Next Article in Journal
99mTc-A1 as a Novel Imaging Agent Targeting Mesothelin-Expressing Pancreatic Ductal Adenocarcinoma
Next Article in Special Issue
The Histone Demethylase LSD1/ΚDM1A Mediates Chemoresistance in Breast Cancer via Regulation of a Stem Cell Program
Previous Article in Journal
Three-Dimensional Conformal Radiotherapy-Based or Intensity-Modulated Radiotherapy-Based Concurrent Chemoradiotherapy in Patients with Thoracic Esophageal Squamous Cell Carcinoma
Previous Article in Special Issue
LncRNAs as Chromatin Regulators in Cancer: From Molecular Function to Clinical Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Drugs for Cancer and microRNAs: A Focus on Histone Deacetylase Inhibitors

CRCINA, INSERM, Université d’Angers, Université de Nantes, 44007 Nantes, France
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(10), 1530; https://doi.org/10.3390/cancers11101530
Submission received: 7 August 2019 / Revised: 9 September 2019 / Accepted: 3 October 2019 / Published: 10 October 2019
(This article belongs to the Special Issue Epigenetic Dysregulation in Cancer: From Mechanism to Therapy)

Abstract

:
Over recent decades, it has become clear that epigenetic abnormalities are involved in the hallmarks of cancer. Histone modifications, such as acetylation, play a crucial role in cancer development and progression, by regulating gene expression, such as for oncogenes or tumor suppressor genes. Therefore, histone deacetylase inhibitors (HDACi) have recently shown efficacy against both hematological and solid cancers. Designed to target histone deacetylases (HDAC), these drugs can modify the expression pattern of numerous genes including those coding for micro-RNAs (miRNA). miRNAs are small non-coding RNAs that regulate gene expression by targeting messenger RNA. Current research has found that miRNAs from a tumor can be investigated in the tumor itself, as well as in patient body fluids. In this review, we summarized current knowledge about HDAC and HDACi in several cancers, and described their impact on miRNA expression. We discuss briefly how circulating miRNAs may be used as biomarkers of HDACi response and used to investigate response to treatment.

1. Introduction

In recent decades, non-coding RNAs have been described as key regulators of cellular functions and differentiation. This includes long non-coding RNAs with a size above 200 nucleotides (nt) and small non-coding RNAs (under 200 nt) consisting of numerous subtypes. Micro-RNAs (miRNAs) are endogenous small non-coding RNA of about 19 to 22 nucleotides that modulate gene expression through translational repression, or degradation of the target messenger RNA (mRNA) [1]. A single miRNA has the capacity to inhibit numerous different mRNA targets [2] explaining why miRNAs are potent regulators of gene expression. miRNAs are also important regulators since more than 60% of human genes are regulated by them, as demonstrated by Friedman et al. [3]. In cancer, miRNAs can act as tumor suppressors (TS-miR) or oncogenes (oncomiR), depending on their targets. Recent research has found that miRNAs can not only be detected in tissues but also in all body fluids such as blood, saliva, urine, and milk [4], where they can be used as biomarkers [5]. MicroRNAs harbor attractive features for uses ranging from translation to clinical practices, such as an easy extraction from body fluids, a resistance to molecular degradation by their encapsulation in exosomes, or by their interaction with lipids and proteins, and their easy quantification by different methods including quantitative PCR [6].
In the following sections, we will discuss how miRNAs are regulated by epigenetic drugs, such as histone deacetylase inhibitors (HDACi) used in cancer. We will also succinctly discuss the use of circulating miRNAs as a predictor of response to epigenetic clinical therapies.

2. Epigenetic Drugs in Cancer

Epigenetic drugs consist of compounds that inhibit proteins implicated in the writing, the reading, or the erasing of epigenetic marks such as DNA methylation or post-translational modifications (PTM) of histones. Concerning DNA methylation, epigenetic drugs include, for example, the food and drug administration (FDA)-approved decitabine targeting DNMT1 (DNA methyltransferase 1), or AG-221 (or enasidenib), currently tested in a phase III clinical trial (NCT02577406), targeting IDH2 (Isocitrate DeHydrogenase 2), an enzyme providing cofactor for the DNA methylation eraser protein TET1 (ten eleven translocation 1). Concerning PTM, most of the focus has been on histone acetylation erasers that will be described below, but some of them have also been developed against histone methylation writers or erasers, as well as histone acetylation readers, i.e., bromodomain-containing proteins (see review [7]). In this review, we decided to focus on the largest class of epigenetic drugs, the histone deacetylase inhibitors. It is a family of promising epigenetic agents for cancer treatments. Indeed, during cancer initiation, a decrease of histone acetylation leads to the repression of genes resulting in uncontrolled cell proliferation, differentiation and decreased apoptosis. Later, during cancer progression, increasing histone deacetylases (HDAC) activity leads to a loss of cell adhesion, resulting in cell migration, invasion and angiogenesis.

2.1. Histone Deacetylase

Previous works have identified 18 deacetylases. These enzymes are classified in four categories depending on homologies with yeast deacetylases, function, localization and substrates (Table 1, more details in the review [8]). Essentially, nucleic HDAC removes the acetyl group on the N-ε-lysine side chain of the histone N-terminal tail, increasing its positive charge, and stabilizing DNA-histone complexes by electrostatic interactions. This induces chromatin compaction and transcription repression. Cytoplasmic HDACs can deacetylate non-histone proteins [9,10,11,12,13,14,15,16,17,18].

2.2. Histone Deacetylase Inhibitors

HDACi were first identified from natural sources, currently however, new molecules have been developed with an improved activity and specificity. To date, a high number of compounds are available and evaluated in preclinical or clinical studies. HDACi are classified in four classes according to their chemical structure [19], hydroxamates is the largest one. These compounds are usually pan-HADCi acting in the range of micro to nanomolar concentrations. The well-known members of this family are vorinostat (SAHA), belinostat (PDX101) and panobinostat (LBH589). All of these are approved by the USA food and drug administration (FDA) for the treatment of respectively (i) cutaneous T-cell lymphoma (CTCL) [20], (ii) patients with relapse or refractory peripheral T-cell lymphoma (PTCL) [21], or (iii) multiple myeloma (MM) [22]. Trichostatin A (TSA), the first natural hydroxamate, was excluded from clinical uses due to its high toxicity [23] despite its interesting effects at nanomolar concentrations on cancer cells. The two other groups are benzamides and cyclic peptides which target mainly class I HDAC. The prototypes of these families are entinostat (MS-275) and romidepsin (FK2208) respectively. Romidepsin was approved by FDA for the treatment of CTCL [24] and PTCL [25]. Finally, short chain carboxylic acids, such as valproic acid (VPA) or sodium butyrate (NaBu), inhibit class I and class IIa HDACs.

2.3. FDA-Approved Histone Deacetylase Inhibitors

Vorinostat. Vorinostat or suberoylanilide hydroxamic acid (SAHA) is a HDACi belonging to the hydroxamate family, acting on class I and class II HDAC (Table 1 and Table 2). This compound is probably the most used HDACi for preclinical and clinical evaluations. In October 2006, Vorinostat was approved in the USA by the FDA for the treatment of CTCL [26]. When used as a single agent, a poor efficacy was observed on solid tumours [27]. Thus, combination strategies have been or are tested (approximately 134 phase II clinical trials and nine phase III clinical trials in progress in 2019, ClinicalTrials.gov). For examples, Vorinostat is currently evaluated in phase III clinical trials in combination with alkylating agents, proteasome inhibitors, anthracyclines, anti-angiogenics and/or antimetabolites.
Romidepsin. Romidepsin is a bicyclic peptide (Table 2) isolated from a bacteria named Chromobacterium violaceum [28,29]. This molecule inhibits mainly class I HDACs (Table 1). Romidepsin is a prodrug that has to be activated in cells to be efficient by the reduction of the disulphide bond included in its structure with the zinc ion present in the HDAC catalytic site. This molecule was approved by the FDA in 2009 for the treatment of patients with CTCL who have received at least one prior systemic therapy [30]. In 2011, the FDA approved romidepsin for the treatment of patients with PTCL who have failed or who were refracted to at least one prior systemic therapy [31]. As for Vorinostat, a poor activity was observed on solid tumours leading to the evaluation of combination strategies in clinic (52 phase II clinical trials and four phase III clinical trials, ClinicalTrials.gov).
Belinostat. Belinostat, a hydroxamate HDACi, presents a broad-spectrum of action (class I and class II HDACi). Belinostat was approved by FDA in 2014 for the treatment of patients with PTCL that was refractory or had relapsed after prior treatment [21,32]. A second phase II clinical trials confirmed these results and showed a better activity of belinostat on PTCL compared to CTCL [33]. The poor activity of belinostat on solid tumor [34] has led to the evaluation of this HDACi in combination with current chemotherapeutic agents (24 phase II clinical trials, ClinicalTrials.gov), notably alkylating agents (cisplatin and carboplatin).
Panobinostat. Panobinostat is a pan-HDACi of the hydroxamate family. A phase III clinical trials, named PANORAMA1, was at the origin of the approval of panobinostat by FDA in 2015, in combination with bortezomib and dexamethasone, for the treatment of patients with multiple myeloma who have received at least two prior regimens, including bortezomib and an immunomodulatory agent [35]. Numerous phase II or III clinical trials, on different cancers, were conducted or are in progress to evaluate the efficacy of this molecule alone or in combination.

3. Effect of Histone Deacetylase Inhibitors on Tumor Cells

According to the large number of genes regulated by HDAC, HDACi can affect numerous cellular mechanisms implicated in oncogenic properties of cancer cells. It was notably shown that these molecules induce proliferation arrest, sensitivity to apoptosis, decrease angiogenesis and affect DNA damage repair machinery (Figure 1). Here, we will present only the major pathways affected by HDACi (for more details, see reviews [36,37]).

3.1. Cell Cycle

HDACi induced a cell cycle arrest in G0/G1, G1/S or G2/M phase depending on the cancer cell line and on the used HDACi [38]. Induction of expression of the cyclin-dependent kinase (CDK) inhibitor gene CDKN1A, coding for p21, seems to be a major mechanism in the cell cycle arrest effect of HDACi even if other CDK inhibitors genes are induced by these molecules [39]. The protein p53 was described as a regulator of p21 expression through binding to its promotor [40]. However, the induction of p21 following HDACi treatment is independent on p53 status of cells [41,42,43] whereas some studies have described an activation of p53 after HDAC inhibition [44,45]. Other mechanisms could explain this observation such as dephosphorylation of retinoblasma protein (Rb) [46,47,48] and inhibition of E2F transcriptional activity [49].

3.2. Cell Death

HDACi modulates both the intrinsic and extrinsic pathways of apoptosis. Concerning the extrinsic pathway, HDACi increased Death Receptor (DR4, DR5) expression in cancer cells [49,50,51,52]. Interaction of DR4 and DR5 with tumor necrosis factor (TNF)-super family receptor ligands (Fas-L, TRAIL (TNF-Related Apoptosis Inducing Ligand), TNFα) induced apoptosis by the activation of caspase 8 and 10. Additionally to these regulations, HDACi can also modulate the level of intracellular adaptor molecules, such as the inhibitor of apoptosis named cellular FLICE (Caspase 8)-inhibitory protein (c-FLIP) [50,52,53], or by modulating the interaction between Fas-associated death domain (FADD) and the death-inducing signaling complex (DISC) [50,54]. Intrinsic apoptotic pathways are classically activated by cellular stress stimuli such as free radicals, misfolded proteins or DNA damages. Chemotherapeutic agents can also induce these stress stimuli leading to an increased permeability of the mitochondria and to caspases activation following the release of pro-apoptotic proteins. Intrinsic apoptosis in cells is regulated by the balance of expression of pro-apoptotic (Bak and Bax) and anti-apoptotic BCL-2 proteins (BCL-2, BCL-XL, MCL-1). BH3-only proteins (Bad, Bik, Bid, Bim, Puma, Noxa), a third family of pro-apoptotic proteins, are sensors of cellular stress, and fine tune apoptosis in cells. It is now well established that HDAC inhibition leads to an increasing expression of the pro-apoptotic BCL-2 protein members or BH3-only proteins, such as Bim [55].

3.3. Angiogenesis

HDACi have a mainly anti-angiogenic action, modulating angiogenesis by decreasing VEGF expression and hypoxia-inducible factor-1α (HIF1α), but also inducing VEGF (vascular endothelial growth factor) expression in several models of cancers [56,57,58,59,60]. Additional mechanisms were described such as an upregulation of the tumor suppressor gene von Hippel Lindau (VHL) and an alteration of the HSP90 (Heat Shock Protein 90) chaperone function, by modification of its acetylation, all leading to the degradation of HIF1α [61,62]. A direct action of HDACi on the HIF1α stability was described as well, through its acetylation [12]. Finally, HDACi can also affect the capacity of endothelial cells to induce angiogenesis in functional tests [63,64,65,66,67].

3.4. DNA Damage

Sensitivity of cancer cells to chemotherapeutic agents, such as alkylating agents or topoisomerase inhibitors, and radiotherapies, can depend on DNA damage repair (DDR) machinery. An increase of the duration of DNA damage induced by irradiation of cancer cells was observed following treatments with HDACi such as VPA, NaBu, vorinostat and MS-275. This demonstrates the incapacity of cells to repair double strand break (DSB) following HDAC inhibition [68,69,70,71]. These observations can be explained by the capacity of HDACi to repress proteins such as Rad50, Ku70 and Ku80, implicated in DDR [71,72]. Others studies showed that TSA, vorinostat and abexinostat can repress BRCA1 (Breast Cancer 1) and RAD51 (Recombination Protein A) expressions [73,74] and thereby inhibit the homologous recombination and the non-homologous recombination end joining DDR mechanisms [70,74,75,76]. Finally, cancer cells treatment with HDACi leads to the induction of reactive oxygen species (ROS) which cooperate with the DDR inhibition to induce DNA damages [77,78]. Proposed mechanisms for the induction of ROS by HDACi are a (i) downregulation of the expression of thioredoxin (TRX), reducing protein, (ii) an induction of the expression of the thioredoxin-binding protein-2 (TBP-2) gene as shown in prostate cancer cells [79], and (iii) the induction of the thioredoxin-interacting protein (TXNIP), an inhibitor of TRX, as demonstrated in human gastric cancer cells and HeLa cells [80,81].

4. Effect of Histone Deacetylase Inhibitors on microRNA Expressions in Cancer

HDACi treatments can modulate miRNA expressions in tumor cells. Indeed, the first step of miRNA biogenesis is the transcription of the miRNA gene. As classical genes, miRNAs, located outside or inside a coding gene, have their own promoter, TSS (transcription start site), and terminator signals, that are sensitive to epigenetic modifications, such as lysine acetylation which classically opens chromatin structure and enhances transcription activation.

4.1. microRNAs Dysregulated in Cancer

miRNA dysregulation in cancer was first reported in 2002, when miR-15 and miR-16 were identified at 13q14.3, a frequently deleted region in chronic lymphocytic leukemia (CLL), leading to the overexpression of their target, i.e., BCL-2 (B cell lymphoma 2) [82]. Different miRNAs have been then labeled as TS-miR (tumor Suppressor miR) or oncomiR based on the nature of their target mRNAs. OncomiRs can repress expression of protein-coding tumor suppressor genes and are frequently upregulated in cancer, whereas TS-miRs target cancer-promoting genes and are downregulated [83].
Let-7c is a one of the most described TS-miRs. It belongs to the let-7 family, highly conserved between species [84]. Let-7c is frequently downregulated in cancer, or even deleted since it is located in a region of frequent homozygous deletion [82]. Let-7c targets various oncogenes and cancer related genes such as IL6-R (interleukin-6 receptor) [85] or E2F5 (E2F transcription factor 5) [86] (Table 3). Its downregulation is also associated with poor prognosis in non-small cell lung carcinoma (NSCLC) [87], in colorectal cancer, or in metastatic prostate cancer [88].
The miR-17-92 cluster highly conserved among species, comprises six miRNAs (miR-17-5p, miR-18a, miR-19a, miR-20a, miR19b-1 and miR-92a-1), that are overexpressed in many human cancers. miR-18a is one of the most expressed of this cluster, and is considered as an oncomiR. It has been found to be upregulated in breast cancer, head and neck squamous cell carcinoma, esophageal squamous cell carcinoma, gastric carcinoma, pancreatic carcinoma, hepatocellular, and colorectal carcinoma [89]. Interestingly the concentration of miR-18a in plasma or serum of patients with cancer is much higher than that of healthy persons [89]. So aberrant expression of miRNA might serve as a biomarker of cancer or to evaluate cancer response to treatment in non-invasive liquid biopsy.
Over these two examples, numerous miRNAs are dysregulated in malignancies and many data are currently available on their expression for diagnostic or prognostic uses (see review [90]).

4.2. miRNA Regulated by Histone Deacetylase Inhibitors

A few years back, there were discrepancies about miRNA expression modifications by HDACi in tumors [100,101] probably resulting from different parameters such as cell lines and/or concentrations used. Since then, involvement of miRNAs in HDACi effects on tumors have been well documented. In vitro, miRNAs have an important role in HDACi effects such as in colorectal cancer, where it has been demonstrated that vorinostat modulates not less than 275 miRNAs resulting in a myriad of possible targets and pathways affected [102]. Moreover, in some cases, miRNAs modulated by HDACi have been correlated with tumor stage or clinical outcome. In this section, we will describe the main miRNAs involved in HDACi effects on tumors. In the current state of the art, it is challenging to find a link between miRNAs, HDACi and/or a specific cancer or pathways, and so the following section is ordered regarding both HDACi approval and actions of miRNAs involved (namely, TS-miRs and oncomiRs).

4.2.1. FDA-Approved HDACi and miRNAs

As mentioned earlier, only four HDACi have been approved by the FDA, namely: Vorinostat (SAHA), Panobinostat (LBH589), Belinostat (PXD101), and Romidepsin (FK288). Several studies on various tumor models have been led to understand the mechanisms of these molecules and especially, the importance of miRNAs in tumor-suppressive effects of these HDACi (Table 4).
HDACi-induced TS-miRs. Treatment by HDACi leads to an increase expression of TS-miR from the let-7 family in several models. Similarly, in hepatocellular cancer, in vitro and in vivo treatment with vorinostat or panobinostat triggers let-7b upregulation, leading to the downregulation of BCL-XL, TRAIL (tumor necrosis factor (ligand) superfamily, member 10) or the oncoprotein HMG2A (high mobility group box 3) [103] (Figure 2). Other upregulation, induced by vorinostat, of almost all let-7 family members have been reported in ovarian cancer by Balch et al. [104] as well as in renal cancer by Pili et al. [105]. Conversely, studies described downregulation induced by vorinostat of let-7 family members (let-7b, let-7c, let-7f) in other types of tumors such as lung [106] and thyroid cancers [107]. However, these last studies only described miRNAs modification without going further into let-7-related mechanisms and functions. These discrepancies can also be a consequence of the methods used to purify and screen miRNAs.
Another example of TS-miR is the miR-200 family, consisting of five members divided into two clusters, namely, miR-200b, -200a, and -429 (cluster I); and miR-200c and -141 (cluster II). They are often found to be lost in cancers with different pathways involved [108]. This family appears to be linked with HDACi effects especially in breast cancers were two studies described upregulation of miR-200a and miR-200c induced by vorinostat resulting in (i) an upregulation of antioxidant pathway Nrf2 and (ii) a decrease of proliferation, invasion and migration in tumor cell lines [109,110].
Other miRNAs have been described as playing a crucial role in these HDACi-induced modifications depending on cancer type. Panobinostat treatment has lead to increased cell senescence through miR-31 in breast cancer cells [111]. In pancreatic cancer cells, vorinostat induced many modifications of cell phenotype through miR-34a [112]. One of the most common mechanisms described in the literature is the ability by several HDACi to increase apoptosis in various tumor cell lines in a miRNA-dependent manner (leukemia, lymphoma, pancreatic cancer). This mechanism has been explained by a HDACi-induced upregulation of several miRNAs such as miR-15a, miR-16, miR-34a or miR-195 leading to a downregulation of their target mRNAs mainly (but not only) from the BCL-2 family in vitro and in vivo in mice [104,113,114] (Figure 2).
HDACi-induced oncomiR. The role of miR-17~92 cluster members in promoting tumorigenesis has been widely demonstrated and thus, effects of HDACi on these miRNAs has also been evaluated. Even though the oncogenic role of the miR-17~92 cluster has been largely described, the six miRNAs composing this cluster are not equivalent when it comes to promoting tumorigenesis. Consistently, HDACi affect these miRNAs towards repressing the tumor-promoting tendency of this cluster. In the literature, vorinostat mechanisms often seem to rely on miR-17~92 miRNAs. Indeed, in lymphoma, vorinostat decreases miR-17-5p and miR-18 through c-myc, leading to more sensitivity to apoptosis of tumor cells [115] (Figure 2). In another hematopoietic cancer, Lepore et al. demonstrated that vorinostat in human leukemia cell lines, leads to increasing apoptosis through repression of BARD-1 (BRCA1 associated RING domain 1) protein. This vorinostat-induced BARD-1 repression was due to the modulation of several miRNAs within the cell including especially, and surprisingly, an upregulation of miR-19a and miR-19b [116]. This highlights the fact that despite its role as an onco-miR in some cases, treatment mechanisms involving miR-19 are diverse and still need to be fully elucidated. Modulation of this cluster by HDACi has also been confirmed in solid tumors [117]. miR-20a and other miRNAs from the miR-17~92 cluster showed an altered expression in hepatocellular cancer, by vorinostat resulting in an upregulation of MICA protein levels and a better recognition of these tumor cells by innate immune cells and especially NK cells [118]. Moreover in colorectal and renal cancer, the decreased cell proliferation induced by vorinostat have been linked to a repression of the miR-17~92 cluster expression [119,120].

4.2.2. Other HDACi and miRNAs

There are plenty of HDACi that have not yet been approved by the FDA. Some of them are involved in phase III clinical trials such as Valproic acid to treat cervical and ovarian cancers or Tacedinaline for multiple myeloma and lung cancers [124]. Others are in earlier stages but nonetheless, interesting studies have been done to strengthen the close relationship between HDACi effects and miRNA-related mechanisms (Table 5).
Firstly, and expectedly, some non-approved HDACi act on similar pathways and miRNA clusters that the ones authorized by the FDA. In lung cancer, the let-7 family miRNAs are also upregulated by TSA, leading to increased cell cycle arrest and apoptosis of tumor cells compared to adjacent non-tumorous lung tissue [125]. Similarly, in lymphoma, let-7a alongside with other miRNAs are upregulated by Romidepsin, decreasing anti-apoptotic proteins such as BCL-2 (B-cell CLL/lymphoma 2) and BCL-XL (BCL2-like 1) [126]. The miR-17~92 cluster members have been described to be regulated by butyrate in colorectal cancer [127] and the miR-200 family is involved in reducing tumor cell proliferation in NSCLC and SCLC (Small Cell Cancer of the Lung) treated with Entinostat (MS275) [128]. miR-34 and miR-15a, described below, are also upregulated by AR42 in ovarian cancer, which trigger a cascade of pathways leading to a decrease of Wnt receptor signaling and EMT (epithelial mesenchymal transition), and an increase of negative regulation processes of cell cycle and apoptosis [104]. The same HDACi in pancreatic cancer, decreases p53 and cyclins protein levels thanks to variation of miR-30d, miR-33, and miR-125b, leading to the inhibition of cell proliferation, invasion and tumor growth, and to an increase of ROS generation, DNA damage and apoptosis [129]. Mocetinostat, a clinical phase II HDACi, has been described as involving miR-31 in the inhibition of E2F6 (E2F transcription factor 6), leading to apoptosis of the prostate tumor cells [130]. Another HDACi, OBP-801, has been described as inducing, both in vitro and in vivo (mice), a decrease of tumor cell growth involving an upregulation of miR-320a [131]. In the same study, they also identified that miR-320a was almost not modified by other HDACi such as SAHA or TSA, highlighting the mechanistic specificities of HDACi.

4.2.3. Clinical Relevance

Interestingly, miRNAs modulated by HDACi have been proven to have importance for clinical outcomes, such as miR-200c and miR-203 in pancreatic adenocarcinomas directly resected from patients. Indeed, the group with no recurrence within six months exerted a much higher level of these two miRNAs than the “recurrence group” [146]. In primary resected tumors, it has been demonstrated that miR-200c and miR-203 may also have a biomarker relevance. Indeed, Hibino et al. showed a significant association between non-recurrence and a high expression of miR-203 and miR-200c [114]. In a clinical study on renal cancer patients, miR-605 was directly targeted by a combination of vorinostat and bevacizumab, an antibody targeting growth factors. They demonstrated that this miRNA was upregulated in treatment responders at baseline and that it was downregulated after treatment ([105]; clinical trial NCT00324870). This is explained by the fact that these miRNAs, modified by epigenetic drugs such as HDACi, crosstalk with other proteins such as p53 for instance and are, therefore, able to shift pathways into anti-tumor outcomes for the cell. To further confirm the importance of miRNAs and their relevance in clinics, several ongoing trials plan to investigate miRNA involvement in HDACi-related effects such as Belinostat in carcinoma patients (NCT00926640), or vorinostat in bladder and renal cancers (NCT00926640).
Overall, as previously noticed, it appears difficult to bring out common mechanisms whether it is regarding HDACi molecules, tumor types or miRNAs involved. Conventional clusters such as let-7, miR-17~92, miR-200 are often described to be modified by HDACi but other less studied miRNAs have also been recently described. As expected, effects described are consistent with pathway modifications described in Section 3 of this review. Finally, most of the aforementioned articles have functionally tested miRNAs (mostly with miRNA mimic and/or anti-miR), describing both the importance and the need of these miRNAs to be involved in HDACi-induced mechanisms.

4.3. Histone Deacetylase Inhibitors and Circulating miRNAs

As mentioned earlier, miRNAs modulated by HDACi can also be screened in body fluids even if few studies have investigated this characteristic. As a proof of concept, Pili et al. evaluated the modulation of circulating miRNAs in clear-cell renal cell carcinoma (ccRCC) patients under a combinatory treatment of vorinostat and bevacizumab (a humanised monoclonal antibody that neutralises VEGF) [105]. They observed in responder patients an upregulation of miR-20a and miR-let-7b and a downregulation of miR-142-3p, miR-154, miR-605 and miR-199a-5p after treatment. Conversely, miR-605 was upregulated after treatment in progressor patients. Interestingly, this miRNA participates in the p53 network [147] and is frequently upregulated or mutated in cancers [148,149]. To our knowledge, this is the only study about the use of circulating miRNAs as a prognostic biomarker of HDACi response, even if it is a conventional approach for other anti-tumor treatments, as recently described in the review of Najminejad et al. [150] and Pardini et al. [151]. We believe however, that it can be a promising approach since miRNAs are stable and easily detectable in all body fluids. Indeed, circulating miRNA are protected from RNase activity through their conjugation with proteins, their inclusion in lipid or lipoprotein complexes or through their loading in exosomes/microvesicles. Exosomes are small intraluminal vesicles that are 50–150 nm in diameter. They are generated inside multivesicular endosomes (MVB) [152] that fuse with cell membranes and release the vesicles into the extracellular space. Exosomal miRNAs participate in intercellular communication (Figure 3). Uptake by normal cells of the exosome cargo secreted by cancer cells can affect the behavior of recipient cells in various ways that provide benefits to the tumor. Several studies have described how these exosomal miRNAs, induced or not by treatment, participate in tumor immune escape [153,154] or drug resistance [155,156].

5. Conclusions

Many miRNAs have shown different expression levels in response to HDACi treatment. Some of them can potentiate the anti-tumor response, or on the contrary, decrease it. Since tumor cells release miRNAs through exosomes that can be detected in all body fluids, such as plasma, urine or saliva, analysis of circulating miRNAs in patient liquid biopsies provides promising biomarkers to monitor drugs in patients. However, to date, it is still challenging to accurately identify clinically relevant miRNAs due to the lack of standardization in their extraction or in the conservation of biopsy, which greatly affects the stability of miRNAs.

Author Contributions

Conceptualization, D.F.; writing—original draft preparation, P.A., C.B. and D.F.; writing—review and editing, D.F. and C.B.; funding acquisition, D.F. and C.B.

Funding

Our research was funded by grant from Ligue contre le Cancer, comités 22, 29, 35, 44, 56 and from Cancéropole Grand Ouest—AO Structurant—ExomiR. P.A. was supported by a grant from Région Pays de la Loire—Episavmen.

Acknowledgments

In this section you can acknowledge any support given which is not covered by the author contribution or funding sections. This may include administrative and technical support, or donations in kind (e.g., materials used for experiments).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gebert, L.F.R.; MacRae, I.J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell Biol. 2019, 20, 21–37. [Google Scholar] [CrossRef]
  2. Selbach, M.; Schwanhäusser, B.; Thierfelder, N.; Fang, Z.; Khanin, R.; Rajewsky, N. Widespread changes in protein synthesis induced by microRNAs. Nature 2008, 455, 58–63. [Google Scholar] [CrossRef] [PubMed]
  3. Friedman, R.C.; Farh, K.K.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef]
  4. Chen, X.; Liang, H.; Zhang, J.; Zen, K.; Zhang, C.-Y. Secreted microRNAs: A new form of intercellular communication. Trends Cell Biol. 2012, 22, 125–132. [Google Scholar] [CrossRef]
  5. Qin, X.; Xu, H.; Gong, W.; Deng, W. The Tumor Cytosol miRNAs, Fluid miRNAs, and Exosome miRNAs in Lung Cancer. Front. Oncol. 2014, 4, 357. [Google Scholar] [CrossRef]
  6. Chen, C.; Tan, R.; Wong, L.; Fekete, R.; Halsey, J. Quantitation of microRNAs by real-time RT-qPCR. Methods Mol. Biol. 2011, 687, 113–134. [Google Scholar]
  7. Kelly, A.D.; Issa, J.-P.J. The promise of epigenetic therapy: Reprogramming the cancer epigenome. Curr. Opin. Genet. Dev. 2017, 42, 68–77. [Google Scholar] [CrossRef] [PubMed]
  8. Seto, E.; Yoshida, M. Erasers of Histone Acetylation: The Histone Deacetylase Enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, L.; Fischle, W.; Verdin, E.; Greene, W.C. Duration of nuclear NF-kappaB action regulated by reversible acetylation. Science 2001, 293, 1653–1657. [Google Scholar] [CrossRef] [PubMed]
  10. Gaughan, L.; Logan, I.R.; Cook, S.; Neal, D.E.; Robson, C.N. Tip60 and histone deacetylase 1 regulate androgen receptor activity through changes to the acetylation status of the receptor. J. Biol. Chem. 2002, 277, 25904–25913. [Google Scholar] [CrossRef] [PubMed]
  11. Gu, W.; Roeder, R.G. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 1997, 90, 595–606. [Google Scholar] [CrossRef]
  12. Jeong, J.W.; Bae, M.K.; Ahn, M.Y.; Kim, S.H.; Sohn, T.K.; Bae, M.H.; Yoo, M.A.; Song, E.J.; Lee, K.J.; Kim, K.W. Regulation and destabilization of HIF-1alpha by ARD1-mediated acetylation. Cell 2002, 111, 709–720. [Google Scholar] [CrossRef]
  13. Martinez-Balbas, M.A.; Bauer, U.M.; Nielsen, S.J.; Brehm, A.; Kouzarides, T. Regulation of E2F1 activity by acetylation. EMBO J. 2000, 19, 662–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Patel, J.H.; Du, Y.; Ard, P.G.; Phillips, C.; Carella, B.; Chen, C.J.; Rakowski, C.; Chatterjee, C.; Lieberman, P.M.; Lane, W.S.; et al. The c-MYC oncoprotein is a substrate of the acetyltransferases hGCN5/PCAF and TIP60. Mol. Cell. Biol. 2004, 24, 10826–10834. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, C.; Fu, M.; Angeletti, R.H.; Siconolfi-Baez, L.; Reutens, A.T.; Albanese, C.; Lisanti, M.P.; Katzenellenbogen, B.S.; Kato, S.; Hopp, T.; et al. Direct acetylation of the estrogen receptor alpha hinge region by p300 regulates transactivation and hormone sensitivity. J. Biol. Chem. 2001, 276, 18375–18383. [Google Scholar] [CrossRef] [PubMed]
  16. Kovacs, J.J.; Murphy, P.J.; Gaillard, S.; Zhao, X.; Wu, J.T.; Nicchitta, C.V.; Yoshida, M.; Toft, D.O.; Pratt, W.B.; Yao, T.P. HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor. Mol. Cell 2005, 18, 601–607. [Google Scholar] [CrossRef] [PubMed]
  17. Yuan, Z.L.; Guan, Y.J.; Chatterjee, D.; Chin, Y.E. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science 2005, 307, 269–273. [Google Scholar] [CrossRef] [PubMed]
  18. Zhang, Y.; Li, N.; Caron, C.; Matthias, G.; Hess, D.; Khochbin, S.; Matthias, P. HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo. EMBO J. 2003, 22, 1168–1179. [Google Scholar] [CrossRef] [Green Version]
  19. Martinet, N.; Bertrand, P. Interpreting clinical assays for histone deacetylase inhibitors. Cancer Manag. Res. 2011, 3, 117–141. [Google Scholar] [Green Version]
  20. Marks, P.A. Discovery and development of SAHA as an anticancer agent. Oncogene 2007, 26, 1351–1356. [Google Scholar] [CrossRef] [Green Version]
  21. Poole, R.M. Belinostat: First global approval. Drugs 2014, 74, 1543–1554. [Google Scholar] [CrossRef] [PubMed]
  22. Richardson, P.G.; Laubach, J.P.; Lonial, S.; Moreau, P.; Yoon, S.S.; Hungria, V.T.; Dimopoulos, M.A.; Beksac, M.; Alsina, M.; San-Miguel, J.F. Panobinostat: A novel pan-deacetylase inhibitor for the treatment of relapsed or relapsed and refractory multiple myeloma. Expert Rev. Anticancer Ther. 2015, 15, 737–748. [Google Scholar] [CrossRef] [PubMed]
  23. Vanhaecke, T.; Papeleu, P.; Elaut, G.; Rogiers, V. Trichostatin A-like hydroxamate histone deacetylase inhibitors as therapeutic agents: Toxicological point of view. Curr. Med. Chem. 2004, 11, 1629–1643. [Google Scholar] [CrossRef] [PubMed]
  24. Grant, C.; Rahman, F.; Piekarz, R.; Peer, C.; Frye, R.; Robey, R.W.; Gardner, E.R.; Figg, W.D.; Bates, S.E. Romidepsin: A new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors. Expert Rev. Anticancer Ther. 2010, 10, 997–1008. [Google Scholar] [CrossRef] [PubMed]
  25. Iyer, S.P.; Foss, F.F. Romidepsin for the Treatment of Peripheral T-Cell Lymphoma. Oncologist 2015, 20, 1084–1091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Mann, B.S.; Johnson, J.R.; Cohen, M.H.; Justice, R.; Pazdur, R. FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007, 12, 1247–1252. [Google Scholar] [CrossRef]
  27. Krug, L.M.; Kindler, H.L.; Calvert, H.; Manegold, C.; Tsao, A.S.; Fennell, D.; Ohman, R.; Plummer, R.; Eberhardt, W.E.; Fukuoka, K.; et al. Vorinostat in patients with advanced malignant pleural mesothelioma who have progressed on previous chemotherapy (VANTAGE-014): A phase 3, double-blind, randomised, placebo-controlled trial. Lancet Oncol. 2015, 16, 447–456. [Google Scholar] [CrossRef]
  28. Nakajima, H.; Kim, Y.B.; Terano, H.; Yoshida, M.; Horinouchi, S. FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor. Exp. Cell Res. 1998, 241, 126–133. [Google Scholar] [CrossRef] [PubMed]
  29. Ueda, H.; Nakajima, H.; Hori, Y.; Goto, T.; Okuhara, M. Action of FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum no. 968, on Ha-ras transformed NIH3T3 cells. Biosci. Biotechnol. Biochem. 1994, 58, 1579–1583. [Google Scholar] [CrossRef]
  30. Piekarz, R.L.; Frye, R.; Turner, M.; Wright, J.J.; Allen, S.L.; Kirschbaum, M.H.; Zain, J.; Prince, H.M.; Leonard, J.P.; Geskin, L.J.; et al. Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. J. Clin. Oncol. 2009, 27, 5410–5417. [Google Scholar] [CrossRef]
  31. Coiffier, B.; Pro, B.; Prince, H.M.; Foss, F.; Sokol, L.; Greenwood, M.; Caballero, D.; Borchmann, P.; Morschhauser, F.; Wilhelm, M.; et al. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J. Clin. Oncol. 2012, 30, 631–636. [Google Scholar] [CrossRef] [PubMed]
  32. O’Connor, O.A.; Horwitz, S.; Masszi, T.; Van Hoof, A.; Brown, P.; Doorduijn, J.; Hess, G.; Jurczak, W.; Knoblauch, P.; Chawla, S.; et al. Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma: Results of the Pivotal Phase II BELIEF (CLN-19) Study. J. Clin. Oncol. 2015, 33, 2492–2499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Foss, F.; Advani, R.; Duvic, M.; Hymes, K.B.; Intragumtornchai, T.; Lekhakula, A.; Shpilberg, O.; Lerner, A.; Belt, R.J.; Jacobsen, E.D.; et al. A Phase II trial of Belinostat (PXD101) in patients with relapsed or refractory peripheral or cutaneous T-cell lymphoma. Br. J. Haematol. 2015, 168, 811–819. [Google Scholar] [CrossRef] [PubMed]
  34. Ramalingam, S.S.; Belani, C.P.; Ruel, C.; Frankel, P.; Gitlitz, B.; Koczywas, M.; Espinoza-Delgado, I.; Gandara, D. Phase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesothelioma. J. Thorac. Oncol. 2009, 4, 97–101. [Google Scholar] [CrossRef] [PubMed]
  35. San-Miguel, J.F.; Hungria, V.T.; Yoon, S.S.; Beksac, M.; Dimopoulos, M.A.; Elghandour, A.; Jedrzejczak, W.W.; Gunther, A.; Nakorn, T.N.; Siritanaratkul, N.; et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: A multicentre, randomised, double-blind phase 3 trial. Lancet Oncol. 2014, 15, 1195–1206. [Google Scholar] [CrossRef]
  36. 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] [PubMed]
  37. Eckschlager, T.; Plch, J.; Stiborova, M.; Hrabeta, J. Histone Deacetylase Inhibitors as Anticancer Drugs. Int. J. Mol. Sci. 2017, 18, 1414. [Google Scholar] [CrossRef] [PubMed]
  38. Mottamal, M.; Zheng, S.; Huang, T.L.; Wang, G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 2015, 20, 3898–3941. [Google Scholar] [CrossRef] [PubMed]
  39. Archer, S.Y.; Meng, S.; Shei, A.; Hodin, R.A. p21(WAF1) is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc. Natl. Acad. Sci. USA 1998, 95, 6791–6796. [Google Scholar] [CrossRef] [PubMed]
  40. El-Deiry, W.S.; Tokino, T.; Velculescu, V.E.; Levy, D.B.; Parsons, R.; Trent, J.M.; Lin, D.; Mercer, W.E.; Kinzler, K.W.; Vogelstein, B. WAF1, a potential mediator of p53 tumor suppression. Cell 1993, 75, 817–825. [Google Scholar] [CrossRef]
  41. Saito, A.; Yamashita, T.; Mariko, Y.; Nosaka, Y.; Tsuchiya, K.; Ando, T.; Suzuki, T.; Tsuruo, T.; Nakanishi, O. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc. Natl. Acad. Sci. USA 1999, 96, 4592–4597. [Google Scholar] [CrossRef] [PubMed]
  42. Gui, C.Y.; Ngo, L.; Xu, W.S.; Richon, V.M.; Marks, P.A. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc. Natl. Acad. Sci. USA 2004, 101, 1241–1246. [Google Scholar] [CrossRef] [PubMed]
  43. Sowa, Y.; Orita, T.; Minamikawa-Hiranabe, S.; Mizuno, T.; Nomura, H.; Sakai, T. Sp3, but not Sp1, mediates the transcriptional activation of the p21/WAF1/Cip1 gene promoter by histone deacetylase inhibitor. Cancer Res. 1999, 59, 4266–4270. [Google Scholar] [PubMed]
  44. Condorelli, F.; Gnemmi, I.; Vallario, A.; Genazzani, A.A.; Canonico, P.L. Inhibitors of histone deacetylase (HDAC) restore the p53 pathway in neuroblastoma cells. Br. J. Pharmacol. 2008, 153, 657–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhao, Y.; Lu, S.; Wu, L.; Chai, G.; Wang, H.; Chen, Y.; Sun, J.; Yu, Y.; Zhou, W.; Zheng, Q.; et al. Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21(Waf1/Cip1). Mol. Cell. Biol. 2006, 26, 2782–2790. [Google Scholar] [CrossRef] [PubMed]
  46. Strait, K.A.; Dabbas, B.; Hammond, E.H.; Warnick, C.T.; Iistrup, S.J.; Ford, C.D. Cell cycle blockade and differentiation of ovarian cancer cells by the histone deacetylase inhibitor trichostatin A are associated with changes in p21, Rb, and Id proteins. Mol. Cancer Ther. 2002, 1, 1181–1190. [Google Scholar] [PubMed]
  47. Greenberg, V.L.; Williams, J.M.; Cogswell, J.P.; Mendenhall, M.; Zimmer, S.G. Histone deacetylase inhibitors promote apoptosis and differential cell cycle arrest in anaplastic thyroid cancer cells. Thyroid 2001, 11, 315–325. [Google Scholar] [CrossRef] [PubMed]
  48. Florenes, V.A.; Skrede, M.; Jorgensen, K.; Nesland, J.M. Deacetylase inhibition in malignant melanomas: Impact on cell cycle regulation and survival. Melanoma Res. 2004, 14, 173–181. [Google Scholar] [CrossRef]
  49. Fandy, T.E.; Shankar, S.; Ross, D.D.; Sausville, E.; Srivastava, R.K. Interactive effects of HDAC inhibitors and TRAIL on apoptosis are associated with changes in mitochondrial functions and expressions of cell cycle regulatory genes in multiple myeloma. Neoplasia 2005, 7, 646–657. [Google Scholar] [CrossRef]
  50. Guo, F.; Sigua, C.; Tao, J.; Bali, P.; George, P.; Li, Y.; Wittmann, S.; Moscinski, L.; Atadja, P.; Bhalla, K. Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer Res. 2004, 64, 2580–2589. [Google Scholar] [CrossRef]
  51. Singh, T.R.; Shankar, S.; Srivastava, R.K. HDAC inhibitors enhance the apoptosis-inducing potential of TRAIL in breast carcinoma. Oncogene 2005, 24, 4609–4623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Shankar, S.; Singh, T.R.; Fandy, T.E.; Luetrakul, T.; Ross, D.D.; Srivastava, R.K. Interactive effects of histone deacetylase inhibitors and TRAIL on apoptosis in human leukemia cells: Involvement of both death receptor and mitochondrial pathways. Int. J. Mol. Med. 2005, 16, 1125–1138. [Google Scholar] [CrossRef] [PubMed]
  53. Iacomino, G.; Medici, M.C.; Russo, G.L. Valproic acid sensitizes K562 erythroleukemia cells to TRAIL/Apo2L-induced apoptosis. Anticancer Res. 2008, 28, 855–864. [Google Scholar] [PubMed]
  54. Inoue, S.; Harper, N.; Walewska, R.; Dyer, M.J.; Cohen, G.M. Enhanced Fas-associated death domain recruitment by histone deacetylase inhibitors is critical for the sensitization of chronic lymphocytic leukemia cells to TRAIL-induced apoptosis. Mol. Cancer Ther. 2009, 8, 3088–3097. [Google Scholar] [CrossRef] [PubMed]
  55. Matthews, G.M.; Newbold, A.; Johnstone, R.W. Intrinsic and extrinsic apoptotic pathway signaling as determinants of histone deacetylase inhibitor antitumor activity. Adv. Cancer Res. 2012, 116, 165–197. [Google Scholar] [PubMed]
  56. Mie Lee, Y.; Kim, S.H.; Kim, H.S.; Jin Son, M.; Nakajima, H.; Jeong Kwon, H.; Kim, K.W. Inhibition of hypoxia-induced angiogenesis by FK228, a specific histone deacetylase inhibitor, via suppression of HIF-1alpha activity. Biochem. Biophys. Res. Commun. 2003, 300, 241–246. [Google Scholar] [CrossRef]
  57. Sawa, H.; Murakami, H.; Ohshima, Y.; Murakami, M.; Yamazaki, I.; Tamura, Y.; Mima, T.; Satone, A.; Ide, W.; Hashimoto, I.; et al. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A inhibit vascular endothelial growth factor (VEGF) secretion from human glioblastoma cells. Brain Tumor Pathol. 2002, 19, 77–81. [Google Scholar] [CrossRef] [PubMed]
  58. Sasakawa, Y.; Naoe, Y.; Noto, T.; Inoue, T.; Sasakawa, T.; Matsuo, M.; Manda, T.; Mutoh, S. Antitumor efficacy of FK228, a novel histone deacetylase inhibitor, depends on the effect on expression of angiogenesis factors. Biochem. Pharmacol. 2003, 66, 897–906. [Google Scholar] [CrossRef]
  59. Zgouras, D.; Becker, U.; Loitsch, S.; Stein, J. Modulation of angiogenesis-related protein synthesis by valproic acid. Biochem. Biophys. Res. Commun. 2004, 316, 693–697. [Google Scholar] [CrossRef]
  60. Heider, U.; Kaiser, M.; Sterz, J.; Zavrski, I.; Jakob, C.; Fleissner, C.; Eucker, J.; Possinger, K.; Sezer, O. Histone deacetylase inhibitors reduce VEGF production and induce growth suppression and apoptosis in human mantle cell lymphoma. European J. Haematol. 2006, 76, 42–50. [Google Scholar] [CrossRef]
  61. Kim, M.S.; Kwon, H.J.; Lee, Y.M.; Baek, J.H.; Jang, J.E.; Lee, S.W.; Moon, E.J.; Kim, H.S.; Lee, S.K.; Chung, H.Y.; et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat. Med. 2001, 7, 437–443. [Google Scholar] [CrossRef] [PubMed]
  62. Qian, D.Z.; Kachhap, S.K.; Collis, S.J.; Verheul, H.M.; Carducci, M.A.; Atadja, P.; Pili, R. Class II histone deacetylases are associated with VHL-independent regulation of hypoxia-inducible factor 1 alpha. Cancer Res. 2006, 66, 8814–8821. [Google Scholar] [CrossRef] [PubMed]
  63. Cheng, H.T.; Hung, W.C. Inhibition of proliferation, sprouting, tube formation and Tie2 signaling of lymphatic endothelial cells by the histone deacetylase inhibitor SAHA. Oncol. Rep. 2013, 30, 961–967. [Google Scholar] [CrossRef] [PubMed]
  64. Hellebrekers, D.M.; Castermans, K.; Vire, E.; Dings, R.P.; Hoebers, N.T.; Mayo, K.H.; Oude Egbrink, M.G.; Molema, G.; Fuks, F.; van Engeland, M.; et al. Epigenetic regulation of tumor endothelial cell anergy: Silencing of intercellular adhesion molecule-1 by histone modifications. Cancer Res. 2006, 66, 10770–10777. [Google Scholar] [CrossRef] [PubMed]
  65. Hellebrekers, D.M.; Melotte, V.; Vire, E.; Langenkamp, E.; Molema, G.; Fuks, F.; Herman, J.G.; Van Criekinge, W.; Griffioen, A.W.; van Engeland, M. Identification of epigenetically silenced genes in tumor endothelial cells. Cancer Res. 2007, 67, 4138–4148. [Google Scholar] [CrossRef]
  66. Kwon, H.J.; Kim, M.S.; Kim, M.J.; Nakajima, H.; Kim, K.W. Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int. J. Cancer 2002, 97, 290–296. [Google Scholar] [CrossRef]
  67. Srivastava, R.K.; Kurzrock, R.; Shankar, S. MS-275 sensitizes TRAIL-resistant breast cancer cells, inhibits angiogenesis and metastasis, and reverses epithelial-mesenchymal transition in vivo. Mol. Cancer Ther. 2010, 9, 3254–3266. [Google Scholar] [CrossRef]
  68. Camphausen, K.; Burgan, W.; Cerra, M.; Oswald, K.A.; Trepel, J.B.; Lee, M.J.; Tofilon, P.J. Enhanced radiation-induced cell killing and prolongation of gammaH2AX foci expression by the histone deacetylase inhibitor MS-275. Cancer Res. 2004, 64, 316–321. [Google Scholar] [CrossRef]
  69. Camphausen, K.; Cerna, D.; Scott, T.; Sproull, M.; Burgan, W.E.; Cerra, M.A.; Fine, H.; Tofilon, P.J. Enhancement of in vitro and in vivo tumor cell radiosensitivity by valproic acid. Int. J. Cancer 2005, 114, 380–386. [Google Scholar] [CrossRef]
  70. Munshi, A.; Kurland, J.F.; Nishikawa, T.; Tanaka, T.; Hobbs, M.L.; Tucker, S.L.; Ismail, S.; Stevens, C.; Meyn, R.E. Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity. Clin. Cancer Res. 2005, 11, 4912–4922. [Google Scholar] [CrossRef]
  71. Munshi, A.; Tanaka, T.; Hobbs, M.L.; Tucker, S.L.; Richon, V.M.; Meyn, R.E. Vorinostat, a histone deacetylase inhibitor, enhances the response of human tumor cells to ionizing radiation through prolongation of gamma-H2AX foci. Mol. Cancer Ther. 2006, 5, 1967–1974. [Google Scholar] [CrossRef] [PubMed]
  72. Perona, M.; Thomasz, L.; Rossich, L.; Rodriguez, C.; Pisarev, M.A.; Rosemblit, C.; Cremaschi, G.A.; Dagrosa, M.A.; Juvenal, G.J. Radiosensitivity enhancement of human thyroid carcinoma cells by the inhibitors of histone deacetylase sodium butyrate and valproic acid. Mol. Cell. Endocrinol. 2018, 478, 141–150. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, Y.; Carr, T.; Dimtchev, A.; Zaer, N.; Dritschilo, A.; Jung, M. Attenuated DNA damage repair by trichostatin A through BRCA1 suppression. Radiat. Res. 2007, 168, 115–124. [Google Scholar] [CrossRef] [PubMed]
  74. Adimoolam, S.; Sirisawad, M.; Chen, J.; Thiemann, P.; Ford, J.M.; Buggy, J.J. HDAC inhibitor PCI-24781 decreases RAD51 expression and inhibits homologous recombination. Proc. Natl. Acad. Sci. USA 2007, 104, 19482–19487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kachhap, S.K.; Rosmus, N.; Collis, S.J.; Kortenhorst, M.S.; Wissing, M.D.; Hedayati, M.; Shabbeer, S.; Mendonca, J.; Deangelis, J.; Marchionni, L.; et al. Downregulation of homologous recombination DNA repair genes by HDAC inhibition in prostate cancer is mediated through the E2F1 transcription factor. PLoS ONE 2010, 5, e11208. [Google Scholar] [CrossRef] [PubMed]
  76. Koprinarova, M.; Botev, P.; Russev, G. Histone deacetylase inhibitor sodium butyrate enhances cellular radiosensitivity by inhibiting both DNA nonhomologous end joining and homologous recombination. DNA Repair 2011, 10, 970–977. [Google Scholar] [CrossRef] [PubMed]
  77. Rosato, R.R.; Almenara, J.A.; Grant, S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res. 2003, 63, 3637–3645. [Google Scholar] [PubMed]
  78. Ruefli, A.A.; Bernhard, D.; Tainton, K.M.; Kofler, R.; Smyth, M.J.; Johnstone, R.W. Suberoylanilide hydroxamic acid (SAHA) overcomes multidrug resistance and induces cell death in P-glycoprotein-expressing cells. Int. J. Cancer 2002, 99, 292–298. [Google Scholar] [CrossRef] [PubMed]
  79. Butler, L.M.; Zhou, X.; Xu, W.S.; Scher, H.I.; Rifkind, R.A.; Marks, P.A.; Richon, V.M. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl. Acad. Sci. USA 2002, 99, 11700–11705. [Google Scholar] [CrossRef] [Green Version]
  80. Lee, J.H.; Jeong, E.G.; Choi, M.C.; Kim, S.H.; Park, J.H.; Song, S.H.; Park, J.; Bang, Y.J.; Kim, T.Y. Inhibition of histone deacetylase 10 induces thioredoxin-interacting protein and causes accumulation of reactive oxygen species in SNU-620 human gastric cancer cells. Mol. Cells 2010, 30, 107–112. [Google Scholar] [CrossRef]
  81. Ungerstedt, J.; Du, Y.; Zhang, H.; Nair, D.; Holmgren, A. In vivo redox state of human thioredoxin and redox shift by the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA). Free Radic. Biol. Med. 2012, 53, 2002–2007. [Google Scholar] [CrossRef] [PubMed]
  82. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed]
  83. Esquela-Kerscher, A.; Slack, F.J. Oncomirs—micrornas with a role in cancer. Nat. Rev. Cancer 2006, 6, 259–269. [Google Scholar] [CrossRef] [PubMed]
  84. Roush, S.; Slack, F.J. The let-7 family of microRNAs. Trends Cell Biol. 2008, 18, 505–516. [Google Scholar] [CrossRef] [PubMed]
  85. Lin, K.-Y.; Ye, H.; Han, B.-W.; Wang, W.-T.; Wei, P.-P.; He, B.; Li, X.-J.; Chen, Y.-Q. Genome-wide screen identified let-7c/miR-99a/miR-125b regulating tumor progression and stem-like properties in cholangiocarcinoma. Oncogene 2016, 35, 3376–3386. [Google Scholar] [CrossRef] [PubMed]
  86. Huang, M.; Gong, X. Let-7c Inhibits the Proliferation, Invasion, and Migration of Glioma Cells via Targeting E2F5. Oncol. Res. 2018, 26, 1103–1111. [Google Scholar] [CrossRef] [PubMed]
  87. Zhao, B.; Han, H.; Chen, J.; Zhang, Z.; Li, S.; Fang, F.; Zheng, Q.; Ma, Y.; Zhang, J.; Wu, N.; et al. MicroRNA let-7c inhibits migration and invasion of human non-small cell lung cancer by targeting ITGB3 and MAP4K3. Cancer Lett. 2014, 342, 43–51. [Google Scholar] [CrossRef] [PubMed]
  88. Leite, K.R.M.; Sousa-Canavez, J.M.; Reis, S.T.; Tomiyama, A.H.; Camara-Lopes, L.H.; Sañudo, A.; Antunes, A.A.; Srougi, M. Change in expression of miR-let7c, miR-100, and miR-218 from high grade localized prostate cancer to metastasis. Urol. Oncol. Semin. Orig. Investig. 2011, 29, 265–269. [Google Scholar] [CrossRef] [PubMed]
  89. Komatsu, S.; Ichikawa, D.; Takeshita, H.; Morimura, R.; Hirajima, S.; Tsujiura, M.; Kawaguchi, T.; Miyamae, M.; Nagata, H.; Konishi, H.; et al. Circulating miR-18a: A Sensitive Cancer Screening Biomarker in Human Cancer. In Vivo 2014, 28, 293–297. [Google Scholar]
  90. Sun, Z.; Shi, K.; Yang, S.; Liu, J.; Zhou, Q.; Wang, G.; Song, J.; Li, Z.; Zhang, Z.; Yuan, W. Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer 2018, 17, 147. [Google Scholar] [CrossRef]
  91. Tang, H.; Ma, M.; Dai, J.; Cui, C.; Si, L.; Sheng, X.; Chi, Z.; Xu, L.; Yu, S.; Xu, T.; et al. miR-let-7b and miR-let-7c suppress tumourigenesis of human mucosal melanoma and enhance the sensitivity to chemotherapy. J. Exp. Clin. Cancer Res. 2019, 38, 212. [Google Scholar] [CrossRef] [PubMed]
  92. Johnson, S.M.; Grosshans, H.; Shingara, J.; Byrom, M.; Jarvis, R.; Cheng, A.; Labourier, E.; Reinert, K.L.; Brown, D.; Slack, F.J. RAS Is Regulated by the let-7 MicroRNA Family. Cell 2005, 120, 635–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Xie, Y.; Zhang, H.; Guo, X.-J.; Feng, Y.-C.; He, R.-Z.; Li, X.; Yu, S.; Zhao, Y.; Shen, M.; Zhu, F.; et al. Let-7c inhibits cholangiocarcinoma growth but promotes tumor cell invasion and growth at extrahepatic sites. Cell Death Dis. 2018, 9, 249. [Google Scholar] [CrossRef] [PubMed]
  94. Cui, S.-Y.; Huang, J.-Y.; Chen, Y.-T.; Song, H.-Z.; Feng, B.; Huang, G.-C.; Wang, R.; Chen, L.-B.; De, W. Let-7c Governs the Acquisition of Chemo- or Radioresistance and Epithelial-to-Mesenchymal Transition Phenotypes in Docetaxel-Resistant Lung Adenocarcinoma. Mol. Cancer Res. 2013, 11, 699–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Zhang, W.; Zeng, Q.; Ban, Z.; Cao, J.; Chu, T.; Lei, D.; Liu, C.; Guo, W.; Zeng, X. Effects of let-7c on the proliferation of ovarian carcinoma cells by targeted regulation of CDC25a gene expression. Oncol. Lett. 2018, 16, 5543–5550. [Google Scholar] [CrossRef] [PubMed]
  96. Zhu, X.; Wu, L.; Yao, J.; Jiang, H.; Wang, Q.; Yang, Z.; Wu, F. MicroRNA let-7c Inhibits Cell Proliferation and Induces Cell Cycle Arrest by Targeting CDC25A in Human Hepatocellular Carcinoma. PLoS ONE 2015, 10, e0124266. [Google Scholar] [CrossRef] [PubMed]
  97. Han, H.-B.; Gu, J.; Zuo, H.-J.; Chen, Z.-G.; Zhao, W.; Li, M.; Ji, D.-B.; Lu, Y.-Y.; Zhang, Z.-Q. Let-7c functions as a metastasis suppressor by targeting MMP11 and PBX3 in colorectal cancer. J. Pathol. 2012, 226, 544–555. [Google Scholar] [CrossRef] [PubMed]
  98. Mortazavi, D.; Sharifi, M. Antiproliferative effect of upregulation of hsa-let-7c-5p in human acute erythroleukemia cells. Cytotechnology 2018, 70, 1509–1518. [Google Scholar] [CrossRef]
  99. Fu, X.; Fu, X.; Mao, X.; Mao, X.; Wang, Y.; Wang, Y.; Ding, X.; Ding, X.; Li, Y.; Li, Y. Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer. Oncol. Rep. 2017, 38, 1851–1856. [Google Scholar] [CrossRef] [Green Version]
  100. Diederichs, S.; Haber, D.A. Sequence Variations of MicroRNAs in Human Cancer: Alterations in Predicted Secondary Structure Do Not Affect Processing. Cancer Res 2006, 66, 6097–6104. [Google Scholar] [CrossRef] [Green Version]
  101. Scott, G.K.; Mattie, M.D.; Berger, C.E.; Benz, S.C.; Benz, C.C. Rapid alteration of microRNA levels by histone deacetylase inhibition. Cancer Res. 2006, 66, 1277–1281. [Google Scholar] [CrossRef] [PubMed]
  102. Shin, S.; Lee, E.-M.; Cha, H.J.; Bae, S.; Jung, J.H.; Lee, S.-M.; Yoon, Y.; Lee, H.; Kim, S.; Kim, H.; et al. MicroRNAs that respond to histone deacetylase inhibitor SAHA and p53 in HCT116 human colon carcinoma cells. Int. J. Oncol. 2009, 35, 1343–1352. [Google Scholar] [PubMed] [Green Version]
  103. Di Fazio, P.; Montalbano, R.; Neureiter, D.; Alinger, B.; Schmidt, A.; Merkel, A.L.; Quint, K.; Ocker, M. Downregulation of HMGA2 by the pan-deacetylase inhibitor panobinostat is dependent on hsa-let-7b expression in liver cancer cell lines. Exp. Cell Res. 2012, 318, 1832–1843. [Google Scholar] [CrossRef]
  104. Balch, C.; Naegeli, K.; Nam, S.; Ballard, B.; Hyslop, A.; Melki, C.; Reilly, E.; Hur, M.-W.; Nephew, K.P. A unique histone deacetylase inhibitor alters microRNA expression and signal transduction in chemoresistant ovarian cancer cells. Cancer Biol. Ther. 2012, 13, 681–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Pili, R.; Liu, G.; Chintala, S.; Verheul, H.; Rehman, S.; Attwood, K.; Lodge, M.A.; Wahl, R.; Martin, J.I.; Miles, K.M.; et al. Combination of the histone deacetylase inhibitor vorinostat with bevacizumab in patients with clear-cell renal cell carcinoma: A multicentre, single-arm phase I/II clinical trial. Br. J. Cancer 2017, 116, 874–883. [Google Scholar] [CrossRef]
  106. Lee, E.M.; Shin, S.; Cha, H.J.; Yoon, Y.; Bae, S.; Jung, J.H.; Lee, S.M.; Lee, S.J.; Park, I.C.; Jin, Y.W.; et al. Suberoylanilide hydroxamic acid (SAHA) changes microRNA expression profiles in A549 human non-small cell lung cancer cells. Int. J. Mol. Med. 2009, 24, 45–50. [Google Scholar] [PubMed] [Green Version]
  107. Borbone, E.; De Rosa, M.; Siciliano, D.; Altucci, L.; Croce, C.M.; Fusco, A. Up-regulation of miR-146b and down-regulation of miR-200b contribute to the cytotoxic effect of histone deacetylase inhibitors on ras-transformed thyroid cells. J. Clin. Endocrinol. Metab. 2013, 98, E1031–E1040. [Google Scholar] [CrossRef] [PubMed]
  108. Humphries, B.; Yang, C. The microRNA-200 family: Small molecules with novel roles in cancer development, progression and therapy. Oncotarget 2015, 6, 6472–6498. [Google Scholar] [CrossRef]
  109. Eades, G.; Yang, M.; Yao, Y.; Zhang, Y.; Zhou, Q. miR-200a regulates Nrf2 activation by targeting Keap1 mRNA in breast cancer cells. J. Biol. Chem. 2011, 286, 40725–40733. [Google Scholar] [CrossRef]
  110. Bian, X.; Liang, Z.; Feng, A.; Salgado, E.; Shim, H. HDAC inhibitor suppresses proliferation and invasion of breast cancer cells through regulation of miR-200c targeting CRKL. Biochem. Pharmacol. 2018, 147, 30–37. [Google Scholar] [CrossRef]
  111. Cho, J.-H.; Dimri, M.; Dimri, G.P. MicroRNA-31 Is a Transcriptional Target of Histone Deacetylase Inhibitors and a Regulator of Cellular Senescence. J. Biol. Chem. 2015, 290, 10555–10567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Nalls, D.; Tang, S.-N.; Rodova, M.; Srivastava, R.K.; Shankar, S. Targeting epigenetic regulation of miR-34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS ONE 2011, 6, e24099. [Google Scholar] [CrossRef] [PubMed]
  113. Sampath, D.; Liu, C.; Vasan, K.; Sulda, M.; Puduvalli, V.K.; Wierda, W.G.; Keating, M.J. Histone deacetylases mediate the silencing of miR-15a, miR-16, and miR-29b in chronic lymphocytic leukemia. Blood 2012, 119, 1162–1172. [Google Scholar] [CrossRef] [PubMed]
  114. Hibino, S.; Saito, Y.; Muramatsu, T.; Otani, A.; Kasai, Y.; Kimura, M.; Saito, H. Inhibitors of enhancer of zeste homolog 2 (EZH2) activate tumor-suppressor microRNAs in human cancer cells. Oncogenesis 2014, 3, e104. [Google Scholar] [CrossRef] [PubMed]
  115. Kretzner, L.; Scuto, A.; Dino, P.M.; Kowolik, C.M.; Wu, J.; Ventura, P.; Jove, R.; Forman, S.J.; Yen, Y.; Kirschbaum, M.H. Combining histone deacetylase inhibitor vorinostat with aurora kinase inhibitors enhances lymphoma cell killing with repression of c-Myc, hTERT, and microRNA levels. Cancer Res. 2011, 71, 3912–3920. [Google Scholar] [CrossRef] [PubMed]
  116. Lepore, I.; Dell’Aversana, C.; Pilyugin, M.; Conte, M.; Nebbioso, A.; De Bellis, F.; Tambaro, F.P.; Izzo, T.; Garcia-Manero, G.; Ferrara, F.; et al. HDAC inhibitors repress BARD1 isoform expression in acute myeloid leukemia cells via activation of miR-19a and/or b. PLoS ONE 2013, 8, e83018. [Google Scholar] [CrossRef] [PubMed]
  117. Talbert, D.R.; Wappel, R.L.; Moran, D.M.; Shell, S.A.; Bacus, S.S. The Role of Myc and the miR-17~92 Cluster in Histone Deacetylase Inhibitor Induced Apoptosis of Solid Tumors. J. Cancer Ther. 2013, 4, 907–918. [Google Scholar] [CrossRef] [Green Version]
  118. Yang, H.; Lan, P.; Hou, Z.; Guan, Y.; Zhang, J.; Xu, W.; Tian, Z.; Zhang, C. Histone deacetylase inhibitor SAHA epigenetically regulates miR-17-92 cluster and MCM7 to upregulate MICA expression in hepatoma. Br. J. Cancer 2015, 112, 112–121. [Google Scholar] [CrossRef]
  119. Humphreys, K.J.; Cobiac, L.; Le Leu, R.K.; Van der Hoek, M.B.; Michael, M.Z. Histone deacetylase inhibition in colorectal cancer cells reveals competing roles for members of the oncogenic miR-17-92 cluster. Mol. Carcinog. 2013, 52, 459–474. [Google Scholar] [CrossRef]
  120. Schiffgen, M.; Schmidt, D.H.; von Rücker, A.; Müller, S.C.; Ellinger, J. Epigenetic regulation of microRNA expression in renal cell carcinoma. Biochem. Biophys. Res. Commun. 2013, 436, 79–84. [Google Scholar] [CrossRef]
  121. Bamodu, O.A.; Kuo, K.-T.; Yuan, L.-P.; Cheng, W.-H.; Lee, W.-H.; Ho, Y.-S.; Chao, T.-Y.; Yeh, C.-T. HDAC inhibitor suppresses proliferation and tumorigenicity of drug-resistant chronic myeloid leukemia stem cells through regulation of hsa-miR-196a targeting BCR/ABL1. Exp. Cell Res. 2018, 370, 519–530. [Google Scholar] [CrossRef] [PubMed]
  122. Lai, T.-H.; Ewald, B.; Zecevic, A.; Liu, C.; Sulda, M.; Papaioannou, D.; Garzon, R.; Blachly, J.S.; Plunkett, W.; Sampath, D. HDAC Inhibition Induces MicroRNA-182, which Targets RAD51 and Impairs HR Repair to Sensitize Cells to Sapacitabine in Acute Myelogenous Leukemia. Clin. Cancer Res. 2016, 22, 3537–3549. [Google Scholar] [CrossRef] [PubMed]
  123. Seol, H.S.; Akiyama, Y.; Shimada, S.; Lee, H.J.; Kim, T.I.; Chun, S.M.; Singh, S.R.; Jang, S.J. Epigenetic silencing of microRNA-373 to epithelial-mesenchymal transition in non-small cell lung cancer through IRAK2 and LAMP1 axes. Cancer Lett. 2014, 353, 232–241. [Google Scholar] [CrossRef] [PubMed]
  124. Suraweera, A.; O’Byrne, K.J.; Richard, D.J. Combination Therapy with Histone Deacetylase Inhibitors (HDACi) for the Treatment of Cancer: Achieving the Full Therapeutic Potential of HDACi. Front. Oncol. 2018, 8, 92. [Google Scholar] [CrossRef]
  125. Chen, C.; Chen, C.; Chen, J.; Zhou, L.; Xu, H.; Jin, W.; Wu, J.; Gao, S. Histone deacetylases inhibitor trichostatin A increases the expression of Dleu2/miR-15a/16-1 via HDAC3 in non-small cell lung cancer. Mol. Cell. Biochem. 2013, 383, 137–148. [Google Scholar] [CrossRef]
  126. Adams, C.M.; Hiebert, S.W.; Eischen, C.M. Myc Induces miRNA-Mediated Apoptosis in Response to HDAC Inhibition in Hematologic Malignancies. Cancer Res. 2016, 76, 736–748. [Google Scholar] [CrossRef]
  127. Hu, S.; Dong, T.S.; Dalal, S.R.; Wu, F.; Bissonnette, M.; Kwon, J.H.; Chang, E.B. The Microbe-Derived Short Chain Fatty Acid Butyrate Targets miRNA-Dependent p21 Gene Expression in Human Colon Cancer. PLoS ONE 2011, 6, e16221. [Google Scholar] [CrossRef]
  128. Murray-Stewart, T.; Hanigan, C.L.; Woster, P.M.; Marton, L.J.; Casero, R.A. Histone Deacetylase Inhibition Overcomes Drug Resistance through a miRNA-Dependent Mechanism. Mol. Cancer Ther. 2013, 12, 2088–2099. [Google Scholar] [CrossRef] [Green Version]
  129. Chen, Y.-J.; Wang, W.-H.; Wu, W.-Y.; Hsu, C.-C.; Wei, L.-R.; Wang, S.-F.; Hsu, Y.-W.; Liaw, C.-C.; Tsai, W.-C. Novel histone deacetylase inhibitor AR-42 exhibits antitumor activity in pancreatic cancer cells by affecting multiple biochemical pathways. PLoS ONE 2017, 12, e0183368. [Google Scholar] [CrossRef]
  130. Zhang, Q.; Sun, M.; Zhou, S.; Guo, B. Class I HDAC inhibitor mocetinostat induces apoptosis by activation of miR-31 expression and suppression of E2F6. Cell Death Discov. 2016, 2, 16036. [Google Scholar] [CrossRef] [Green Version]
  131. Sato, S.; Katsushima, K.; Shinjo, K.; Hatanaka, A.; Ohka, F.; Suzuki, S.; Naiki-Ito, A.; Soga, N.; Takahashi, S.; Kondo, Y. Histone Deacetylase Inhibition in Prostate Cancer Triggers miR-320–Mediated Suppression of the Androgen Receptor. Cancer Res. 2016, 76, 4192–4204. [Google Scholar] [CrossRef] [PubMed]
  132. Collins-Burow, B. The histone deacetylase inhibitor trichostatin A alters microRNA expression profiles in apoptosis-resistant breast cancer cells. Oncol. Rep. 2011, 27, 10–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Hsieh, T.-H.; Hsu, C.-Y.; Tsai, C.-F.; Long, C.-Y.; Wu, C.-H.; Wu, D.-C.; Lee, J.-N.; Chang, W.-C.; Tsai, E.-M. HDAC Inhibitors Target HDAC5, Upregulate MicroRNA-125a-5p, and Induce Apoptosis in Breast Cancer Cells. Mol. Ther. 2015, 23, 656–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Jung, D.E.; Park, S.B.; Kim, K.; Kim, C.; Song, S.Y. CG200745, an HDAC inhibitor, induces anti-tumour effects in cholangiocarcinoma cell lines via miRNAs targeting the Hippo pathway. Sci. Rep. 2017, 7, 10921. [Google Scholar] [CrossRef] [PubMed]
  135. Bandres, E.; Agirre, X.; Bitarte, N.; Ramirez, N.; Zarate, R.; Roman-Gomez, J.; Prosper, F.; Garcia-Foncillas, J. Epigenetic regulation of microRNA expression in colorectal cancer. Int. J. Cancer 2009, 125, 2737–2743. [Google Scholar] [CrossRef] [PubMed]
  136. Ribas, J.; Ni, X.; Castanares, M.; Liu, M.M.; Esopi, D.; Yegnasubramanian, S.; Rodriguez, R.; Mendell, J.T.; Lupold, S.E. A novel source for miR-21 expression through the alternative polyadenylation of VMP1 gene transcripts. Nucl. Acids Res. 2012, 40, 6821–6833. [Google Scholar] [CrossRef] [Green Version]
  137. Tsukamoto, Y.; Nakada, C.; Noguchi, T.; Tanigawa, M.; Nguyen, L.T.; Uchida, T.; Hijiya, N.; Matsuura, K.; Fujioka, T.; Seto, M.; et al. MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res. 2010, 70, 2339–2349. [Google Scholar] [CrossRef] [PubMed]
  138. 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]
  139. Xie, H.; Zhang, Q.; Zhou, H.; Zhou, J.; Zhang, J.; Jiang, Y.; Wang, J.; Meng, X.; Zeng, L.; Jiang, X. microRNA-889 is downregulated by histone deacetylase inhibitors and confers resistance to natural killer cytotoxicity in hepatocellular carcinoma cells. Cytotechnology 2018, 70, 513–521. [Google Scholar] [CrossRef]
  140. Trécul, A.; Morceau, F.; Gaigneaux, A.; Schnekenburger, M.; Dicato, M.; Diederich, M. Valproic acid regulates erythro-megakaryocytic differentiation through the modulation of transcription factors and microRNA regulatory micro-networks. Biochem. Pharmacol. 2014, 92, 299–311. [Google Scholar] [CrossRef]
  141. Mazar, J.; DeBlasio, D.; Govindarajan, S.S.; Zhang, S.; Perera, R.J. Epigenetic regulation of microRNA-375 and its role in melanoma development in humans. FEBS Lett. 2011, 585, 2467–2476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Canella, A.; Nieves, H.C.; Sborov, D.W.; Cascione, L.; Radomska, H.S.; Smith, E.; Stiff, A.; Consiglio, J.; Caserta, E.; Rizzotto, L.; et al. HDAC inhibitor AR-42 decreases CD44 expression and sensitizes myeloma cells to lenalidomide. Oncotarget 2015, 6, 31134–31150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jung, H.M.; Benarroch, Y.; Chan, E.K.L. Anti-Cancer Drugs Reactivate Tumor Suppressor miR-375 Expression in Tongue Cancer Cells: miR-375 REACTIVATION BY ANTI-CANCER DRUGS. J. Cell. Biochem. 2015, 116, 836–843. [Google Scholar] [CrossRef] [PubMed]
  144. Saito, Y.; Liang, G.; Egger, G.; Friedman, J.M.; Chuang, J.C.; Coetzee, G.A.; Jones, P.A. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell 2006, 9, 435–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Good, K.V.; Martínez de Paz, A.; Tyagi, M.; Cheema, M.S.; Thambirajah, A.A.; Gretzinger, T.L.; Stefanelli, G.; Chow, R.L.; Krupke, O.; Hendzel, M.; et al. Trichostatin A decreases the levels of MeCP2 expression and phosphorylation and increases its chromatin binding affinity. Epigenetics 2017, 12, 934–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Meidhof, S.; Brabletz, S.; Lehmann, W.; Preca, B.-T.; Mock, K.; Ruh, M.; Schüler, J.; Berthold, M.; Weber, A.; Burk, U.; et al. ZEB1-associated drug resistance in cancer cells is reversed by the class I HDAC inhibitor mocetinostat. EMBO Mol. Med. 2015, 7, 831–847. [Google Scholar] [CrossRef]
  147. Xiao, J.; Lin, H.; Luo, X.; Luo, X.; Wang, Z. miR-605 joins p53 network to form a p53: miR-605: Mdm2 positive feedback loop in response to stress. EMBO J. 2011, 30, 524–532. [Google Scholar] [CrossRef]
  148. Zhou, Y.-J.; Yang, H.-Q.; Xia, W.; Cui, L.; Xu, R.-F.; Lu, H.; Xue, Z.; Zhang, B.; Tian, Z.-N.; Cao, Y.-J.; et al. Down-regulation of miR-605 promotes the proliferation and invasion of prostate cancer cells by up-regulating EN2. Life Sci. 2017, 190, 7–14. [Google Scholar] [CrossRef]
  149. Danesh, H.; Hashemi, M.; Bizhani, F.; Hashemi, S.M.; Bahari, G. Association study of miR-100, miR-124-1, miR-218-2, miR-301b, miR-605, and miR-4293 polymorphisms and the risk of breast cancer in a sample of Iranian population. Gene 2018, 647, 73–78. [Google Scholar] [CrossRef]
  150. Najminejad, H.; Kalantar, S.M.; Abdollahpour-Alitappeh, M.; Karimi, M.H.; Seifalian, A.M.; Gholipourmalekabadi, M.; Sheikhha, M.H. Emerging roles of exosomal miRNAs in breast cancer drug resistance. IUBMB Life 2019. [Google Scholar] [CrossRef]
  151. Pardini, B.; Sabo, A.A.; Birolo, G.; Calin, G.A. Noncoding RNAs in Extracellular Fluids as Cancer Biomarkers: The New Frontier of Liquid Biopsies. Available online: https://www-ncbi-nlm-nih-gov.gate2.inist.fr/pubmed/31416190 (accessed on 6 September 2019).
  152. Colombo, M.; Raposo, G.; Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  153. Schmittgen, T.D. Exosomal miRNA Cargo as Mediator of Immune Escape Mechanisms in Neuroblastoma. Cancer Res. 2019, 79, 1293–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Kapetanakis, N.-I.; Baloche, V.; Busson, P. Tumor exosomal microRNAs thwarting anti-tumor immune responses in nasopharyngeal carcinomas. Ann. Trans. Med. 2017, 5, 164. [Google Scholar] [CrossRef] [PubMed]
  155. Kulkarni, B.; Kirave, P.; Gondaliya, P.; Jash, K.; Jain, A.; Tekade, R.K.; Kalia, K. Exosomal miRNA in chemoresistance, immune evasion, metastasis and progression of cancer. Drug Discov. Today 2019. [Google Scholar] [CrossRef] [PubMed]
  156. Bach, D.-H.; Hong, J.-Y.; Park, H.J.; Lee, S.K. The role of exosomes and miRNAs in drug-resistance of cancer cells. Int. J. Cancer 2017, 141, 220–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The main cellular processes affected in cancer cells by HDACi treatments. The decrease of histone acetylation by HDACi leads to the modification of the expression of several genes implicated in oncogenic properties of cancer cells. From top left to bottom right, HDACi reduces angiogenesis and tumor growth, HDACi improves treatments by inhibiting DNA repair, HDACi induces cell cycle arrest and stimulates apoptosis.
Figure 1. The main cellular processes affected in cancer cells by HDACi treatments. The decrease of histone acetylation by HDACi leads to the modification of the expression of several genes implicated in oncogenic properties of cancer cells. From top left to bottom right, HDACi reduces angiogenesis and tumor growth, HDACi improves treatments by inhibiting DNA repair, HDACi induces cell cycle arrest and stimulates apoptosis.
Cancers 11 01530 g001
Figure 2. miRNAs modulated by HDACi treatments in cancer. HDACi upregulate TS-miR and downregulate oncomiR to inhibit proliferation and metastasis and to favor apoptosis.
Figure 2. miRNAs modulated by HDACi treatments in cancer. HDACi upregulate TS-miR and downregulate oncomiR to inhibit proliferation and metastasis and to favor apoptosis.
Cancers 11 01530 g002
Figure 3. miRNA biogenesis pathway. miRNA is transcribed in the nucleus and then cleaved numerous times to conduct to a mature single strand miRNA included in the RISC complex. miRNA may regulate gene expression in the cell but also in other cells by their encapsulation in microvesicles such as exosomes. miRNA may also be disseminated through the bloodstream. MVB: endosomal MultiVesicular bodies, RISC: RNA-induced silencing complex.
Figure 3. miRNA biogenesis pathway. miRNA is transcribed in the nucleus and then cleaved numerous times to conduct to a mature single strand miRNA included in the RISC complex. miRNA may regulate gene expression in the cell but also in other cells by their encapsulation in microvesicles such as exosomes. miRNA may also be disseminated through the bloodstream. MVB: endosomal MultiVesicular bodies, RISC: RNA-induced silencing complex.
Cancers 11 01530 g003
Table 1. Classification of histone deacetylase inhibitors.
Table 1. Classification of histone deacetylase inhibitors.
ClassTargeted Histone Deacetylases (HDACs)LocalizationZn2+Expression
I1, 2, 3, 8NucleusYesUbiquitous
IIa4, 5, 7, 9Nucleus and cytoplasmYesTissue specific
IIb6, 10CytoplasmYesTissue specific
IIISirtuins 1–7Nucleus, cytoplasm and mitochondriaNoVariable
IV11Nucleus and cytoplasmYesUbiquitous
Table 2. Structure and applications of the four food and drug administration (FDA)-approved histone deacetylase inhibitors.
Table 2. Structure and applications of the four food and drug administration (FDA)-approved histone deacetylase inhibitors.
NameStructureYear of ApprovalApplication
Vorinostat Cancers 11 01530 i0012006Cutaneous T Cell Lymphoma
Romidepsin Cancers 11 01530 i0022009
2011
Cutaneous T Cell Lymphoma
Peripheral T Cell Lymphoma
Belinostat Cancers 11 01530 i0032014Peripheral T Cell Lymphoma
Panobinostat Cancers 11 01530 i0042015Multiple Myeloma
Table 3. Let-7c target genes described in cancer diseases.
Table 3. Let-7c target genes described in cancer diseases.
DiseaseTargetsFunctionReference
GliomaE2F5Control of cell cycle[86]
MelanomaCALUProtein folding and sorting[91]
Lung cancerRASOncogene[92]
NSCLCITGB3/MAP4K3Metastatic abilities[87]
Cholangiocarcinoma (CCA)IL6-RImmune response[85]
EZH2/DVL3/βcateninMetastatic abilities[93]
Oral squamous cell carcinomaIL8Immune response[93]
Lung adenocarcinomaBCL-XLInhibitor of cell death[94]
Ovarian carcinomaCDC25AControl of cell cycle[95]
Hepatocellular carcinoma (HCC)CDC25AControl of cell cycle[96]
Colorectal cancerMMP11/PBX3Metastatic abilities[97]
ErythroleukemiaPBX2Transcription[98]
Breast cancerERCC6Transcription/excision repair[99]
E2F5: E2F transcription factor 5, CALU: calumenin, ITGB3: integrin beta 3, MAP4K3: mitogen-activated protein kinase kinase kinase kinase 3, IL6-R: Interleukin 6 receptor, EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit, DVL3: dishevelled segment polarity protein 3, IL8: Interleukin 8, BCL-XL: BCL2-like 1, CDC25A: cell division cycle 25A, MMP11: matrix metallopeptidase 11, PBX3: pre-B-cell leukemia homeobox 3, PBX2: pre-B-cell leukemia homeobox 2, ERCC6: excision repair cross-complementation group 6
Table 4. microRNAs regulated by the four FDA-approved histone deacetylase inhibitors in cancers.
Table 4. microRNAs regulated by the four FDA-approved histone deacetylase inhibitors in cancers.
CancersHDACimiRNAsmiRNA TargetsPathwaysRef.
BreastVorinostat Cancers 11 01530 i005 miR-200a Cancers 11 01530 i006 Keap1 Cancers 11 01530 i005 Nrf2 antioxidant pathway[109]
Cancers 11 01530 i005 miR-200C Cancers 11 01530 i006 CRKL Cancers 11 01530 i006 Invasion[110]
Cancers 11 01530 i006 Migration
Panobinostat Cancers 11 01530 i005 miR-31, miR-125a, miR-125b, miR-205, miR-141, miR-200c Cancers 11 01530 i006 NF-kB inducing kinase, ITGA5, SEPHS1, RSBN1, TFDP1 Cancers 11 01530 i005 Cellular senescence[111]
Cancers 11 01530 i006 BMI1 and EZH2 (indirect)
ColorectalVorinostatChanges in 275 out of the 723 studied human miRNAssee article for predicted targets[102]
Cancers 11 01530 i006 miR-17-92 cluster Cancers 11 01530 i005 PTENProliferation (opposite effects depending on members of the cluster)[119]
Cancers 11 01530 i006 mRNA levels of c-MYC, E2F1, E2F2 and E2F3
HCCVorinostat
Panobinostat
Cancers 11 01530 i005 let-7b Cancers 11 01530 i005 p21 Cancers 11 01530 i006 E2F1 transcriptional activity[103]
Cancers 11 01530 i006 MYC, MET, HMGA2, TRAIL, BCLX Cancers 11 01530 i006 Cell proliferation
Vorinostat Cancers 11 01530 i006 miR-17, miR-18a, miR-19a, miR-20a, miR-93, miR-106b Cancers 11 01530 i005 MICA, MICB Cancers 11 01530 i005 Recognition of tumor by innate immune cells[118]
LeukemiaVorinostat
Romidepsin
Cancers 11 01530 i005 miR-15a, miR16, miR29b Cancers 11 01530 i006 MCL1, BCL-2 Cancers 11 01530 i005 Apoptosis[113]
Vorinostat Cancers 11 01530 i005 23 miR (e.g. miR-19a, miR-19b) Cancers 11 01530 i006 BARD-1 Cancers 11 01530 i005 Sensitivity to vorinostat[116]
Cancers 11 01530 i006 26 miR (see article) Cancers 11 01530 i005 Apoptosis
Cancers 11 01530 i005 miR-196a Cancers 11 01530 i006 BCR/ABL Cancers 11 01530 i006 Transcriptional activity of the pluripotency factors[121]
Cancers 11 01530 i006 Cell cycle progression genes
Cancers 11 01530 i005 Sentivity to imatinib mesylate (a Tyrosine Kinase inhibitor)
PanobinostatmiR-26a, miR-133a, miR-181b, miR-182, miR-200c, miR-211, miR-320a, miR-320c, miR-423-5p, miR-638, miR-877, miR-1307, miR-1308, miR-1268
miR-516a-3p, miR-605
Cancers 11 01530 i006 Homologous recombination repair pathway (RAD51, BRCA1, NBS1) Cancers 11 01530 i006 Homologous recombination repairdelay DNA repair
Cancers 11 01530 i005 Sensitivity to CNDAC (prodrug used in AML)
[122]
LungVorinostat Cancers 11 01530 i006 let7b, miR-17*, miR-92aexpected targets for each miR listed in the article[106]
Cancers 11 01530 i005 miR-373 Cancers 11 01530 i006 LAMP1, VSP4B, IRAK2, BRMS1L, SYDE1, CYBRD1, PDIK1L, C10orf46, TGFBR2Associated with poorer disease-free survival[123]
LymphomaVorinostat Cancers 11 01530 i006 miR-15b, miR-17-3p, miR-17-5p, miR-18, miR-34a, miR-155 Cancers 11 01530 i006 c-myc Cancers 11 01530 i005 Sensitivity to apoptosis[115]
OvarianVorinostat Cancers 11 01530 i005 Let-7, miR-99, miR-100, miR-125… (see figure in article)[104]
PancreaticVorinostat Cancers 11 01530 i005 miR-34a Cancers 11 01530 i006 Cyclin D1, CDK6, SIRT1, survivin, BCL-2, VEGF, Notch pathway Cancers 11 01530 i006 Cell proliferation, stem cell renewal, invasivness[112]
Cancers 11 01530 i005 p21/CIP1, acetylated p53, PUMA Cancers 11 01530 i005 Apoptosis, cell cycle arrest
CRKL: v-crk avian sarcoma virus CT10 oncogene homolog-like, NF-kB: nuclear factor of kappa light polypeptide gene enhancer in B-cells 1, ITGA5: integrin, alpha 5, SEPHS1: selenophosphate synthetase 1, RSBN1: round spermatid basic protein 1, TFDP1: transcription factor Dp-1, BMI1: BMI1 proto-oncogene, polycomb ring finger, EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit, PTEN: phosphatase and tensin homolog, E2F: E2F transcription factor, p21/CIP1: cyclin-dependent kinase inhibitor 1A, MET: MET proto-oncogene, receptor tyrosine kinase, HMGA2: high mobility group AT-hook 2, TRAIL: tumor necrosis factor (ligand) superfamily, member 10, BCLX: BCL2-like 1, MICA/B: MHC class I polypeptide-related sequence A/B, MCL1: myeloid cell leukemia 1, BCL-2: B-cell CLL/lymphoma 2, BARD-1: BRCA1 associated RING domain 1, BCR: breakpoint cluster region, ABL: ABL proto-oncogene 1, non-receptor tyrosine kinase, RAD51: RAD51 recombinase, BRCA1: breast cancer 1, early onset, NBS1: nibrin, LAMP1: lysosomal-associated membrane protein 1, IRAK2: interleukin-1 receptor-associated kinase 2, BRMS1L: breast cancer metastasis-suppressor 1-like, SYDE1: synapse defective 1, Rho GTPase, homolog 1, CYBRD1: cytochrome b reductase 1, PDIK1L: PDLIM1 interacting kinase 1 like, TGFBR2: transforming growth factor, beta receptor II, CDK6: cyclin-dependent kinase 6, SIRT1: sirtuin 1, VEGF: vascular endothelial growth factor A, PUMA: BCL2 binding component 3. Arrows indicate decrease ( Cancers 11 01530 i006) or increase ( Cancers 11 01530 i005) of either miRNA or target and their associated pathway.
Table 5. microRNAs modulated by histone deacetylase inhibitors used in cancer models.
Table 5. microRNAs modulated by histone deacetylase inhibitors used in cancer models.
CancersHDACimiRNAsmiRNA TargetsPathwaysRef.
BreastLAQ824 Cancers 11 01530 i006 miR27a (≈40% of miRNAs modulated) Cancers 11 01530 i005 RYBP/DEDAF, ZBTB10/RINZF [101]
TSA Cancers 11 01530 i005 22 miR among which: miR-1, miR-143, miR-144, miR-191-3p, miR-202-5p…(predicted targets for each miRNAs provided in the article)[132]
Cancers 11 01530 i006 10 miR among which: miR-500, miR-645, miR-512-3p, miR-613…
(see article for complete listing)
TSA, VPA NaBu… Cancers 11 01530 i005 miR125-a Cancers 11 01530 i006 HDAC5 mRNA Cancers 11 01530 i005 apoptosis[133]
CCACG200746 Cancers 11 01530 i005 miR-22-3p, miR-22-5p, miR-194-3p, miR-194-5p, miR-210-3p, miR-509-3pexpression induced in treated cells Cancers 11 01530 i006 tumor growth
Cancers 11 01530 i006 proliferation
[134]
ColorectalPBA Cancers 11 01530 i005 miR-9, miR-127, miR-129, miR-137 [135]
Butyrate Cancers 11 01530 i005 18 miRNAs Cancers 11 01530 i005 p21 protein expression Cancers 11 01530 i006 proliferation[127]
Cancers 11 01530 i006 26 miRNAs (among which miR-17-92a, miR-18b-106 and miR25-106b clusters)
Entinostat (MS-275) Cancers 11 01530 i005 pri and mature miR-21 [136]
Gastric carcinomaTSA Cancers 11 01530 i005 miR-375 Cancers 11 01530 i006 PDK1, XIAP, 14-3-3ζ (YWHAZ), cIAP-2 (BIRC3) Cancers 11 01530 i006 Tumor cell viability[137]
BCL2L11 (Bim) Cancers 11 01530 i005 apoptosis
HCCTSA Cancers 11 01530 i005 miR-449 Cancers 11 01530 i005 c-MET Cancers 11 01530 i006 cell proliferation
Cancers 11 01530 i005 apoptosis
[138]
Sodium valproate Cancers 11 01530 i006 miR-889 Cancers 11 01530 i005 MICB Cancers 11 01530 i005 sensitivity to NK cell-mediated lysis[139]
Leukemiavalpromide
(=VPA analog)
Cancers 11 01530 i006 miR-144, miR-451, miR-155 (all cells) Cancers 11 01530 i006 GATA-1 Cancers 11 01530 i005 erythropoiesis impairment[140]
Cancers 11 01530 i005 GATA-2
Cancers 11 01530 i006 miR-15a, miR-16, miR-222 (some cells) Cancers 11 01530 i005 ETS family (PU.1, ETS-1, GABP-α, Fli-1) Cancers 11 01530 i005 megakaryocyte features
LungEntinostat (MS275) Cancers 11 01530 i005 miR-200a Cancers 11 01530 i006 KEAP1/NRF2 Cancers 11 01530 i006 cell growth[128]
TSA Cancers 11 01530 i005 Let-7, miR-15a, miR-16-1 Cancers 11 01530 i006 Cancers 11 01530 i005 proliferation and apoptosis[125]
induce cell cycle arrest
LymphomaRGFP966 Cancers 11 01530 i005 miR-15a, miR-195, let-7a (in vitro and in vivo) Cancers 11 01530 i006 BCL-2, BCL-XL Cancers 11 01530 i005 apoptosis[126]
Melanoma4PBA (or 5Aza, 5Aza + 4PBS) Cancers 11 01530 i005 miR-34b, miR-132, miR-142-3p, miR-200a, miR-375, miR-489 Cancers 11 01530 i006 Proliferation, invasion[141]
Cancers 11 01530 i005 wound healing
changes in cell morphology
Multiple MyelomaAR-42 Cancers 11 01530 i005 miR-9-5p Cancers 11 01530 i006 CD44 [142]
OvarianAR42 Cancers 11 01530 i005 miR-15a, miR-34, …
(see figure in article)
Cancers 11 01530 i005 WT1, PAX2, GATA6, APO2/TRAIL…
(see article)
Cancers 11 01530 i006 EMT, Canonical Wnt R signaling
Cancers 11 01530 i005 Negative regulation of cell cycle processes, extrinsic apoptosis
[104]
PancreaticAR-42 Cancers 11 01530 i005 miR-30d, miR-33, miR-125b Cancers 11 01530 i006 p53, cyclin B2, CDC25B Cancers 11 01530 i006 Invasion, tumor growth[129]
ProstateMocetinostat Cancers 11 01530 i005 miR-31 Cancers 11 01530 i006 E2F6 Cancers 11 01530 i005 apoptosis[130]
OBP-801 Cancers 11 01530 i005 miR-320a in vitro and in vivo (rat) Cancers 11 01530 i006 PSA, androgen receptor Cancers 11 01530 i006 Viability, cell growth, cell proliferation, prostate tumorigenesis (in vivo)[131]
TongueTSA (or Doxorubicin, 5-fluorouracil, etoposide treatments) Cancers 11 01530 i005 miR-375 Cancers 11 01530 i006 CIP2A, MYC, 14-3-3z, E6AP, E6, E7 Cancers 11 01530 i006 cell proliferation, migration and invasion[143]
Cancers 11 01530 i005 p21, p53, RB
Various modelsPBA (and 5-Aza-CdR) Cancers 11 01530 i005 17 miR/313 studied (see article for details) Cancers 11 01530 i006 BCL6 (suggested) [144]
TSA Cancers 11 01530 i005 miR132/212 Cancers 11 01530 i006 MeCP2 [145]
NaBu, Sodium Butyrate, E2F: E2F transcription factor, p21/CIP1: cyclin-dependent kinase inhibitor 1A, MET: MET proto-oncogene, receptor tyrosine kinase, BCL-2: B-cell CLL/lymphoma 2, RYBP/DEDAF: RING1 and YY1 binding protein, ZBTB10/RINZF: zinc finger and BTB domain containing 10, HDAC5: histone deacetylase 5, PDK1: pyruvate dehydrogenase kinase, isozyme 1, XIAP: X-linked inhibitor of apoptosis, 14-3-3ζ (YWHAZ): tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta, cIAP-2 (BIRC3): baculoviral IAP repeat containing 3, BCL2L11 (Bim): BCL2-like 11, MICB: MHC class I polypeptide-related sequence B, GATA: globin transcription factor, KEAP1/NRF2: kelch-like ECH-associated protein 1, BCL-XL: BCL2-like 1, WT1: Wilms tumor 1, PAX2: paired box 2, APO2/TRAIL: tumor necrosis factor receptor superfamily, member 10a, PU.1: Spi-1 proto-oncogene, ETS-1: v-ets avian erythroblastosis virus E26 oncogene homolog 1, GABP-α: GA binding protein transcription factor, alpha subunit 60kDa, CDC25B: cell division cycle 25B, PSA: prostate specific antigen, CIP2A: cancerous inhibitor of PP2A, RB: retinoblastoma 1, BCL6: B-cell CLL/lymphoma 6, MeCP2: methyl CpG binding protein 2. Arrows indicate decrease ( Cancers 11 01530 i006) or increase ( Cancers 11 01530 i005) of either miRNA or target and their associated pathway.

Share and Cite

MDPI and ACS Style

Autin, P.; Blanquart, C.; Fradin, D. Epigenetic Drugs for Cancer and microRNAs: A Focus on Histone Deacetylase Inhibitors. Cancers 2019, 11, 1530. https://doi.org/10.3390/cancers11101530

AMA Style

Autin P, Blanquart C, Fradin D. Epigenetic Drugs for Cancer and microRNAs: A Focus on Histone Deacetylase Inhibitors. Cancers. 2019; 11(10):1530. https://doi.org/10.3390/cancers11101530

Chicago/Turabian Style

Autin, Pierre, Christophe Blanquart, and Delphine Fradin. 2019. "Epigenetic Drugs for Cancer and microRNAs: A Focus on Histone Deacetylase Inhibitors" Cancers 11, no. 10: 1530. https://doi.org/10.3390/cancers11101530

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