computer-aided molecular design of compounds …csbj.org/articles/e2015031.pdfmini review...

8
Mini Review Computer-aided Molecular Design of Compounds Targeting Histone Modifying Enzymes Federico Andreoli a , Alberto Del Rio a,b, a Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italy b Institute of Organic Synthesis and Photoreactivity, National Research Council, Via P. Gobetti, 101 40129 Bologna, Italy abstract article info Article history: Received 13 October 2014 Received in revised form 24 April 2015 Accepted 30 April 2015 Available online 7 May 2015 Keywords: Epigenetics Post-translational modications Histone Computer-aided molecular design Drug design Drug discovery Growing evidences show that epigenetic mechanisms play crucial roles in the genesis and progression of many physiopathological processes. As a result, research in epigenetic grew at a fast pace in the last decade. In partic- ular, the study of histone post-translational modications encountered an extraordinary progression and many modications have been characterized and associated to fundamental biological processes and pathological con- ditions. Histone modications are the catalytic result of a large set of enzyme families that operate covalent mod- ications on specic residues at the histone tails. Taken together, these modications elicit a complex and concerted processing that greatly contribute to the chromatin remodeling and may drive different pathological conditions, especially cancer. For this reason, several epigenetic targets are currently under validation for drug discovery purposes and different academic and industrial programs have been already launched to produce the rst pre-clinical and clinical outcomes. In this scenario, computer-aided molecular design techniques are of- fering important tools, mainly as a consequence of the increasing structural information available for these tar- gets. In this mini-review we will briey discuss the most common types of known histone modications and the corresponding operating enzymes by emphasizing the computer-aided molecular design approaches that can be of use to speed-up the efforts to generate new pharmaceutically relevant compounds. © 2015 Andreoli, Del Rio. Published by Elsevier B.V. on behalf of the Research Network of Computational and Structural Biotechnology. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358 2. Type of Histone Modications and Their Biological and Clinical Relevance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359 3. Chemical Mechanisms of Histone Modications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360 4. Molecular Design Techniques Applied to Histone Modifying Enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361 5. Summary and Outlook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363 Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363 1. Introduction In the last decades, major efforts have been done in genetics though it is evident that unraveling complex biological mechanisms like cancer requires a larger framework of research that recently evolved toward the better understanding of the immune system regulation [1], the re- discovery of metabolism [2] and, importantly, the role of epigenetics [3]. The term epigenetics refers to the mechanisms of temporal and spa- tial control of gene activity that do not entail modication of the DNA sequence but inuence the physiological and pathological development of an organism. The molecular mechanisms by which epigenetic chang- es occur are complex and cover a wide range of processes [4]. Epigenetic mechanisms can occur at biochemical level in three different ways: Computational and Structural Biotechnology Journal 13 (2015) 358365 Corresponding author at: Institute of Organic Synthesis and Photoreactivity, National Research Council, Via P. Gobetti, 101 40129 Bologna, Italy. Tel.: +39 051 6398308; fax: +39 051 6398349. E-mail addresses: [email protected], [email protected] (A. Del Rio). http://dx.doi.org/10.1016/j.csbj.2015.04.007 2001-0370/© 2015 Andreoli, Del Rio. Published by Elsevier B.V. on behalf of the Research Network of Computational and Structural Biotechnology. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Contents lists available at ScienceDirect journal homepage: www.elsevier.com/locate/csbj

Upload: lybao

Post on 03-Apr-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

Computational and Structural Biotechnology Journal 13 (2015) 358–365

Contents lists available at ScienceDirect

journa l homepage: www.e lsev ie r .com/ locate /csb j

Mini Review

Computer-aided Molecular Design of Compounds Targeting HistoneModifying Enzymes

Federico Andreoli a, Alberto Del Rio a,b,⁎a Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Via S. Giacomo 14, 40126 Bologna, Italyb Institute of Organic Synthesis and Photoreactivity, National Research Council, Via P. Gobetti, 101 40129 Bologna, Italy

⁎ Corresponding author at: Institute of Organic SynthesResearch Council, Via P. Gobetti, 101 40129 Bologna,fax: +39 051 6398349.

E-mail addresses: [email protected], alberto.de

http://dx.doi.org/10.1016/j.csbj.2015.04.0072001-0370/© 2015 Andreoli, Del Rio. Published by ElsevieCC BY license (http://creativecommons.org/licenses/by/4.

a b s t r a c t

a r t i c l e i n f o

Article history:Received 13 October 2014Received in revised form 24 April 2015Accepted 30 April 2015Available online 7 May 2015

Keywords:EpigeneticsPost-translational modificationsHistoneComputer-aided molecular designDrug designDrug discovery

Growing evidences show that epigenetic mechanisms play crucial roles in the genesis and progression of manyphysiopathological processes. As a result, research in epigenetic grew at a fast pace in the last decade. In partic-ular, the study of histone post-translational modifications encountered an extraordinary progression and manymodifications have been characterized and associated to fundamental biological processes and pathological con-ditions. Histonemodifications are the catalytic result of a large set of enzyme families that operate covalent mod-ifications on specific residues at the histone tails. Taken together, these modifications elicit a complex andconcerted processing that greatly contribute to the chromatin remodeling and may drive different pathologicalconditions, especially cancer. For this reason, several epigenetic targets are currently under validation for drugdiscovery purposes and different academic and industrial programs have been already launched to producethe first pre-clinical and clinical outcomes. In this scenario, computer-aided molecular design techniques are of-fering important tools, mainly as a consequence of the increasing structural information available for these tar-gets. In this mini-review we will briefly discuss the most common types of known histone modifications andthe corresponding operating enzymes by emphasizing the computer-aided molecular design approaches thatcan be of use to speed-up the efforts to generate new pharmaceutically relevant compounds.© 2015 Andreoli, Del Rio. Published by Elsevier B.V. on behalf of the Research Network of Computational and

Structural Biotechnology. This is an open access article under the CC BY license(http://creativecommons.org/licenses/by/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3582. Type of Histone Modifications and Their Biological and Clinical Relevance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3593. Chemical Mechanisms of Histone Modifications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3604. Molecular Design Techniques Applied to Histone Modifying Enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3615. Summary and Outlook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363

1. Introduction

In the last decades, major efforts have been done in genetics thoughit is evident that unraveling complex biological mechanisms like cancer

is and Photoreactivity, NationalItaly. Tel.: +39 051 6398308;

[email protected] (A. Del Rio).

r B.V. on behalf of the Research Netw0/).

requires a larger framework of research that recently evolved towardthe better understanding of the immune system regulation [1], the re-discovery of metabolism [2] and, importantly, the role of epigenetics[3]. The term epigenetics refers to themechanisms of temporal and spa-tial control of gene activity that do not entail modification of the DNAsequence but influence the physiological and pathological developmentof an organism. Themolecularmechanisms bywhich epigenetic chang-es occur are complex and cover awide range of processes [4]. Epigeneticmechanisms can occur at biochemical level in three different ways:

ork of Computational and Structural Biotechnology. This is an open access article under the

359F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

i) through histone post-translational modifications (PTMs), which willbe the object of this mini-review, as well as the molecular recognitionof non-catalytic readers of histones [5], ii) through the DNA methyla-tion, i.e. the methylation of cytosines to 5-methylcytosines, which isthe object of recent reviews [6–8], and iii) through regulation of geneexpression by non-coding RNA (ncRNA), which is also an emergingtopic of research, covered by recent reviews [9–11]. All these processescontribute to define the epigenetic mechanisms by which gene expres-sion is activated or silenced [12–16].

Post-translational modifications of histones occur at the N-terminaltails of the protein chains and consist in covalent modifications thatare catalyzed by different classes of enzymes [17,18]. The ensemble ofthesemodifications is commonly referred as to be the histone code refer-ring to the idea that all histone PTMs determine the activity state of anunderlying gene [19]. One of the hallmarks of the histone code is thatit can be positively or negatively correlatedwith specific transcriptionalstates or organization of chromatin [20–23]. This is accomplishedthrough a fine regulation of histone PTMs controlled by an enzymaticmachinery, which existence and function have been elucidated partly,but with an extraordinary progression in the last years [23–29]. Impor-tantly, further understanding of epigenetic phenomena occurring onhistone proteins is critical to shed light on biological processes that areprogressively translating into the development of new medical options[29–31]. In this direction, different studies have highlighted how thehistone alterations contribute to the onset and growth of a variety ofcancers [7,23,24,27,32–41], amongother pathologies. Consequently, en-zymes operating PTMs on histones are constituting attractive therapeu-tic targets for the development of new therapies [13,31,42–44]. Itshould be noted that, while the resulting effects on chromatin collec-tively depend on the ensemble of histone PTMs, these are operated byprecise variations of physicochemical properties that we recentlyreviewed [17]. For these reasons, large efforts from both academic andindustrial settings have been dedicated in the last year to identify andevaluate new biologically active compounds against histone modifyingenzymes. Fuelled by the increasing availability of structural information,several endeavors have been initiated and helped by the usage ofcomputer-aided molecular design techniques. Thus, in this mini-review, we aim to highlight the aspects relating histone modificationsin the light of the future applications of computational techniques tothe research of new probe or lead-like epigenetic modulators.

2. Type of Histone Modifications and Their Biological andClinical Relevance

To understand the relevance of computational techniques inhistone-related epigenetic targets, it is important to highlightthat these post-translational modifications are functionalizations/defunctionalizations of specific residues, which are lysine, arginine, ser-ine, threonine, histidine, tyrosine, cysteine and glutamic acid, located atthe N-terminal tails of each chain. Fig. 1 summarizes all the most com-mon PTMs that can occur on histones. By far, lysine represents the res-idues with most chemical versatility, as it is capable to undergo severalkinds and grades of modifications. Consequently, histone methyltrans-ferases, demethylases, acetyltransferases and deacetylases have beenrecently ascribed an important role as new classes of biological targetsfor drug discovery [18,45–49]. Arginine represents also a residue thatis modified by enzymes recognized for drug development, in particularhistone methyltransferases. Differently to these previous cases, en-zymes thatmodify histone serines, threonines, histidines, tyrosines, cys-teines and glutamic acids have not been exploited yet for the discoveryof newmodulating compounds. Nevertheless, it is expected that furtherelucidation of their biological role and protein structure will spur suchendeavors. It is worth to note that other kinds of modifications likepropionylation, butyrylation, crotonylation, 2-hydroxyisobutyrylationhave been reported [50].

Different studies elucidate the impact that PTMs have on chromatinand their relevance in human physiology and pathology [16,18,25,26,31,51–57]. Interestingly, their biological role greatly varies, dependingon the kind of modification. Therefore, for instance, the acetylation ap-pears to be the most promiscuous histone modification and is alwaysassociated to transcriptional activity. Conversely, histone methylationhas a high degree of selectivity toward specific histone residues andcan be associated with both repression and transcription [58,59]. In ad-dition PTMs can “cross-talk”, meaning that modifications can occur in aconcerted or a subsequent manner [25,60–63].

In the last decade, epigenetic modifications of histones have beenmainly studied in the context of cancer, particularly histone deacetylasesof classes I, II and IV (HDACs) [64]. Indeed, abnormal activity of the en-zymes responsible for deacetylation of histones, modification that altersthe chromatin structure repressing transcription, has been shown to beimplicated in several diseases [18,65]. Because of these compelling evi-dences, HDACs have been recognized as consolidated drug target, in par-ticular for breast cancer, colorectal cancer, leukemia, lymphoma, ovarianand prostate cancer. In addition, another class of histone deacetylasesnamed sirtuins (or class III deacetylases), which uses NAD+ to catalyzethe removal of an acetyl group, also came into the light as new therapeutictargets [66,67]. This family of enzymes, in fact,was found to be involved inrelevant physiological and pathological processes, as well as in aging-related disorders, metabolic and inflammatory conditions and processesinvolving DNA regulation and integrity, including cancer [68–71]. Inter-estingly, also the correspondent families of enzymes that revert the cata-lytic activity of histone deacetylation, i.e. acetyltransferases (HAT), havealso met a great deal of interest. Two classes of HAT exist and consist ofenzymes able to acetylatemultiple sites in the histone tails and additionalsites on the globular histone core, for the first class, while the second classmostly consists of cytoplasmic enzymes able to acetylate newly synthe-sized histones prior to their deposition into chromatin. Abnormal regula-tion of HATs has been linked to leukemia and several studies connectingthem to prostate and gastric cancers have been realized [72,73].

The secondmost studied histone modification is methylation. As an-ticipated above, protein methyltransferases (PMT) emerged recently asnew important targets for cancer therapy, since they were found to beoverexpressed or repressed in several types of cancer, precisely in breastcancer, leukemia,myeloma, ovarian, prostate and kidney cancers. Sever-al recently published reviews describe mechanism and biological rolesof PMTs [6,41,45,46,74–81]. Indeed, due to their importance in differentpathological conditions, several drug discovery programs have beenlaunched in order to design specific compounds able to modulatethese targets [46]. Equally, the recent focus in understanding histonemethylation led to the characterization of histone demethylases(HDM). The first described protein has been the lysine specificdemethylase 1 (LSD1) [82]. Soon after, a new class of proteins havingdemethylase activity, the JMJC (Jumonji C) domain family, was discov-ered and characterized [83]. LSD and JMJC demethylases have been re-ported to be regulators of various cellular processes. Therefore, aspecial effort, as in the case of PMTs, is currently made, aimed to the dis-covery of small-molecule inhibitors with therapeutic potential [47,83].

Beside the above classes of enzymes, there is a mounting evidencethat also other type of modifications can constitute important paradigmin epigenetics and may underlie new biological target for therapeuticpurposes. For instance, some studies highlighted specific roles ofubiquitination [84–87], poly-ADP-ribosylation [88–96] and glycosyla-tion [61,97–101] to the epigenetic code. However, the therapeutic po-tential of these modifications still needs to be validated for thetherapeutic and drug discovery point of view. Equally, other modifica-tions like histone phosphorylation, citrullination (deamination) [102],biotinylation [103,104], tail clipping and proline isomerization [25],are still poorly understood and their role in human pathologies remainslargely unclear [25]. In the next paragraph,we aim to describe the state-of-the-art of these modifications, analyzing, from a chemical point ofview, their role on the dynamics of histone proteins.

Fig. 1.Most common type of post-translational modifications occurring on histone proteins.

360 F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

3. Chemical Mechanisms of Histone Modifications

The histone code collectively depends on the ensemble of post-translational modifications that is operated at the chromatin level, bysingle variations of physicochemical properties of the modified resi-dues. Consequently, the microenvironment and the biochemical differ-ences obtained by PTMs depend on the attachment or removal ofchemical functional groups, whose enzymes, mechanisms of actionand cofactors are overviewed in Table 1. We recently proposed that,from a chemical functionality point of view, histone PTMs can be divid-ed in two main groups [17]. The first group I encompassing PTMs lead-ing to the addition or the removal of monofunctional, generally small,organic substituents and a second group including polyfunctional, andin some case elaborate and large, organic molecules. Both groups arecomposed of writer or eraser enzymes, i.e. which add or remove fromhistone residues specific substituents.

Modifications of the first group are acetylation, methylation, phos-phorylation, deimination and palmitoylation (acylation). As seenabove, acetylation is the most common and studied PTM and consistsin the addition of an acetyl group on a lysine residue mediated by twomajor classes of histone acetyltransferases (HAT), Type-A (which

includes GNAT, p300/CBP, and MYST) and Type-B [18,65,72]. This func-tion can be removed by two categories of different catalytic activity en-zymes: classes I, II and IV histone deacetylases (HDAC) and class IIIdeacetylases, also known as sirtuins (SIRT) that work with a NAD+-dependent mechanism [105].

Methylation is the second most common PTM and lysine, as seenabove, is the residue that undergoes the widest number of methylationreactions. Indeed, lysine can bemono-, di-, or tri-methylated by histonemethyltransferases (HMT, also known as PKMT). HMTs can also mono-or di-methylate arginines and this class of enzymes is commonly re-ferred to as protein arginine methyltransferases (PRMT) [106]. Methylgroups can be erased only from lysines through the action of histonedemethylases (HDM or, in the case of lysines, KDM). Phosphorylationof histone has been observed on serine, threonine [107], tyrosine[107] and histidine [108] sites while phosphatases, which hydrolyzethe phosphoric monoesters or monoamides restoring the original resi-dues, represent the erasers of this histonemodification. Deimination oc-curs on arginines and methylarginines through the catalysis ofpeptidylarginine deiminases (PAD) [109].

Modifications of the second group are ubiquitylation, SUMOylation,biotinylation, glycosylation and ADP-ribosylation. While, in principle,

Table 1Mechanisms of histone modifications.

Histone modification Cofactor Leaving group Target residue Substituents Operating enzyme Type of enzyme

Acetylation Acetyl-CoA CoA Lys HAT WriterDeacetylation Zn2+, Fe2+, Co2+, Mn2+ CH3COO− Ac-Lys HDAC Eraser

NAD+ OAADPr Ac-Lys SIRT EraserPalmitoylation – – Cys – Writer

Methylation SAM SAH Arg, Lys PMT WriterDemethylation FAD HCHO Me-Lys LSD Eraser

Me2-LysFe2+, α-KG HCHO Me-Lys PHF8 Eraser

Me2-Lys JmjDMe3-Lys JHDM

Citrullination/deimination Ca2+ NH3 Arg PAD WriterMe-Arg

Phosphorylation ATP ADP Ser Kinase WriterThrTyrHis

Dephosphorylation – – Phos-Ser PhosphatasePhos-Thr

ADP-ribosylation NAD+ NAM Lys ADP-ribosyltransferase, SIRT4, SIRT6 WriterGlu

Glycosylation (O-GlcNAcylation) UDP-GlcNAc UDP Ser OGT WriterThr

Biotinylation – – Lys Biotin-protein ligase Writer

Ubiquitination ATP – Lys E1, E2, E3 proteins WriterDeubiquitination – – Ub-Lys Histone H2A deubiquitinase Eraser

361F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

these modifications are catalyzed by writers and eraser enzymes, itis worth to note that decitrullination, depalmitoylation, deADP-ribosylation, debiotinylation and deglycosylation have not been de-scribed, yet. Mono- or poly-ADP-ribosylation is catalyzed by ADP-ribosyltransferases (ART), clostridia-toxin-like (ARTC) or diphtheriatoxin-like (ARTD), which are most commonly known as PARP, andsome sirtuin isoforms (Table 1) [88,110,111]. Biotinylation and glyco-sylation are still poorly characterized and have been linked to the his-tone code recently [97,99,112,113]. Ubiquitylation takes place by theaction of E1, E2, and E3 enzymes,which catalyze the addition of anubiq-uitin (Ub) molecule to the target lysine via an isopeptide bond. The re-versal of this action is operated by deubiquitylating enzymes (DUB), sofar demonstrated to act only on histones H2A and H2B. SUMO (SmallUbiquitin-like MOdifier) proteins can be added on lysine residues in asimilar way.

4. Molecular Design Techniques Applied to HistoneModifying Enzymes

Computer-aided molecular techniques are widely used in academiaand industrial settings to assist the selection of new compounds that canmodulate biological targets. Several examples testify their successful ap-plications in the development of new chemical entities [114–116] and awide range of disciplines, including chemoinformatics, computationalchemistry, structural biology, biophysics, medicinal chemistry, organicchemistry and pharmacology, is at disposal of scientists working inthese fields. When applied to the discovery of compounds supposed tobecome future drugs, these techniques are commonly referred ascomputer-aided drug design (CADD) techniques. Certainly, amongthem, virtual screening acquired the greatest popularity due to its abil-ity to screen rapidly and cost-effectively large libraries of chemical com-pounds [117–119]. CADD techniques can be divided in two categories:ligand-based and structure-based drug design techniques (LBDD andSBDD), even if this classification is becoming nowadays loose, as severaltechniques offer technical advantages that are proper of both categories.

The first category usually takes advantage of the information of knownbioactive compounds (ligands), while the second usually exploit three-dimensional structure of the biological target (protein), in order to iden-tify new small-moleculemodulators of the protein activity. The growingavailability of protein structures resolved by X-ray crystallography orNMR technique has progressively raised the possibility to deploySBDD.However, ligand-based techniques still constitute useful tools, es-pecially when the structural information of a biological target is missingor when the molecular design effort is not directed toward a target-centric approach. Studies aimed at the modulation of cellular pathwaysor specific phenotypic traits without a precise knowledge of the mech-anism of action are representative of this case.

In the context of epigenetics, the research oriented toward the de-velopment of new therapeutically-relevant molecules has flourishedin the last years. Several works report rationales, targets, drugs, ap-proaches, compounds, tools and methodologies [32,120]. In addition,different reviews reporting the approaches for the discovery of newepidrugs or chemical probes for epigenetic targets have beenwritten re-cently [18,56,57,121,122]. An intriguing aspect in this field of research isthe fast pace aimed to extend the discovery of new compounds toepitargets that are currently not validated and that may offer new per-spective for the generation of new therapeutic agents. In this direction,particular aspect of epigenetics, for instance, the modulation of themicroRNA biogenesis pathway, have been recognized as new possibleway to achieve therapeutic target for human disease, in particular can-cer [9,123–125].

In all this framework of research, computational techniques areplaying a progressive role to help the identification of new molecularentities for epigenetic targets. Different techniques have been used toidentify new modulating compounds and to explain, mechanism of ac-tions, binding modes and protein dynamics [22,80,126–129]. Most ofthe research in this direction has beendone on classes I, II and IV histonedeacetylases. Indeed, a variety of quantitative structure–activity rela-tionship (QSAR) studies and computational works, elucidating retro-spectively protein dynamics, binding modes, binding affinities and

362 F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

selectivity among HDAC isoforms, have been published [130–139]. Forinstance, a recent and prospective work has been done to identify newinhibitors of SchistosomamansoniHDAC8bymeans of homologymodel-ing, molecular dynamics and molecular docking techniques [140]. It isinteresting to note that most of the prospective works have been ap-plied on other epigenetic targets than HDACs, reflecting the idea thatcomputational techniques are useful tool to help the identification ofnew biologically active compounds for investigational epigenetic tar-gets. In this direction two molecular docking studies have been appliedonHAT p300, resulting on the identification of two classes of new inhib-itors: benzothiazines and pyrazolone exomethylene vinyl compounds(1 and2, Fig. 2) [141,142]. Prospective high-throughput docking screen-ing and computational studies with molecular dynamic simulationswere performed on LSD1, leading to the identification of nanomolarN′-(1-phenylethylidene)-benzohydrazides (3, Fig. 2) and novel classesof short peptide inhibitors [143–146]. LSD1was also object of retrospec-tive studies based on virtual screening procedures, aiming at elucidatingligand selectivity with a closely related target, theMAO-B [147] and ex-tended molecular dynamic simulations were used to highlight new po-tential binding regions for drug-like molecules, peptides, proteinpartners and chromatin [144]. Docking studies were also applied toidentify new KDM4B inhibitors, which demonstrated an activity in thelowmicromolar range of concentration (4, Fig. 2), and Sirt1/Sirt2 inhib-itors, like thiobarbiturates (5, Fig. 2) [148,149].We also recently appliedthese techniques to the identification of new and selective inhibitors ofSirt6 (6, Fig. 2) [150]. Interestingly, pharmacophore techniques havebeen used less frequently than docking techniques on histone modify-ing enzymes. A successful applicative example of the first techniquehas been published by Sippl and colleagues for the identification ofnew PRMT1 inhibitors, which were found through a pharmacophore

Fig. 2. Representative molecular entities able to interfere with histone modifying enzymes. M(http://www.chemaxon.com).

hypothesis built on the basis of the PRMT1–allantodapsone interactionmodel (7, Fig. 2) [151].

Computational chemistry and protein modeling is also activelysupporting the basic research on epigenetic targets. A valuable exampleis the set of tools that are currently used to for detect protein plasticity,dynamics of catalytic sites, analysis of allosteric pockets and interactionswith other proteins that may constitute a useful way to study new pro-tein–protein inhibitors (PPI). Some recent reviews discussing these as-pects have been written recently on HDACs and other epigeneticplayers [152–154]. Interestingly, also quantum mechanical methodslike QM/MM or DFT have found utility in this field, especially to explorethe role metal-containing enzymes and their influence on catalytic ac-tivities and the inhibition mechanism by small-molecule modulators[155–157]. Combined computational techniques have been also suc-cessfully exploited for the de novo design of inhibitory peptides for his-tone methyltransferase [158] and chemoinformatic data mining toolslike self-organizing maps (SOM) have been exploited for the computa-tional prediction of common non-epigenetic drugs as epigenetic modu-lators [159]. It is worth to note also that emerging works driven bycomputational-based techniques are also starting to focus on food andnatural components, known to be able to influence the epigeneticcode [37,160–164]. Indeed, dietary components like complementaryand/or alternative medicines from green tea, genistein from soybean,isothiocyanates from plant foods, curcumin from turmeric, resveratrolfrom grapes, and sulforaphane from cruciferous vegetables, have beenstudied for their ability to target the epigenome, especially in differentcancer pathologies [18]. Nevertheless, the mechanisms of action ofthese compounds are still poorly understood and computer-aided tech-niques are expected to help the comprehension of thesemechanisms. Inparticular, we believe that the availability of compound databases of

arvin was used for drawing and displaying chemical structures, Marvin 6.0.0, ChemAxon

363F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

natural and dietary sources could constitute an effective step toward theidentification, development and pharmacological definition of naturaland dietary-derived components, affecting epigenetic mechanism, thathold the advantage of pharmaceutical formulation based on natural-occurring scaffold.

5. Summary and Outlook

Major research efforts are currently directed toward the discovery ofnew small-molecules able tomodulate epigenetic writers or erases thatare involved in chromatin remodeling. Recent successful stories docu-ment the possibility to interfere with the epigenetic codewith small or-ganic molecules. Indeed, first pre-clinical and clinical results, especiallyfor HDACs, testify thatmany other epidrugsmight be effective as combi-nation therapies to control the process of genesis and progression ofseveral forms of cancer, among other pathologies. It is unquestionablethat epigenetics framework will play a major role in the near future todevelop new therapies against these diseases and molecular designtechniques offer the chance to tackle these challenges.

Acknowledgments

This work was supported by the Italian Association for CancerResearch (Start Up grant N.6266).

References

[1] Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini P-L. The immune hallmarks ofcancer. Cell 2011;60:319–26.

[2] Semenza GL. A return to cancer metabolism. J Mol Med (Berl) 2011;89:203–4.[3] Tsai H-C, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine.

Cell Res 2011;21:502–17.[4] Martin C, Zhang Y. Mechanisms of epigenetic inheritance. Curr Opin Cell Biol 2007;

19:266–72.[5] Dawson MA, Kouzarides T, Huntly BJP. Targeting epigenetic readers in cancer.

N Engl J Med 2012;367:647–57.[6] Fandy TE. Development of DNA methyltransferase inhibitors for the treatment of

neoplastic diseases. Curr Med Chem 2009;16:2075–85.[7] Kulis M, Esteller M. DNA methylation and cancer. Adv Genet 2010;70:27–56.[8] Brueckner B, Kuck D, Lyko F. DNA methyltransferase inhibitors for cancer therapy.

Cancer J 2007;13:17–22.[9] De Santa F, Iosue I, Del Rio A, Fazi F. microRNA biogenesis pathway as therapeutic

target for human disease and cancer. Curr Pharm Des 2013;19:745–64.[10] Fabbri M, Calin GA. Epigenetics and miRNAs in human cancer. 1st ed. Elsevier Inc.;

2010[vol. 70].[11] Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for

anticancer drug development. Nat Rev Drug Discov 2013;12:847–65.[12] Herceg Z, Ushijima T. Introduction: epigenetics and cancer. 1st ed. Elsevier Inc.;

2010[vol. 70].[13] Baylin SB. The molecular basis of cancer. Elsevier; 2008[vol. 2].[14] Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007;128:683–92.[15] Kouzarides T. Chromatin modifications and their function. Cell 2007;128:693–705.[16] Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets.

Nat Biotechnol 2010;28:1069–78.[17] Andreoli F, Del Rio A. Physicochemical modifications of histones and their impact

on epigenomics. Drug Discov Today 2014;19:1372–9.[18] Andreoli F, Barbosa AJM, Parenti MD, Del Rio A.Modulation of epigenetic targets for

anticancer therapy: clinicopathological relevance, structural data and drug discov-ery perspectives. Curr Pharm Des 2013;19:578–613.

[19] Henikoff S, Shilatifard A. Histone modification: cause or cog? Trends Genet 2011;27:389–96.

[20] Suganuma T, Workman JL. Crosstalk among histone modifications. Cell 2008;135:604–7.

[21] CruickshankMN, Besant P, Ulgiati D. The impact of histone post-translational mod-ifications on developmental gene regulation. Amino Acids 2010;39:1087–105.

[22] Sippl W, Jung M. Epigenetic targets in drug discovery; 2009.[23] Chi P, Allis CD, Wang GG. Covalent histone modifications — miswritten,

misinterpreted and mis-erased in human cancers. Nat Rev Cancer 2010;10:457–69.

[24] Santos-Rosa H, Caldas C. Chromatin modifier enzymes, the histone code and can-cer. Eur J Cancer 2005;41:2381–402.

[25] Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. CellRes 2011;21:381–95.

[26] Golbabapour S, Abdulla MA, Hajrezaei M. A concise review on epigeneticregulation: insight into molecular mechanisms. Int J Mol Sci 2011;12:8661–94.

[27] Rius M, Lyko F. Epigenetic cancer therapy: rationales, targets and drugs. Oncogene2012;31:4257–65.

[28] Copeland RA, Olhava EJ, Scott MP. Targeting epigenetic enzymes for drug discovery.Curr Opin Chem Biol 2010;14:505–10.

[29] Dhanak D. Cracking the code: the promise of epigenetics. ACS Med Chem Lett2012;3:521–3.

[30] Best JD, Carey N. Epigenetic therapies for non-oncology indications. Drug DiscovToday 2010;15:1008–14.

[31] Best JD, Carey N. Epigenetic opportunities and challenges in cancer. Drug DiscovToday 2010;15:65–70.

[32] Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin mod-ifications. Mol Cancer Ther 2009;8:1409–20.

[33] Altucci L, Minucci S. Epigenetic therapies in haematological malignancies:searching for true targets. Eur J Cancer 2009;45:1137–45.

[34] Herranz M, Esteller M. New therapeutic targets in cancer: the epigenetic connec-tion. Clin Transl Oncol 2006;8:242–9.

[35] Graham JS, Kaye SB, Brown R. The promises and pitfalls of epigenetic therapies insolid tumours. Eur J Cancer 2009;45:1129–36.

[36] Rodríguez-ParedesM, Esteller M. Cancer epigenetics reachesmainstream oncology.Nat Med 2011;17:330–9.

[37] Meeran SM, Ahmed A, Tollefsbol TO. Epigenetic targets of bioactive dietary compo-nents for cancer prevention and therapy. Clin Epigenetics 2010;1:101–16.

[38] Ljungman M. Targeting the DNA damage response in cancer. Chem Rev 2009;109:2929–50.

[39] Claes B, Buysschaert I, Lambrechts D. Pharmaco-epigenomics: discovering thera-peutic approaches and biomarkers for cancer therapy. Heredity (Edinb) 2010;105:152–60.

[40] Pollock RM, Richon VM. Epigenetic approaches to cancer therapy. Drug DiscovToday Ther Strateg 2009;6:71–9.

[41] Spannhoff A, Sippl W, Jung M. Cancer treatment of the future: inhibitors of histonemethyltransferases. Int J Biochem Cell Biol 2009;41:4–11.

[42] Sala A, Corona DFV. Epigenetics: more than genetics. Fly (Austin) 2008;2:165–8.[43] Lohrum M, Stunnenberg HG, Logie C. The new frontier in cancer research:

deciphering cancer epigenetics. Int J Biochem Cell Biol 2007;39:1450–61.[44] Inche AG, La Thangue NB. Chromatin control and cancer-drug discovery: realizing

the promise. Drug Discov Today 2006;11:97–109.[45] Albert M, Helin K. Histone methyltransferases in cancer. Semin Cell Dev Biol 2010;

21:209–20.[46] Copeland RA, SolomonME, Richon VM. Protein methyltransferases as a target class

for drug discovery. Nat Rev Drug Discov 2009;8:724–32.[47] Heightman TD. Chemical biology of lysine demethylases. Curr Chem Genomics

2011;5:62–71.[48] Natoli G, Testa G, De Santa F. The future therapeutic potential of histone

demethylases: a critical analysis. Curr Opin Drug Discov Devel 2009;12:607–15.[49] Lohse B, Kristensen JL, Kristensen LH, Agger K, Helin K, Gajhede M, et al. Inhibitors

of histone demethylases. Bioorg Med Chem 2011;19:3625–36.[50] Huang H, Sabari BR, Garcia BA, Allis CD, Zhao Y. SnapShot: histone modifications.

Cell 2014;159:458-458.e1.[51] Jenuwein T, Allis CD. Translating the histone code. Science 2001;293:1074–80.[52] Bannister AJ, Kouzarides T. Reversing histone methylation. Nature 2005;436:

1103–6.[53] Karberg S. Switching on epigenetic therapy. Cell 2009;139:1029–31.[54] Hamm CA, Costa FF. The impact of epigenomics on future drug design and New

therapies. Drug Discov Today 2011;16:626–35.[55] Adwan L, Zawia NH. Epigenetics: a novel therapeutic approach for the treatment of

Alzheimer's disease. Pharmacol Ther 2013;139:41–50.[56] Drewes G. Future strategies in epigenetic drug discovery. Drug Discov Today Ther

Strateg 2012;9:e121–7.[57] Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families:

a new frontier for drug discovery. Nat Rev Drug Discov 2012;11:384–400.[58] Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inher-

itance. Nat Rev Genet 2012;13:343–57.[59] