Review

1.

Introduction

2.

HDAC classes and their involvement in cancerogenesis

3.

Cooperative miRNA-HDAC

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

regulation in cancer 4.

pathways of HDACs and miRNAs The analyzed pathways and their clinical relevance in detail 6.

Stefan Swierczynski, Eckhard Klieser, Romana Illig, Beate Alinger-Scharinger, Tobias Kiesslich & Daniel Neureiter† †

Paracelsus Medical University, Salzburger Landeskliniken, Institute of Pathology, Salzburg, Austria

Elaboration of the hypothesis based on shared downstream

5.

Histone deacetylation meets miRNA: epigenetics and post-transcriptional regulation in cancer and chronic diseases

HDACis in clinical trials and therapeutic possibilities

7.

Conclusion

8.

Expert opinion

Introduction: Epigenetic regulation via DNA methylation, histone acetylation, as well as by microRNAs (miRNAs) is currently in the scientific focus due to its role in carcinogenesis and its involvement in initiation, progression and metastasis. While many target genes of DNA methylation, histone acetylation and miRNAs are known, even less information exists as to how these mechanisms cooperate and how they may regulate each other in a specific pathological context. For further development of therapeutic approaches, this review presents the current status of the crosstalk of histone acetylation and miRNAs in human carcinogenesis and chronic diseases. Areas covered: This article reviews information from comprehensive PubMed searches to evaluate relevant literature with a focus on possible association between histone acetylation, miRNAs and their targets. Our analysis identified specific miRNAs which collaborate with histone deacetylases (HDACs) and cooperatively regulate several relevant target genes. Expert opinion: Fourteen miRNAs could be linked to the expression of eight HDACs influencing the a-(1,6)-fucosyltransferase, polycystin-2 and the fibroblast-growth-factor 2 pathways. Focusing on the complex linkage of miRNA and HDAC expression could give deeper insights in new ‘druggable’ targets and might provide possible novel therapeutic approaches in future. Keywords: epigenetics, histone deacetylase, microRNA Expert Opin. Biol. Ther. [Early Online]

1.

Introduction

Carcinogenesis is not restricted to genomic alterations such as amplifications, translocations, deletions and point mutations. It is also associated with epigenetic events which summarize the mechanisms of regulation of gene expression levels without alterations at the DNA sequence level. The central epigenetic mechanisms in humans are DNA methylation and post-translational histone modifications [1]. Many developmental and tissue remodeling processes such as epithelial mesenchymal transition (EMT) are epigenetically regulated by an interplay of epigenetic modifications such as DNA methylation and other ways of transcriptional reprogramming [2]. Currently, silencing of tumor-suppressor genes by promoter hypermethylation are the most widely studied epigenetic modifications in human cancers. In the last years, there was a significant increase in the knowledge and understanding of the involvement of histone modifications in cancer development [1]. The level of histone acetylation plays a crucial role in the regulation of gene transcription [3]. Histone deacetylation increases the ionic interactions between the positive histones and the negative DNA by deacetylation of "-amino groups of lysine residues in all four histone proteins in histone tails. These structural changes yield 10.1517/14712598.2015.1025047 © 2015 Informa UK, Ltd. ISSN 1471-2598, e-ISSN 1744-7682 All rights reserved: reproduction in whole or in part not permitted

1

S. Swierczynski et al.

Article highlights. .

. . .

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

.

Histone deacetylases (HDAC) are overexpressed in many types of cancer such as colorectal cancer, ovarian cancer, breast cancer, pancreatic cancer and chronic disease. HDACs and miRNAs (microRNA) have been shown to interact with each other. There are distinct pathways (FUT8, PC2, FGF2) that show a HDAC-miRNA crosstalk. HDAC and miRNA driven interactions could lead to establishment of new biomarkers and new therapeutic approaches. Combination of selective HDAC inhibitors and new miRNA silencing techniques could form a new perspective in epigenetics-based therapies.

This box summarizes key points contained in the article.

to a repression of gene transcription by limiting the accessibility of the transcription system due to chromatin compaction. By histone modification, histone deacetylase (HDAC) enzymes can interact with numerous transcription factors and can regulate a large number of genes [1]. HDACs, in general, are considered to be transcriptional repressive as they stabilize condense chromosome structure and make it less accessible for transcription factors. Furthermore, these enzymes take part in co-repressor protein complexes. Apoptosis is negatively regulated by HDACs as HDAC inhibitors (HDACi) causes upregulation of proapoptotic proteins such as Bad and downregulation of antiapoptotic proteins like Bcl-2 [4]. DNA damage repair is also supported and regulated by HDACs which interact with DNA damage-responsive factors and promote DNA repair. By inhibition of the cooperating HDACs an increase of ROS (reactive oxygen species) production accompanied by suppression of DNA repair proteins is observed [5,6]. Replication of DNA requires global chromatin reconstruction which is governed by HDACs as key regulators including other aspects of cell cycle regulation via control of cyclins and cyclin-dependent-kinases. In yeast, HDACs are centrally involved in the temporal expression sequence of cell cycle genes. Accordingly, inhibition of HDACs leads to cell cycle arrest at certain phases and a delayed proliferation [5-7]. Additionally, HDACs are involved in many heterogeneous biological functions of the cell including autophagy, metabolism and heat-shock processes as well as angiogenesis: i) the influence of HDACs in autophagy which results in selfdegradation of unnecessary or dysfunctional cellular components is still under investigation; ii) HDACs have also been shown to influence metabolic processes like fatty acid metabolism, glucose utilization and oxidative phosphorylation [8]; iii) Furthermore, oncogenes and tumor suppressor genes are influenced by HDACs via the molecular chaperone heatshock protein 90; and iv) The last, currently important area of HDAC activity is the field of angiogenesis where the use 2

of HDAC inhibitors (HDACis) showed a very profound linkage to angiogenesis-stimulating factors where they block angiogenesis by downregulating angiogenesis-stimulating factor, hypoxia-inducible factor 1a (HIF-1a) and vascular endothelial growth [9]. In summary, histone de-acetylation by HDACs has been demonstrated as an important epigenetic mechanism in many research areas. In contrast, the dysregulation of cell pathways interacting with HDAC function is observed in a variety of human epithelial and hematological tumors and the therapeutic approach to treat such tumors with HDACis seems promising (Figure 1). Early clinical trials were conducted in defined tumor (sub-)types such as myeloid leukemia [10,11]. Currently, a large body of evidence indicates similarly important roles of microRNAs (miRNA) in (dys-) regulation of cellular functions in the context of cancer initiation and progression. As some of these miRNA have been shown to interact with HDAC-mediated epigenetic mechanisms, knowledge about these particular mechanisms could lead to new and meaningful combinatory treatment strategies compared to monotherapies targeting single HDACs by specific inhibitors (as discussed in Sections 6 and 7). Therefore, as many diseases share common pathways of gene or/and protein regulation by HDACs and miRNAs, a multitarget approach on HDAC and miRNA regulation could be beneficial for both, treatment of chronic diseases and cancer.

HDAC classes and their involvement in cancerogenesis

2.

The family of HDAC enzymes is divided into four classes including subclasses and comprise 18 members based on their sequence homology to the corresponding yeast HDAC [1,12]. For a better understanding of the importance of each HDAC, we first give a short description of the role of each HDAC member in human malignancies focusing on: i) target downstream pathways; ii) the tumor entity; and iii) parameters of clinical outcome (recurrence, metastasis and survival). We further give an overview of the published literature of the main HDAC members and their interactions with miRNA functions. Class I HDACs HDAC1, 2 and 3 are members of protein complexes which are necessary for transcriptional repression and epigenetic landscaping. These large multi-protein complexes silence target genes through removal of the acetyl group from the lysine residues in histone tails. In a recent series of 140 colorectal cancer samples, high HDAC1, 2, 3 expression levels were associated with a significantly shorter survival, with HDAC2 expression being an independent prognostic factor. High HDAC1, 2, 3 expressions was also associated with dedifferentiation and enhanced proliferation of pancreatic cancer cells [13,14]. 2.1

Expert Opin. Biol. Ther. (2015) 15(5)

HDACs and miRNAs

Motility (HDAC6)

Autophagy (HDAC1,6,10)

Cell cycle/proliferaton (HDAC1,2,3)

Apoptosis (HDAC1,2,4)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

Immune response (HDAC1,2,6,7,11)

Angiogenesis (HDAC1,4,6,7)

Differentiation (HDAC3,8,9)

Metabolism (HDAC1,2,3)

HDAC inhibitors

DNA damage repair (HDAC1,2,3,4,6,9,10 )

• Vorinostat (SAHA) • Romidepsin • Panobinostat • Valproic acid • Belinostat • Mocetinostat • Abexinostat • Entinostat • SB939 • Resminostat • Givinostat • Quisinostat

Heat shock protein 90 (HDAC6,10)

HDACs

Figure 1. HDAC inhibitors (HDACi) and HDACs. This illustration shows the conventional areas of HDAC inhibition based on Limoene et al. with a list of FDA-approved HDACi [83]. 2.2

Class II HDACs

This class is further subdivided in two subfamilies: Class IIa is characterized by a long functionally important N-terminal domain regulating nuclear-cytoplasmic shuttling and specific DNA-binding. Class IIb contains of two tandem deacetylase domains and a C-terminal zinc finger [13]. Class IIa HDAC4 is associated with the repression of differentiationrelated genes such as Pax7 which stimulates the differentiation of satellite cells and regulates HIF-1 a transcriptional activity in renal carcinoma. HDAC5 expression has been shown to be upregulated in colorectal cancer. Its interaction with the transcription factor GATA-1 leads to murine erythroleukemia cell differentiation. HDAC7 is highly expressed in colorectal cancer, whereas it shows only moderate expression in bladder, renal and breast cancer. HDAC7 silencing in endothelial cells (EC) altered their migration and their capacity to form capillary tube like structures in vitro while it has no effect on cell adhesion, proliferation or apoptosis. This led to the conclusion that HDAC7 might be a target for anti-angiogenesis in 2.2.1

cancer therapy. HDAC9 promotes angiogenesis in EC by transcriptional repression of miRNA 17 -- 92 cluster [13-15]. Class IIb HDAC6 expression was significantly higher in squamous cell carcinoma versus normal oral squamous tissue. HDAC6 expression was correlated in breast cancer samples with better survival. High HDAC6 mRNA tended to be more responsive to endocrine treatment and was a prognostic indicator for breast cancer progression. In vitro, HDAC6 overexpression leads to an increased migration of embryonic fibroblasts while after inhibition the migration levels decreased. HDAC6 was found to play a role in epidermal growth factor-induced nuclear localization of b-catenin and subsequent c-Myc activation in colon carcinoma. HDACs 4 and 6 were shown to regulate and bind HIF-1a transcriptional activity, which is a key regulator for adaption to the hypoxic microenvironment. These two HDACs may therefore represent an important target of anti-angiogenesic therapies. Knockdown of HDAC6 and HDAC10 via siRNA transfection induced a depletion of VEGF receptor 2.2.2

Expert Opin. Biol. Ther. (2015) 15(5)

3

S. Swierczynski et al.

1/2 proteins. HDAC6 is also involved in epithelialmesenchymal transition (EMT) in metastasis of bronchial carcinoma by an interaction with the TGF-b smad3 cascade. The knockdown of HDAC10 reduces VEGF receptor 1/2 expression and is involved in autophagy and the cooperation of metalloproteases 2 and 9 [13,14,16]. Class III HDACs (Sirtuins) Like other HDACs, the group of sirtuins is (over-)expressed in many types of cancer. SIRT1 is upregulated in acute myeloid leukemia, prostate and non-melanoma skin cancer, whereas it is downregulated in colorectal cancer. SIRT4 is reported to be involved in the DNA damage response pathway. SIRT3 and SIRT7 are upregulated in breast cancer. SIRT2 is downregulated in gliomas and gastric carcinoma. Up-to-now, altered expression of SIRT5 and SIRT6 in cancer has not been reported [17,18].

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

2.3

Class IV HDACs The loss of HDAC11 leads to cell death and inhibition of metabolic activity in ovarian cancer cell lines. Contrarily, HDAC11 is overexpressed in colon, prostate, breast and ovarian cancer cell lines [19]. In Hodgkin lymphoma, HDAC11 has been found to be involved in the regulation of OX40, which is one mediator of antitumor immunity [20]. Tumorigenic mis-regulation of HDAC expression may depend on ‘classical’ activation through DNA mutations such as point mutations. While these findings are commonly accepted, another possible regulative mechanism has been introduced by CM Wendtner in 2012: He demonstrated that inhibition of HDAC that are overexpressed in chronic lymphocytic leukemia (CLL) induces acetylation of a histone complex which influences the promoter activity of critical miRNAs involved in CLL. This finding raised the question of whether miRNAs are cooperatively linked with HDACs during tumorigenesis and how this cooperation influences downstream targets or signaling pathways. By taking up this hypothesis, the following sections summarize the available literature on HDAC and miRNA interactions and their role for carcinogenesis [21]. 2.4

Cooperative miRNA-HDAC regulation in cancer 3.

By now, the mechanisms of miRNA regulation are not fully understood. Alterations in miRNA expression in tumorigenesis and metastasis have been described within several publications. miRNAs are short, non-coding RNA molecules that influence and regulate gene expression on the posttranscriptional level. They can silence genes by binding to the 3´-untranslated region (3¢UTR) of target mRNAs, which leads to their degradation or the repression of translation. In tumor cells, specific miRNAs are down- or upregulated: for example, tumor-supressive miRNA might be downregulated during tumorigenesis, while oncogenic miRNAs (onco-miRs) 4

that target, for example, Growth-inhibiting genes are upregulated. Of special importance, it has been shown that miRNAs also regulate genes encoding for particular HDACs [11,22]. In the next paragraphs, we try to summarize current information on the complex recursive interactions between HDACs and miRNA: in particular, we focus the discussion on putatively affected downstream pathways and tumor entities associated with clinical outcome variables as far published data are available. HDAC1 The tumor suppressor miR-34, a transcriptional target of TP53, activates TP53 in a positive feedback loop via p21 (Cip1/Waf1). Interestingly, HDAC1 is a direct target of miR-34 and the repression of HDAC1 leads to an induction of p21 (Cip1/Waf1), which is mimicking the positive feedback loop as miR-34. In the absence of TP53, miR-34 is capable of inducing p21 (Cip1/Waf1). This shows that both miR-34 and HDAC1 can lead to a p21 (Cip1/Waf1) induction. Second, miR-449a is directly targeted by HDAC1 in prostate cancer tissue and this miRNA represses HDAC1 and thus growth and viability in prostate cancer cells. In summary, HDAC1 and miR-34 or miR-449a interactions are linked to cell cycle arrest, apoptosis and senescent-like phenotype via cell-cycle-dependent kinase inhibitors like p21 (Cip1/Waf1) or p27 (Kip1) as well as other related proteins such as p53, PUMA, bcl-2, CDK6 or Sirt1 [23-25]. 3.1

HDAC2 The depletion of HDAC2 in neuroblastoma models caused an increased miR-183 expression. This could also be mimicked by treating neuroblastoma cell lines with small molecules inhibiting HDAC2 enzyme activity. Complete HDAC2 depletion increased histone H4 pan-acetylation of the miR-183 promoter region indicating increased transcriptional activation which leads to a tumor suppressive effect through miR-183 in neuroblastoma. The molecular linkage between HDAC2 and miR-183 is the proto-oncogen MYCN regulating the transcriptional activation and repression of multiple genes involved in human cancerogenesis [26]. 3.2

HDAC3 Knockdown of HDAC3 with specific siRNA results in an increased hyperacetylation of the Dleu/miR-15a/16-1promoter region. This upregulation increased the expression of miR-15a/16-1, which suppressed cell growth and colony formation in lung cancer cells, which is obviously regulated via Bcl-2, a master anti-apoptotic gene. In hepatocellular cancer, inhibition of HDAC1 and HDAC3 significantly enhanced the transcription of miR-224 leading to increased cell proliferation and apoptosis as well as cell migration and invasion in liver cells [27,28]. 3.3

Expert Opin. Biol. Ther. (2015) 15(5)

HDACs and miRNAs

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

3.4

HDAC4

In B-cell leukemia, miR-155 directly targets HDAC4 a corepressor of Bcl-6 which regulates the expression and activity of Bcl-6 by promoting cell survival and proliferation markers such as inhibitor of differentiation (Id2), IL-6 (IL6), cMyc, Cyclin D1, and Mip1a/ccl3 [29]. Additionally, miR-206 inhibits HDAC4 in mice muscle cells; this affects the endogenous levels of HDAC expression in the mouse model modulating the myogenic differentiation via the TGF-b-signaling pathway as already shown for HDAC3 [30,31]. Several miRNAs were characterized to modulate HDAC4 including miR-1, miR-29, miR-140, miR-200a and miR-365 in different cell types. MiR-200a directly targets the 3¢-UTR of the HDAC4 mRNA and represses expression of HDAC4. MiR-1 and miR-499 in cardiomyocyte progenitor cells reduced proliferation rate, and enhanced differentiation into cardiomyocytes in human cardiomyocyte progenitor cells and embryonic stem cells via the repression of HDAC4. MiR-29 has been implicated in muscle differentiation is downregulated by TGF-b. TGF-b treatment of myogenic cells is associated with increased expression of HDAC4. MiR-365, a mechanically responsive miRNA, was associated with modulation of chondrocyte differentiation via directly targeting HDAC4 and miR-140 demonstrated the phenotype of dwarf as a consequence of impaired chondrocytes [32]. HDAC5 HDAC5 is targeted by miR-2861 in osteogenesis. The relevant downstream pathway partner of both is the bone morphogenetic protein 2. Experimental regulation of HDAC5 and miR-2861 is linked to primary osteoporosis in humans [33]. 3.5

LAMC1 and PAX6 in tongue squamous cell carcinoma (TSCC). Therefore, the HDAC7 and miR-140-5p-associated extracellular matrix interactions via ADAM10 influence the invasive and migrative potency of TSCC [37]. HDAC8 In human colon cancer stem cells, negative regulation of HDAC8 via miR-93 suppresses proliferation and cell colony formation [38]. Thereby, the repressive effect of miR-93 on HDAC8 is paralleled with other miRNA targets such as BAMBI, CCND2, CDKN1A, KIF23, MAP3K9, MAP3K11, MYCN, PPARD, TLE4 and ZDHHC1. The mentioned pathways are partially linked to the APC/b-Catenin-pathway, for example, BAMBI (Akiyama et al. 2004), CDKN1A (Ben-Neriah et al. 2011), MCYN (Yochum et al. 2014), PPARD (Faivre et al. 2002) and TL4 (Jones et al. 2006) indicating how HDAC and miRNA is cooperatively linked to colorectal cancer by downstream interactions of different sub-pathways [39-43]. 3.8

HDAC9 HDAC9 regulates miRNAs in EC as silencing of HDAC9 stopped sprouting in vitro and reduces vessel growth in a zebrafish model in vivo. The pro-angiogenic effect of HDAC9 is mediated by repressing the miR-17-92 cluster. The endothelial cell tube formation and sprouting is linked to the JAK-1 pathway which could be used as possible common therapeutic target in cancer vasculogenesis [44]. 3.9

HDAC10 Until now, no miRNA data are published (01/2015). 3.10

HDAC11 Downregulation of miR-145 by IFN-I targets HDAC11 and leads to increased IL-10 expression in macrophages. This crosstalk between HDAC11 and miR-145 via IFN-I and IL-10 influences the innate immune response which is linked again with the JAK1-Stat1 signal cascade in HDAC9 [45]. To summarize these published data, inhibition of miRNAs and their influence on the HDACs showed similar or better effects on the cellular effects (like proliferation/apoptosis, inflammation/immune response, differentiation/motility -- see Figure 1) compared to the inhibition via HDAC inhibitors. As reviewed by Shi et al., it could be postulated that by using proper biomarkers, identifying tumors and stratifying patients into groups that may undergo an improved clinical response to HDAC inhibitor-based therapy we can make an individual therapy for the benefit of the patients, although it would gain more precision to incorperate miRNAs, too [46]. In 2012, La Thangue et al. suggested that rationally designed therapy in different types of cancers, probably in combination with other anticancer agents for targeting the pathway for each type of HDAC could be promising for development of more effective therapies [47]. From these thoughts combined with the abovementioned known associations of miRNA and HDAC 3.11

3.6

HDAC6

In a case report of a family with X-linked dominant chondrodysplasia, it was shown that this gene variant is located in the HDAC6 genomic sequence matching the seed of miR-433. This HDAC variant completely abrogates the posttranscriptional regulation normally exerted by this miRNA on an HDAC6 3¢-UTR-bearing transgene in transduction experiments. The pathogenetic effect of HDAC6 inhibition via has-miR-433 is an increased level of a-tubulin associated with decreased histone acetylation at the a-tubulin locus [34]. MiR-22 acts as a critical regulator of balance between adipogenic and osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing its target HDAC6 [35]. Increased expression of miR-548 by ectopic miR-548 expression leads to a downregulation of HDAC6 mRNA and protein expression in lymphoma cells. In contrast, knockdown of miR-548 by transfection of antimiR-548 increased HDAC6 expression [36]. HDAC7 HDAC7 is a direct target of miR-140-5p, which plays a role in inhibition of migration-related genes such as ADAM10, 3.7

Expert Opin. Biol. Ther. (2015) 15(5)

5

S. Swierczynski et al.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

• Literature combinations of HDACs and miRNAs. • miRNA from the literature searched for their target genes.

2. Searching cycle • Target genes searched for other miRNAs. • HDACs searched for their theoretical target miRNAs from databases (mirbase.org/ DIANAMICROT).

1. Searching cycle

• Combination of the miRNAs from the HDACs, the target genes and their miRNAs as well as the miRNAs from literature search. • Literature query for documented conformities.

Final step

Figure 2. Strategies for data mining and database search (PubMed, mirbase.org and DIANA-MICROT).

functions, we propose that combined inhibitory strategies targeting both these epigenetic regulators might be especially worth exploring in future research. 4. Elaboration of the hypothesis based on shared downstream pathways of HDACs and miRNAs

For substantiation of our hypothesis of a multitarget therapy based on simultaneous inhibition of HDAC and miRNA function, PubMed was searched for literature to investigate any published linkage between HDACs and miRNAs. We searched in two different miRNA and gene databases (mirbase.org and DIANA-MICROT) for overlap of the published HDAC-miRNA combinations within their theoretical gene output and classified the results in three categories: i) direct interaction of miRNAs and HDACs; ii) the interaction of these miRNAs on targets (genes); and iii) and the genes that directly or indirectly interact with HDACs. As outlined in Figure 2, combination of HDACs and miRNAs were searched in the first step of data mining, combining target genes of both players in the second step and the consecutive pathways were characterized for the association of HDACs and miRNAs (Figure 2). These three overlapping queries led to an index (Figure 3) where we collected all miRNA target genes of every HDAC (database software DIANA-MICROT v3.0) with a precision almost higher than 0.8 in the database. After detailed consideration, we formed a hypothesis on the connections of HDACs with their miRNA targets and vice versa. This formed a cycle of regulations between genes, HDACs, miRNAs and the attached protein expression 6

(illustrated in Figure 4), which shows the perceptual weighting of direct and indirect linkage between HDACs and miRNAs in the literature. In comparison to the current literature, which rather describes indirect regulation (ii/iii) effects of HDACs and miRNAs, available data on the direct regulation (i) is scarce as also mentioned by Noonan et al. as well as by other authors [23,48-51]. Figure 4 illustrates that there is evidence that the regulation of HDACs and miRNA is linked on different molecular levels as shown by Delcuve et al. [52]. To substantiate this hypothesis, we searched in every direction of the regulatory HDAC-miRNA cycle in order to find evidence for such connections. We searched all sides of HDAC interaction, although direct or indirect with miRNAs and their targets (genes/proteins) and after that we filtered only the few that give a complete signaling cycle out. The remaining genes were again searched in PubMed for the published data to validate our hypothesis [52,53]. For the first data set, we searched for any evaluated HDAC and for all genes displayed in Figure 3. We looked for both, the estimated miRNA targets and the one connected with them. After that we did another literature search for the 12 genes that displayed a HDAC-gene couple. The a-(1,6)-fucosyltransferase 8 (FUT8), polycystin 2 (PC2) and fibroblast-growth-factor 2 (FGF2) pathways revealed a prominent connection by database and literature search and were selected for further investigation (whole search list not shown due to the extensive data set of genes). All three genes (Figure 5A -- C) demonstrated a function in proliferation and could be linked with regulation of growth factors.

Expert Opin. Biol. Ther. (2015) 15(5)

mir15a

mir224

HDAC3

HDAC3

Expert Opin. Biol. Ther. (2015) 15(5)

mir17

HDAC9

Traget Genes Functional Areas Sirt1 SATB2 Apoptosis ZNF281 TGIF2 Receptor Function (GABA) GABRA3 BRPF3 Malignancy/Invasivness FUT8 E2F3 Oncogenic Transformation RRAS Protein Modification FAM76A Development FAM70A Cell Cycle Modulation ACSC4 Development (Cleft Palate) GOLPH3 Mitotic Cycle CPEB2 SALL1 Translation CCNE1 KIF23 Mitotic Cycle FGF2 WIPI2 Growth Factor PLAG1 AKT3 Development WEE1 C1ORF21 Organelle Transport STOX2 Multiprotein Complex SLC20A2 STK33 ZBTB34 C2ORF21 API5L1 Histone Acetyltransferase Nuclear Transcription MYST3 API5 Oncogenic Transformation AFF3 YOD1 Mitochondrial Membran AFF2 UBE2D3 Apoptosis KCTD12 DDX34 Ubiquitin Ligation BAZ2A FAM49B MARCH5 GGNBP2 XKR3 SLMAP Extracellular matrix FNDC3A HIVEP3 Migration RNF165 NRP1 Differnetiation GJA1 CD2AP Angiogenesis TNKS2 SFRS9 Invasion FNDC3B RNA Binding UST Transcription Factor TMSB4X Splicing Factor HNRNPU Gap Junction CAGE1 Cell-Cell Contact RNF165 C11ORF61 Actinremodeling FNDC3A RSBN1 Alternative Splicing TNKS2 CAGE1 GJA1 CD2AP UST HNRNPU HIVEP3 TMSB4X SFRS9 Mitotic Cycle TITY5 AP3B1 Development UBR1 SAT1 Neuronal Excitability CEP135 PDZRN3 EPHA7 ZC3H6 C2ORF3 KCNIP2 SORBS CHD9 TTC8 Development SERF1A E2F5 Oncogenic Transformation SERF1B C7ORF43 MAPK-Pathway ZNFX ITGB8 Cell-Cell Contact PLEKHA3 VSX1 Polycystic Kidney Disease SCC35E2 SLITRK3 MYCN PKD2 MAP3K2 GPR137C MAP3K9 Growth Factors SNX2 RNF19A Leucine Zipper Factor Family GPR161 HAND2 Cardiac Morphogenesis MMD ZNF800 Immunoglobulin Superfamily C15ORF29 GALNTL1 Sorting Nexin Family TSSK2 CNTN1 BCL11A G-protein coupled receptor 161 PDGFRA YOD1 C14ORF101 BACH1

HDAC Member miRNA

Traget Genes FUT8 E2F3 RRAS LEF1 HDAC1 mir449a ZNF281 SYT1 SATB2 SLC25A27 GABRA3 FAM76A GOLPH3L FAM70A TGIF2 Breast Cancer PSEN2 LRP6 Chronic Lymphotic Leukemia DUSP10 PTPN4 Myeloid Leukemia HDAC2 mir183 PFN2 PPR2CB Ovarian Carcinoma PPR2CA SAC5 Basal Ganglia Calcification KCNK10 GNG5 Tones-Brock Syndrome TMPO Glioma AKAP12 MAL2 BTG1 Myeloid Leukemia ZNF585a ZNF678 Diabetes Type2 GMFB ZNF283 Parkinson HDAC3 mir16 ZNF588 GOLGA7 ZNF107 GOLGA8G ZNF117 GOLGA8F ZNF138 SEPT2 HDAC1/7a IKZF2 WDR72 RNF6 Multiple Sclerosis JARID2 LDLRAD3 Urinary Bladder Carcinoma TSHZ3 MBTD1 Parkinson HDAC4 mir155 IRF2BP2 ZBTB38 Melanoma RNF123 DUSP14 Colorectal Cancer WEE1 FGF7P2 DHX40 FGF7 LRP1B ARID2 FOS Multiple Sclerosis SIRPA PALM Parkinson PTPRC Arthritis HDAC5 mir2861 COLEC10 Urinary Bladder Carcinoma PI3 Alzheimer Disease FAM62A Interstitial Fibrosis Disease GRIK2 RRBP1 CLIP3 HOXB4 Alzheimer Disease SERF1A TNKS2 Johanson-Blizzard Syndrome SERF1B MAGI3 Glioblastoma HDAC8 mir93 MYCN RAB11FIPS Hyperpigmentation ZVFX1 GPR6 Cardiac Disease PLEKMA3 MYT1L Foot-and-Mouth Disease MAP3K2 mir183 SLC35E2 GPR137C HDAC2 SLITRK3 Neuroblastoma ATN1 CLCN6 Pulmonary Hypertension PPME1 TNPO1 Lung Cancer HDAC9 mir92 BCAM NANP Ovarian Cancer ZBTB34 DRP2 Synapse Development ZNF3 PPFIA4 Polycystic Kidney Disease HKR1 MB SIX4 WFDC2 TPP1 Leukemia DST LGALS14 Thyroid Disorders ATRX Lung Cancer HDAC11 mir145 GRIK2 Proliferative Vitreoretinopathy SMG7 Breast cancer PDZK1P2 Hears Disease PRAMEF19 Squamous Cell Carcinoma PDZK1 RAD51AP1 SHOC2 NFIA All data were collected with mirbase.org and evaluated with DIANA-MICROT.

Clinical Relevance Alzheimer Disease Blood Pressure Pulmonary Emphysema Huntington Disease Hepatocellular Carcinoma Osteoporosis Prostate cancer

Myeloid Leukemia Huntington Disease

Zinc Finger Protein Glia Maturation IKFZ2+IKFZ1--Hdac7a, Hdac1 Golgi Organisation DNA Damage Response

Panceatic Neuroend. Tumor Noonan-Syndrom Myeloid Disease

Huntington Disease Alzheimer Disease Sickle Cell Disease Ovarian Cancer Neuronal C. Lipofuscinosis

Exctracellular Matrix Modulation Transcription Factor Lysosome Function Ion Chanel Regulation

Chromatin Remodeling DNA Repair Transcription Factors Membrane Regulation

Neuroblastoma Neuropsychiatric Disorders Colon Cancer Alzheimer Disease Depression

Melanoma Leukemia

Development Ras Pathway Oncogenic Transformation MAPK Pathway PTEN Interaction Transmembrane Signaling

Growth Receptor Signaling Mitotic Cycle Oncogenic Transformation RNA Editing mRNA Modulation Development

Myeloid Leukemia Depression Ovarian Cancer Alzheimer Disease Pulmonary Disease

Alzheimer Disease Huntington Disease Ovarian Cancer Colorectal Cancer Thyroid Carcinoma

Cell growth (MAPK/ERK/PKA) Actin Polymerization Regulation of Ca/K CD90 Induction Transcytose WNT-Signaling (LRP) Mitotic Cycle Oncogenic Transformation

Cell Proliferation Neural Development Mitotic Cycle Cellular Processes (Splicing) LDL Receptor Cell Survival Environmental Stress

Clinical Relevance Pulmonary Emphysema Huntington Disease Colorectal Cancer Osteoporosis Hepatocellular Carcinoma Glioblastoma Lung cancer

Functional Areas Ras Pathway Zinc Finger Protein Development (Cleft Palate) Neurotransmitter Golgi Organisation Oncogenic Transformation Mitotic Cycle

Figure 3. Overview of data mining results. This figure shows the regulations between HDACs and distinct miRNAs including common (?) target genes. The functional areas and the clinical relevance indicate possible therapeutic fields for several cancers including lung and breast cancer as well as neuropsychiatric disorders such as depression.

? HDAC7

HDAC2

? mir140

mir433

HDAC6

mir183

mir1

HDAC4

HDAC4

mir206

mir34

HDAC1

HDAC1

miRNA

HDAC Member

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

HDACs and miRNAs

7

S. Swierczynski et al.

Direct regulation between HDAC – miRNA (10% of literature)

HDAC

Indirect regulation through gene – protein level (90% of literature)

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

miRNA Genes ↑↓ Proteins ↑↓

Figure 4. Distribution of direct and indirect regulation within the miRNA-HDAC axis. This illustration places special emphasis on indirect regulations between HDACs and miRNAs. Additionally, a few studies prove a direct interaction which demonstrate that both ways of HDAC-miRNA regulation are possible (see Figure 3 for details) [23,48-51]. 4.1

FUT8 pathway

FUT8 is attached with HDAC1 and HDAC2 which are imparted over the miR-34a which is described to be involved in p53 interaction [54-59]. PC2 pathway PC2 is closely connected with the miRNA cluster 17 -- 92, and this cluster is regulated by HDAC9. The silencing of HDAC9 in EC increased the expression of miRNA cluster 17 -- 92. Furthermore, the inhibition of miRNA cluster 17 -- 92 rescued the impairment of EC sprouting induced by HDAC9 silencing in culture and partly reversed the effect. MiRNA cluster 17 -- 92 is also the link between HDAC9 and BMPR2 [60-63]. 4.2

FGF2 pathway FGF2 is in a very central position in the cycle and is counter affected from the HDAC1/9 and also HDAC3. FGF2 also is regulated indirect over miRNA-15a, which is connected with HDAC3 [64]. 4.3

The analyzed pathways and their clinical relevance in detail 5.

a-(1,6)-fucosyltransferase 8 (FUT8) The FUT family is involved in cell-surface antigen synthesis during various biological processes such as tumor multi-drug resistance (MDR). Cheng et al. showed in hepatocellular carcinoma cell lines (HCC) that the members of the FUT family were highly expressed in MDR cell lines [65]. 5.1

8

The altered level of FUT8 was involved in a change of the drug resistance of the cells. The regulation of FUT8 had an impact on the phosphoinositide 3 kinase (PI3K)/Akt signaling pathway [65]. Tonetti et al. showed in hepatocarcinoma cell lines that forced expression of miR-122 and miR-34a is able to induce a decrease of FUT8 levels and also to affect core fucosylation of secreted proteins [66]. Fang et al. showed that overexpression of miR-198 in CRC cell lines decreased FUT8 levels as shown by immunofluorescence analysis and inhibited cell proliferation, migration and invasion [67]. Polycystin-2 pathway Polycystin-2 (PC2) is emerging as a major player in mechanotransduction and therefore relevant for tumor cellular invasion and metastasis during cancer progression. The protein expression of PC2 in colorectal cancer showed that its overexpression results in upregulation of the mTOR pathway and PC2 overexpression is connected with unfavorable pathological parameters, including invasiveness and mucinous phenotypes. Low expression of PC2 leads to an overall poor survival [68]. Dysregulated miRNA expression is observed in PC2, but whether miRNAs are directly involved in kidney cyst formation and growth is not known. Igarashi et al. show that kidney-specific inactivation of miR-17-92 in a mouse model of PC2 retards kidney cyst growth, improves renal function and prolongs survival [69]. As reviewed by Li et al., pharmacological inhibition of HDAC 6 and 5 activities reduces the progression of cyst formation and slows down the decline of kidney function in PC1-conditional knockout mice and PC2-knockout mice [70]. 5.2

Expert Opin. Biol. Ther. (2015) 15(5)

HDACs and miRNAs

A.

TP53 HDAC1 mir-34a

a-(1,6)fucosyltransferase 8

mir-449a FUT8

mir-3148

mir-34a

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

HDAC2

• Responsible for α1,6-fucosylation of N-glycans • Core fucosylation has been demonstrated to be essential for signaling of several growth factors and adhesion molecules, such as EGF, E-cadherins, TGF-β, and integrins • Activity and expression are increased in colorectal cancer and hepatocarcinoma cells

C.

B. HDAC8

BMPR2

mir-15a HDAC9

mir-3143

mir-17(-92 cluster)

HDAC 1/9

HDAC3

mir-3163

mir-16

PKD2

Polycystic kidney disease (polycystin 2) Fibroblast growth factor 2

• Presence of numerous fluid-filled cysts in the renal parenchyma • Cyst epithelial cells secrete excessive fluid • Monogenic human diseases • Proliferation

FGF2

• Proliferation • Wound healing • Differentiation

Figure 5. Detailed interaction map of HDAC and miRNAs. A. Regulation of FUT8: a-(1,6)-fucosyltransferase 8 is regulated by three miRNAs (mir-34a, mir-449a, mir-3148) which are connected with HDACs 1 and 2. HDAC1 and mir-34a are also involved in the feedback loop regulation with TP53 over p21 (Cip1/Waf1) for cell cycle control [54-59]. B. Regulation of PKD2/PC2: The cycle of PKD2/PC2 shows an involvement of the miRNA cluster 17 -- 92 which regulates HDAC9 and acts by repression of proangiogenic factors. The cluster is also involved in BMPR2 signaling which is connected to metastasis in breast and colorectal cancer [60-63]. C. Regulation of FGF2: The regulation of FGF2 is mainly performed by mir-16 and mir-15a which are in a feedback loop with HDAC3. HDAC 1 and HDAC9 play only secondary roles in FGF2 regulation through mir-3163 but may also be important as a therapeutic target [64].

5.3

Fibroblast growth facter-2 pathway

Fibroblast growth factor-2 (FGF-2) is an important trigger for EMT. In particular, FGF-2 triggers the motility of proximal tubular epithelium cells through the expression of vimentin (VIM) and a-smooth muscle actin (SMA), release of matrix metallopoteinases 9 and 2 and reduced expression of

cytokeratin and E-cadherin in proximal tubular epithelium. Intracellular signaling by FGF-2 is transported via the MAP kinase pathway, the PI3K/akt pathway and the phospholipase C g pathway. The PI3K/Akt pathway has been linked direct to FGF-2-induced EMT [71]. Bonci et al. showed that downregulation of miR-15 and miR-16 in cancer-associated

Expert Opin. Biol. Ther. (2015) 15(5)

9

S. Swierczynski et al.

Table 1. Overview of current clinical trials and possible miRNA targets, based on [84-92]. Clinical trial phase appr.

III

HDACi

HDAC classes or HDAC members

Vorinostat I, II(a,b), IV (SAHA) Romidepsin 1, 2, 4, 6 Panobinostat I, II(a,b), IV Valproic acid I, IIa

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

II

Belinostat Mocetinostat Abexinostat Entinostat SB939 Resminostat Givinostat Quisinostat

I, II(a,b), IV 1, 2, 3, 11 I, IIb 1, 2, 3, 9 Unknown Unknown I, II Unknown (Takai et al. 2010)

Related miRNAs miR-19a and miR-19b (Altucci et al. 2013)/miRNAs 21 and 17 -- 92 (Humpreys et al. 2013) n.a. miR-19a, miR-19b1, mir-30a (di Fazio et al. 2013)/miR-15a and miR-16 (Keating et al. 2012) miR-129 (Stamm et al. 2013)/miR-331 and miR-885-3p (Chuang et al. 2012)/ miR-16a, 18c, 122, 132, 457b, and 724 (Hahn et al. 2013) n.a. n.a. miR-21 (Kiguchi et al. 2012) miR-125a, miR-125b, and miR-205 (Liu et al. 2013) n.a. n.a. n.a. n.a.

appr.: FDA-approved; HDACi: HDAC inhibitors; n.a.: No data available.

fibroblasts in prostate tumors promoted tumor growth and progression through reduced post-transcriptional repression of FGF-2 and its receptor FGFr1, which act on both stromal and tumor cells to enhance cancer cell survival, proliferation and migration [72]. The three pathways FUT8, PC2 and FGF2 contribute together with the important stages of the hallmarks of cancer for drug resistance, invasiveness and transformation postulated by Weinberg and Hanahan [73].

Sungkwan An et al. reviewed that treatment of human lung epithelial cells with vorinostat significantly changes the miRNA expression in this cell type and evaluated the profile of several regulated miRNAs in detail [79]. Combination of HDACis with miRNA inhibition is not yet established in any clinical trial, and based on the data summarized in this review, this approach is highly interesting to establish a new therapies for diseases associated with specific deregulations of HDACs and miRNAs [79].

6. HDACis in clinical trials and therapeutic possibilities 7.

HDACis are used as anticancer agents that inhibit all functional processes of HDACs [74]. They have emerged as powerful new class of small molecule inhibitors for modulation the acetylation state of histone proteins and none-histone proteins. Over 490 clinical trials have been initiated in the last 10 years but only a few in Europe and the US are actually concerning possible interactions on miRNAs [75]. HDACis cause apoptosis, differentiation and cell growth arrest both in vitro as well as in vivo. The HDACis are divided in four chemical classes: hydroxamic acid derivatives, cyclic peptides, short-chain aliphatic acids and benzamides (summarized in Table 1) [76]. As an example for a current cell culture study, the HDACi vorinistat showed in the human colon cancer cell line (HCT116) with or without p53 changes in the miRNA expression of 144 miRNAs out of 275 target miRNAs. [77]. In colorectal cancer cells, the miRNAs 21 and 17 -- 92 clusters have oncogenic properties, and Humpreys et al. demonstrated that vorinostat resulted in a decrease in cell growth associated with altered expression of these particular miRNAs [78]. 10

Conclusion

HDACs and miRNA are interconnected, which indicates that epigenetic regulation and post-transcriptional regulation play an important role and influence each other in various ways during cancerogenesis. Additionally, HDACis are reported to influence the expression levels of a defined number of miRNAs. The feedback between miRNAs and HDACs often plays an important role in cancer progression and cancer inhibition. This knowledge could lead to specific cancer therapies, which would employ HDACis and simultaneously modify miRNAs. Many HDACs showed interactions with a variety of miRNAs in many different human malignancies, and in each case, the modulation of these led to significant changes in cancer progression. Silencing of miRNA by the hypermethylation of CpG islands of tumor-suppressor miRNAs contributes to cancerogenesis and methylation signatures of miRNAs are associated with metastasis. Therefore, it might make sense to evaluate these methylation signatures to detect new biomarkers for therapeutic applications in the future [11,22,80-82].

Expert Opin. Biol. Ther. (2015) 15(5)

HDACs and miRNAs

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

8.

Expert opinion

Biomarker determination should be the first step to tailor a personalized treatment for each and every patient (Illig et al. 2013). The observation of miRNA and HDAC interactions in cancer progression and drug resistance should be evaluated for combination treatments. There are an increasing number of clinical trials with HDACis in different tumor entities (e.g., lymphoma, pancreatic cancer) in Phases I to III, whereby possible miRNA crosstalk is currently analyzed only in a limited number. In these trials, miRNAs play only a secondary role in the evaluation of clinical parameters which are directly affected by HDACis. Interestingly, several in vitro studies showed a relevant interaction of HDACs and miRNAs involved in established cancer-associated pathways (e.g., TGF-b-signaling, APC/b-Catenin pathway, MYC pathway and cell-cycle signaling [p21]). This justifies studying these two possible partners in the context where they interact with each other during tumorigenesis. As already shown by our group (Kiesslich et al. 2014), combination of small molecule inhibitors of the hedgehog pathway and conventional chemotherapeutics could open new therapeutic windows in orphans and lethal cancer disease such as biliary tract cancer. We are aware that the non-histone modifications by HDACs could complicate this simplistic view of our hypothesis, see also Stintzing et al. 2011. In our opinion, the combination of silencing specific miRNAs in combination with approved HDACis could be a very promising approach in the field of epigenetic therapy. Furthermore, the mentioned HDAC and miRNA interaction partners could serve as additional biomarkers to stratify patients with particular chronic diseases and cancer types for optimal therapeutic outcome. For this approach, still more specific and also clinically approved HDACis are needed as well as better ways to directly target miRNAs. As discussed by Faroogi et al. 2014, specific targeting of miRNAs is a Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

.

2.

.

Ropero S, Esteller M. The role of histone deacetylases (HDACs) in human cancer. Mol Oncol 2007;1(1):19-25 A very good work which describes all relevant fields of histone deacetylase (HDAC) functions. Kiesslich T, Pichler M, Neureiter D. Epigenetic control of epithelial-mesenchymal-transition in human cancer. Mol Clin Oncol 2013;1(1):3-11 Shows the epigenetic regulative mechanisms of epithelial mesenchymal transition and cancer development.

complex task -- nevertheless, it was shown in vitro and in vivo (xenografts) to silence miR-122 and miR-21 with a transfer system of liposomses or via polyarginine-peptide nucleic acid (PNA).This is still one of the biggest limitations in realizing a combination therapy of HDACs and miRNAs in the clinical setting. The illustrated pathways (FUT8, PC2, FGF-2) should be analyzed in more detail both in vitro and in vivo to confirm our hypothesis. Evaluation of new miRNA sets in various cancer types could help to manage this goal a lot more quicker and the establishment of new HDACis or better said more HDAC class specific inhibitors could in the future help to refine some therapeutically hypothesis in the field of HDAC and miRNA science. For further investigations, it is of importance to rather treat the network of HDACs and miRNAs than a single HDAC. Which HDAC-miRNA network is necessary for which tumor has to be further evaluated as well as interactions of those with the hallmarks of cancer as reviewed by Weinberg and Hanahan in 2011. We hope that in the next approaches/trials there will be more attention given to the evaluation of miRNA sets with distinct focus on interactions with HDACs or HDACis. This would simplify many considerations in HDAC and miRNA science in the near future and would open doors to new and powerful therapeutic tools and therapies.

Declaration of interest The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

3.

Stasevich TJ, Hayashi-Takanaka Y, Sato Y, et al. Regulation of RNA polymerase II activation by histone acetylation in single living cells. Nature 2014;516(7530):272-5

4.

Sawa H, Murakami H, Ohshima Y, et al. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol 2001;18:109-14

5.

Wilting RH, Yanover E, Heideman MR, et al. Overlapping functions of Hdac1 and Hdac2 in cell cycle regulation and haematopoiesis. EMBO J 2010;29:2586-97

Expert Opin. Biol. Ther. (2015) 15(5)

6.

Brehm A, Miska EA, McCance DJ, et al. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature 1998;391:597-601

7.

Bernstein BE, Tong JK, Schreiber SL. Genomewide studies of histone deacetylase function in yeast. Proc Natl Acad Sci USA 2000;97:13708-13

8.

Medina V, Edmonds B, Young GP, et al. Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway. Cancer Res 1997;57:3697-707

11

S. Swierczynski et al.

9.

..

10.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

..

11.

Li Z, Zhu WG. Targeting histone deacetylases for cancer therapy: from molecular mechanisms to clinical implications. Int J Biol Sci 2014;10(7):757-70 Gives a very good overview of HDAC implementation in clinical therapies. Stintzing S, Kemmerling R, Kiesslich T, et al. Myelodysplastic syndrome and histone deacetylase inhibitors: “to be or not to be acetylated”? J Biomed Biotechnol 2011;2011:214143 This paper reviewed the functions of HDACs on non-histone proteins and their further modulations. Lujambio A, Calin GA, Villanueva A, et al. DNA methylation signature for human cancer metastasis. Proc Natl Acad Sci USA 2008;105(36):13556-61

12.

Ropero S, Fraga MF, Ballestar E, et al. A truncating mutation of HDAC2 in human cancers confers resistance to histone deacetylase inhibition. Nat Genet 2006;38(5):566-9

13.

Witt O, Deubzer HE, Milde T, Oehme I. HDAC family: what are the cancer relevant targets? Cancer Lett 2009;277(1):8-21 A very good review on all cancer relevant fields of HDAC interactions and the HDAC classes.

.

14.

15.

16.

17.

20.

21.

..

