often deemed non-coding RNA molecules, has been of great interest in epigenetic regulation and chromatin remodeling, specifically in HF [64]. Many RNA molecules have been studied in pathological cardiac models, microRNAs (miRNAs), circular RNA (circRNA), and long non-coding RNAs (lncRNAs) being the primary focus due to influential and novel data [65,66,67,68].
It has been shown that the lncRNAs are regulated by ATP-dependent chromatin remodeling factors in both mouse and human hearts during HF [40]. The lncRNAs have also been used as biomarkers for HF [65,66]. Current research has indicated that these lncRNAs may be the master regulators of disease progression in HF via chromatin remodeling [69,70,71]. Many lncRNA can bind and regulate chromatin remodeling through epigenetic modifications and therefore regulate various cardiac gene expression levels during pathological stress [61,68]. For example, an lncRNA labeled AHIT was found to regulate cardiac hypertrophy by binding to SUZ12, which is a part of the polycomb repressive complex 2 (PRC2), known to regulate histone modifications and chromatin remodeling [72]. Another study found that the lncRNA NEAT1 increased cardiac fibrosis through EZH2, which is an epigenetic modifier known for cardiac gene repression [73]. Interestingly, the lncRNA CHAIR was found to be cardioprotective by modulating DNA methylation. Specifically, CHAIR inhibited the DNA binding complex, which limited the pathological response in the heart [74]. Finally, the lncRNA labeled MEG3 was found to regulate chromatin remodeling, which specifically induced matrix metalloproteinase 2 (MMP2), increasing cardiac fibrosis [75]. The role of lncRNAs in cardiac chromatin remodeling is an expanding source of innovative research for the treatment of HF.
The circular RNAs (circRNA) have also been of interest due to their ability to act as a sponge as well as a transcriptional regulator throughout cardiac tissue [76]. Because of their unique role in cardiac tissue, circRNAs have been proposed as major biomarkers for HF [77]. For example, one study found that the circRNA named circNCX1 played a major role in cardiomyocyte death during cardiac injury. Specifically, there was an increase in this circNCX1 with an increase in reactive oxygen species (ROS). The knockdown of this circRNA decreased the amount of cardiomyocyte cell death in mouse hearts [78]. Often circRNAs do not act completely alone. One study found that the circRNA circ-HIPK3 interacted with the miRNA miR-17-3p to regulate calcium signaling during HF. Specifically, the downregulation of the circRNA seemed to lessen the fibrotic response and the progression of HF in adult mice [79].
Another major RNA molecule studied in cardiac biology is the miRNAs, known for extensive signaling networks and chromatin regulation in the heart [80]. Because of the interconnected signaling, miRNAs are often connected to other RNA molecules and other signal transduction. One study found a connection between the miRNA miR-193b and ROS. Specifically, there was an increase in both the miR-193b and ROS in HF connected to a metabolic syndrome that increased pulmonary dysfunction in exercise-induced pulmonary hypertension [81]. The gene NRF2 has been associated with antioxidant redox balance that often becomes dysregulated in HF. Many myocardial miRNAs have been associated with the dysregulation of NRF2 during HF [82]. Overall, RNA molecules have great potential for unlocking biomarkers and signaling associated with chromatin remodeling during HF, which could provide an invaluable resource with therapeutic potential.

3.4. Chromatin Remodelers and Complexes

Unlike RNA molecules and cardiac gene transcription, chromatin remodelers play a direct role in regulating chromatin architecture. When the chromatin remodelers are dysregulated, the chromatin structure is altered, which can lead to devasting pathological responses that cause increased cardiac cell death, inflammatory signaling, and stress responses found in HF [28]. One study found that the chromatin remodeling protein BRG1 and p300 had stage-specific regulation of histone acetylation. Specifically, these chromatin remodelers were upregulated during HF but not during left ventricular hypertrophy, indicating a step-wise transition regulated by chromatin regulators [83]. The chromatin regulator, identified as SETD7, known for the methylation of histone 3 at lysine 4 (H3K4me1), was found to regulate inflammation pathways in obese patients with HF [84]. It was found that the loss of SETD7 in murine cardiomyocytes protected against hypertrophy and further cardiac dysfunction that was directly associated with the regulation of inflammatory genes [84]. Another study found that the interaction between a histone lysine methyltransferase named G9a and its downstream target Brain Derived Neurotrophic Factor (BDNF) regulated histone epigenetic modifications, therefore altering chromatin remodeling. It was found that G9a, which inhibits BDNF and increases cardiomyocyte death, is overexpressed in HF [85]. Another study found that ZNHIT1, a major regulator of a chromatin remodeling complex, was necessary for heart function [86]. Specifically, the loss of this chromatin regulator caused rapid HF and dysregulated calcium signaling. It was determined that the ZNHIT1 regulated CASQ1, a major regulator of calcium signaling in the sarcoplasmic reticulum, by altering the histone 2A variant and therefore chromatin regulation [86]. It was also found that BAF60c, a chromatin remodeling complex also called SMARCD3, regulates cardiomyocyte growth through MEF2 and myocardin gene expression [87].
Two major chromatin remodeling complexes are histone deacetylases (HDACs) and histone acetyltransferases (HATs). Both of these are known to regulate lysine acetylation in many cell types, including cardiac cells [88,89]. The dysregulation of both HATs and HDACs has been associated with increased HF phenotypes and cardiac disease progression [90]. Pharmaceutical inhibitors of these complexes have been suggested as a therapeutic approach to cardiac diseases [91]. Many studies have connected HDAC inhibition with lessening cardiac fibrosis seen in HF [92,93,94]. This HDAC inhibition has directly improved heart function [95], making it a major therapeutic target for HF. Additionally, HDAC regulation has been associated with DNA damage mechanisms associated with aging. The SIRT1, a class III HDAC, was found to regulate cellular senescence in cardiac cells, which was associated with increased stress and cardiac injury [27].
It has also been well studied that the bromodomain and extraterminal (BET) chromatin regulators, such as BRD4, play a major role in maintaining cardiomyocyte health, specifically via mitochondrial function, during HF [96]. Other studies have seen similar advancements and chromatin remodeling therapeutic potential using BET regulation in HF [92,93,97]. One study found that BET inhibitors (BETi) caused increased DNA damage responses and altered overall chromatin structure, resulting in better cell viability. This was connected to BRD4 colocalizing with YY1 at a Topologically Associating Domain Boundary (TADB) [98]. Similarly to HDACs, BET inhibition has been a major topic for drug-based chromatin remodeling interventions.
