Next Article in Journal
PLEKHM1 Overexpression Impairs Autophagy and Exacerbates Neurodegeneration in rAAV-α-Synuclein Mice
Previous Article in Journal
From Aberrant Brainwaves to Altered Plasticity: A Review of QEEG Biomarkers and Neurofeedback in the Neurobiological Landscape of ADHD
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Regulation Through Histone Deacetylation: Implications and Therapeutic Potential in Hepatocellular Carcinoma

Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 37134 Verona, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2025, 14(17), 1337; https://doi.org/10.3390/cells14171337
Submission received: 10 July 2025 / Revised: 21 August 2025 / Accepted: 22 August 2025 / Published: 29 August 2025

Abstract

Hepatocellular carcinoma (HCC) is a leading cause of global cancer-related mortality worldwide. Increasing evidence indicates that epigenetic mechanisms, which are potentially reversible and modifiable by environmental and nutritional factors, play a key role in hepatocarcinogenesis. Histone deacetylases (HDACs) are fundamental epigenetic modulators that regulate chromatin dynamics and ultimately gene transcription with important pathophysiological implications and promising therapeutic perspectives. The role of HDACs is gaining interest for the understanding of HCC development mechanisms and for the potential therapeutic implications of their natural and synthetic inhibitors. This review provides an overview on HDACs classification and their peculiar expression patterns in HCC, with a focus on zinc-dependent histone deacetylases (HDACs). HDAC inhibitors (HDACis), both synthetic and natural-derived compounds, are also discussed for their emerging effects in optimizing the anticancer efficacy of the current therapeutic strategies. Novel dietary-derived and bioactive compounds-based interventions are discussed in the context of HCC management as promising nutri-epigenetic avenues. Targeting HDACs bears a significant therapeutic potential for HCC management while further confirmatory clinical investigation is warranted.

1. Introduction

1.1. Epigenetic Landscape

Epigenetics refers to hereditary changes in gene expression that occur without alterations in the underlying DNA sequence [1,2]. Epigenetic modifications involve heritable changes that allows specific DNA regions to interact or repel regulatory proteins, such as transcription factors or transcription enhancers/silencers, thereby causing the activation or repression of the expression of specific genes in different cell types [1]. Broadly studied epigenetic mechanisms include DNA methylation, histone modifications, and non-coding RNAs (ncRNAs). The main ncRNAs involved in gene regulation and transcription include microRNA, small interfering RNA, long ncRNA, and piwi-interacting RNA [3]. DNA methylation is considered one of the major epigenetic modifications of DNA and was extensively studied in different pathologies [4,5,6,7]. The role of epigenetic features of DNA are especially fascinating because of their potential reversibility by environmental and nutritional factors [8,9,10]. DNA methylation patterns play a dual role in cancer, encompassing both gene-specific and genomic alterations [4,6]. Promoter hypermethylation is a hallmark of transcriptional repression and is often observed in the context of silencing tumor suppressor genes (TSGs) that are critical for cell cycle regulation. On the contrary, global genomic hypomethylation, occurring specifically in repetitive elements, impacts genomic stability and peculiar oncogene activation [11,12]. Recent studies have further emphasized the importance of this methylation pattern in maintaining cellular homeostasis [13,14], liver plasticity [15], iron metabolism and epigenetic regulation in hepatocellular carcinoma (HCC) [4,5,16,17]. The interplay between histone acetylation and deacetylation patterns play a key role in the regulation of the transcriptional machinery in different carcinoma including liver cancer [18,19], colorectal cancer [20] and pancreatic cancer [21]. These epigenetic modifications have been significantly implicated in the development and progression of malignancies through several molecular pathways [22,23,24]. Such alterations are recognized as significant contributors to disease heterogeneity and drug resistance. The epigenetic landscape is dynamically regulated, enabling changes in cell fate through transcriptional reprogramming. Epigenetic modifications are reversible, thus presenting an opportunity for anticancer therapies [25,26] that can target, for example, different histone modifications [27]. This review aims to present an up-to-date understanding of post-translational histone modifications, particularly histone acetylation/deacetylation, in the context of HCC, and to explore their potential implications for the development of targeted therapeutic strategies. Moreover, by considering the key role of nutritional factors in the regulation of epigenetic mechanisms such as those related to HDACs function, this review addresses also the current knowledge on the interaction between HDACs inhibitors and nutrients, highlighting their contribution in current therapeutic strategies and opening to precision therapeutic targeting.

1.2. Post-Translational Histone Modifications

Histone proteins are key players in epigenetics. Histones are bound to DNA in the nucleus and belong to a family of basic proteins with five main components, i.e., H1, H2A, H2B, H3, and H4. Nucleosome is the fundamental unit of chromatin which comprises approximately 147bp DNA wrapped around a histone octamer composed of two copies of H3, H4, H2A and H2B proteins. These nucleosomal histones can be chemically modified [28,29]. Chromatin structure and function is regulated through the action of histone post-translational modifications (PTMs). Almost half a century has passed since Vincent Allfrey first explained the presence of histone acetylation and methylation [30] while their functional significance remained uncertain for many years. Although fundamental breakthroughs have been made in understanding the function of histone PTMs through the identification of the protein machineries that encompass add (write), eliminate (erase) and bind (read) modifications, there are still open questions on how histone PTMs act in chromatin regulation. Histone modifications have been recognized as a pivotal mechanism governing gene transcription that substantially effects cancer induction, progression and metastasis [31]. Extensively studied PTMs include histone lysine acetylation and deacetylation [32,33], lysine and arginine methylation [34,35,36], arginine citrullination, lysine ubiquitination and lysine sumoylation [37,38,39,40,41]. Histone acetylation and deacetylation play a critical and prominent role in epigenetic modulation and regulation of gene expression. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are two opposing classes of enzymes that play crucial functions in the regulation of transcription either by changing chromatin structure or by modulating the activity of specific transcription factors [42].

2. Dynamics of Histone Acetylation/Deacetylation

The chemical properties of histones, which carry a positive charge, promote their strong interaction with the negatively charged DNA, leading to tight binding between the two. Histone acetylation occurs mostly at the N-terminal lysine residues of histones H3 and H4 and leads to transcriptional active chromatin, i.e., open chromatin or euchromatin. Histone acetyltransferases (HATs) transfer the acetyl group of acetyl coenzyme A to histone proteins within the N-terminal tail protruding from the histone core at specific lysine residues, thereby neutralizing the positive charge of histones ultimately weakening the interaction between DNA and histones and relaxing the structure of the nucleosome (Figure 1) and it leads to transcriptional active chromatin, i.e., open chromatin or euchromatin. HATs are classified into two general classes based on their cellular origin and function. Cytoplasmic HATs include B-type HATs that likely catalyze acetylation events associated with the transfer of newly synthesized histones from the cytoplasm to the nucleus. On the contrary, nuclear HATs are A-type HATs catalyzing transcription-related acetylation events [43,44]. In contrast, the action of Histone deacetylases (HDACs), which remove acetyl groups from histone tails, allows histones to tightly bind to negatively charged DNA and repressing gene transcription, leading to a more compact chromatin structure associated with transcriptional repression, i.e., heterochromatin [45,46] (Figure 1). HDACs play a critical role in epigenetic regulation through precise molecular interactions and catalytic mechanisms. These enzymes mediate chromatin remodeling orchestrating intricate cellular processes through sophisticated protein complex formations, including interactions with co-repressors like Sin3A and NuRD complexes [47,48], modulating gene expression across different cellular contexts. HDACs have recently been shown to modify a variety of other proteins that are involved in different cellular processes [26,49,50,51]. The enzymatic mechanism of HDACs involves a nuanced zinc-dependent hydrolysis process, characterized by substrate-specific recognition, conformational changes, and differential regulation across four distinct classes. Each HDAC class exhibits unique structural characteristics and functional specificity, ranging from ubiquitously expressed nuclear Class I HDACs to specialized metabolic sensing Class III sirtuins, ultimately contributing to complex epigenetic networks that regulate transcription, cell cycle progression, and cellular differentiation through precise molecular interactions and catalytic activities [52,53]. Disruption of the balance between HATs and HDACs activities can result in aberrant expression of specific genes. Inhibition of HAT activity may delay the proper timing of target gene expression, whereas blocking HDAC activity could result in prolonged or continuous expression of the same gene. The imbalance between HATs and HDACs has been described having a role in the silencing of TSGs and in cancer induction [22,54,55]. Therefore, tightly regulating the functions of HATs and HDACs is essential to ensure accurate and timely expression of genes involved in signal transduction, as well as in processes such as cell proliferation and death.

3. HDACs Classification

The known histone deacetylase homologs in mammals are eighteen. Histone deacetylases are divided into four classes: class I (HDACs 1, 2, 3, and 8), class II (HDACs 4, 5, 6, 7, 9, and 10), class III (sirtuins, SIRT1-7), and class IV (HDAC11). Figure 2 explains the classification and domain architecture of all HDACs. The first human HDAC was discovered in 1998 and was named HDAC1 [56], followed by HDAC2 [57] and HDAC3 [58]. In humans, the HDAC family characterization was last updated in 2002 with the discovery of HDAC11 [59]. All classes of HDACs are numbered according to their chronological order of discovery and are grouped according to their homology to yeast orthologues. Histone deacetylases from 1 to 11 are zinc dependent while Class III sirtuins require NAD+ as a cofactor [60]. HDAC1 is considered the prototype of the HDAC family. Class I and II HDACs are widely studied and reported to be associated with cancer pathogenesis, thus, this literature review is focused on these zinc-dependent histone deacetylases.

3.1. Class I HDACs

Class I mammalian HDACs include HDAC 1, 2, 3, and 8. These HDACs are composed of an entirely conserved deacetylase domain and possess sequence similarity to the yeast HDAC, reduced potassium dependency protein (Rpd3), which is responsible for the deacetylation of lysine residues on the N-terminal part of the core histones in yeast [61]. Class I HDACs are primarily located in the nucleus, where they present strong deacetylase activity toward histones. They mainly operate in groups where multiple HDACs like HDAC1 and HDAC2 exist together in at least three different multiprotein complexes: nucleosome remodeling and deacetylase complex (NuRD), corepressor of REST (CoREST) and transcriptional regulatory protein Sin3A. The non-conserved C-terminal region of HDAC3 is required for both deacetylase activity and transcriptional repression. HDAC3 activity depends on SMRT (silencing mediator for retinoic acid and thyroid hormone receptors) and N-CoR (nuclear receptor co-repressor) which contain deacetylase activating domains [62]. The last member of HDAC class I is HDAC8 with a structure simpler than other class I HDACs, consisting mainly of a catalytic domain with a central nuclear localization signal (NLS) which functions alone [63,64,65]. Nevertheless, its regulatory complex and protein interactions remain unknown. In addition to histones, Class I HDACs deacetylate several non-histone proteins because of their predominant nuclear localization. Table 1 shows the histone and non-histone substrates as well as the cellular localization of all HDACs.

3.2. Class II HDACs

Class II HDACs have sequence homology with yeast HDAC1, the putative catalytic subunit of histone deacetylase complex in yeast and exhibit a conserved deacetylase domain at their C-terminus [66]. Generally, class II is subdivided into two sub-classes: Class IIa and Class IIb, based on sequence analysis. HDAC class IIa involves HDAC4, 5, 7, and 9 which possess a unique adapter domain in the N-terminus that forms a binding site for the DNA-binding transcription factor and myocyte enhancer factor 2 (MEF2). Nucleocytoplasmic shuttling is the distinctive characteristic of class II HDACs which demonstrates cell type specificity and signal dependence. Class IIa HDACs also form a large complex with the SMRT/N-CoR-HDAC3. Intrinsically, this class has low enzymatic activity. Class I HDACs have a conserved tyrosine residue in the catalytic site whereas in class IIa HDACs this is substituted with histidine [67,68]. Thus, class IIa HDACs may function as deacetylases with low enzymatic activity or may possess specific targets that have yet to be identified. Ectopic expression studies of HDAC5 and HDAC7 revealed novel nuclear structures called matrix associated deacetylase (MAD) bodies whose formation is determined by deacetylase activity. These MAD bodies contain several proteins and also components of the NuRD/Mi2/NRD and Sin3/HDAC complexes [69]. HDAC9 catalytic domain is located on the N-terminus, and it also possesses a conserved deacetylase domain. When HDAC9 is recruited to a promotor, this results in the repression of gene activity through deacetylation of histones. Additionally, HDAC9 also interacts with MEF2 [70]. There are three reported HDAC9 splice variants with different protein functions distinguished as HDAC9a, HDAC9b, and HDRP/HDAC9c [71].
Class IIb HDACs comprise HDAC6 and HDAC10. HDAC6 contains two deacetylase domains and a C-terminal zinc finger ubiquitin-binding domain commonly known as HDAC6, USP3-, Brap2-related zinc finger motif (HUB) [72]. It comprises distinct structural domains such as nuclear localization signal region (NLS), tandem deacetylation catalytic regions (DD1, DD2), serine–glutamate-containing tetradecapeptide repeat region (SE14), leucine-rich nuclear export signal regions (NES1, NES2) and ubiquitin-binding zinc finger structure (ZnF-UBP). Despite containing a nuclear localization signal, HDAC6 cytoplasmic localization is primarily governed by NES and SE14, facilitating its translocation and anchoring in the cytoplasm [73,74]. Structurally, HDAC10 features an N-terminal catalytic domain, a nuclear export signal (NES), and a possible second catalytic domain at its C-terminus. It also contains two putative Rb binding domains. HDAC10 also operates as a transcriptional repressor, capable of shuttling between the nucleus and cytoplasm. Analysis of protein sequence identity shows that HDAC10 is most closely related (37% overall similarity) to HDAC6 [75]. Although HDAC10 demonstrates autonomous deacetylase activity when recombinantly expressed, its capability to interact with multiple HDACs (1, 2, 3, 4, 5, and 7) but not with HDAC6 proposes the idea that it might primarily function as a recruiting protein.

3.3. Class III HDACs

The class III HDACs share homology with the yeast silent information regulator 2 (Sir2), a protein essential for transcriptional silencing, and are evolutionarily conserved across a broad range of species from bacteria to humans. A defining feature of class III HDACs is the presence of a deoxyhypusine synthase-like domain capable of binding NAD and FAD cofactors, distinguishing them from other HDAC classes. In humans, seven Sir2 homologs, collectively known as sirtuins (SIRT1-7), have been identified. Notably, sirtuins possess additional enzymatic functions, including mono-ADP-ribosyltransferase activity, beyond their histone deacetylase role. Sirtuins share 22–50% overall amino acid sequence similarity and 27–88% resemblance in their conserved catalytic domains [53,60]. These proteins are localized in various cellular compartments, including the nucleus (SIRT1, SIRT2, SIRT3, SIRT6, and SIRT7), the cytoplasm (SIRT1 and SIRT2), and the mitochondria (SIRT3, SIRT4, and SIRT5) [76].

3.4. Class IV HDACs

Class IV histone deacetylase includes only HDAC11, which is homologous to yeast Hos3 and shares a catalytic domain with both HDACs of class I and class II. HDAC11 exhibits sequence homology to Rpd3 and HDAC1 proteins [61]. The highly conserved dynamic residues of HDAC11 in the catalytic core region also share similarity with both class I and II HDACs. HDAC11 is the shortest isoform and is primarily composed of the core catalytic domain that exhibits exclusive deacetylase activity [77]. In vivo, HDAC11 can also form complexes with HDAC6, but the 3D structure is not yet available. The expression of HDAC11 can be regulated by temperature [78].

4. HDACs Expression Patterns in Hepatocellular Carcinoma

Liver cancer is the sixth most diagnosed cancer worldwide and the third leading cause of cancer-related deaths with over 750,000 deaths annually. The incidence and mortality rate of liver cancer are two to three times higher in men than women [79].
Hepatocellular carcinoma is the most common type of primary liver cancer comprising the 75–85% of liver cancer cases. The age of HCC occurrence varies globally but the median age considered for its onset is >60 years [80]. The primary risk factors for HCC include HBV or HCV chronic infection, aflatoxin exposure, heavy alcohol consumption, excess body weight, type 2 diabetes, and smoking [79]. Although hepatocellular carcinoma predominantly develops in cirrhotic livers, a notable proportion of cases (approximately 15–20%) arises in non-cirrhotic individuals, particularly in the context of metabolic dysfunction-associated steatotic liver disease [81]. The pathogenesis of HCC is a complex, multifactorial process influenced by genetic alterations, epigenetic modifications, and environmental exposures that collectively contribute to malignant transformation. The treatment options range from locoregional treatments, including ablation, and surgical resection, to liver transplantation and systemic therapy for more advanced stages cases [82].
Several studies have reported that histone deacetylases are linked with tumor progression in different cancer types including HCC, where HDACs are usually highly overexpressed [83,84]. Overall, HDACs upregulation, in particular that of zinc-dependent HDACs, is associated with cancer cell proliferation, promotion of angiogenesis, and metastases and inhibition of T-cell tumor infiltration (Figure 3). HDAC1 is the most widely studied member of the HDAC family, demonstrating anomalous high expression in HCC tissues and cell lines [85,86,87]. Likewise, Buurman and colleagues have reported upregulated expression of HDAC1-3 in both HCC patients and cell lines. They demonstrated that the HDAC inhibition in HCC cells through Trichostatin A induces apoptosis and decreases proliferation, partly by upregulating miR-449 leading to reduced ERK1/2 signaling [88]. Similarly, elevated HDAC1 and HDAC2 expression has been associated with increased mortality in HCC patients and in vitro studies showed that simultaneous knockdown of HDAC1/2 resulted in reduced cell proliferation, colony formation, and survival in HCC cell lines [89]. Lachenmayer and colleagues reported the significant upregulated expression of HDAC2, HDAC4, and HDAC11 in a cohort of liver cancer patients, whereas HDAC3 and HDAC5 were found to be associated with copy number gains in human HCC [90]. Another study reported the elevated expression of HDAC5 in HCC and its downregulation in HCC cell lines contributed to apoptosis induction and cell cycle arrest [91]. Furthermore, HDAC5 has been shown to facilitate HCC metastasis under hypoxia by repressing HIPK2, stabilizing hypoxia-inducible factor 1α (HIF1α), and promoting epithelial–mesenchymal transition (EMT) and angiogenesis [92]. LukS-PV, a leucocidin, inhibited HCC cell migration by downregulating HDAC6, thus enhancing α-tubulin acetylation in a concentration-dependent manner [93]. AR420626, a selective agonist of G-protein coupled receptor GPR41/FFA3, suppressed the growth of HCC cells by inducing apoptosis thereby reducing the expression of HDAC2-7 with an increase in histone H3 acetylation [94]. High expression of HDAC8 was observed in both tumor tissues and liver cancer cell lines while its suppression effectively disrupted cancer cell dynamics by reducing cell proliferation and triggering programmed cell death [95]. Moreover, Yang and colleagues highlighted that HDAC8 inhibition can increase T cell infiltration into tumors and decrease regulatory T cells (Tregs), thus improving the immune response against HCC [96]. An in vivo study reported a positive correlation between HDAC9 and programmed death-ligand 1 (PD-L1) expression levels. Moreover, the combination of high levels of HDAC9 and of PD-L1 was associated with a decreasing overall survival (OS) in patients with HCC [97]. HDAC11 deacetylates Egr-1, consequently suppressing the expression of p53 and promoting liver cancer progression [98] (Figure 3). Nevertheless, studies investigating the role of HDAC11 in liver cancer are limited.
Current research has highlighted the profound significance of epigenetic regulation in cancer pathogenesis, particularly liver cancer. Epigenetic alterations have displayed a unique therapeutic opportunity, offering a promising avenue for developing novel strategies that can potentially interrupt or reverse cancer development at the molecular level [99]. Therefore, altered expression of HDACs could play an active role in tumor onset and progression and make them attractive candidate targets for anticancer drugs and therapies.

5. Therapeutic Implications: Promising Synthetic HDAC Inhibitors in HCC

HDACs are among the most promising therapeutic targets for cancer treatment. HDAC inhibitors are the compounds targeting epigenetic dysregulation—a hallmark of cancer. HDAC inhibitors have direct anti-tumor effects, modulating the tumor microenvironment. Therefore, HDAC inhibitors have been targeted for the development of anticancer strategies as well as therapies for human diseases [100,101,102]. The utilization of multiple therapeutic agents in the treatment of cancer is another useful anticancer strategy [103,104]. Currently, clinical trials are demonstrating the effectiveness of HDAC inhibitors (HDACis) alone and in combination with other drugs [105,106]. A few HDAC inhibitors have been approved by the Food and Drug Administration (FDA) for the treatment of different types of cancer [107]. Although, at present, no HDACi is approved for the treatment of HCC, some are currently the object of clinical or preclinical trials for HCC treatment. In particular Resminostat and several HDACis, when used in combination with other treatment modalities, have also demonstrated enhanced therapeutic efficacy against HCC [108,109,110]. Treatment with HDAC inhibitors was shown to upregulate the expression of checkpoint inhibitors in tumor cells. A selective HDAC1/2/3 inhibitor (CXD101) has overcome resistance to immune checkpoint therapy in liver cancer by activating an IFNγ/STAT1 signaling and GSDME-mediated pyroptosis circuit, effectively converting immune-excluded tumors into inflamed ones that respond better to treatment. This finding has progressed to a phase-II clinical trial and could provide a new treatment strategy for patients with immunotherapy-resistant liver cancer [111]. HDAC inhibitors have also been investigated in combination with oncolytic virotherapy to promote tumor immunogenicity in hepatocellular carcinoma [112,113].
Another emerging field of research focuses on nicotinamide N-methyltransferase (NNMT), a NAD regulator, that influences the activity of sirtuins, NAD-dependent histone deacetylases. NNMT could be the target of specific inhibitors that can be used in liver cancer treatment [114,115]. These inhibitors function through multiple mechanisms including immunomodulation, apoptosis induction and cell cycle arrest with current clinical trials testing both monotherapy and combination approaches with standard chemotherapy, immunotherapy and targeted agents [116,117].
In the following paragraphs, we will illustrate the recent advances found in the literature regarding selected synthetic HDACis with, a focus on those targeting zinc-dependent HDACs, highlighting their antitumoral function and clinical relevance related to HCC treatment. Table 2 illustrates the most important HDACis with related characteristics.

5.1. Resminostat

Resminostat is an oral, potent bioavailable pan-inhibitor of class I and II HDACs. It is a member of benzenes, a hydroxamic acid, a member of pyrroles, a sulfonamide, a tertiary amino compound, and an enamide. Resminostat binds to and inhibits HDACs leading to an accumulation of highly acetylated histones. This results in the induction of chromatin remodeling, transcription of tumor suppressor genes, tumor cell apoptosis, and inhibition of tumor cell division. Resminostat has shown particular promise in addressing the epigenetic dysregulation common in HCC development and has been used in clinical trials studying the treatment of many cancers [133,134,135]. Clinical studies with Resminostat in HCC have produced encouraging results, particularly in combination with Sorafenib. Its development program has specifically focused on HCC patients, with trials examining its potential in both first line and second-line settings, making it one of the most thoroughly studied HDAC in the context of liver cancer. In earlier studies, Resminostat showed additive or synergistic activities in combination with other novel pharmaceutical agents and conventional chemotherapeutic agents for HCC treatment [136]. The SHELTER study where Resminostat was investigated as a second-line therapy for advanced HCC reported that combination of Resminostat with Sorafenib improved progression-free survival (PFS) to 6.5 months and overall survival (OS) to 8.0 months as compared to 1.8 months PFS and 4.1 months OS with Resminostat alone, suggesting its potential in overcoming therapy resistance and exhibiting promising clinical outcomes in terms of safety and survival [118]. Resminostat also exhibited potent anti-HCC activity by activating the mitochondrial permeability transition pore (mPTP) dependent apoptosis pathway, which led to cytochrome C release and caspase-9 activation in HCC cell lines and primary HCC cells. When combined with Sorafenib, Resminostat synergistically enhanced the anti-tumor effects through amplified activation of the mitochondrial apoptosis pathway [119]. In another study, Resminostat was found to stimulate mesenchymal HCC cells to have a more epithelial phenotype with lower invasive activity, downregulating CD44 (a cancer stem cell marker) that may contribute to the sensitization to Sorafenib-induced apoptosis, preventing HCC cells growth in vitro [120]. It is worth noting that in a Phase I/II trial comparing first-line Sorafenib plus Resminostat versus Sorafenib alone in East Asian patients with advanced HCC, no significant improvement in overall survival with the combination therapy was found. However exploratory analyses suggested potential benefits in patients with normal to high baseline platelet counts or HBV-related HCC [137]. Previously, researchers demonstrated that inhibiting mTOR significantly enhanced the anticancer effects of Resminostat in hepatocellular carcinoma cells, with the combination activating the mitochondrial apoptosis pathway more potently. This finding revealed that mTOR activation may be a key resistance mechanism against HDAC inhibitor therapy in liver cancer, suggesting that combining HDACis with mTOR inhibitors could be a promising therapeutic strategy [138].

5.2. Vorinostat

Vorinostat is a histone deacetylase inhibitor of class I and II, a hydroxamic acid, and a dicarboxylic acid diamide. It has a role as an apoptosis inducer and an antineoplastic agent. It binds to the zinc atom in the catalytic domain of HDAC enzymes and projects its phenyl ring out of the catalytic site onto the surface of HDAC enzymes. Thus, HDACs cannot remove the acetyl group of histone and non-histone proteins, provoking the accumulation of acetylated proteins with effects in many cellular functions, such as cell cycle arrest in cancer cells. It has been approved by FDA for the treatment of cutaneous T-cell lymphoma [139]. Vorinostat, also known as suberoylanilide hydroxamic acid (SAHA), effectively overcomes Lenvatinib resistance in HCC by suppressing the PI3K/AKT signaling pathway, revealing that AKT activation contributes to Lenvatinib resistance, while HDAC inhibition restores sensitivity to treatment. Both in vitro and in vivo findings support the potential of Vorinostat, and Lenvatinib combination therapy, as a promising strategy to enhance therapeutic efficacy and counteract drug resistance in HCC [121]. Vorinostat combined with the DNMT inhibitor Decitabine has also shown effective results in solid tumors, including HCC. It has synergistically enhanced the antitumor effects in HCC cells by inducing apoptosis and autophagy mediated by caspase-3 activation, Bcl-2 downregulation, and autophagic flux upregulation [122]. Studies demonstrating Vorinostat ability to induce apoptosis and prevent cell growth in HCC cell lines include that by Li and colleagues [123] who reported that Vorinostat triggers apoptosis through caspase activation and downregulation of anti-apoptotic proteins, while Sanaei and colleagues [140] showed its role in inhibiting HCC cell proliferation by modulating key oncogenic pathways.

5.3. Panobinostat

Panobinostat is a hydroxamic acid, a member of cinnamamides, a secondary amino compound, and a methylindole. It acts as an HDACi, an antineoplastic agent, and an angiogenesis-modulating agent, and it has been approved by the FDA in 2015. It is used for the treatment of relapsed/refractory (R/R) multiple myeloma [141], and recently it has been used to also treat brain cancer [142]. Some studies have reported Panobinostat’s promising anti-HCC efficacy both in vitro and in vivo with minimal adverse effects. Liu and colleagues [124] have reported that Panobinostat treatment, particularly when combined with NAT10 (a histone acetyltransferase) silencing, significantly reduced HCC cell lung metastasis as well as noticeably decreased liver tumor growth in xenograft and lung metastasis mouse models. Panobinostat also showed enhanced anti-tumor effect in HCC cells when combined with radiotherapy [143]. Some studies report the enhanced induction of apoptosis as well as proton sensitization when treated with Panobinostat in HCC cells [125,126] in addition to autophagy-induced cell death [144]. Panobinostat treatment led to the silencing of 5 out of 6 miRNA of the oncogenic miR17-92 cluster in HCC cell lines ultimately reducing carcinogenesis [145].

5.4. Romidepsin

Romidepsin is a cyclodepsipeptide classified as an organic disulfide and a heterocyclic antibiotic, which has been approved by FDA in the treatment of cutaneous T-cell lymphoma and peripheral T-cell lymphoma [146]. The possible effectiveness of Romidepsin in the treatment of HCC is still under investigation but preliminary studies have yielded promising results. In vitro studies demonstrate that Romidepsin inhibits cell cycle and induces apoptosis [127,147] in HCC cell lines. These findings were corroborated in tumor xenograft mouse models where Romidepsin effectively suppressed HCC cell growth. Further studies on diethylnitrosamine (DEN)-induced HCC mouse models have shown that Romidepsin treatment led to a significant suppression of hepatocellular tumorigenesis [128,148]. A retrospective study demonstrated the pro-apoptotic effects of Romidepsin in HCC cells by mediating G2/M arrest through the activation of JNK/MAPK and Erk/MAPK pathways as shown in an in vivo mouse model and in an in vitro drug sensitivity assay [149]. A recent study on the combination therapy approach, involving the co-administration of Romidepsin with PARP inhibitor Olaparib, showed an efficient decrease in HCC growth both in vivo and in vitro [129].

5.5. Belinostat

Belinostat is a hydroxamic acid, a sulfonamide, and an olefinic compound with antineoplastic activity. In 2014, it has been approved by the FDA, and it is widely used to treat several cancers such as T-cell lymphoma, soft tissue sarcoma, mesothelioma, colorectal cancer, B-cell lymphoma, liver cancer and refractory or relapsed PTCL [139]. Belinostat in combination with another drug, Bortezomib, has shown antiproliferative and cytotoxicity properties towards HCC cell lines [130]. Moreover, a previous study also demonstrated that Belinostat inhibits growth and induces apoptosis in HCC cell lines with the partial restoration in expression of some silenced tumor suppressor genes [131]. Belinostat has also shown to enhance antitumor immunity when combined with the immune checkpoint inhibitor anti-CTL4 in murine HCC models [132]. Belinostat also showed enhanced cytotoxicity against HCC cell lines even in over-expressed ornithine decarboxylase 1 (ODC1) HCC cells [150].