Barneda-Zahonero B, Parra M. Histone deacetylases and cancer. Mol Oncol 2012;6(6):579-89 Wendtner CM. Cocktail of eternity: HDAC meets miR. Blood 2012;119(5):1095-6 The relevant work from Wendtner, which helps us to form the hypothesis for this review.

22.

Chatterjee N, Wang WL, Conklin T, et al. Histone deacetylase inhibitors modulate miRNA and mRNA expression, block metaphase, and induce apoptosis in inflammatory breast cancer cells. Cancer Biol Ther 2013;14(7):658-71

23.

Noonan EJ, Place RF, Pookot D, et al. miR-449a targets HDAC-1 and induces growth arrest in prostate cancer Function of miR-449a in prostate cancer. Oncogene 2009;28:1714-24

24.

Zhao J, Lammers P, Torrance CJ, Bader AG. TP53-independent function of miR-34a via HDAC1 and p21(CIP1/ WAF1.). Mol Ther 2013;21(9):1678-86

31.

Winbanks CE, Beyer C, Hagg A, et al. miR-206 represses hypertrophy of myogenic cells but not muscle fibers via inhibition of HDAC4. PLoS One 2013;8(9):e73589

32.

Wang Z, Qin G, Zhao TC. HDAC4: mechanism of regulation and biological functions. Epigenomics 2014;6(1):139-50

33.

Li H, Xie H, Liu W, et al. A novel microRNA targeting HDAC5 regulates osteoblast differentiation in mice and contributes to primary osteoporosis in humans. J Clin Invest 2009;119(12):3666-77

34.

Simon D, Laloo B, Barillot M, et al. A mutation in the 3’-UTR of the HDAC6 gene abolishing the posttranscriptional regulation mediated by hsa-miR-433 is linked to a new form of dominant X-linked chondrodysplasia. Hum Mol Genet 2010;19(10):2015-27

35.

Huang S, Wang S, Bian C, et al. Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression. Stem Cells Dev. 2012;21(13):2531-40

25.

Millard CJ, Watson PJ, Celardo I, et al. Class I HDACs share a common mechanism of regulation by inositol phosphates. Mol Cell 2013;51(1):57-67

Nalls D, Tang SN, Rodova M, et al. Targeting epigenetic regulation of miR34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS ONE 2011;6(8):e24099

26.

36.

Kaluza D, Kroll J, Gesierich S, et al. Histone deacetylase 9 promotes angiogenesis by targeting the antiangiogenic microRNA-17-92 cluster in endothelial cells. Arterioscler Thromb Vasc Biol 2013;33(3):533-43

Lodrini M, Oehme I, Schroeder C, et al. MYCN and HDAC2 cooperate to repress miR-183 signaling in neuroblastoma. Nucleic Acids Res 2013;41(12):6018-33

Lwin T, Zhao X, Cheng F, et al. A microenvironment-mediated c-Myc/ miR-548m/HDAC6 amplification loop in non-Hodgkin B cell lymphomas. J Clin Invest 2013;123(11):4612-26

27.

Chen CQ, Chen CS, Chen JJ, et al. Histone deacetylases inhibitor trichostatin A increases the expression of Dleu2/miR15a/16-1 via HDAC3 in non-small cell lung cancer. Mol Cell Biochem 2013;383(1-2):137-48

37.

28.

Wang Y, Toh HC, Chow P, et al. MicroRNA-224 is up-regulated in hepatocellular carcinoma through epigenetic mechanisms. FASEB J. 2012;26(7):3032-41

Kai Y, Peng W, Ling W, et al. Reciprocal effects between microRNA-140-5p and ADAM10 suppress migration and invasion of human tongue cancer cells. Biochem Biophys Res Commun 2014;448(3):308-14

38.

Yu XF, Zou J, Bao ZJ, Dong J. miR-93 suppresses proliferation and colony formation of human colon cancer stem cells. World J Gastroenterol 2011;17(42):4711-17

39.

Sekiya T, Adachi S, Kohu K, et al. Identification of BMP and activin membrane-bound inhibitor (BAMBI), an inhibitor of transforming growth factorbeta signaling, as a target of the betacatenin pathway in colorectal tumor cells. J Biol Chem 2004;279(8):6840-6

Morris MJ, Monteggia LM. Unique functional roles for class I and class II histone deacetylases in central nervous system development and function. Int J Dev Neurosci 2013;31(6):370-81 Yuan H, Su L, Chen WY. The emerging and diverse roles of sirtuins in cancer: a clinical perspective. Onco Targets Ther 2013;6:1399-416

18.

de Oliveira MV, Andrade JM, Paraı´so AF, Santos SH. Sirtuins and cancer: new insights and cell signaling. Cancer Invest 2013;31(10):645-53

19.

Deubzer HE, Schier MC, Oehme I, et al. HDAC11 is a novel drug target in

12

29 to control myogenic differentiation through regulation of HDAC4. J Biol Chem 2011;286(16):13805-14

carcinomas. Int J Cancer 2013;132(9):2200-8

29.

30.

Sandhu SK, Volinia S, Costinean S, et al. miR-155 targets histone deacetylase 4 (HDAC4) and impairs transcriptional activity of B-cell lymphoma 6 (BCL6) in the Eµ-miR-155 transgenic mouse model. Proc Natl Acad Sci USA 2012;109(49):20047-52 Winbanks CE, Wang B, Beyer C, et al. TGF-beta regulates miR-206 and miR-

Expert Opin. Biol. Ther. (2015) 15(5)

HDACs and miRNAs

40.

41.

42.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

43.

44.

45.

46.

.

47.

..

48.

49.

50.

Elyada E, Pribluda A, Goldstein RE, et al. CKIa ablation highlights a critical role for p53 in invasiveness control. Nature 2011;470(7334):409-13 Konsavage WM Jr, Yochum GS. The myc 3’ wnt-responsive element suppresses colonic tumorigenesis. Mol Cell Biol 2014;34(9):1659-69 Piard F, Martin L, Chapusot C, et al. [Genetic pathways in colorectal cancer: interest for the pathologist]. Ann Pathol 2002;22(4):277-88 Sierra J, Yoshida T, Joazeiro CA, Jones KA. The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev 2006;20(5):586-600 Kaluza D, Kroll J, Gesierich S, et al. Histone deacetylase 9 promotes angiogenesis by targeting the antiangiogenic microRNA-17-92 cluster in endothelial cells. Arterioscler Thromb Vasc Biol 2013;33(3):533-43 Lin L, Hou J, Ma F, et al. Type I IFN inhibits innate IL-10 production in macrophages through histone deacetylase 11 by downregulating microRNA-145. J Immunol 2013;191(7):3896-904 Shi B, Xu W. The development and potential clinical utility of biomarkers for HDAC inhibitors. Drug Discov Ther 2013;7(4):129-36 Relevant work for the importance of HDACs as biomarkers in clinical routine. New M, Olzscha H, La Thangue NB. HDAC. inhibitor-based therapies: can we interpret the code? Mol Oncol 2012;6(6):637-56 Review about the main used HDAC inhibitors and their function. Saccone V, Consalvi S, Giordani L, et al. HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles. Genes Dev 2014;28(8):841-57 Cheng TL, Wang Z, Liao Q, et al. MeCP2 suppresses nuclear microRNA processing and dendritic growth by regulating the DGCR8/ Drosha complex. Dev Cell 2014;28(5):547-60 Pushpavalli SN, Ramaiah MJ, Lavanya A, et al. Imidazo-benzothiazoles a potent microRNA modulator involved

in cell proliferation. Bioorg Med Chem Lett 2012;22(20):6418-24

62.

Sato Y. Is histone deacetylase-9MicroRNA-17~92 cluster a novel axis for angiogenesis regulation? Arterioscler Thromb Vasc Biol 2013;33(3):445-6

51.

Hughes DP. How the NOTCH pathway contributes to the ability of osteosarcoma cells to metastasize. Cancer Treat Res 2009;152:479-96

63.

52.

Delcuve GP, Khan DH, Davie JR. Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Epigenetics 2012;4(1):5

Patel V, Williams D, Hajarnis S, et al. miR-17~92 miRNA cluster promotes kidney cyst growth in polycystic kidney disease. Proc Natl Acad Sci USA 2013;110(26):10765-70

64.

53.

Ververis K, Hiong A, Karagiannis TC, Licciardi PV. Histone deacetylase inhibitors (HDACIs): multitargeted anticancer agents. Biologics 2013;7:47-60

Musumeci M, Coppola V, Addario A, et al. Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene 2011;30(41):4231-42

54.

Muinelo-Romay L, Villar-Portela S, Cuevas Alvarez E, et al. a(1,6) Fucosyltransferase expression is an independent prognostic factor for diseasefree survival in colorectal carcinoma. Hum Pathol 2011;42(11):1740-50

65.

Cheng L, Luo S, Jin C, et al. FUT family mediates the multidrug resistance of human hepatocellular carcinoma via the PI3K/Akt signaling pathway. Cell Death Dis 2013;4:e923

66.

55.

Bernardi C, Soffientini U, Piacente F, Tonetti MG. Effects of microRNAs on fucosyltransferase 8 (FUT8) expression in hepatocarcinoma cells. PLoS One 2013;8(10):e76540

Bernardi C, Soffientini U, Piacente F, Tonetti MG. Effects of microRNAs on fucosyltransferase 8 (FUT8) expression in hepatocarcinoma cells. PLoS ONE 2013;8(10):e76540

67.

Wang M, Wang J, Kong X, et al. MiR-198 represses tumor growth and metastasis in colorectal cancer by targeting fucosyl transferase 8. Sci Rep 2014;4:6145

68.

Gargalionis AN, Korkolopoulou P, Farmaki E, et al. Polycystin-1 and polycystin-2 are involved in the acquisition of aggressive phenotypes in colorectal cancer. Int J Cancer 2015;136(7):1515-27

56.

Mizuno S, Yasuo M, Bogaard HJ, et al. Inhibition of histone deacetylase causes emphysema. Am J Physiol Lung Cell Mol Physiol 2011;300(3):L402-13

57.

Ji Q, Hao X, Zhang M, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS ONE 2009;4(8):e6816

58.

Zhao J, Lammers P, Torrance CJ, Bader AG. TP53-independent function of miR-34a via HDAC1 and p21(CIP1/ WAF1.). Mol Ther 2013;21(9):1678-86

69.

Zhang HS, Chen XY, Wu TC, et al. MiR-34a is involved in Tat-induced HIV-1 long terminal repeat (LTR) transactivation through the SIRT1/NFjB pathway. FEBS Lett 2012;586(23):4203-7

Patel V, Williams D, Hajarnis S, et al. miR-17~92 miRNA cluster promotes kidney cyst growth in polycystic kidney disease. Proc Natl Acad Sci USA 2013;110(26):10765-70

70.

Li X. Epigenetics and autosomal dominant polycystic kidney disease. Biochim Biophys Acta 2011;1812(10):1213-18

71.

Masola V, Gambaro G, Tibaldi E, et al. Heparanase and syndecan-1 interplay orchestrates fibroblast growth factor-2-induced epithelial-mesenchymal transition in renal tubular cells. J Biol Chem 2012;287(2):1478-88

72.

Musumeci M, Coppola V, Addario A, et al. Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene 2011;30(41):4231-42

59.

60.

61.

Mogilyansky E, Rigoutsos I. The miR17/92 cluster: a comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ 2013;20(12):1603-14 Luo T, Cui S, Bian C, Yu X. Crosstalk between TGF-b/Smad3 and BMP/ BMPR2 signaling pathways via miR-17-92 cluster in carotid artery restenosis. Mol Cell Biochem 2014;389(1-2):169-76

Expert Opin. Biol. Ther. (2015) 15(5)

13

S. Swierczynski et al.

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646-74

74.

Borbone E, De Rosa M, Siciliano D, et al. 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(6):E1031-40

82.

Matthias P. Too much or too little, how much HDAC activity is good for you? Blood 2013;121(11):1930-1

83.

Lemoine M, Younes A. Histone deacetylase inhibitors in the treatment of lymphoma. Discov Med 2010;10(54):462-70

Gryder BE, Sodji QH, Oyelere AK. Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Future Med Chem 2012;4(4):505-24

84.

75.

Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by University of Connecticut on 04/02/15 For personal use only.

microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene 2011;30(41):4231-42

73.

76.

77.

78.

79.

80.

81.

14

Shi B, Xu W. The development and potential clinical utility of biomarkers for HDAC inhibitors. Drug Discov Ther 2013;7(4):129-36 Shin S, Lee EM, Cha HJ, et al. MicroRNAs that respond to histone deacetylase inhibitor SAHA and p53 in HCT116 human colon carcinoma cells. Int J Oncol 2009;35(6):1343-52 Humphreys KJ, Cobiac L, Le Leu RK, et al. Histone deacetylase inhibition in colorectal cancer cells reveals competing roles for members of the oncogenic miR-17-92 cluster. Mol Carcinog 2013;52(6):459-74 Lee EM, Shin S, Cha HJ, 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(1):45-50 New M, Olzscha H, La Thangue NB. HDAC inhibitor-based therapies: can we interpret the code? Mol Oncol 2012;6(6):637-56 Musumeci M, Coppola V, Addario A, et al. Control of tumor and

85.

Lepore I, Dell’Aversana C, Pilyugin M, 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(12):e83018 Henrici A, Montalbano R, Neureiter D, et al. The pan-deacetylase inhibitor panobinostat suppresses the expression of oncogenic miRNAs in hepatocellular carcinoma cell lines. Mol Carcinog 2013. [Epub ahead of print]

86.

Sampath D, Liu C, Vasan K, et al. Histone deacetylases mediate the silencing of miR-15a, miR-16, and miR29b in chronic lymphocytic leukemia. Blood 2012;119(5):1162-72

87.

Zhang Z, Convertini P, Shen M, et al. Valproic acid causes proteasomal degradation of DICER and influences miRNA expression. PLoS ONE 2013;8(12):e82895

88.

Hunsberger JG, Fessler EB, Wang Z, et al. Post-insult valproic acid-regulated microRNAs: potential targets for cerebral ischemia. Am J Transl Res 2012;4(3):316-32

89.

Aluru N, Deak KL, Jenny MJ, Hahn ME. Developmental exposure to valproic acid alters the expression of microRNAs involved in

Expert Opin. Biol. Ther. (2015) 15(5)

neurodevelopment in zebrafish. Neurotoxicol Teratol 2013;40:46-58 90.

Kitamura T, Connolly K, Ruffino L, et al. The therapeutic effect of histone deacetylase inhibitor PCI-24781 on gallbladder carcinoma in BK5. erbB2 mice. J Hepatol 2012;57(1):84-91

91.

Wang S, Huang J, Lyu H, et al. Functional cooperation of miR-125a, miR-125b, and miR-205 in entinostatinduced downregulation of erbB2/ erbB3 and apoptosis in breast cancer cells. Cell Death Dis 2013;4:e556

92.

Takai N, Narahara H. Preclinical studies of chemotherapy using histone deacetylase inhibitors in endometrial cancer. Obstet Gynecol Int 2010;2010:923824

Affiliation Stefan Swierczynski1,2, Eckhard Klieser2, Romana Illig2, Beate Alinger-Scharinger2, Tobias Kiesslich3,4 & Daniel Neureiter†2 † Author for correspondence 1 Paracelsus Medical University, Salzburger Landeskliniken, Department of Surgery, Salzburg, Austria 2 Paracelsus Medical University, Salzburger Landeskliniken, Institute of Pathology, Salzburg, Austria Tel: +0043 0662 4482 4737; Fax: +0043 0662 4482 882; E-mail: [email protected] 3 Paracelsus Medical University, Institute of Physiology and Pathophysiology, Salzburg, Austria 4 Paracelsus Medical University, Salzburger Landeskliniken, Department of Internal Medicine I, Salzburg, Austria

Histone deacetylation meets miRNA: epigenetics and post-transcriptional regulation in cancer and chronic diseases.

Epigenetic regulation via DNA methylation, histone acetylation, as well as by microRNAs (miRNAs) is currently in the scientific focus due to its role ...
688KB Sizes 1 Downloads 7 Views