As more and more chromatin remodeling complexes have been uncovered, the role of super-enhancers within those complexes has been discovered as well. Super-enhancers are often areas in the genome that have an increased number of chromatin interactions [99]. These super-enhancers have been studied regarding chromatin remodeling during HF as a major target for cardiac disease progression inhibition [100]. Likewise, it has been studied that ATP-dependent chromatin remodeling complexes are extremely important in regulating pro-inflammatory responses, making them interesting targets for biomarkers of hypertension during HF [101,102]. There have also been connections made between chromatin remodelers and global chromatin architectural changes, specifically ones associated with heart failure and aging mechanisms. Chromatin remodeling dysregulation was specifically connected with skeletal muscle changes in aging models [103]. Overall, chromatin remodelers are a promising strategy for uncovering epigenetic modulation during HF.
Table 1. Summary of chromatin remodeling mechanisms during heart failure.
Table 1. Summary of chromatin remodeling mechanisms during heart failure.
Chromatin AlterationsSummary of FunctionSource
Target Genes
Med1
Gata4
Nxk2.5
CTCF
AP-1
MEF2c
Tbx5
BRG1
BHB
Transcriptional regulation, caridac development, metabolism, cardiac fibrosis, cell death regulation, epigenetic regulation, chromatin remodeling[51,52,53,54,55,56,57,58,59,60,61,62,63]
RNA Molecules
lncRNAs
AHIT
CHAIR
MEG3
circRNA
circNCX1
circHIPK3
miRNA
miR-17-3p
miR-193b
Regulating epigenetic marks, cardiac gene expression, chromatin remodeling, apoptoic regulation, redox regulation[64,65,66,67,68,70,72,73,74,75,76,77,78,79,80,81]
Chromatin Remodelers
BRG1
P300
SETD7
G9a
Znhit1
Baf60c
HDACs
HATs
BETs/BRD4
Direct chromatin architecture regulation, adding and removing epigenetic modifications, cardiac disease regulation, DNA damage respones, pro-inflammatory response regulation, aging mechanisms[28,83,84,85,86,87,88,89,90,91,92,93,94,95,96,98,99,100,101]

4. Current Clinical Application of Chromatin Remodeling for the Treatment of Heart Failure

Because of the influential and groundbreaking research on chromatin remodeling in HF, some research has progressed into clinical applications as a form of treatment for the disease. Many have suggested using epigenetic modifications and chromatin remodeling dysregulation as biomarkers for HF [102]. More recently, the use of the epitranscriptiome, which consists of RNA epigenetic coding, has been suggested. This incorporates different RNA molecules and further gene expression [89,90]. One study suggested the “EPi-transgeneratIonal networK mOdeling-STratificatiOn of heaRt Morbidity” (EPIKO-STORM) platform, which helps log the history of HF. The suggested application of it is to document the epigenetic history of patients to help better predict familial related HF risk factors that are not as common [104]. As described earlier, the use of multiple omics, or multiomic, approaches has also been applied to clinical applications [104,105,106,107]. There is also a great benefit to using genomics-based approaches to diagnose any hereditary based cardiac diseases [31]. Through a combination of many different omics approaches, a more comprehensive understanding is continuously developing to uncover the genetic and epigenetic mechanisms of HF in patients.
Because of the lack of a cure for HF currently, individualized medicine is at the forefront of clinical applications [108]. This has led to “epi-drugs” or epigenetic-based pharmaceutic interventions for patients with HF [94,95]. It is thought that epi-drugs could alter a specific epigenetic profile by adding or removing an epigenetic mark and therefore alter chromatin remodeling in a pathological setting. Interestingly, these epi-drugs have been approved for other diseases [109], specifically some cancers [110]. Some epi-drugs that have been considered and studied included targeting the chromatin remodeling complexes. For example, it has been shown that HDAC inhibitors can be used to help lessen inflammatory responses, pathological gene expression, and fibrosis during HF [111,112,113,114,115]. Clinical applications are looking at DNA methylation modifiers [116,117], BET inhibitors [118], and even chromatin remodeling miRNAs [102,111]. Similarly, it has been suggested that interventions, such as the epi-drugs, could be used to target transcriptional-based machinery use in chromatin remodeling [118]. Finally, there is evidence to suggest that altering the gut microbiota can even change chromatin remodeling in cardiac related genes [108,119,120]. It was specifically found that HDAC3 activity could be altered with microbiota-based metabolites [119]. Overall, there are a variety of promising therapeutic techniques being studied and developed for HF interventions using chromatin remodeling.

5. Conclusions

Chromatin remodeling is a promising strategy not only for understanding HF but also for potentially treating it. Because of the influential role of the genomic architecture and the regulation of gene expression, targeting chromatin remodeling is an encouraging approach for altering cardiac genes during stress or injury. However, with all the great advancements, there are still major hurdles to overcome. Additionally, the scope of this review focused on chromatin remodeling. It is important to address that epigenetic regulation in cardiac disease progression encompasses more than just chromatin remodeling, though this is not addressed specifically in this review. Many of the approaches have limitations that still need to be addressed, including massive genomics-based interventions, which can overlook details and create correlations that are not necessarily connected. Pharmaceutical-based chromatin remodeling interventions, such as RNA molecules and epi-drugs, have the advantage of being less invasive but still need to address adverse effects, the timing of administration, and the specificity [108,121]. In the future, using the techniques as biomarkers is one of the most promising applications, and with more regulation, consistency, and standardization, they could be pivotal in addressing HF in patients before it becomes deadly [121,122,123]. As therapeutic advancements are made, the use of chromatin remodeling as a pharmaceutical intervention and as a biomarker for cardiac disease states would be monumental for heart failure patients. Overall, chromatin remodeling is an exciting and encouraging approach to HF that continues to grow to eventually cure this disease.

Author Contributions

Conceptualization, L.K., methodology, L.K.; investigation, L.K. and B.B., writing—original draft preparation, L.K., writing—review and editing, B.B. and L.K.; visualization, L.K.; supervision, L.K.; project administration, L.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to thank Alvernia’s O’Pake Institute of Economic Development and Entrepreneurship, and specifically its Academic Services division, for their student-powered research support for this project through the O’Pake Fellows program.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mc Namara, K.; Alzubaidi, H.; Jackson, J.K. Cardiovascular disease as a leading cause of death: How are pharmacists getting involved? Integr. Pharm. Res. Pract. 2019, 8, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cardiovascular Diseases (CVDs). Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 17 February 2020).
  3. Ciumărnean, L.; Milaciu, M.V.; Negrean, V.; Orășan, O.H.; Vesa, S.C.; Sălăgean, O.; Iluţ, S.; Vlaicu, S.I. Cardiovascular Risk Factors and Physical Activity for the Prevention of Cardiovascular Diseases in the Elderly. Int. J. Environ. Res. Public Health 2022, 19, 207. [Google Scholar] [CrossRef] [PubMed]
  4. Lifestyle Strategies for Risk Factor Reduction, Prevention and Treatme. Available online: https://www.taylorfrancis.com/chapters/edit/10.1201/9781315201108-2/lifestyle-strategies-risk-factor-reduction-prevention-treatment-cardiovascular-disease-james-rippe-theodore-angelopoulos (accessed on 6 January 2023).
  5. Zhao, D.; Liu, J.; Wang, M.; Zhang, X.; Zhou, M. Epidemiology of cardiovascular disease in China: Current features and implications. Nat. Rev. Cardiol. 2019, 16, 203–212. [Google Scholar] [CrossRef] [PubMed]
  6. Savarese, G.; Lund, L.H. Global Public Health Burden of Heart Failure. Card. Fail. Rev. 2017, 3, 7–11. [Google Scholar] [CrossRef] [PubMed]
  7. Kurmani, S.; Squire, I. Acute Heart Failure: Definition, Classification and Epidemiology. Curr. Heart Fail. Rep. 2017, 14, 385–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. CDC Heart Failure|cdc.gov. Centers for Disease Control and Prevention. 8 September 2020. Available online: https://www.cdc.gov/heartdisease/heart_failure.htm (accessed on 12 October 2020).
  9. Lippi, G.; Sanchis-Gomar, F. Global epidemiology and future trends of heart failure. AME Med. J. 2020, 5, 1–6. [Google Scholar] [CrossRef]
  10. Rossignol, P.; Hernandez, A.F.; Solomon, S.D.; Zannad, F. Heart failure drug treatment. Lancet 2019, 393, 1034–1044. [Google Scholar] [CrossRef]
  11. Smits, J.M.; Samuel, U.; Laufer, G. Bridging the gap in heart transplantation. Curr. Opin. Organ. Transplant. 2017, 22, 221–224. [Google Scholar] [CrossRef]
  12. Rheault-Henry, M.; White, I.; Grover, D.; Atoui, R. Stem cell therapy for heart failure: Medical breakthrough, or dead end? World J. Stem Cells 2021, 13, 236–259. [Google Scholar] [CrossRef]
  13. Segers, V.F.M.; Lee, R.T. Stem-cell therapy for cardiac disease. Nature 2008, 451, 937–942. [Google Scholar] [CrossRef]
  14. Alexanian, M.; Padmanabhan, A.; McKinsey, T.A.; Haldar, S.M. Epigenetic therapies in heart failure. J. Mol. Cell Cardiol. 2019, 130, 197–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kim, S.Y.; Morales, C.; Gillette, T.G.; Hill, J.A. Epigenetic Regulation in Heart Failure. Curr. Opin. Cardiol. 2016, 31, 255–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Chaturvedi, P.; Tyagi, S.C. Epigenetic mechanisms underlying cardiac degeneration and regeneration. Int. J. Cardiol. 2014, 173, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Liu, C.-F.; Tang, W.H.W. Epigenetics in Cardiac Hypertrophy and Heart Failure. J. Am. Coll. Cardiol. Basic Trans. Sci. 2019, 4, 976–993. [Google Scholar] [CrossRef]
  18. McKinsey, T.A.; Vondriska, T.M.; Wang, Y. Epigenomic regulation of heart failure: Integrating histone marks, long noncoding RNAs, and chromatin architecture. F1000Res 2018, 7, F1000. [Google Scholar] [CrossRef] [Green Version]
  19. Kimball, T.H.; Vondriska, T.M. Metabolism, Epigenetics, and Causal Inference in Heart Failure. Trends Endocrinol. Metab. 2020, 31, 181–191. [Google Scholar] [CrossRef]
  20. Shi, Y.; Zhang, H.; Huang, S.; Yin, L.; Wang, F.; Luo, P.; Huang, H. Epigenetic regulation in cardiovascular disease: Mechanisms and advances in clinical trials. Sig. Transduct. Target. 2022, 7, 200. [Google Scholar] [CrossRef]
  21. McGinty, R.K.; Tan, S. Histone, Nucleosome, and Chromatin Structure. In Fundamentals of Chromatin; Workman, J.L., Abmayr, S.M., Eds.; Springer: New York, NY, USA, 2014; pp. 1–28. ISBN 978-1-4614-8624-4. [Google Scholar]
  22. Shah, M.; He, Z.; Rauf, A.; Beikoghli Kalkhoran, S.; Heiestad, C.M.; Stensløkken, K.-O.; Parish, C.R.; Soehnlein, O.; Arjun, S.; Davidson, S.M.; et al. Extracellular histones are a target in myocardial ischaemia–reperfusion injury. Cardiovasc. Res. 2021, 118, 1115–1125. [Google Scholar] [CrossRef]
  23. Gilsbach, R.; Schwaderer, M.; Preissl, S.; Grüning, B.A.; Kranzhöfer, D.; Schneider, P.; Nührenberg, T.G.; Mulero-Navarro, S.; Weichenhan, D.; Braun, C.; et al. Distinct epigenetic programs regulate cardiac myocyte development and disease in the human heart in vivo. Nat. Commun. 2018, 9, 391. [Google Scholar] [CrossRef] [Green Version]
  24. Zhang, Q.-J.; Liu, Z.-P. Histone methylations in heart development, congenital and adult heart diseases. Epigenomics 2015, 7, 321–330. [Google Scholar] [CrossRef] [Green Version]
  25. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
  26. Kraus, L. Targeting Epigenetic Regulation of Cardiomyocytes through Development for Therapeutic Cardiac Regeneration after Heart Failure. Int. J. Mol. Sci. 2022, 23, 11878. [Google Scholar] [CrossRef] [PubMed]
  27. Han, P.; Hang, C.T.; Yang, J.; Chang, C.-P. Chromatin remodeling in cardiovascular development and physiology. Circ. Res. 2011, 108, 378–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Han, P.; Yang, J.; Shang, C.; Chang, C.-P. Chromatin Remodeling in Heart Failure. In Epigenetics in Cardiac Disease; Backs, J., McKinsey, T.A., Eds.; Cardiac and Vascullar Biology; Springer International Publishing: Cham, Switzerland, 2016; pp. 103–124. ISBN 978-3-319-41457-7. [Google Scholar]
  29. Lorch, Y.; Maier-Davis, B.; Kornberg, R.D. Mechanism of chromatin remodeling. Proc. Natl. Acad. Sci. USA 2010, 107, 3458–3462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Chromatin Remodeling in Eukaryotes|Learn Science at Scitable. Available online: http://www.nature.com/scitable/topicpage/chromatin-remodeling-in-eukaryotes-1082 (accessed on 6 January 2023).
  31. Pasipoularides, A. Implementing genome-driven personalized cardiology in clinical practice. J. Mol. Cell. Cardiol. 2018, 115, 142–157. [Google Scholar] [CrossRef]
  32. Khomtchouk, B.B.; Tran, D.-T.; Vand, K.A.; Might, M.; Gozani, O.; Assimes, T.L. Cardioinformatics: The nexus of bioinformatics and precision cardiology. Brief. Bioinform. 2020, 21, 2031–2051. [Google Scholar] [CrossRef]
  33. Hunt, C.; Montgomery, S.; Berkenpas, J.W.; Sigafoos, N.; Oakley, J.C.; Espinosa, J.; Justice, N.; Kishaba, K.; Hippe, K.; Si, D.; et al. Recent Progress of Machine Learning in Gene Therapy. Curr. Gene Ther. 2022, 22, 132–143. [Google Scholar] [CrossRef]
  34. Tsompana, M.; Buck, M.J. Chromatin accessibility: A window into the genome. Epigenet. Chromatin 2014, 7, 33. [Google Scholar] [CrossRef] [Green Version]
  35. Xu, W.; Wen, Y.; Liang, Y.; Xu, Q.; Wang, X.; Jin, W.; Chen, X. A plate-based single-cell ATAC-seq workflow for fast and robust profiling of chromatin accessibility. Nat. Protoc. 2021, 16, 4084–4107. [Google Scholar] [CrossRef]
  36. Bhattacharyya, S.; Kollipara, R.K.; Orquera-Tornakian, G.; Goetsch, S.; Zhang, M.; Perry, C.; Li, B.; Shelton, J.M.; Bhakta, M.; Duan, J.; et al. Global chromatin landscapes identify candidate noncoding modifiers of cardiac rhythm. J. Clin. Investig. 2022, 133, 3. [Google Scholar] [CrossRef]
  37. Jia, G.; Preussner, J.; Chen, X.; Guenther, S.; Yuan, X.; Yekelchyk, M.; Kuenne, C.; Looso, M.; Zhou, Y.; Teichmann, S.; et al. Single cell RNA-seq and ATAC-seq analysis of cardiac progenitor cell transition states and lineage settlement. Nat. Commun. 2018, 9, 4877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. McKinsey, T.A.; Olson, E.N. Toward transcriptional therapies for the failing heart: Chemical screens to modulate genes. J. Clin. Investig. 2005, 115, 538–546. [Google Scholar] [CrossRef] [Green Version]
  39. Centore, R.C.; Sandoval, G.J.; Soares, L.M.M.; Kadoch, C.; Chan, H.M. Mammalian SWI/SNF Chromatin Remodeling Complexes: Emerging Mechanisms and Therapeutic Strategies. Trends Genet. 2020, 36, 936–950. [Google Scholar] [CrossRef] [PubMed]
  40. Han, P.; Chang, C.-P. Long non-coding RNA and chromatin remodeling. RNA Biol. 2015, 12, 1094–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Han, P.; Li, W.; Lin, C.-H.; Yang, J.; Shang, C.; Nuernberg, S.T.; Jin, K.K.; Xu, W.; Lin, C.-Y.; Lin, C.-J.; et al. A long noncoding RNA protects the heart from pathological hypertrophy. Nature 2014, 514, 102–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kuppe, C.; Ramirez Flores, R.O.; Li, Z.; Hayat, S.; Levinson, R.T.; Liao, X.; Hannani, M.T.; Tanevski, J.; Wünnemann, F.; Nagai, J.S.; et al. Spatial multi-omic map of human myocardial infarction. Nature 2022, 608, 766–777. [Google Scholar] [CrossRef] [PubMed]
  43. Chapski, D.J.; Cabaj, M.; Morselli, M.; Mason, R.J.; Soehalim, E.; Ren, S.; Pellegrini, M.; Wang, Y.; Vondriska, T.M.; Rosa-Garrido, M. Early adaptive chromatin remodeling events precede pathologic phenotypes and are reinforced in the failing heart. J. Mol. Cell. Cardiol. 2021, 160, 73–86. [Google Scholar] [CrossRef]
  44. Wang, J.; Wang, Y.; Duan, Z.; Hu, W. Hypoxia-induced alterations of transcriptome and chromatin accessibility in HL-1 cells. IUBMB Life 2020, 72, 1737–1746. [Google Scholar] [CrossRef]
  45. Zhou, X.; Zhang, S.; Zhao, Y.; Wang, W.; Zhang, H. A multi-omics approach to identify molecular alterations in a mouse model of heart failure. Theranostics 2022, 12, 1607–1620. [Google Scholar] [CrossRef]
  46. Arvanitis, M.; Tampakakis, E.; Zhang, Y.; Wang, W.; Auton, A.; Dutta, D.; Glavaris, S.; Keramati, A.; Chatterjee, N.; Chi, N.C.; et al. Genome-wide association and multi-omic analyses reveal ACTN2 as a gene linked to heart failure. Nat. Commun. 2020, 11, 1122. [Google Scholar] [CrossRef] [Green Version]
  47. Rosa-Garrido, M.; Chapski, D.J.; Schmitt, A.D.; Kimball, T.H.; Karbassi, E.; Monte, E.; Balderas, E.; Pellegrini, M.; Shih, T.-T.; Soehalim, E.; et al. High-Resolution Mapping of Chromatin Conformation in Cardiac Myocytes Reveals Structural Remodeling of the Epigenome in Heart Failure. Circulation 2017, 136, 1613–1625. [Google Scholar] [CrossRef] [PubMed]
  48. Meder, B.; Haas, J.; Sedaghat-Hamedani, F.; Kayvanpour, E.; Frese, K.; Lai, A.; Nietsch, R.; Scheiner, C.; Mester, S.; Bordalo, D.M.; et al. Epigenome-Wide Association Study Identifies Cardiac Gene Patterning and a Novel Class of Biomarkers for Heart Failure. Circulation 2017, 136, 1528–1544. [Google Scholar] [CrossRef] [PubMed]