6. Natural Dietary Components as HDAC Inhibitors

Natural compounds from various dietary and plant sources can serve as effective histone deacetylase inhibitors (Figure 4), providing a novel avenue for chemoprevention and therapy [151,152,153,154]. These bioactive compounds show both selective and multiple target actions against HDAC enzymes while maintaining low toxic effects (Table 3). Intersection of nutrition and epigenetics ascertains that several dietary components function as molecular switches, controlling gene expression patterns in different pathologies including cancer.

6.1. Short-Chain Fatty Acids (SCFAs)

Short-chain fatty acids are produced by gut microbiota through the fermentation of dietary fibers. SCFAs are organic acids with carbon chain containing less than six carbon atoms. Acetate, butyrate, propionate and valeric acid are the most common SCFAs. They act as an endogenous HDAC inhibitors [155]. Acetate suppressed IL-17A production by type 3 innate lymphoid cells through the inhibitory activity on HDAC and induction of Sox13 (SRY-box transcription factor 13) acetylation in a dose-dependent manner. Acetate administration with PD-1 therapy delayed tumor growth and improved anticancer immunity in the HCC mouse model [156]. Propionate upregulated activation of caspase-3 and cisplatin-induced apoptosis through a TNF-α-induced pathway. Augmented acetylation of H3 by reducing the expression of HDACs via GPCR activation is also reported. Sodium propionate in combination with cisplatin significantly increased apoptosis and suppressed growth of HCC xenografts [157]. A high concentration of sodium butyrate (NaB) in sorafenib-resistant HCC cells showed reduced cell viability and increased cell apoptosis as well as the decreased expression of miRNA-7641 and miRNA-199 [158]. Butyrate in combination with chloroquine exerted cytotoxic activity in HCC tumors and increased the production of reactive oxygen species. Variation in the levels of autophagy markers are also reported [159]. The NaB and DNMT inhibitor indicated inhibition in cell growth and induction in apoptosis while upregulating the expression of some TSGs and apoptotic regulators [160]. The HDAC inhibitor NaB upregulated the expression of tumor suppressor gene Cyld in HCC cells through enhanced histone acetylation [161]. Increased expression of sarco-endoplasmic reticulum Ca2+-ATPase 3 (SERCA3) is detected through NaB and TSA treatment in HCC cells with an increase in H3 acetylation on the respective SERCA3 gene [162]. Butyrate, selectively, induces apoptosis to cancerous cells while supporting normal cells, with effects varying according to cell type and dose. It is evident that at high concentrations (2mM), butyrate exhibited apoptosis of both cancerous and non-cancerous colonocytes. Low availability has limited its clinical use but strategies like nanodelivery, structural analogs, coupling with phytochemicals, and serine conjugation can improve its therapeutic potential [163,164]. Valeric acid (VA) has shown significant anticancer effects including reduced colony formation, migration, and invasion of liver cancer cells. This study also investigated the role of encapsulated valeric acid for liver cancer cells in xenograft mouse models, indicating efficient results. Both VA and encapsulated VA showed reduced HDAC activity as well [165]. As short-chain fatty acids have notable anticancer and anti-inflammatory properties and improve physiological function, studies are reported regarding SCFA oral supplementation as a therapeutic strategy. However, several factors can reduce the bioavailability of SCFAs over time, due to their potential decline in the gut, such as aging, metabolic syndrome, and cardiovascular diseases [166,167,168].

6.2. Curcumin

Curcumin is a polyphenolic compound, naturally occurring in turmeric spice, that also acts as a natural HDAC inhibitor. Curcumin exerted its tumor-initiating cell (TIC) depleting activity partially through inhibiting NF-kB signaling. Curcumin-sensitive HCC cells showed reduced stemness and tumorigenicity by downregulating HDAC class I and II [169]. Curcumin and Trichostatin A can reduce cell growth and induce apoptosis in HCC cells. Curcumin showed dose- and time-dependent substantial antiproliferative effects, more significant apoptotic effect than TSA and the upregulation of ERα gene expression [170]. The bioavailability of curcumin is compromised, due to its rapid metabolism and its low serum and tissue levels. Studies are being conducted to enhance its bioavailability using delivery approaches including nanoparticles, liposomes, or its derivatives. Curcumin has shown no harmful consequences at high dosage. Curcuminoids are FDA-recognized as safe and well-tolerated in clinical trials at doses of 4000–8000 mg [171,172].

6.3. Sulforaphane

Sulforaphane is an isothiocyanate found mostly in cruciferous vegetables and functions as a dietary HDACi. Sulforaphane significantly downregulated the expression of HDAC5 and HDAC11 leading to the upregulation of several genes involved in DNA damage response and cell cycle. Sulforaphane also determined reduced cell viability and induced apoptosis in HCC cells and it possibly regulates oncogenic transcription factors via methylation of their DNA binding motifs [173]. In acute liver failure mouse models, sulforaphane has been shown to reduce HDAC6 expression by inhibiting ferroptosis through activation of NRF2 [174]. Since NRF2 plays a central role in triggering the antioxidant response in various cell types, this mechanism is particularly relevant given the key involvement of oxidative stress regulation in the ferroptosis process [175,176,177,178].
Systemic availability of sulforaphane is highly dependent on the form of administration as well as the presence and activity of myrosinase, an enzyme for plant defense. Fresh broccoli sprouts retain active myrosinase and are the most effective dietary source of sulforaphane [179,180]. No adverse side effect has been observed with the high dose of sulforaphane (50 μM) on HCC cells except mild reversible effects exhibiting minimal toxicity [181].

6.4. Resveratrol

Resveratrol is a polyphenol abundant in grapes and berries, and it acts as an HDAC suppressing compound. Resveratrol suppressed all eleven human HDACs, from HDAC1 to 11 in a dose-dependent manner in an in vitro analysis and showed antiproliferative effects on HCC cell lines. Interestingly, Resveratrol showed significant HDAC inhibition and consequently histone hyperacetylation in HepG2 cells. Further testing on human blood detected a resveratrol-mediated inhibition of HDAC in PBMCs, demonstrating its high concentration in blood can modulate HDAC activity [182]. Nowrasteh and colleagues evaluated the effect of nutrient rich fruit extract including trans-resveratrol in cancer induced mice models and demonstrated the significant reduction in expression of HDAC2, HDAC3 and a few DNMTs [183]. Although resveratrol possesses several beneficial biological activities [184], it has low bioavailability and solubility. A dose of 100–500 mg is recommended for its supplementation to avoid potential risks. Resveratrol bioavailability enhancement of oral administration when in combination with other compounds is under investigation [185,186].

6.5. Genistein

Genistein is an isoflavone, a type of flavonoid and a natural HDACi present in soy products and legumes. It has shown apoptotic and antiproliferative effects alone and in combination with TSA on HCC cell lines. In addition, it also directed the re-expression of silenced gene estrogen receptor alpha ERα in the HCC cell line [187]. Sanaei and colleagues had also shown the restoration of gene expression of some DNA methyltransferases through Genistein and TSA in addition to cell growth inhibition and apoptosis induction. They also studied the effects of Genistein alone and in combination with Valproic acid [188,189]. According to the US-FDA, daily consumption of 25 g of soy containing genistein is recommended without side effects. Despite showing promising anticancer effects [190,191], pharmacokinetic analysis of genistein has shown low oral bioavailability, which is a major hurdle in its application in clinical studies [192].
Table 3. Common natural HDAC inhibitors studied in HCC models.
Table 3. Common natural HDAC inhibitors studied in HCC models.
Compound and StructureMolecular
Formula (MW)
SourceStudy ModelKey FindingsRef.
Acetic acid
Cells 14 01337 i006
C2H4O2
(60.05 g/mol)
Natural SCFA;
Bifidobacteria
In vivo and ex vivoAnti-tumor activity, efficient immunity[156]
Propionic acid
Cells 14 01337 i007
C3H6O2
(74.08 g/mol)
Natural SCFA; BacteroidetesCell lines; Xenograft mouse modelTumor regression, enhanced apoptosis and H3 acetylation[157]
Butyric acid
Cells 14 01337 i008
C4H8O2
(88.11 g/mol)
Natural SCFA; FirmicutesCell linesCytotoxic activity, increased
acetylation
[159,161,162]
Valeric acid
Cells 14 01337 i009
C5H10O2
(102.13 g/mol)
Natural SCFA;
Clostridia
Cell lines; Xenograft mouse modelAntitumor effects[165]
Curcumin
Cells 14 01337 i010
C21H20O6
(368.4 g/mol)
Turmeric
(Curcuma longa)
Cell linesApoptotic and antiproliferative activity[169,170]
Sulforaphane
Cells 14 01337 i011
C6H11NOS2
(177.3 g/mol)
Cruciferous
vegetables
Cell linesApoptosis
induction, gene
regulation
[173]
Resveratrol
Cells 14 01337 i012
C14H12O3
(228.24 g/mol)
Red grapesIn vitro and ex vivoAntiproliferative
effects
[182]
Genistein
Cells 14 01337 i013
C15H10O5
(270.24 g/mol)
Soy productsCell linesCell proliferation
inhibition, increase apoptosis
[187,188]

7. Future Perspectives and Conclusions

Recent advances in the development of novel epigenetic-targeted drugs demonstrated significant potential in preclinical and clinical trials [26,27]. These therapeutic approaches aim to modulate aberrant epigenetic modifications and restore physiological cellular function, offering a promising frontier in cancer treatment.
Despite the plethora of studies reported in this review, some questions remain uncovered and are still matters of debate such as the cytoplasmic effects of HDACi, the interplay between their anti-apoptotic and autophagic effects, overall effect on epigenetic crosstalk during treatment, etc. Another critical issue is related to non-histone targets of HDACs, since HDAC inhibition can trigger widespread downstream effects, limiting therapeutic specificity. Moreover, recently, a study reported that radiotherapy can impart a long-term epigenetic memory in skin cells. It is an open and critical question whether tumor cells similarly retain an epigenetic memory of treatment. This memory could be detrimental or potentially advantageous and its implications are completely unknown [193]. Regarding a more general perspective, while the goal is often “epigenome normalization,” it is still not proven whether this is the inhibitors’ main mechanism of action due to their pleiotropic nature.
Nevertheless, several challenges remain in optimizing HDAC inhibitor therapy regardless of promising results. Studies on natural HDAC inhibitors are predominantly focused on in vivo and in vitro experimentations, lacking robust and randomized trials in humans. Their heterogenous mechanism of action, low bioavailability, rapid metabolism, and short half-lives interfere with their efficacy and applicability. Conversely, synthetic HDAC inhibitors are extensively evaluated in clinical studies but face several challenges like isoform selectivity, pharmacokinetics and epigenetic plasticity. Furthermore, limited efficacy as a monotherapy, off-target interactions, and disparity in clinical trial design are other aspects of their limited therapeutic capability. Another major limitation in the real-life use of HDACs inhibitors refers to the limited knowledge in defining the most appropriate dosage as well as timing, meaning the stadium of disease, in which HDACs could reach the most beneficial effects.
Current research should focus on developing more selective and natural HDACis, identifying reliable biomarkers for patient stratification and optimizing combination strategies. The emergence of precision medicine approaches, coupled with advanced molecular profiling techniques, is enabling more personalized therapeutic strategies, potentially leading to improved clinical outcomes for hepatocellular carcinoma patients. The future of HDAC-targeted therapy likely lies in rational combination strategies incorporating HDACis with conventional chemotherapeutics, immunotherapies, oncolytic viral therapy and other epigenetic regulators. Translational research should also explore next-generation HDACis with enhanced selectivity and reduced off-target effects ensuring better clinical applicability. Healthy lifestyle and nutrient rich diet based preventive approaches could also contribute to epigenetic resilience and reduced cancer risk. Characterization of the cellular and molecular mechanisms by which specific epigenomic reprogramming elicits anti-tumor immunity is instrumental in the rational development and clinical translation of selective HDAC-targeted immunotherapy. Conclusively, HDAC inhibition remains a promising therapeutic avenue for hepatocellular carcinoma. However, maximizing clinical efficacy demands further elucidation of mechanisms of resistance, patient stratification and identification of synergistic drug combinations. Given the complexity of the epigenetic network and sheer number of potential targets, a systematic approach identifying and validating potential drug targets is imperative to focus on drug development and confirm the potential of this strategy.

Author Contributions

Writing—original draft preparation, K.S., S.U., F.A., P.P., R.B., G.A. and M.M.; writing—review and editing, S.F., A.C. and S.M.; visualization, K.S., F.A. and S.U. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

This study was performed, in part, in the LURM (Laboratorio Universitario di Ricerca Medica) Research Center, University of Verona, Italy.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CoRESTCorepressor of REST
DENDiethynitrosamine
EMTEpithelial–mesenchymal transition
HATsHistone acetyltransferases
HCCHepatocellular carcinomas
HIF1αHypoxia-inducible factor 1 α
HDACiHistone deacetylase inhibitor
HDACHistone deacetylase
MADMatrix associated deacetylase
MEF2Myocyte enhancer factor 2
mPTPMitochondrial permeability transition pore
N-CoRNuclear receptor co-repressor
NaBSodium butyrate
ncRNAnon-coding RNA
NESNuclear export signal
NLSNuclear localization signal
NuRDNucleosome remodeling and deacetylase complex
ODC1Ornithine decarboxylase 1
OSOverall survival
PD-L1Programmed death ligand 1
PFSProgression free survival
PTMPost-translational modification
Rpd3Reduced potassium dependency protein 3
SAHASuberoylanilide hydroxamic acid
SCFAShort-chain fatty acid
Sir2Silent information regulator 2
TregRegulatory T cell
TSATrichostatin A
TSGTumor suppressor gene
VAValeric acid
ZnFZinc finger domain

References

  1. Choi, S.-W.; Friso, S. Epigenetics: A New Bridge between Nutrition and Health. Adv. Nutr. 2010, 1, 8–16. [Google Scholar] [CrossRef]
  2. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic Modifications: Basic Mechanisms and Role in Cardiovascular Disease. Circulation 2011, 123, 2145–2156. [Google Scholar] [CrossRef]
  3. Li, Y. Modern Epigenetics Methods in Biological Research. Methods 2021, 187, 104–113. [Google Scholar] [CrossRef]
  4. Udali, S.; Guarini, P.; Moruzzi, S.; Ruzzenente, A.; Tammen, S.A.; Guglielmi, A.; Conci, S.; Pattini, P.; Olivieri, O.; Corrocher, R.; et al. Global DNA Methylation and Hydroxymethylation Differ in Hepatocellular Carcinoma and Cholangiocarcinoma and Relate to Survival Rate. Hepatology 2015, 62, 496–504. [Google Scholar] [CrossRef]
  5. Udali, S.; Guarini, P.; Ruzzenente, A.; Ferrarini, A.; Guglielmi, A.; Lotto, V.; Tononi, P.; Pattini, P.; Moruzzi, S.; Campagnaro, T.; et al. DNA Methylation and Gene Expression Profiles Show Novel Regulatory Pathways in Hepatocellular Carcinoma. Clin. Epigenetics 2015, 7, 43. [Google Scholar] [CrossRef]
  6. Udali, S.; De Santis, D.; Ruzzenente, A.; Moruzzi, S.; Mazzi, F.; Beschin, G.; Tammen, S.A.; Campagnaro, T.; Pattini, P.; Olivieri, O.; et al. DNA Methylation and Hydroxymethylation in Primary Colon Cancer and Synchronous Hepatic Metastasis. Front. Genet. 2018, 8, 229. [Google Scholar] [CrossRef]
  7. Friso, S.; Carvajal, C.A.; Fardella, C.E.; Olivieri, O. Epigenetics and Arterial Hypertension: The Challenge of Emerging Evidence. Transl. Res. 2015, 165, 154–165. [Google Scholar] [CrossRef]
  8. Moruzzi, S.; Guarini, P.; Udali, S.; Ruzzenente, A.; Guglielmi, A.; Conci, S.; Pattini, P.; Martinelli, N.; Olivieri, O.; Tammen, S.A.; et al. One-Carbon Genetic Variants and the Role of MTHFD1 1958G>A in Liver and Colon Cancer Risk According to Global DNA Methylation. PLoS ONE 2017, 12, e0185792. [Google Scholar] [CrossRef]
  9. Friso, S.; Lotto, V.; Corrocher, R.; Choi, S.W. Vitamin B6 and Cardiovascular Disease. Subcell. Biochem. 2012, 56, 265–290. [Google Scholar] [CrossRef]
  10. Moruzzi, S.; Udali, S.; Ruzzenente, A.; Guglielmi, A.; Guarini, P.; Martinelli, N.; Conci, S.; Mazzi, F.; Pattini, P.; Tammen, S.A.; et al. The RFC1 80G>A, among Common One-Carbon Polymorphisms, Relates to Survival Rate According to DNA Global Methylation in Primary Liver Cancers. PLoS ONE 2016, 11, e0167534. [Google Scholar] [CrossRef]
  11. Liu, J.; Huang, B.; Ding, F.; Li, Y. Environment Factors, DNA Methylation, and Cancer. Environ. Geochem. Health 2023, 45, 7543–7568. [Google Scholar] [CrossRef] [PubMed]
  12. Nishiyama, A.; Nakanishi, M. Navigating the DNA Methylation Landscape of Cancer. Trends Genet. 2021, 37, 1012–1027. [Google Scholar] [CrossRef] [PubMed]
  13. Deng, S.; Zhang, J.; Su, J.; Zuo, Z.; Zeng, L.; Liu, K.; Zheng, Y.; Huang, X.; Bai, R.; Zhuang, L.; et al. RNA m6A Regulates Transcription via DNA Demethylation and Chromatin Accessibility. Nat. Genet. 2022, 54, 1427–1437. [Google Scholar] [CrossRef]
  14. Yamaguchi, K.; Chen, X.; Rodgers, B.; Miura, F.; Bashtrykov, P.; Bonhomme, F.; Salinas-Luypaert, C.; Haxholli, D.; Gutekunst, N.; Aygenli, B.Ö.; et al. Non-Canonical Functions of UHRF1 Maintain DNA Methylation Homeostasis in Cancer Cells. Nat. Commun. 2024, 15, 2960. [Google Scholar] [CrossRef]
  15. Salerno, D.; Peruzzi, G.; Pascucci, G.R.; Levrero, M.; Belloni, L.; Pediconi, N. miRNA-27a-3p Is Involved in the Plasticity of Differentiated Hepatocytes. Gene 2024, 913, 148387. [Google Scholar] [CrossRef]
  16. Udali, S.; Castagna, A.; Corbella, M.; Ruzzenente, A.; Moruzzi, S.; Mazzi, F.; Campagnaro, T.; De Santis, D.; Franceschi, A.; Pattini, P.; et al. Hepcidin and DNA Promoter Methylation in Hepatocellular Carcinoma. Eur. J. Clin. Investig. 2018, 48, e12870. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, Y.; Feng, J.; Mi, Y.; Fan, W.; Qin, R.; Mei, Y.; Jin, G.; Mao, J.; Zhang, H. Epigenetic Activation of Cytochrome P450 1A2 Sensitizes Hepatocellular Carcinoma Cells to Sorafenib. Drug Metab. Dispos. 2024, 52, 555–564. [Google Scholar] [CrossRef]
  18. Liu, C.; Liu, L.; Shan, J.; Shen, J.; Xu, Y.; Zhang, Q.; Yang, Z.; Wu, L.; Xia, F.; Bie, P.; et al. Histone Deacetylase 3 Participates in Self-Renewal of Liver Cancer Stem Cells through Histone Modification. Cancer Lett. 2013, 339, 60–69. [Google Scholar] [CrossRef]
  19. Liu, K.-Y.; Wang, L.-T.; Hsu, S.-H. Modification of Epigenetic Histone Acetylation in Hepatocellular Carcinoma. Cancers 2018, 10, 8. [Google Scholar] [CrossRef]
  20. Seo, Y.; Kim, D.K.; Park, J.; Park, S.J.; Park, J.J.; Cheon, J.H.; Kim, T.I. A Comprehensive Understanding of Post-Translational Modification of Sox2 via Acetylation and O-GlcNAcylation in Colorectal Cancer. Cancers 2024, 16, 1035. [Google Scholar] [CrossRef]
  21. Xu, X.; Ding, Y.; Jin, J.; Xu, C.; Hu, W.; Wu, S.; Ding, G.; Cheng, R.; Cao, L.; Jia, S. Post-Translational Modification of CDK1-STAT3 Signaling by Fisetin Suppresses Pancreatic Cancer Stem Cell Properties. Cell Biosci. 2023, 13, 176. [Google Scholar] [CrossRef] [PubMed]
  22. El Omari, N.; Bakrim, S.; Khalid, A.; Abdalla, A.N.; Almalki, W.H.; Lee, L.-H.; Ardianto, C.; Ming, L.C.; Bouyahya, A. Molecular Mechanisms Underlying the Clinical Efficacy of Panobinostat Involve Stochasticity of Epigenetic Signaling, Sensitization to Anticancer Drugs, and Induction of Cellular Cell Death Related to Cellular Stresses. Biomed. Pharmacother. 2023, 164, 114886. [Google Scholar] [CrossRef] [PubMed]
  23. Fattahi, S.; Amjadi-Moheb, F.; Tabaripour, R.; Ashrafi, G.H.; Akhavan-Niaki, H. PI3K/AKT/mTOR Signaling in Gastric Cancer: Epigenetics and Beyond. Life Sci. 2020, 262, 118513. [Google Scholar] [CrossRef]
  24. Wils, L.J.; Bijlsma, M.F. Epigenetic Regulation of the Hedgehog and Wnt Pathways in Cancer. Crit. Rev. Oncol./Hematol. 2018, 121, 23–44. [Google Scholar] [CrossRef]
  25. Minucci, S.; Pelicci, P.G. Histone Deacetylase Inhibitors and the Promise of Epigenetic (and More) Treatments for Cancer. Nat. Rev. Cancer 2006, 6, 38–51. [Google Scholar] [CrossRef]
  26. Wu, C.; Song, Q.; Gao, S.; Wu, S. Targeting HDACs for Diffuse Large B-Cell Lymphoma Therapy. Sci. Rep. 2024, 14, 289. [Google Scholar] [CrossRef]
  27. Kawaf, R.R.; Ramadan, W.S.; El-Awady, R. Deciphering the Interplay of Histone Post-Translational Modifications in Cancer: Co-Targeting Histone Modulators for Precision Therapy. Life Sci. 2024, 346, 122639. [Google Scholar] [CrossRef]
  28. Kouzarides, T. Chromatin Modifications and Their Function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef]
  29. Zhou, V.W.; Goren, A.; Bernstein, B.E. Charting Histone Modifications and the Functional Organization of Mammalian Genomes. Nat. Rev. Genet. 2011, 12, 7–18. [Google Scholar] [CrossRef]
  30. Allfrey, V.G.; Faulkner, R.; Mirsky, A.E. Acetylation and Methylation of Histones and Their Possible Role in the Regulation of Rna Synthesis. Proc. Natl. Acad. Sci. USA 1964, 51, 786–794. [Google Scholar] [CrossRef] [PubMed]
  31. Neganova, M.E.; Klochkov, S.G.; Aleksandrova, Y.R.; Aliev, G. Histone Modifications in Epigenetic Regulation of Cancer: Perspectives and Achieved Progress. Semin. Cancer Biol. 2022, 83, 452–471. [Google Scholar] [CrossRef]
  32. Archana, B.; D’Cruze, L.; Sundaram, S.; Ramanathan, K.; Ganesh, K. Immunohistochemical Expression of Histone Modification Pattern in Adult Glioblastoma. J. Cancer Res. Ther. 2024, 20, 52–56. [Google Scholar] [CrossRef]
  33. Lu, Y.; Wang, Y.; Zhang, L.; Ma, Z.; Yu, K.; Shu, Y.; Zou, X.; Yang, J.; Liu, X.; Wang, C.; et al. KAT7 Enhances the Proliferation and Metastasis of Head and Neck Squamous Carcinoma by Promoting the Acetylation Level of LDHA. Cancer Lett. 2024, 590, 216869. [Google Scholar] [CrossRef]
  34. Gu, R.; Kim, T.-D.; Song, H.; Sui, Y.; Shin, S.; Oh, S.; Janknecht, R. SET7/9-Mediated Methylation Affects Oncogenic Functions of Histone Demethylase JMJD2A. JCI Insight 2023, 8, e164990. [Google Scholar] [CrossRef]
  35. Roy, A.; Niharika; Chakraborty, S.; Mishra, J.; Singh, S.P.; Patra, S.K. Mechanistic Aspects of Reversible Methylation Modifications of Arginine and Lysine of Nuclear Histones and Their Roles in Human Colon Cancer. Prog. Mol. Biol. Transl. Sci. 2023, 197, 261–302. [Google Scholar] [CrossRef]
  36. Sawesi, S.; Malkaram, S.A.; Abd Elmageed, Z.Y.; Fandy, T.E. Modulation of the Activity of Histone Lysine Methyltransferases and Demethylases by Curcumin Analog in Leukaemia Cells. J. Cell Mol. Med. 2022, 26, 5624–5633. [Google Scholar] [CrossRef]
  37. Cheng, C.; Yao, H.; Li, H.; Liu, J.; Liu, Z.; Wu, Y.; Zhu, L.; Hu, H.; Fang, Z.; Wu, L. Blockade of the Deubiquitinating Enzyme USP48 Degrades Oncogenic HMGA2 and Inhibits Colorectal Cancer Invasion and Metastasis. Acta Pharm. Sin. B 2024, 14, 1624–1643. [Google Scholar] [CrossRef]
  38. Cook, K.W.; Xue, W.; Symonds, P.; Daniels, I.; Gijon, M.; Boocock, D.; Coveney, C.; Miles, A.K.; Shah, S.; Atabani, S.; et al. Homocitrullination of Lysine Residues Mediated by Myeloid-Derived Suppressor Cells in the Tumor Environment Is a Target for Cancer Immunotherapy. J. Immunother. Cancer 2021, 9, e001910. [Google Scholar] [CrossRef]
  39. Liu, L.; Yin, S.; Gan, W. TRAF6 Promotes PRMT5 Activity in a Ubiquitination-Dependent Manner. Cancers 2023, 15, 2501. [Google Scholar] [CrossRef]
  40. Lyu, Y.; Yang, Y.; Talwar, V.; Lu, H.; Chen, C.; Salman, S.; Wicks, E.E.; Huang, T.Y.-T.; Drehmer, D.; Wang, Y.; et al. Hypoxia-Inducible Factor 1 Recruits FACT and RNF20/40 to Mediate Histone Ubiquitination and Transcriptional Activation of Target Genes. Cell Rep. 2024, 43, 113972. [Google Scholar] [CrossRef]
  41. Yang, H.; Du, Y.; Fei, X.; Huang, S.; Yimiti, M.; Yang, X.; Ma, J.; Li, S.; Tuoheniyazi, H.; Zhao, Y.; et al. SUMOylation of the Ubiquitin Ligase Component KEAP1 at K39 Upregulates NRF2 and Its Target Function in Lung Cancer Cell Proliferation. J. Biol. Chem. 2023, 299, 105215. [Google Scholar] [CrossRef]
  42. Marks, P.A.; Rifkind, R.A.; Richon, V.M.; Breslow, R.; Miller, T.; Kelly, W.K. Histone Deacetylases and Cancer: Causes and Therapies. Nat. Rev. Cancer 2001, 1, 194–202. [Google Scholar] [CrossRef]
  43. Haery, L.; Thompson, R.C.; Gilmore, T.D. Histone Acetyltransferases and Histone Deacetylases in B- and T-Cell Development, Physiology and Malignancy. Genes. Cancer 2015, 6, 184–213. [Google Scholar] [CrossRef]
  44. Sun, X.-J.; Man, N.; Tan, Y.; Nimer, S.D.; Wang, L. The Role of Histone Acetyltransferases in Normal and Malignant Hematopoiesis. Front. Oncol. 2015, 5, 108. [Google Scholar] [CrossRef]
  45. Gray, S.G.; Teh, B.T. Histone Acetylation/Deacetylation and Cancer: An “Open” and “Shut” Case? Curr. Mol. Med. 2001, 1, 401–429. [Google Scholar] [CrossRef]
  46. Wang, X.; Li, N.; Zheng, M.; Yu, Y.; Zhang, S. Acetylation and Deacetylation of Histone in Adipocyte Differentiation and the Potential Significance in Cancer. Transl. Oncol. 2024, 39, 101815. [Google Scholar] [CrossRef] [PubMed]
  47. Adams, G.E.; Chandru, A.; Cowley, S.M. Co-Repressor, Co-Activator and General Transcription Factor: The Many Faces of the Sin3 Histone Deacetylase (HDAC) Complex. Biochem. J. 2018, 475, 3921–3932. [Google Scholar] [CrossRef]
  48. Basta, J.; Rauchman, M. The Nucleosome Remodeling and Deacetylase (NuRD) Complex in Development and Disease. Transl. Res. 2015, 165, 36–47. [Google Scholar] [CrossRef]
  49. Chen, L.; He, J.; Zhang, Y.; Li, Y.; Zhang, T.; Wang, R.; Bai, L.; Zhao, S.; Liu, E.; Wang, W. Regulation of Endothelial-to-Mesenchymal Transition by Histone Deacetylase 3 Posttranslational Modifications in Neointimal Hyperplasia. Ann. Transl. Med. 2023, 11, 207. [Google Scholar] [CrossRef]
  50. Zhang, H.; Zhou, P.; Xing, W.; Chen, L.; Zhou, Y.; Yang, H.; Fu, K.; Liu, Z. GLIS2 Prevents Hepatic Fibrosis by Competitively Binding HDAC3 to Inhibit Hepatic Stellate Cell Activation. Cell Mol. Gastroenterol. Hepatol. 2023, 15, 355–372. [Google Scholar] [CrossRef]
  51. Zhao, H.; Zhang, X.-M.; Xiao, S.; Wu, Z.-R.; Shi, Y.-J.; Xie, M.-J. HDAC11 Is Related to Breast Cancer Prognosis and Inhibits Invasion and Proliferation of Breast Cancer Cells. Int. J. Clin. Exp. Pathol. 2023, 16, 172. [Google Scholar]
  52. Gray, S.G.; Ekström, T.J. The Human Histone Deacetylase Family. Exp. Cell Res. 2001, 262, 75–83. [Google Scholar] [CrossRef]
  53. Sacconnay, L.; Carrupt, P.-A.; Nurisso, A. Human Sirtuins: Structures and Flexibility. J. Struct. Biol. 2016, 196, 534–542. [Google Scholar] [CrossRef]
  54. Cheung, C.H.Y.; Cheng, C.K.; Leung, K.T.; Zhang, C.; Ho, C.Y.; Luo, X.; Kam, A.Y.F.; Xia, T.; Wan, T.S.K.; Pitts, H.A.; et al. C-Terminal Binding Protein (CTBP2) Is a Novel Tumor Suppressor Targeting the MYC-IRF4 Axis in Multiple Myeloma. Blood Adv. 2024, 8, 2217–2234. [Google Scholar] [CrossRef]
  55. Sanaei, M.; Kavoosi, F. Histone Deacetylase Inhibitors, Intrinsic and Extrinsic Apoptotic Pathways, and Epigenetic Alterations of Histone Deacetylases (HDACs) in Hepatocellular Carcinoma. Iran. J. Pharm. Res. 2021, 20, 324–336. [Google Scholar] [CrossRef] [PubMed]
  56. Stiegler, P.; De Luca, A.; Bagella, L.; Giordano, A. The COOH-Terminal Region of pRb2/P130 Binds to Histone Deacetylase 1 (HDAC1), Enhancing Transcriptional Repression of the E2F-Dependent Cyclin A Promoter1. Cancer Res. 1998, 58, 5049–5052. [Google Scholar] [PubMed]
  57. Betz, R.; Gray, S.G.; Ekström, C.; Larsson, C.; Ekström, T.J. Human Histone Deacetylase 2, HDAC2 (Human RPD3), Is Localized to 6q21 by Radiation Hybrid Mapping. Genomics 1998, 52, 245–246. [Google Scholar] [CrossRef] [PubMed]
  58. Emiliani, S.; Fischle, W.; Van Lint, C.; Al-Abed, Y.; Verdin, E. Characterization of a Human RPD3 Ortholog, HDAC3. Proc. Natl. Acad. Sci. USA 1998, 95, 2795–2800. [Google Scholar] [CrossRef]
  59. Gao, L.; Cueto, M.A.; Asselbergs, F.; Atadja, P. Cloning and Functional Characterization of HDAC11, a Novel Member of the Human Histone Deacetylase Family. J. Biol. Chem. 2002, 277, 25748–25755. [Google Scholar] [CrossRef] [PubMed]
  60. Seto, E.; Yoshida, M. Erasers of Histone Acetylation: The Histone Deacetylase Enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef]
  61. Yang, X.-J.; Seto, E. The Rpd3/Hda1 Family of Lysine Deacetylases: From Bacteria and Yeast to Mice and Men. Nat. Rev. Mol. Cell Biol. 2008, 9, 206–218. [Google Scholar] [CrossRef] [PubMed]
  62. Karagianni, P.; Wong, J. HDAC3: Taking the SMRT-N-CoRrect Road to Repression. Oncogene 2007, 26, 5439–5449. [Google Scholar] [CrossRef] [PubMed]
  63. Buggy, J.J.; Sideris, M.L.; Mak, P.; Lorimer, D.D.; McIntosh, B.; Clark, J.M. Cloning and Characterization of a Novel Human Histone Deacetylase, HDAC8. Biochem. J. 2000, 350 Pt 1, 199–205. [Google Scholar] [CrossRef]
  64. Takami, Y.; Nakayama, T. N-Terminal Region, C-Terminal Region, Nuclear Export Signal, and Deacetylation Activity of Histone Deacetylase-3 Are Essential for the Viability of the DT40 Chicken B Cell Line. J. Biol. Chem. 2000, 275, 16191–16201. [Google Scholar] [CrossRef]
  65. Yang, W.-M.; Tsai, S.-C.; Wen, Y.-D.; Fejér, G.; Seto, E. Functional Domains of Histone Deacetylase-3. J. Biol. Chem. 2002, 277, 9447–9454. [Google Scholar] [CrossRef]
  66. Grozinger, C.M.; Hassig, C.A.; Schreiber, S.L. Three Proteins Define a Class of Human Histone Deacetylases Related to Yeast Hda1p. Proc. Natl. Acad. Sci. USA 1999, 96, 4868–4873. [Google Scholar] [CrossRef]
  67. Hess, L.; Moos, V.; Lauber, A.A.; Reiter, W.; Schuster, M.; Hartl, N.; Lackner, D.; Boenke, T.; Koren, A.; Guzzardo, P.M.; et al. A Toolbox for Class I HDACs Reveals Isoform Specific Roles in Gene Regulation and Protein Acetylation. PLoS Genet. 2022, 18, e1010376. [Google Scholar] [CrossRef]
  68. Wright, L.H.; Menick, D.R. A Class of Their Own: Exploring the Nondeacetylase Roles of Class IIa HDACs in Cardiovascular Disease. Am. J. Physiol.-Heart Circ. Physiol. 2016, 311, H199–H206. [Google Scholar] [CrossRef]
  69. Khochbin, S.; Kao, H.-Y. Histone Deacetylase Complexes: Functional Entities or Molecular Reservoirs. FEBS Lett. 2001, 494, 141–144. [Google Scholar] [CrossRef] [PubMed]
  70. Haberland, M.; Arnold, M.A.; McAnally, J.; Phan, D.; Kim, Y.; Olson, E.N. Regulation of HDAC9 Gene Expression by MEF2 Establishes a Negative-Feedback Loop in the Transcriptional Circuitry of Muscle Differentiation. Mol. Cell Biol. 2007, 27, 518–525. [Google Scholar] [CrossRef] [PubMed]
  71. Zhou, X.; Marks, P.A.; Rifkind, R.A.; Richon, V.M. Cloning and Characterization of a Histone Deacetylase, HDAC9. Proc. Natl. Acad. Sci. USA 2001, 98, 10572–10577. [Google Scholar] [CrossRef]
  72. Bertos, N.R.; Wang, A.H.; Yang, X.J. Class II Histone Deacetylases: Structure, Function, and Regulation. Biochem. Cell Biol. 2001, 79, 243–252. [Google Scholar] [CrossRef]
  73. Curcio, A.; Rocca, R.; Alcaro, S.; Artese, A. The Histone Deacetylase Family: Structural Features and Application of Combined Computational Methods. Pharmaceuticals 2024, 17, 620. [Google Scholar] [CrossRef]
  74. Liu, P.; Xiao, J.; Wang, Y.; Song, X.; Huang, L.; Ren, Z.; Kitazato, K.; Wang, Y. Posttranslational Modification and beyond: Interplay between Histone Deacetylase 6 and Heat-Shock Protein 90. Mol. Med. 2021, 27, 110. [Google Scholar] [CrossRef]
  75. Guardiola, A.R.; Yao, T.-P. Molecular Cloning and Characterization of a Novel Histone Deacetylase HDAC10. J. Biol. Chem. 2002, 277, 3350–3356. [Google Scholar] [CrossRef]
  76. Schwer, B.; Verdin, E. Conserved Metabolic Regulatory Functions of Sirtuins. Cell Metab. 2008, 7, 104–112. [Google Scholar] [CrossRef]
  77. Khatun, S.; Prasad Bhagat, R.; Dutta, R.; Datta, A.; Jaiswal, A.; Halder, S.; Jha, T.; Amin, S.A.; Gayen, S. Unraveling HDAC11: Epigenetic Orchestra in Different Diseases and Structural Insights for Inhibitor Design. Biochem. Pharmacol. 2024, 225, 116312. [Google Scholar] [CrossRef]
  78. Tian, Y.; Lv, W.; Li, X.; Wang, C.; Wang, D.; Wang, P.G.; Jin, J.; Shen, J. Stabilizing HDAC11 with SAHA to Assay Slow-Binding Benzamide Inhibitors. Bioorganic Med. Chem. Lett. 2017, 27, 2943–2945. [Google Scholar] [CrossRef] [PubMed]
  79. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A Global View of Hepatocellular Carcinoma: Trends, Risk, Prevention and Management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef] [PubMed]
  81. Sato-Espinoza, K.; Valdivia-Herrera, M.; Chotiprasidhi, P.; Diaz-Ferrer, J. Hepatocellular Carcinoma in Patients without Cirrhosis. World J. Gastroenterol. 2025, 31, 107100. [Google Scholar] [CrossRef] [PubMed]
  82. Amin, N.; Anwar, J.; Sulaiman, A.; Naumova, N.N.; Anwar, N. Hepatocellular Carcinoma: A Comprehensive Review. Diseases 2025, 13, 207. [Google Scholar] [CrossRef]
  83. Fan, C.; Kam, S.; Ramadori, P. Metabolism-Associated Epigenetic and Immunoepigenetic Reprogramming in Liver Cancer. Cancers 2021, 13, 5250. [Google Scholar] [CrossRef]
  84. Kim, H.S.; Shen, Q.; Nam, S.W. Histone Deacetylases and Their Regulatory MicroRNAs in Hepatocarcinogenesis. J. Korean Med. Sci. 2015, 30, 1375–1380. [Google Scholar] [CrossRef] [PubMed]
  85. Xie, J.; Liu, R.; Cai, Y.; Liu, D. HDAC1: A Promising Target for Cancer Treatment: Insights from a Thorough Analysis of Tumor Functions. Transl. Cancer Res. 2024, 13, 5300–5315. [Google Scholar] [CrossRef]
  86. Freese, K.; Seitz, T.; Dietrich, P.; Lee, S.M.L.; Thasler, W.E.; Bosserhoff, A.; Hellerbrand, C. Histone Deacetylase Expressions in Hepatocellular Carcinoma and Functional Effects of Histone Deacetylase Inhibitors on Liver Cancer Cells In Vitro. Cancers 2019, 11, 1587. [Google Scholar] [CrossRef]
  87. Zhou, H.; Xu, J.; Zhang, C.; Wen, Y. Aberrant Histone Deacetylase 1 Expression Upregulates Vimentin Expression via an NF-κB-Dependent Pathway in Hepatocellular Carcinoma. Oncol. Lett. 2019, 18, 339–347. [Google Scholar] [CrossRef]
  88. Buurman, R.; Gürlevik, E.; Schäffer, V.; Eilers, M.; Sandbothe, M.; Kreipe, H.; Wilkens, L.; Schlegelberger, B.; Kühnel, F.; Skawran, B. Histone Deacetylases Activate Hepatocyte Growth Factor Signaling by Repressing MicroRNA-449 in Hepatocellular Carcinoma Cells. Gastroenterology 2012, 143, 811–820.e15. [Google Scholar] [CrossRef]
  89. Ler, S.Y.; Leung, C.H.W.; Khin, L.W.; Lu, G.-D.; Salto-Tellez, M.; Hartman, M.; Iau, P.T.C.; Yap, C.T.; Hooi, S.C. HDAC1 and HDAC2 Independently Predict Mortality in Hepatocellular Carcinoma by a Competing Risk Regression Model in a Southeast Asian Population. Oncol. Rep. 2015, 34, 2238–2250. [Google Scholar] [CrossRef] [PubMed]
  90. Lachenmayer, A.; Toffanin, S.; Cabellos, L.; Alsinet, C.; Hoshida, Y.; Villanueva, A.; Minguez, B.; Tsai, H.-W.; Ward, S.C.; Thung, S.; et al. Combination Therapy for Hepatocellular Carcinoma: Additive Preclinical Efficacy of the HDAC Inhibitor Panobinostat with Sorafenib. J. Hepatol. 2012, 56, 1343–1350. [Google Scholar] [CrossRef]
  91. Fan, J.; Lou, B.; Chen, W.; Zhang, J.; Lin, S.; Lv, F.; Chen, Y. Down-Regulation of HDAC5 Inhibits Growth of Human Hepatocellular Carcinoma by Induction of Apoptosis and Cell Cycle Arrest. Tumor Biol. 2014, 35, 11523–11532. [Google Scholar] [CrossRef]
  92. Ye, M.; Fang, Z.; Gu, H.; Song, R.; Ye, J.; Li, H.; Wu, Z.; Zhou, S.; Li, P.; Cai, X.; et al. Histone Deacetylase 5 Promotes the Migration and Invasion of Hepatocellular Carcinoma via Increasing the Transcription of Hypoxia-Inducible Factor-1α under Hypoxia Condition. Tumour Biol. 2017, 39, 1010428317705034. [Google Scholar] [CrossRef]
  93. Xu, X.; Ding, P.; Shi, L.; Wu, G.; Ma, X. LukS-PV Inhibits Hepatocellular Carcinoma Cells Migration by Downregulating HDAC6 Expression. BMC Cancer 2022, 22, 630. [Google Scholar] [CrossRef] [PubMed]
  94. Mikami, D.; Kobayashi, M.; Uwada, J.; Yazawa, T.; Kamiyama, K.; Nishimori, K.; Nishikawa, Y.; Nishikawa, S.; Yokoi, S.; Taniguchi, T.; et al. AR420626, a Selective Agonist of GPR41/FFA3, Suppresses Growth of Hepatocellular Carcinoma Cells by Inducing Apoptosis via HDAC Inhibition. Ther. Adv. Med. Oncol. 2020, 12, 1758835920913432. [Google Scholar] [CrossRef] [PubMed]
  95. Wu, J.; Du, C.; Lv, Z.; Ding, C.; Cheng, J.; Xie, H.; Zhou, L.; Zheng, S. The Up-Regulation of Histone Deacetylase 8 Promotes Proliferation and Inhibits Apoptosis in Hepatocellular Carcinoma. Dig. Dis. Sci. 2013, 58, 3545–3553. [Google Scholar] [CrossRef]
  96. Yang, W.; Feng, Y.; Zhou, J.; Cheung, O.K.-W.; Cao, J.; Wang, J.; Tang, W.; Tu, Y.; Xu, L.; Wu, F.; et al. A Selective HDAC8 Inhibitor Potentiates Antitumor Immunity and Efficacy of Immune Checkpoint Blockade in Hepatocellular Carcinoma. Sci. Transl. Med. 2021, 13, eaaz6804. [Google Scholar] [CrossRef]
  97. Yang, Z.; Zhang, L.; Liu, J.; Yang, L.; Xue, H.; Bai, S.; Wang, K. PD-L1 Combined with HDAC9 Is a Useful Prognostic Predictor in Hepatocellular Carcinoma. Transl. Cancer Res. 2021, 10, 2305. [Google Scholar] [CrossRef] [PubMed]
  98. Gong, D.; Zeng, Z.; Yi, F.; Wu, J. Inhibition of Histone Deacetylase 11 Promotes Human Liver Cancer Cell Apoptosis. Am. J. Transl. Res. 2019, 11, 983–990. [Google Scholar]
  99. Bates, S.E. Epigenetic Therapies for Cancer. N. Engl. J. Med. 2020, 383, 650–663. [Google Scholar] [CrossRef]
  100. Cheng, B.; Pan, W.; Xiao, Y.; Ding, Z.; Zhou, Y.; Fei, X.; Liu, J.; Su, Z.; Peng, X.; Chen, J. HDAC-Targeting Epigenetic Modulators for Cancer Immunotherapy. Eur. J. Med. Chem. 2024, 265, 116129. [Google Scholar] [CrossRef]
  101. Tiwari, V.; Banerjee, S.K. Therapeutic Potential of HDAC Inhibitors in the Treatment of Cardiac Diseases: A Short Review. Curr. Drug Targets 2023, 24, 718–727. [Google Scholar] [CrossRef]
  102. Patel, V.K.; Shirbhate, E.; Tiwari, P.; Kore, R.; Veerasamy, R.; Mishra, A.; Rajak, H. Multi-Targeted HDAC Inhibitors as Anticancer Agents: Current Status and Future Prospective. Curr. Med. Chem. 2023, 30, 2762–2795. [Google Scholar] [CrossRef] [PubMed]
  103. Waitman, K.; Parise-Filho, R. New Kinase and HDAC Hybrid Inhibitors: Recent Advances and Perspectives. Future Med. Chem. 2022, 14, 745–766. [Google Scholar] [CrossRef]
  104. Shirbhate, E.; Singh, V.; Jahoriya, V.; Mishra, A.; Veerasamy, R.; Tiwari, A.K.; Rajak, H. Dual Inhibitors of HDAC and Other Epigenetic Regulators: A Novel Strategy for Cancer Treatment. Eur. J. Med. Chem. 2024, 263, 115938. [Google Scholar] [CrossRef]
  105. Karagianni, F.; Piperi, C.; Valero-Diaz, S.; Amato, C.; Vaque, J.P.; Casar, B.; Papadavid, E. Combination of JAKi and HDACi Exerts Antiangiogenic Potential in Cutaneous T-Cell Lymphoma. Cancers 2024, 16, 3176. [Google Scholar] [CrossRef]
  106. Huang, W.; Zhu, Q.; Shi, Z.; Tu, Y.; Li, Q.; Zheng, W.; Yuan, Z.; Li, L.; Zu, X.; Hao, Y.; et al. Dual Inhibitors of DNMT and HDAC Induce Viral Mimicry to Induce Antitumour Immunity in Breast Cancer. Cell Death Discov. 2024, 10, 143. [Google Scholar] [CrossRef]
  107. Zhao, J.; Gray, S.G.; Greene, C.M.; Lawless, M.W. Unmasking the Pathological and Therapeutic Potential of Histone Deacetylases for Liver Cancer. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 247–256. [Google Scholar] [CrossRef] [PubMed]
  108. Chang, Y.; Lee, Y.B.; Cho, E.J.; Lee, J.-H.; Yu, S.J.; Kim, Y.J.; Yoon, J.-H. CKD-5, a Novel Pan-Histone Deacetylase Inhibitor, Synergistically Enhances the Efficacy of Sorafenib for Hepatocellular Carcinoma. BMC Cancer 2020, 20, 1001. [Google Scholar] [CrossRef] [PubMed]
  109. Streubel, G.; Schrepfer, S.; Kallus, H.; Parnitzke, U.; Wulff, T.; Hermann, F.; Borgmann, M.; Hamm, S. Histone Deacetylase Inhibitor Resminostat in Combination with Sorafenib Counteracts Platelet-Mediated pro-Tumoral Effects in Hepatocellular Carcinoma. Sci. Rep. 2021, 11, 9587. [Google Scholar] [CrossRef]
  110. Wu, B.; Tapadar, S.; Ruan, Z.; Sun, C.Q.; Arnold, R.S.; Olugbami, J.O.; Johnston, A.; Arunsi, U.; Gaul, D.A.; Petros, J.A.; et al. A Novel Liver Cancer-Selective Histone Deacetylase Inhibitor Is Effective Against Hepatocellular Carcinoma and Induces Durable Responses with Immunotherapy. bioRxiv 2024, 7, 3155–3169. [Google Scholar] [CrossRef]
  111. Tu, Y.; Wu, H.; Zhong, C.; Liu, Y.; Xiong, Z.; Chen, S.; Wang, J.; Wong, P.P.-C.; Yang, W.; Liang, Z.; et al. Pharmacological Activation of STAT1-GSDME Pyroptotic Circuitry Reinforces Epigenetic Immunotherapy for Hepatocellular Carcinoma. Gut 2024, 74, 613–627. [Google Scholar] [CrossRef] [PubMed]
  112. Lin, Z.-Z.; Hu, M.C.-T.; Hsu, C.; Wu, Y.-M.; Lu, Y.-S.; Ho, J.-A.A.; Yeh, S.-H.; Chen, P.-J.; Cheng, A.-L. Synergistic Efficacy of Telomerase-Specific Oncolytic Adenoviral Therapy and Histone Deacetylase Inhibition in Human Hepatocellular Carcinoma. Cancer Lett. 2023, 556, 216063. [Google Scholar] [CrossRef]
  113. Xiao, R.; Jin, H.; Huang, F.; Huang, B.; Wang, H.; Wang, Y.-G. Oncolytic Virotherapy for Hepatocellular Carcinoma: A Potent Immunotherapeutic Landscape. World J. Gastrointest. Oncol. 2024, 16, 2867–2876. [Google Scholar] [CrossRef]
  114. Lai, S.-Y.; Zhu, X.-J.; Sun, W.-D.; Bi, S.-Z.; Zhang, C.-Y.; Liu, A.; Li, J.-H. Nicotinamide N-Methyltransferase (NNMT) and Liver Cancer: From Metabolic Networks to Therapeutic Targets. Biomolecules 2025, 15, 719. [Google Scholar] [CrossRef]