  49. Reuter, M.S.; Chaturvedi, R.R.; Liston, E.; Manshaei, R.; Aul, R.B.; Bowdin, S.; Cohn, I.; Curtis, M.; Dhir, P.; Hayeems, R.Z.; et al. The Cardiac Genome Clinic: Implementing genome sequencing in pediatric heart disease. Genet. Med. 2020, 22, 1015–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Hall, D.D.; Spitler, K.M.; Grueter, C.E. Disruption of cardiac Med1 inhibits RNA polymerase II promoter occupancy and promotes chromatin remodeling. Am. J. Physiol.-Heart Circ. Physiol. 2019, 316, H314–H325. [Google Scholar] [CrossRef] [PubMed]
  51. He, A.; Gu, F.; Hu, Y.; Ma, Q.; Yi Ye, L.; Akiyama, J.A.; Visel, A.; Pennacchio, L.A.; Pu, W.T. Dynamic GATA4 enhancers shape the chromatin landscape central to heart development and disease. Nat. Commun. 2014, 5, 4907. [Google Scholar] [CrossRef] [Green Version]
  52. Beisaw, A.; Kuenne, C.; Guenther, S.; Dallmann, J.; Wu, C.-C.; Bentsen, M.; Looso, M.; Stainier, D.Y.R. AP-1 Contributes to Chromatin Accessibility to Promote Sarcomere Disassembly and Cardiomyocyte Protrusion During Zebrafish Heart Regeneration. Circ. Res. 2020, 126, 1760–1778. [Google Scholar] [CrossRef]
  53. de Pater, E.; Clijsters, L.; Marques, S.R.; Lin, Y.-F.; Garavito-Aguilar, Z.V.; Yelon, D.; Bakkers, J. Distinct phases of cardiomyocyte differentiation regulate growth of the zebrafish heart. Development 2009, 136, 1633–1641. [Google Scholar] [CrossRef] [Green Version]
  54. Jiang, W.; Xiong, Y.; Li, X.; Yang, Y. Cardiac Fibrosis: Cellular Effectors, Molecular Pathways, and Exosomal Roles. Front. Cardiovasc. Med. 2021, 8, 715258. [Google Scholar] [CrossRef]
  55. Mathison, M.; Singh, V.P.; Sanagasetti, D.; Yang, L.; Pinnamaneni, J.P.; Yang, J.; Rosengart, T.K. Cardiac Reprogramming Factor Gata4 Reduces Post-Infarct Cardiac Fibrosis through Direct Repression of the Pro-Fibrotic Mediator Snail. J. Thorac. Cardiovasc. Surg. 2017, 154, 1601–1610. [Google Scholar] [CrossRef] [Green Version]
  56. Ieda, M. Key Regulators of Cardiovascular Differentiation and Regeneration: Harnessing the Potential of Direct Reprogramming to Treat Heart Failure. J. Card. Fail. 2020, 26, 80–84. [Google Scholar] [CrossRef]
  57. Yamakawa, H.; Ieda, M. Cardiac regeneration by direct reprogramming in this decade and beyond. Inflamm. Regen. 2021, 41, 20. [Google Scholar] [CrossRef] [PubMed]
  58. McKinsey, T.A.; Foo, R.; Anene-Nzelu, C.G.; Travers, J.G.; Vagnozzi, R.J.; Weber, N.; Thum, T. Emerging epigenetic therapies of cardiac fibrosis and remodeling in heart failure: From basic mechanisms to early clinical development. Cardiovasc. Res. 2022, 118, cvac142. [Google Scholar] [CrossRef]
  59. Chen, Y.; Yang, Z.; Zhao, Z.-A.; Shen, Z. Direct reprogramming of fibroblasts into cardiomyocytes. Stem Cell Res. Ther. 2017, 8, 118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Hu, S.; Vondriska, T.M. How chromatin stiffens fibroblasts. Curr. Opin. Physiol. 2022, 26, 100537. [Google Scholar] [CrossRef] [PubMed]
  61. Yu, J.; Seldin, M.M.; Fu, K.; Li, S.; Lam, L.; Wang, P.; Wang, Y.; Huang, D.; Nguyen, T.L.; Wei, B.; et al. Topological Arrangement of Cardiac Fibroblasts Regulates Cellular Plasticity. Circ. Res. 2018, 123, 73–85. [Google Scholar] [CrossRef] [PubMed]
  62. Li, Z.; Kong, X.; Zhang, Y.; Zhang, Y.; Yu, L.; Guo, J.; Xu, Y. Dual roles of chromatin remodeling protein BRG1 in angiotensin II-induced endothelial–mesenchymal transition. Cell Death Dis. 2020, 11, 549. [Google Scholar] [CrossRef]
  63. Santulli, G.; Jankauskas, S.; Morelli, M.; Gambardella, J. Chromatin remodeling and mitochondrial biogenesis underlie the improved cardiac function in heart failure induced by ketogenic diet and beta-hydroxibutiyrate supplementation. FASEB J. 2021, 35. [Google Scholar] [CrossRef]
  64. Lu, P.; Ding, F.; Xiang, Y.K.; Hao, L.; Zhao, M. Noncoding RNAs in Cardiac Hypertrophy and Heart Failure. Cells 2022, 11, 777. [Google Scholar] [CrossRef]
  65. Li, M.; Duan, L.; Li, Y.; Liu, B. Long noncoding RNA/circular noncoding RNA–miRNA–mRNA axes in cardiovascular diseases. Life Sci. 2019, 233, 116440. [Google Scholar] [CrossRef]
  66. Gong, C.; Zhou, X.; Lai, S.; Wang, L.; Liu, J. Long Noncoding RNA/Circular RNA-miRNA-mRNA Axes in Ischemia-Reperfusion Injury. Biomed. Res. Int. 2020, 2020, 8838524. [Google Scholar] [CrossRef]
  67. Chen, Y.-H.; Zhong, L.-F.; Hong, X.; Zhu, Q.-L.; Wang, S.-J.; Han, J.-B.; Huang, W.-J.; Ye, B.-Z. Integrated Analysis of circRNA-miRNA-mRNA ceRNA Network in Cardiac Hypertrophy. Front. Genet. 2022, 13, 123. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, G.; Dou, L.; Chen, Y. Association of long-chain non-coding RNA MHRT gene single nucleotide polymorphism with risk and prognosis of chronic heart failure. Medicine 2020, 99, e19703. [Google Scholar] [CrossRef] [PubMed]
  69. Hobuß, L.; Bär, C.; Thum, T. Long Non-coding RNAs: At the Heart of Cardiac Dysfunction? Front. Physiol. 2019, 10, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Zhang, Z.; Wan, J.; Liu, X.; Zhang, W. Strategies and technologies for exploring long noncoding RNAs in heart failure. Biomed. Pharmacother. 2020, 131, 110572. [Google Scholar] [CrossRef]
  71. Hermans-Beijnsberger, S.; van Bilsen, M.; Schroen, B. Long non-coding RNAs in the failing heart and vasculature. Noncoding RNA Res. 2018, 3, 118–130. [Google Scholar] [CrossRef]
  72. Yu, J.; Yang, Y.; Xu, Z.; Lan, C.; Chen, C.; Li, C.; Chen, Z.; Yu, C.; Xia, X.; Liao, Q.; et al. Long Noncoding RNA Ahit Protects Against Cardiac Hypertrophy Through SUZ12 (Suppressor of Zeste 12 Protein Homolog)-Mediated Downregulation of MEF2A (Myocyte Enhancer Factor 2A). Circ. Heart Fail. 2020, 13, e006525. [Google Scholar] [CrossRef]
  73. Ge, Z.; Yin, C.; Li, Y.; Tian, D.; Xiang, Y.; Li, Q.; Tang, Y.; Zhang, Y. Long noncoding RNA NEAT1 promotes cardiac fibrosis in heart failure through increased recruitment of EZH2 to the Smad7 promoter region. J. Transl. Med. 2022, 20, 7. [Google Scholar] [CrossRef]
  74. Qian, Y.; Zhang, M.; Zhou, N.; Xu, X.; Zhang, J.; Ding, Q.; Wang, J. A long noncoding RNA CHAIR protects the heart from pathological stress. Clin. Sci. 2020, 134, 1843–1857. [Google Scholar] [CrossRef]
  75. Piccoli, M.-T.; Gupta, S.K.; Viereck, J.; Foinquinos, A.; Samolovac, S.; Kramer, F.L.; Garg, A.; Remke, J.; Zimmer, K.; Batkai, S.; et al. Inhibition of the Cardiac Fibroblast–Enriched lncRNA Meg3 Prevents Cardiac Fibrosis and Diastolic Dysfunction. Circ. Res. 2017, 121, 575–583. [Google Scholar] [CrossRef]
  76. Tan, W.L.W.; Lim, B.T.S.; Anene-Nzelu, C.G.O.; Ackers-Johnson, M.; Dashi, A.; See, K.; Tiang, Z.; Lee, D.P.; Chua, W.W.; Luu, T.D.A.; et al. A landscape of circular RNA expression in the human heart. Cardiovasc. Res. 2017, 113, 298–309. [Google Scholar] [CrossRef]
  77. Sun, C.; Ni, M.; Song, B.; Cao, L. Circulating Circular RNAs: Novel Biomarkers for Heart Failure. Front. Pharmacol. 2020, 11, 560537. [Google Scholar] [CrossRef] [PubMed]
  78. Li, M.; Ding, W.; Tariq, M.A.; Chang, W.; Zhang, X.; Xu, W.; Hou, L.; Wang, Y.; Wang, J. A circular transcript of ncx1 gene mediates ischemic myocardial injury by targeting miR-133a-3p. Theranostics 2018, 8, 5855–5869. [Google Scholar] [CrossRef] [PubMed]
  79. Deng, Y.; Wang, J.; Xie, G.; Zeng, X.; Li, H. Circ-HIPK3 Strengthens the Effects of Adrenaline in Heart Failure by MiR-17-3p-ADCY6 Axis. Int. J. Biol. Sci. 2019, 15, 2484–2496. [Google Scholar] [CrossRef] [PubMed]
  80. Colpaert, R.M.W.; Calore, M. Epigenetics and microRNAs in cardiovascular diseases. Genomics 2021, 113, 540–551. [Google Scholar] [CrossRef]
  81. Satoh, T.; Wang, L.; Espinosa-Diez, C.; Wang, B.; Hahn, S.A.; Noda, K.; Rochon, E.R.; Dent, M.R.; Levine, A.R.; Baust, J.J.; et al. Metabolic Syndrome Mediates ROS-miR-193b-NFYA–Dependent Downregulation of Soluble Guanylate Cyclase and Contributes to Exercise-Induced Pulmonary Hypertension in Heart Failure With Preserved Ejection Fraction. Circulation 2021, 144, 615–637. [Google Scholar] [CrossRef]
  82. Tian, C.; Gao, L.; Zucker, I.H. Regulation of Nrf2 signaling pathway in heart failure: Role of extracellular vesicles and non-coding RNAs. Free Radic. Biol. Med. 2021, 167, 218–231. [Google Scholar] [CrossRef]
  83. Funamoto, M.; Sunagawa, Y.; Katanasaka, Y.; Shimizu, K.; Miyazaki, Y.; Sari, N.; Shimizu, S.; Mori, K.; Wada, H.; Hasegawa, K.; et al. Histone Acetylation Domains Are Differentially Induced during Development of Heart Failure in Dahl Salt-Sensitive Rats. Int. J. Mol. Sci. 2021, 22, 1771. [Google Scholar] [CrossRef]
  84. Costantino, S.; Ambrosini, S.; Mohammed, S.A.; Gorica, E.; Akhmedov, A.; Cosentino, F.; Ruschitzka, F.; Hamdani, N.; Paneni, F. A chromatin mark by SETD7 regulates myocardial inflammation in obesity-related heart failure with preserved ejection fraction. Eur. Heart J. 2022, 43, ehac544.2883. [Google Scholar] [CrossRef]
  85. Yan, F.; Chen, Z.; Cui, W. H3K9me2 regulation of BDNF expression via G9a partakes in the progression of heart failure. BMC Cardiovasc. Disord. 2022, 22, 182. [Google Scholar] [CrossRef]
  86. Shi, Y.; Fan, W.; Xu, M.; Lin, X.; Zhao, W.; Yang, Z. Critical role of Znhit1 for postnatal heart function and vacuolar cardiomyopathy. JCI Insight 2022, 7, e148752. [Google Scholar] [CrossRef]
  87. Sun, X.; Hota, S.K.; Zhou, Y.-Q.; Novak, S.; Miguel-Perez, D.; Christodoulou, D.; Seidman, C.E.; Seidman, J.G.; Gregorio, C.C.; Henkelman, R.M.; et al. Cardiac-enriched BAF chromatin-remodeling complex subunit Baf60c regulates gene expression programs essential for heart development and function. Biol. Open 2018, 7, bio029512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Yang, X.-J.; Seto, E. HATs and HDACs: From structure, function and regulation to novel strategies for therapy and prevention. Oncogene 2007, 26, 5310–5318. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, Y.; Miao, X.; Liu, Y.; Li, F.; Liu, Q.; Sun, J.; Cai, L. Dysregulation of Histone Acetyltransferases and Deacetylases in Cardiovascular Diseases. Oxid. Med. Cell Longev. 2014, 2014, 641979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Yang, M.; Zhang, Y.; Ren, J. Acetylation in cardiovascular diseases: Molecular mechanisms and clinical implications. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2020, 1866, 165836. [Google Scholar] [CrossRef]
  91. Cao, D.J.; Wang, Z.V.; Battiprolu, P.K.; Jiang, N.; Morales, C.R.; Kong, Y.; Rothermel, B.A.; Gillette, T.G.; Hill, J.A. Histone deacetylase (HDAC) inhibitors attenuate cardiac hypertrophy by suppressing autophagy. Proc. Natl. Acad. Sci. USA 2011, 108, 4123–4128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bagchi, R.A.; Weeks, K.L. Histone deacetylases in cardiovascular and metabolic diseases. J. Mol. Cell. Cardiol. 2019, 130, 151–159. [Google Scholar] [CrossRef]
  93. Jiménez-Uribe, A.P.; Gómez-Sierra, T.; Aparicio-Trejo, O.E.; Orozco-Ibarra, M.; Pedraza-Chaverri, J. Backstage players of fibrosis: NOX4, mTOR, HDAC, and S1P; companions of TGF-β. Cell. Signal. 2021, 87, 110123. [Google Scholar] [CrossRef]
  94. Gillette, T.G. HDAC Inhibition in the Heart: Erasing Hidden Fibrosis. Circulation 2021, 143, 1891–1893. [Google Scholar] [CrossRef]
  95. Travers, J.G.; Wennersten, S.A.; Peña, B.; Bagchi, R.A.; Smith, H.E.; Hirsch, R.A.; Vanderlinden, L.A.; Lin, Y.-H.; Dobrinskikh, E.; Demos-Davies, K.M.; et al. HDAC Inhibition Reverses Preexisting Diastolic Dysfunction and Blocks Covert Extracellular Matrix Remodeling. Circulation 2021, 143, 1874–1890. [Google Scholar] [CrossRef]
  96. Alcendor, R.R.; Gao, S.; Zhai, P.; Zablocki, D.; Holle, E.; Yu, X.; Tian, B.; Wagner, T.; Vatner, S.F.; Sadoshima, J. Sirt1 Regulates Aging and Resistance to Oxidative Stress in the Heart. Circ. Res. 2007, 100, 1512–1521. [Google Scholar] [CrossRef]
  97. Ijaz, T.; Burke, M.A. BET Protein-Mediated Transcriptional Regulation in Heart Failure. Int. J. Mol. Sci. 2021, 22, 6059. [Google Scholar] [CrossRef]
  98. Haldar, S.M.; McKinsey, T.A. BET-ting on chromatin-based therapeutics for heart failure. J. Mol. Cell. Cardiol. 2014, 74, 98–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Tsujikawa, L.M.; Kharenko, O.A.; Stotz, S.C.; Rakai, B.D.; Sarsons, C.D.; Gilham, D.; Wasiak, S.; Fu, L.; Sweeney, M.; Johansson, J.O.; et al. Breaking boundaries: Pan BETi disrupt 3D chromatin structure, BD2-selective BETi are strictly epigenetic transcriptional regulators. Biomed. Pharmacother. 2022, 152, 113230. [Google Scholar] [CrossRef] [PubMed]
  100. Huang, J.; Li, K.; Cai, W.; Liu, X.; Zhang, Y.; Orkin, S.H.; Xu, J.; Yuan, G.-C. Dissecting super-enhancer hierarchy based on chromatin interactions. Nat. Commun. 2018, 9, 943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Liu, C.-F.; Abnousi, A.; Bazeley, P.; Ni, Y.; Morley, M.; Moravec, C.S.; Hu, M.; Tang, W.H.W. Global analysis of histone modifications and long-range chromatin interactions revealed the differential cistrome changes and novel transcriptional players in human dilated cardiomyopathy. J. Mol. Cell. Cardiol. 2020, 145, 30–42. [Google Scholar] [CrossRef] [PubMed]
  102. Crosswhite, P.L. ATP-dependent chromatin remodeling complexes in embryonic vascular development and hypertension. Am. J. Physiol.-Heart Circ. Physiol. 2019, 317, H575–H580. [Google Scholar] [CrossRef]
  103. Wang, K.; Liu, H.; Hu, Q.; Wang, L.; Liu, J.; Zheng, Z.; Zhang, W.; Ren, J.; Zhu, F.; Liu, G.-H. Epigenetic regulation of aging: Implications for interventions of aging and diseases. Sig. Transduct. Target. 2022, 7, 374. [Google Scholar] [CrossRef]
  104. Soler-Botija, C.; Gálvez-Montón, C.; Bayés-Genís, A. Epigenetic Biomarkers in Cardiovascular Diseases. Front. Genet. 2019, 10, 950. [Google Scholar] [CrossRef]
  105. Gatsiou, A.; Stellos, K. Dawn of Epitranscriptomic Medicine. Circ. Genom. Precis. Med. 2018, 11, e001927. [Google Scholar] [CrossRef] [Green Version]
  106. Napoli, C.; Benincasa, G.; Donatelli, F.; Ambrosio, G. Precision medicine in distinct heart failure phenotypes: Focus on clinical epigenetics. Am. Heart J. 2020, 224, 113–128. [Google Scholar] [CrossRef]
  107. Ferreira, J.P.; Verdonschot, J.; Collier, T.; Wang, P.; Pizard, A.; Bär, C.; Björkman, J.; Boccanelli, A.; Butler, J.; Clark, A.; et al. Proteomic Bioprofiles and Mechanistic Pathways of Progression to Heart Failure. Circ. Heart Fail. 2019, 12, e005897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Schiano, C.; Costa, V.; Aprile, M.; Grimaldi, V.; Maiello, C.; Esposito, R.; Soricelli, A.; Colantuoni, V.; Donatelli, F.; Ciccodicola, A.; et al. Heart failure: Pilot transcriptomic analysis of cardiac tissue by RNA-sequencing. Cardiol. J. 2017, 24, 539–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Hunter, W.G.; Kelly, J.P.; McGarrah, R.W.; Khouri, M.G.; Craig, D.; Haynes, C.; Ilkayeva, O.; Stevens, R.D.; Bain, J.R.; Muehlbauer, M.J.; et al. Metabolomic Profiling Identifies Novel Circulating Biomarkers of Mitochondrial Dysfunction Differentially Elevated in Heart Failure With Preserved Versus Reduced Ejection Fraction: Evidence for Shared Metabolic Impairments in Clinical Heart Failure. J. Am. Heart Assoc. 2016, 5, e003190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Hamdani, N.; Costantino, S.; Mügge, A.; Lebeche, D.; Tschöpe, C.; Thum, T.; Paneni, F. Leveraging clinical epigenetics in heart failure with preserved ejection fraction: A call for individualized therapies. Eur. Heart J. 2021, 42, 1940–1958. [Google Scholar] [CrossRef]
  111. Gorica, E.; Mohammed, S.A.; Ambrosini, S.; Calderone, V.; Costantino, S.; Paneni, F. Epi-Drugs in Heart Failure. Front. Cardiovasc. Med. 2022, 9, 923014. [Google Scholar] [CrossRef]
  112. Miranda Furtado, C.L.; Dos Santos Luciano, M.C.; Silva Santos, R.D.; Furtado, G.P.; Moraes, M.O.; Pessoa, C. Epidrugs: Targeting epigenetic marks in cancer treatment. Epigenetics 2019, 14, 1164–1176. [Google Scholar] [CrossRef]
  113. Iyer, A.; Fenning, A.; Lim, J.; Le, G.T.; Reid, R.C.; Halili, M.A.; Fairlie, D.P.; Brown, L. Antifibrotic activity of an inhibitor of histone deacetylases in DOCA-salt hypertensive rats. Br. J. Pharmacol. 2010, 159, 1408–1417. [Google Scholar] [CrossRef] [Green Version]
  114. Ngo, V.; Fleischmann, B.K.; Jung, M.; Hein, L.; Lother, A. Histone Deacetylase 6 Inhibitor JS28 Prevents Pathological Gene Expression in Cardiac Myocytes. J. Am. Heart Assoc. 2022, 11, e025857. [Google Scholar] [CrossRef]
  115. McKinsey, T.A. Targeting Inflammation in Heart Failure with Histone Deacetylase Inhibitors. Mol. Med. 2011, 17, 434–441. [Google Scholar] [CrossRef]
  116. Cash, H.L.; McGarvey, S.T.; Houseman, E.A.; Marsit, C.J.; Hawley, N.L.; Lambert-Messerlian, G.M.; Viali, S.; Tuitele, J.; Kelsey, K.T. Cardiovascular disease risk factors and DNA methylation at the LINE-1 repeat region in peripheral blood from Samoan Islanders. Epigenetics 2011, 6, 1257–1264. [Google Scholar] [CrossRef] [Green Version]
  117. Ma, J.; Rebholz, C.M.; Braun, K.V.E.; Reynolds, L.M.; Aslibekyan, S.; Xia, R.; Biligowda, N.G.; Huan, T.; Liu, C.; Mendelson, M.M.; et al. Whole Blood DNA Methylation Signatures of Diet Are Associated With Cardiovascular Disease Risk Factors and All-Cause Mortality. Circ. Genom. Precis. Med. 2020, 13, e002766. [Google Scholar] [CrossRef] [PubMed]
  118. Duan, Q.; McMahon, S.; Anand, P.; Shah, H.; Thomas, S.; Salunga, H.T.; Huang, Y.; Zhang, R.; Sahadevan, A.; Lemieux, M.E.; et al. BET bromodomain inhibition suppresses innate inflammatory and profibrotic transcriptional networks in heart failure. Sci. Transl. Med. 2017, 9, eaah5084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Landmesser, U.; Poller, W.; Tsimikas, S.; Most, P.; Paneni, F.; Lüscher, T.F. From traditional pharmacological towards nucleic acid-based therapies for cardiovascular diseases. Eur. Heart J. 2020, 41, 3884–3899. [Google Scholar] [CrossRef] [PubMed]
  120. Minerath, R.A.; Hall, D.D.; Grueter, C.E. Targeting transcriptional machinery to inhibit enhancer-driven gene expression in heart failure. Heart Fail Rev. 2019, 24, 725–741. [Google Scholar] [CrossRef] [PubMed]
  121. Wu, S.; Hashimoto-Hill, S.; Woo, V.; Eshleman, E.M.; Whitt, J.; Engleman, L.; Karns, R.; Denson, L.A.; Haslam, D.B.; Alenghat, T. Microbiota-derived metabolite promotes HDAC3 activity in the gut. Nature 2020, 586, 108–112. [Google Scholar] [CrossRef]
  122. Tang, W.H.W.; Bäckhed, F.; Landmesser, U.; Hazen, S.L. Intestinal Microbiota in Cardiovascular Health and Disease. J. Am. Coll. Cardiol. 2019, 73, 2089–2105. [Google Scholar] [CrossRef] [PubMed]
  123. Viereck, J.; Thum, T. Circulating Noncoding RNAs as Biomarkers of Cardiovascular Disease and Injury. Circ. Res. 2017, 120, 381–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Summary of the role of chromatin remodeling for the treatment of heart failure.
Figure 1. Summary of the role of chromatin remodeling for the treatment of heart failure.
Biomedicines 11 00579 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kraus, L.; Beavens, B. The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure. Biomedicines 2023, 11, 579. https://doi.org/10.3390/biomedicines11020579

AMA Style

Kraus L, Beavens B. The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure. Biomedicines. 2023; 11(2):579. https://doi.org/10.3390/biomedicines11020579

Chicago/Turabian Style

Kraus, Lindsay, and Brianna Beavens. 2023. "The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure" Biomedicines 11, no. 2: 579. https://doi.org/10.3390/biomedicines11020579

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