  115. Ulanovskaya, O.A.; Zuhl, A.M.; Cravatt, B.F. NNMT Promotes Epigenetic Remodeling in Cancer by Creating a Metabolic Methylation Sink. Nat. Chem. Biol. 2013, 9, 300–306. [Google Scholar] [CrossRef]
  116. Shen, C.; Li, M.; Duan, Y.; Jiang, X.; Hou, X.; Xue, F.; Zhang, Y.; Luo, Y. HDAC Inhibitors Enhance the Anti-Tumor Effect of Immunotherapies in Hepatocellular Carcinoma. Front. Immunol. 2023, 14, 1170207. [Google Scholar] [CrossRef]
  117. Eckschlager, T.; Plch, J.; Stiborova, M.; Hrabeta, J. Histone Deacetylase Inhibitors as Anticancer Drugs. Int. J. Mol. Sci. 2017, 18, 1414. [Google Scholar] [CrossRef]
  118. Bitzer, M.; Horger, M.; Giannini, E.G.; Ganten, T.M.; Wörns, M.A.; Siveke, J.T.; Dollinger, M.M.; Gerken, G.; Scheulen, M.E.; Wege, H.; et al. Resminostat plus Sorafenib as Second-Line Therapy of Advanced Hepatocellular Carcinoma—The SHELTER Study. J. Hepatol. 2016, 65, 280–288. [Google Scholar] [CrossRef] [PubMed]
  119. Fu, M.; Shi, W.; Li, Z.; Liu, H. Activation of mPTP-Dependent Mitochondrial Apoptosis Pathway by a Novel Pan HDAC Inhibitor Resminostat in Hepatocellular Carcinoma Cells. Biochem. Biophys. Res. Commun. 2016, 477, 527–533. [Google Scholar] [CrossRef]
  120. Soukupova, J.; Bertran, E.; Peñuelas-Haro, I.; Urdiroz-Urricelqui, U.; Borgman, M.; Kohlhof, H.; Fabregat, I. Resminostat Induces Changes in Epithelial Plasticity of Hepatocellular Carcinoma Cells and Sensitizes Them to Sorafenib-Induced Apoptosis. Oncotarget 2017, 8, 110367–110379. [Google Scholar] [CrossRef] [PubMed]
  121. Yan, S.; Chen, L.; Zhuang, H.; Yang, H.; Yang, Y.; Zhang, N.; Liu, R. HDAC Inhibition Sensitize Hepatocellular Carcinoma to Lenvatinib via Suppressing AKT Activation. Int. J. Biol. Sci. 2024, 20, 3046–3060. [Google Scholar] [CrossRef] [PubMed]
  122. Salama, B.M.; Helmy, M.W.; Fouad, H.; Shamaa, M.M.; Houssen, M.E. The Synergistic Antitumor Effect of Decitabine and Vorinostat Combination on HepG2 Human Hepatocellular Carcinoma Cell Line via Epigenetic Modulation of Autophagy–Apoptosis Molecular Crosstalk. Curr. Issues Mol. Biol. 2023, 45, 5935–5949. [Google Scholar] [CrossRef] [PubMed]
  123. Li, J.-Y.; Tian, T.; Han, B.; Yang, T.; Guo, Y.-X.; Wu, J.-Y.; Chen, Y.-S.; Yang, Q.; Xie, R.-J. Suberoylanilide Hydroxamic Acid Upregulates Reticulophagy Receptor Expression and Promotes Cell Death in Hepatocellular Carcinoma Cells. World J. Gastroenterol. 2023, 29, 5038–5053. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, H.; Xu, L.; Yue, S.; Su, H.; Chen, X.; Liu, Q.; Li, H.; Liang, H.; Chen, X.; He, J.; et al. Targeting N4-Acetylcytidine Suppresses Hepatocellular Carcinoma Progression by Repressing eEF2-Mediated HMGB2 mRNA Translation. Cancer Commun. 2024, 44, 1018–1041. [Google Scholar] [CrossRef]
  125. Choi, C.; Lee, G.H.; Son, A.; Yoo, G.S.; Yu, J.I.; Park, H.C. Downregulation of Mcl-1 by Panobinostat Potentiates Proton Beam Therapy in Hepatocellular Carcinoma Cells. Cells 2021, 10, 554. [Google Scholar] [CrossRef]
  126. Di Fazio, P.; Schneider-Stock, R.; Neureiter, D.; Okamoto, K.; Wissniowski, T.; Gahr, S.; Quint, K.; Meissnitzer, M.; Alinger, B.; Montalbano, R.; et al. The Pan-Deacetylase Inhibitor Panobinostat Inhibits Growth of Hepatocellular Carcinoma Models by Alternative Pathways of Apoptosis. Cell Oncol. 2010, 32, 285–300. [Google Scholar] [CrossRef]
  127. Sun, W.-J.; Huang, H.; He, B.; Hu, D.-H.; Li, P.-H.; Yu, Y.-J.; Zhou, X.-H.; Lv, Z.; Zhou, L.; Hu, T.-Y.; et al. Romidepsin Induces G2/M Phase Arrest via Erk/cdc25C/Cdc2/cyclinB Pathway and Apoptosis Induction through JNK/c-Jun/Caspase3 Pathway in Hepatocellular Carcinoma Cells. Biochem. Pharmacol. 2017, 127, 90–100. [Google Scholar] [CrossRef]
  128. Afaloniati, H.; Angelopoulou, K.; Giakoustidis, A.; Hardas, A.; Pseftogas, A.; Makedou, K.; Gargavanis, A.; Goulopoulos, T.; Iliadis, S.; Papadopoulos, V.; et al. HDAC1/2 Inhibitor Romidepsin Suppresses DEN-Induced Hepatocellular Carcinogenesis in Mice. Onco Targets Ther. 2020, 13, 5575–5588. [Google Scholar] [CrossRef]
  129. Sun, L.; Liu, Y.; Guo, X.; Cui, T.; Wu, C.; Tao, J.; Cheng, C.; Chu, Q.; Ji, C.; Li, X.; et al. Acetylation-Dependent Regulation of Core Spliceosome Modulates Hepatocellular Carcinoma Cassette Exons and Sensitivity to PARP Inhibitors. Nat. Commun. 2024, 15, 5209. [Google Scholar] [CrossRef]
  130. Spratlin, J.L.; Pitts, T.M.; Kulikowski, G.N.; Morelli, M.P.; Tentler, J.J.; Serkova, N.J.; Eckhardt, S.G. Synergistic Activity of Histone Deacetylase and Proteasome Inhibition Against Pancreatic and Hepatocellular Cancer Cell Lines. Anticancer. Res. 2011, 31, 1093–1103. [Google Scholar]
  131. Ma, B.B.Y.; Sung, F.; Tao, Q.; Poon, F.F.; Lui, V.W.; Yeo, W.; Chan, S.L.; Chan, A.T.C. The Preclinical Activity of the Histone Deacetylase Inhibitor PXD101 (Belinostat) in Hepatocellular Carcinoma Cell Lines. Investig. New Drugs 2010, 28, 107–114. [Google Scholar] [CrossRef]
  132. Llopiz, D.; Ruiz, M.; Villanueva, L.; Iglesias, T.; Silva, L.; Egea, J.; Lasarte, J.J.; Pivette, P.; Trochon-Joseph, V.; Vasseur, B.; et al. Enhanced Anti-Tumor Efficacy of Checkpoint Inhibitors in Combination with the Histone Deacetylase Inhibitor Belinostat in a Murine Hepatocellular Carcinoma Model. Cancer Immunol. Immunother. 2018, 68, 379–393. [Google Scholar] [CrossRef] [PubMed]
  133. Walewski, J.; Paszkiewicz-Kozik, E.; Borsaru, G.; Hellmann, A.; Janikova, A.; Warszewska, A.; Mais, A.; Ammendola, A.; Herz, T.; Krauss, B.; et al. Resminostat in Patients with Relapsed or Refractory Hodgkin Lymphoma: Results of the Phase II SAPHIRE Study. Leuk. Lymphoma 2019, 60, 675–684. [Google Scholar] [CrossRef]
  134. Karagianni, F.; Piperi, C.; Mpakou, V.; Spathis, A.; Foukas, P.G.; Dalamaga, M.; Pappa, V.; Papadavid, E. Ruxolitinib with Resminostat Exert Synergistic Antitumor Effects in Cutaneous T-Cell Lymphoma. PLoS ONE 2021, 16, e0248298. [Google Scholar] [CrossRef]
  135. Ikeda, M.; Ohno, I.; Ueno, H.; Mitsunaga, S.; Hashimoto, Y.; Okusaka, T.; Kondo, S.; Sasaki, M.; Sakamoto, Y.; Takahashi, H.; et al. Phase I Study of Resminostat, an HDAC Inhibitor, Combined with S-1 in Patients with Pre-Treated Biliary Tract or Pancreatic Cancer. Investig. New Drugs 2019, 37, 109–117. [Google Scholar] [CrossRef]
  136. Brunetto, A.T.; Ang, J.E.; Lal, R.; Olmos, D.; Molife, L.R.; Kristeleit, R.; Parker, A.; Casamayor, I.; Olaleye, M.; Mais, A.; et al. First-in-Human, Pharmacokinetic and Pharmacodynamic Phase I Study of Resminostat, an Oral Histone Deacetylase Inhibitor, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2013, 19, 5494–5504. [Google Scholar] [CrossRef] [PubMed]
  137. Tak, W.Y.; Ryoo, B.-Y.; Lim, H.Y.; Kim, D.-Y.; Okusaka, T.; Ikeda, M.; Hidaka, H.; Yeon, J.-E.; Mizukoshi, E.; Morimoto, M.; et al. Phase I/II Study of First-Line Combination Therapy with Sorafenib plus Resminostat, an Oral HDAC Inhibitor, versus Sorafenib Monotherapy for Advanced Hepatocellular Carcinoma in East Asian Patients. Investig. New Drugs 2018, 36, 1072–1084. [Google Scholar] [CrossRef] [PubMed]
  138. Peng, X.; Zhang, D.; Li, Z.; Fu, M.; Liu, H. mTOR Inhibition Sensitizes Human Hepatocellular Carcinoma Cells to Resminostat. Biochem. Biophys. Res. Commun. 2016, 477, 556–562. [Google Scholar] [CrossRef]
  139. Parveen, R.; Harihar, D.; Chatterji, B.P. Recent Histone Deacetylase Inhibitors in Cancer Therapy. Cancer 2023, 129, 3372–3380. [Google Scholar] [CrossRef]
  140. Sanaei, M.; Kavoosi, F.; Pourahmadi, M. Effect of Decitabine (5-Aza-2′-Deoxycytidine, 5-Aza-CdR) in Comparison with Vorinostat (Suberoylanilide Hydroxamic Acid, SAHA) on DNMT1, DNMT3a and DNMT3b, HDAC 1-3, SOCS 1, SOCS 3, JAK2, and STAT3 Gene Expression in Hepatocellular Carcinoma HLE and LCL-PI 11 Cell Lines. Asian Pac. J. Cancer Prev. 2021, 22, 2089–2098. [Google Scholar] [CrossRef] [PubMed]
  141. Eleutherakis-Papaiakovou, E.; Kanellias, N.; Kastritis, E.; Gavriatopoulou, M.; Terpos, E.; Dimopoulos, M.A. Efficacy of Panobinostat for the Treatment of Multiple Myeloma. J. Oncol. 2020, 2020, 7131802. [Google Scholar] [CrossRef]
  142. Sun, H.; Song, J.; Sun, M.; Shan, C. Chapter 3—Histone Deacetylase Inhibitors (HDACi), the Ongoing Epigenetic Agents to Enhance Chemotherapy Sensitization. In Epigenetic Regulation in Overcoming Chemoresistance; Wu, C., Wang, L., Eds.; Cancer Sensitizing Agents for Chemotherapy; Academic Press: Cambridge, MA, USA, 2021; Volume 15, pp. 25–39. [Google Scholar]
  143. Liu, Y.-F.; Chiang, Y.; Hsu, F.-M.; Tsai, C.-L.; Cheng, J.C.-H. Radiosensitization Effect by HDAC Inhibition Improves NKG2D-Dependent Natural Killer Cytotoxicity in Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 1009089. [Google Scholar] [CrossRef] [PubMed]
  144. Di Fazio, P.; Waldegger, P.; Jabari, S.; Lingelbach, S.; Montalbano, R.; Ocker, M.; Slater, E.P.; Bartsch, D.K.; Illig, R.; Neureiter, D.; et al. Autophagy-Related Cell Death by Pan-Histone Deacetylase Inhibition in Liver Cancer. Oncotarget 2016, 7, 28998–29010. [Google Scholar] [CrossRef] [PubMed]
  145. Henrici, A.; Montalbano, R.; Neureiter, D.; Krause, M.; Stiewe, T.; Slater, E.P.; Quint, K.; Ocker, M.; Di Fazio, P. The Pan-Deacetylase Inhibitor Panobinostat Suppresses the Expression of Oncogenic miRNAs in Hepatocellular Carcinoma Cell Lines. Mol. Carcinog. 2015, 54, 585–597. [Google Scholar] [CrossRef] [PubMed]
  146. Bondarev, A.D.; Attwood, M.M.; Jonsson, J.; Chubarev, V.N.; Tarasov, V.V.; Schiöth, H.B. Recent Developments of HDAC Inhibitors: Emerging Indications and Novel Molecules. Br. J. Clin. Pharmacol. 2021, 87, 4577–4597. [Google Scholar] [CrossRef]
  147. Zhou, H.; Cai, Y.; Liu, D.; Li, M.; Sha, Y.; Zhang, W.; Wang, K.; Gong, J.; Tang, N.; Huang, A.; et al. Pharmacological or Transcriptional Inhibition of Both HDAC1 and 2 Leads to Cell Cycle Blockage and Apoptosis via p21Waf1/Cip1 and p19INK4d Upregulation in Hepatocellular Carcinoma. Cell Prolif. 2018, 51, e12447. [Google Scholar] [CrossRef]
  148. Afaloniati, H.; Poutahidis, T.; Giakoustidis, A.; Gargavanis, A.; Giakoustidis, D.; Angelopoulou, K. Romidepsin Hepatocellular Carcinoma Suppression in Mice Is Associated with Deregulated Gene Expression of Bone Morphogenetic Protein and Notch Signaling Pathway Components. Mol. Biol. Rep. 2021, 48, 551–562. [Google Scholar] [CrossRef]
  149. Li, J.; Xiong, X.; Wang, Z.; Zhao, Y.; Shi, Z.; Zhao, M.; Ren, T. In Vitro High-Throughput Drug Sensitivity Screening with Patient-Derived Primary Cells as a Guide for Clinical Practice in Hepatocellular Carcinoma—A Retrospective Evaluation. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 699–710. [Google Scholar] [CrossRef]
  150. Xie, L.; Song, D.; Lan, J.; Liu, P.; Qin, S.; Ning, Y.; Liu, Q. Plasma Protein Levels and Hepatocellular Carcinoma: A Mendelian Randomization Study with Drug Screening Implications. Discov. Oncol. 2025, 16, 567. [Google Scholar] [CrossRef]
  151. Kim, N.; Yang, C. Butyrate as a Potential Modulator in Gynecological Disease Progression. Nutrients 2024, 16, 4196. [Google Scholar] [CrossRef]
  152. Merarchi, M.; Sethi, G.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; Ahn, K.S. Role of Natural Products in Modulating Histone Deacetylases in Cancer. Molecules 2019, 24, 1047. [Google Scholar] [CrossRef]
  153. Singh, A.K.; Bishayee, A.; Pandey, A.K. Targeting Histone Deacetylases with Natural and Synthetic Agents: An Emerging Anticancer Strategy. Nutrients 2018, 10, 731. [Google Scholar] [CrossRef]
  154. Choi, S.-W.; Friso, S. (Eds.) Nutrients and Epigenetics; CRC Press: Boca Raton, FL, USA, 2009; ISBN 978-0-429-14672-5. [Google Scholar]
  155. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  156. Hu, C.; Xu, B.; Wang, X.; Wan, W.; Lu, J.; Kong, D.; Jin, Y.; You, W.; Sun, H.; Mu, X.; et al. Gut Microbiota–Derived Short-chain Fatty Acids Regulate Group 3 Innate Lymphoid Cells in HCC. Hepatology 2023, 77, 48–64. [Google Scholar] [CrossRef] [PubMed]
  157. Kobayashi, M.; Mikami, D.; Uwada, J.; Yazawa, T.; Kamiyama, K.; Kimura, H.; Taniguchi, T.; Iwano, M. A Short-Chain Fatty Acid, Propionate, Enhances the Cytotoxic Effect of Cisplatin by Modulating GPR41 Signaling Pathways in HepG2 Cells. Oncotarget 2018, 9, 31342–31354. [Google Scholar] [CrossRef] [PubMed]
  158. Kumar, M.; Kaur, R.; Kanthaje, S.; Dhiman, R.K.; Chakraborti, A. Bacterial Metabolite Butyrate in Modulating Sorafenib-Targeted microRNAs to Curtail Its Resistance in Hepatocellular Carcinoma. J. Cancer Res. Clin. Oncol. 2023, 149, 5823–5839. [Google Scholar] [CrossRef]
  159. Shalhoub, H.; Gonzalez, P.; Dos Santos, A.; Guillermet-Guibert, J.; Moniaux, N.; Dupont, N.; Faivre, J. Simultaneous Activation and Blockade of Autophagy to Fight Hepatocellular Carcinoma. Autophagy Rep. 2024, 3, 2326241. [Google Scholar] [CrossRef] [PubMed]
  160. Sanaei, M.; Kavoosi, F.; Safari, M. Effect of 5′-Fluoro-2′-Deoxycytidine and Sodium Butyrate on the Gene Expression of the Intrinsic Apoptotic Pathway, P21, P27, and P53 Genes Expression, Cell Viability, and Apoptosis in Human Hepatocellular Carcinoma Cell Lines. Adv. Biomed. Res. 2023, 12, 24. [Google Scholar] [CrossRef]
  161. Kotantaki, P.; Mosialos, G. The Expression of Tumor Suppressor Gene Cyld Is Upregulated by Histone Deacetylace Inhibitors in Human Hepatocellular Carcinoma Cell Lines. Cell Biochem. Funct. 2016, 34, 465–468. [Google Scholar] [CrossRef]
  162. Hernández-Oliveras, A.; Izquierdo-Torres, E.; Meneses-Morales, I.; Rodríguez, G.; Zarain-Herzberg, Á.; Santiago-García, J. Histone Deacetylase Inhibitors Promote ATP2A3 Gene Expression in Hepatocellular Carcinoma Cells: P300 as a Transcriptional Regulator. Int. J. Biochem. Cell Biol. 2019, 113, 8–16. [Google Scholar] [CrossRef]
  163. Cao, S.; Budina, E.; Raczy, M.M.; Solanki, A.; Nguyen, M.; Beckman, T.N.; Reda, J.W.; Hultgren, K.; Ang, P.S.; Slezak, A.J.; et al. A Serine-Conjugated Butyrate Prodrug with High Oral Bioavailability Suppresses Autoimmune Arthritis and Neuroinflammation in Mice. Nat. Biomed. Eng. 2024, 8, 611–627. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, J.; Zhao, K.-N.; Vitetta, L. Effects of Intestinal Microbial–Elaborated Butyrate on Oncogenic Signaling Pathways. Nutrients 2019, 11, 1026. [Google Scholar] [CrossRef] [PubMed]
  165. Han, R.; Nusbaum, O.; Chen, X.; Zhu, Y. Valeric Acid Suppresses Liver Cancer Development by Acting as a Novel HDAC Inhibitor. Mol. Ther. Oncolytics 2020, 19, 8–18. [Google Scholar] [CrossRef]
  166. Mayorga-Ramos, A.; Barba-Ostria, C.; Simancas-Racines, D.; Guamán, L.P. Protective Role of Butyrate in Obesity and Diabetes: New Insights. Front. Nutr. 2022, 9, 1067647. [Google Scholar] [CrossRef]
  167. Longtine, A.G.; Greenberg, N.T.; Gonzalez, A.; Lindquist, A.; VanDongen, N.S.; Mahoney, S.A.; Rahman, G.; Clayton, Z.S.; Ziemba, B.P.; Ludwig, K.R.; et al. Oral Supplementation with the Short-Chain Fatty Acid Acetate Ameliorates Age-Related Arterial Dysfunction in Mice. Aging Biol. 2024, 2, 20240033. [Google Scholar] [CrossRef]
  168. Hirata, S.-I.; Kunisawa, J. Gut Microbiome, Metabolome, and Allergic Diseases. Allergol. Int. 2017, 66, 523–528. [Google Scholar] [CrossRef]
  169. Marquardt, J.U.; Gomez-Quiroz, L.; Camacho, L.O.A.; Pinna, F.; Lee, Y.-H.; Kitade, M.; Domínguez, M.P.; Castven, D.; Breuhahn, K.; Conner, E.A.; et al. Curcumin Effectively Inhibits Oncogenic NF-kB Signaling and Restrains Stemness Features in Liver Cancer. J. Hepatol. 2015, 63, 661–669. [Google Scholar] [CrossRef]
  170. Sanaei, M.; Kavoosi, F.; Arabloo, M. Effect of Curcumin in Comparison with Trichostatin A on the Reactivation of Estrogen Receptor Alpha Gene Expression, Cell Growth Inhibition and Apoptosis Induction in Hepatocellular Carcinoma Hepa 1-6 Cell lLine. Asian Pac. J. Cancer Prev. 2020, 21, 1045–1050. [Google Scholar] [CrossRef] [PubMed]
  171. Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of Curcumin: Problems and Promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef] [PubMed]
  172. Abd El-Hack, M.E.; El-Saadony, M.T.; Swelum, A.A.; Arif, M.; Abo Ghanima, M.M.; Shukry, M.; Noreldin, A.; Taha, A.E.; El-Tarabily, K.A. Curcumin, the Active Substance of Turmeric: Its Effects on Health and Ways to Improve Its Bioavailability. J. Sci. Food Agric. 2021, 101, 5747–5762. [Google Scholar] [CrossRef]
  173. dos Santos, P.W.D.S.; Machado, A.R.T.; De Grandis, R.A.; Ribeiro, D.L.; Tuttis, K.; Morselli, M.; Aissa, A.F.; Pellegrini, M.; Antunes, L.M.G. Transcriptome and DNA Methylation Changes Modulated by Sulforaphane Induce Cell Cycle Arrest, Apoptosis, DNA Damage, and Suppression of Proliferation in Human Liver Cancer Cells. Food Chem. Toxicol. 2020, 136, 111047. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, Y.; Shi, C.; Zhang, D.; Zhang, L.; Wang, L.; Gong, Z. Sulforaphane, an NRF2 Agonist, Alleviates Ferroptosis in Acute Liver Failure by Regulating HDAC6 Activity. J. Integr. Med. 2023, 21, 464–473. [Google Scholar] [CrossRef]
  175. Fantone, S.; Marzioni, D.; Tossetta, G. NRF2/KEAP1 Signaling Inhibitors in Gynecologic Cancers. Expert Rev. Anticancer. Ther. 2024, 24, 1191–1194. [Google Scholar] [CrossRef] [PubMed]
  176. Schiavoni, V.; Emanuelli, M.; Milanese, G.; Galosi, A.B.; Pompei, V.; Salvolini, E.; Campagna, R. Nrf2 Signaling in Renal Cell Carcinoma: A Potential Candidate for the Development of Novel Therapeutic Strategies. Int. J. Mol. Sci. 2024, 25, 13239. [Google Scholar] [CrossRef] [PubMed]
  177. Tossetta, G.; Fantone, S.; Togni, L.; Santarelli, A.; Olivieri, F.; Marzioni, D.; Rippo, M.R. Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants 2024, 13, 1270. [Google Scholar] [CrossRef]
  178. Tossetta, G.; Fantone, S.; Piani, F.; Crescimanno, C.; Ciavattini, A.; Giannubilo, S.R.; Marzioni, D. Modulation of NRF2/KEAP1 Signaling in Preeclampsia. Cells 2023, 12, 1545. [Google Scholar] [CrossRef]
  179. Men, X.; Han, X.; Oh, G.; Im, J.-H.; Lim, J.S.; Cho, G.H.; Choi, S.-I.; Lee, O.-H. Plant Sources, Extraction Techniques, Analytical Methods, Bioactivity, and Bioavailability of Sulforaphane: A Review. Food Sci. Biotechnol. 2023, 33, 539–556. [Google Scholar] [CrossRef]
  180. Okunade, O.; Niranjan, K.; Ghawi, S.K.; Kuhnle, G.; Methven, L. Supplementation of the Diet by Exogenous Myrosinase via Mustard Seeds to Increase the Bioavailability of Sulforaphane in Healthy Human Subjects after the Consumption of Cooked Broccoli. Mol. Nutr. Food Res. 2018, 62, e1700980. [Google Scholar] [CrossRef]
  181. Yan, L.; Yan, Y. Therapeutic Potential of Sulforaphane in Liver Diseases: A Review. Front. Pharmacol. 2023, 14, 1256029. [Google Scholar] [CrossRef]
  182. Venturelli, S.; Berger, A.; Böcker, A.; Busch, C.; Weiland, T.; Noor, S.; Leischner, C.; Schleicher, S.; Mayer, M.; Weiss, T.S.; et al. Resveratrol as a Pan-HDAC Inhibitor Alters the Acetylation Status of Jistone Proteins in Human-Derived Hepatoblastoma Cells. PLoS ONE 2013, 8, e73097. [Google Scholar] [CrossRef]
  183. Nowrasteh, G.; Zand, A.; Raposa, L.B.; Szabó, L.; Tomesz, A.; Molnár, R.; Kiss, I.; Orsós, Z.; Gerencsér, G.; Gyöngyi, Z.; et al. Fruit Extract, Rich in Polyphenols and Flavonoids, Modifies the Expression of DNMT and HDAC Genes Involved in Epigenetic Processes. Nutrients 2023, 15, 1867. [Google Scholar] [CrossRef]
  184. Galiniak, S.; Aebisher, D.; Bartusik-Aebisher, D. Health Benefits of Resveratrol Administration. Acta Biochim. Pol. 2019, 66, 13–21. [Google Scholar] [CrossRef]
  185. de Vries, K.; Strydom, M.; Steenkamp, V. A Brief Updated Review of Advances to Enhance Resveratrol’s Bioavailability. Molecules 2021, 26, 4367. [Google Scholar] [CrossRef]
  186. Szymkowiak, I.; Marcinkowska, J.; Kucinska, M.; Regulski, M.; Murias, M. Resveratrol Bioavailability After Oral Administration: A Meta-Analysis of Clinical Trial Data. Phytother. Res. 2025, 39, 453–464. [Google Scholar] [CrossRef] [PubMed]
  187. Sanaei, M.; Kavoosi, F.; Salehi, H. Genistein and Trichostatin A Induction of Estrogen Receptor Alpha Gene Expression, Apoptosis and Cell Growth Inhibition in Hepatocellular Carcinoma HepG 2 Cells. Asian Pac. J. Cancer Prev. 2017, 18, 3445–3450. [Google Scholar] [CrossRef] [PubMed]
  188. Sanaei, M.; Kavoosi, F.; Roustazadeh, A.; Golestan, F. Effect of Genistein in Comparison with Trichostatin A on Reactivation of DNMTs Genes in Hepatocellular Carcinoma. J. Clin. Transl. Hepatol. 2018, 6, 141–146. [Google Scholar] [CrossRef]
  189. Sanaei, M.; Kavoosi, F. Effect of DNA Methyltransferase in Comparison to and in Combination with Histone Deacetylase Inhibitors on Hepatocellular Carcinoma HepG2 Cell Line. Asian Pac. J. Cancer Prev. 2019, 20, 1119–1125. [Google Scholar] [CrossRef] [PubMed]
  190. El-Far, Y.M.; Khodir, A.E.; Emarah, Z.A.; Ebrahim, M.A.; Al-Gayyar, M.M.H. Chemopreventive and Hepatoprotective Effects of Genistein via Inhibition of Oxidative Stress and the Versican/PDGF/PKC Signaling Pathway in Experimentally Induced Hepatocellular Carcinoma in Rats by Thioacetamide. Redox Rep. 2022, 27, 9–20. [Google Scholar] [CrossRef]
  191. Sharifi-Rad, J.; Quispe, C.; Imran, M.; Rauf, A.; Nadeem, M.; Gondal, T.A.; Ahmad, B.; Atif, M.; Mubarak, M.S.; Sytar, O.; et al. Genistein: An Integrative Overview of Its Mode of Action, Pharmacological Properties, and Health Benefits. Oxid. Med. Cell Longev. 2021, 2021, 3268136. [Google Scholar] [CrossRef]
  192. Setchell, K.D.R.; Brown, N.M.; Desai, P.; Zimmer-Nechemias, L.; Wolfe, B.E.; Brashear, W.T.; Kirschner, A.S.; Cassidy, A.; Heubi, J.E. Bioavailability of Pure Isoflavones in Healthy Humans and Analysis of Commercial Soy Isoflavone Supplements. J. Nutr. 2001, 131, 1362S–1375S. [Google Scholar] [CrossRef]
  193. Bian, X.; Piipponen, M.; Liu, Z.; Luo, L.; Geara, J.; Chen, Y.; Sangsuwan, T.; Maselli, M.; Diaz, C.; Bain, C.A.; et al. Epigenetic Memory of Radiotherapy in Dermal Fibroblasts Impairs Wound Repair Capacity in Cancer Survivors. Nat. Commun. 2024, 15, 9286. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the HAT-HDAC mediated epigenetic transcriptional regulation. HDAC removes the acetyl groups (Ac) from histone tail forming closed chromatin structure (Heterochromatin) thereby reducing transcriptional activity of RNA polymerase II, while HAT catalyzes the transfer of acetyl groups from acetyl-CoA at the N-terminal histone tail resulting in an open chromatin state (Euchromatin) facilitating gene transcription. Created with BioRender.com.
Figure 1. Overview of the HAT-HDAC mediated epigenetic transcriptional regulation. HDAC removes the acetyl groups (Ac) from histone tail forming closed chromatin structure (Heterochromatin) thereby reducing transcriptional activity of RNA polymerase II, while HAT catalyzes the transfer of acetyl groups from acetyl-CoA at the N-terminal histone tail resulting in an open chromatin state (Euchromatin) facilitating gene transcription. Created with BioRender.com.
Cells 14 01337 g001
Figure 2. Classification and domain architecture of the HDACs Family. HDACs can be classified as NAD+ dependent (Class III) and Zinc dependent (Class I, II and IV). ZnF, ubiquitin binding zinc finger domain; Leucine-rich, Leucine-rich region.
Figure 2. Classification and domain architecture of the HDACs Family. HDACs can be classified as NAD+ dependent (Class III) and Zinc dependent (Class I, II and IV). ZnF, ubiquitin binding zinc finger domain; Leucine-rich, Leucine-rich region.
Cells 14 01337 g002
Figure 3. Impact of HDAC upregulation in hepatocellular carcinoma (HCC). HDAC overexpression is linked with cancer cell proliferation, promotion of angiogenesis, and metastases as well as inhibition of T-cell tumor infiltration and cancer cell autophagy and apoptosis. Created with BioRender.com.
Figure 3. Impact of HDAC upregulation in hepatocellular carcinoma (HCC). HDAC overexpression is linked with cancer cell proliferation, promotion of angiogenesis, and metastases as well as inhibition of T-cell tumor infiltration and cancer cell autophagy and apoptosis. Created with BioRender.com.
Cells 14 01337 g003
Figure 4. Role of natural HDAC inhibitors in Hepatocellular carcinoma. Compounds from gut microbiome and dietary sources act as natural histone deacetylase inhibitors by augmenting acetylation and halting tumorigenesis in HCC.
Figure 4. Role of natural HDAC inhibitors in Hepatocellular carcinoma. Compounds from gut microbiome and dietary sources act as natural histone deacetylase inhibitors by augmenting acetylation and halting tumorigenesis in HCC.
Cells 14 01337 g004
Table 1. HDACs characteristics based on data from Human Protein Atlas.
Table 1. HDACs characteristics based on data from Human Protein Atlas.
ProteinGene IDAliasesChrHDAC ClassCellular LocalizationHistone
Substrates
Non-Histone
Substrates
HDAC13065GON-10, HD1, KDAC1, RPD3, RPD3L11p35.2INucleus, CytoplasmLysine residues of H2A, H2B, H3, H4NR1D2, RELA, SP1, SP3, STAT3 and TSHZ3
HDAC23066HD2, KDAC2, RPD3, YAF16q21INucleus, CytoplasmLysine residues of H2A, H2B, H3, H4TSHZ3
HDAC38841HD3, KDAC3, RPD3, RPD3-25q31.3IPlasma membrane, Nucleus, CytoplasmH3K27KAT5, MEF2D, MAPK14, STAT3 and RARA
HDAC49759AHO3, BDMR, HA6116, HD4, HDAC-4, HDAC-A, HDACA, NEDCHF, NEDCHID2q37.3IIaNucleus, CytoplasmLysine residues of H2A, H2B, H3, H4HSPA1A and HSPA1B at Lys-77
HDAC510014HD5, NY-CO-917q21.31IIaNucleus, CytoplasmLysine residues of H2A, H2B, H3, H4RARA
HDAC610013CPBHM, HD6, JM21, KDAC6, PPP1R90Xp11.23IIbPlasma membrane, Nucleus, CytoplasmLysine residues of H2A, H2B, H3, H4Tubulin, α-tubulin, SQSTM1 and CTTN
HDAC751564HD7, HD7AA, HDAC712q13.11IIaNucleus, CytoplasmLysine residues of H2A, H2B, H3, H4RARA and ALKBH5
HDAC855869CDA07, CDLS5, HD8, HDACL1, KDAC8, MRXS6, RPD3, WTSXq13.1IPlasma membrane, Nucleus, CytoplasmLysine residues of H2A, H2B, H3, H4SMC3
HDAC99734ARCND4, HD7, HD7b, HD9, HDAC, HDAC7, HDAC7BB, HDAC9FL, HDRP, MITR7p21.1IIaNucleus, CytoplasmLysine residues of H2A, H2B, H3, H4-
HDAC1083933HD1022q13.33IIbNucleus, CytoplasmLysine residues of H2A, H2B, H3, H4MSH2
HDAC1179885HD113p25.1IVPlasma membrane, Nucleus, CytoplasmLysine residues of H2A, H2B, H3, H4-
NR1D2, Nuclear Receptor Subfamily 1 Group D Member 2; RELA, V-Rel Avian Reticuloendotheliosis Viral Oncogene Homolog A; SP1, specificity protein 1; SP3, specificity protein 3; STAT, signal transducers and activator of transcription 3; TSHZ3, Teashirt Zinc Finger Homeobox 3; KAT5, Lysine Acetyltransferase 5; MEF2D, Myocyte Enhancer Factor 2D; MAPK14, Mitogen-Activated Protein Kinase 14; RARA, Retinoic Acid Receptor Alpha; HSPA1A, Heat Shock Protein Family A (Hsp70) Member 1; HSPA1B, Heat Shock Protein Family A (Hsp70) Member 1B; SQSTM1, Sequestosome 1; CTTN, Cortactin; ALKBH5, AlkB Homolog 5; SMC3, Structural Maintenance Of Chromosomes 3; MSH2, MutS Homolog 2.
Table 2. Comprehensive Profile of Synthetic HDAC Inhibitors in HCC models.
Table 2. Comprehensive Profile of Synthetic HDAC Inhibitors in HCC models.
HDAC Inhibitor Name
(Trade Name) and Structure
AliasesMolecular
Formula
(MW)
Study
Design/Model
Key FindingsRef.
Resminostat (Kinselby)

Cells 14 01337 i001
4SC-201
RAS2410
BYK408740
C16H19N3O4S
(349.4 g/mol)
Phase I/II clinical trial; in vitro studiesApoptosis induction;
enhanced antitumor
activity; improved
efficacy with Sorafenib
[118,119,120]
Vorinostat (Zolinza)

Cells 14 01337 i002
SAHA
MK0683
C14H20N2O3
(264.32 g/mol)
In vitro and in vivo studiesImproved efficiency with Lenvatinib; synergistic anticancer effect via dual induction of apoptosis and autophagy[121,122,123]
Panobinostat (Farydak)

Cells 14 01337 i003
LBH589 C21H23N3O2
(349.4 g/mol)
Xenograft mouse models; cell linesReduction in lung metastasis; enhanced antitumor effect and apoptosis
induction in combination with radiotherapy
[124,125,126]
Romidepsin (Istodax)

Cells 14 01337 i004
Depsipeptide
Chromadax
FK228
C24H36N4O6S2
(540.7 g/mol)
In vitro and in vivo studiesCell cycle arrest; HCC
tumor suppression;
enhanced outcome with Olaparib
[127,128,129]
Belinostat (Beleodaq)

Cells 14 01337 i005
PXD101C15H14N2O4S
(318.3 g/mol)
Cell lines; murine modelsAntiproliferative and
cytotoxic effects with Bortezomib; apoptotic
induction and cell
suppression; enhanced anti-tumor immunity
[130,131,132]
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

Sadia, K.; Castagna, A.; Udali, S.; Ambrosani, F.; Pattini, P.; Beri, R.; Argentino, G.; Masutti, M.; Moruzzi, S.; Friso, S. Epigenetic Regulation Through Histone Deacetylation: Implications and Therapeutic Potential in Hepatocellular Carcinoma. Cells 2025, 14, 1337. https://doi.org/10.3390/cells14171337

AMA Style

Sadia K, Castagna A, Udali S, Ambrosani F, Pattini P, Beri R, Argentino G, Masutti M, Moruzzi S, Friso S. Epigenetic Regulation Through Histone Deacetylation: Implications and Therapeutic Potential in Hepatocellular Carcinoma. Cells. 2025; 14(17):1337. https://doi.org/10.3390/cells14171337

Chicago/Turabian Style

Sadia, Khulah, Annalisa Castagna, Silvia Udali, Francesca Ambrosani, Patrizia Pattini, Ruggero Beri, Giuseppe Argentino, Maria Masutti, Sara Moruzzi, and Simonetta Friso. 2025. "Epigenetic Regulation Through Histone Deacetylation: Implications and Therapeutic Potential in Hepatocellular Carcinoma" Cells 14, no. 17: 1337. https://doi.org/10.3390/cells14171337

APA Style

Sadia, K., Castagna, A., Udali, S., Ambrosani, F., Pattini, P., Beri, R., Argentino, G., Masutti, M., Moruzzi, S., & Friso, S. (2025). Epigenetic Regulation Through Histone Deacetylation: Implications and Therapeutic Potential in Hepatocellular Carcinoma. Cells, 14(17), 1337. https://doi.org/10.3390/cells14171337

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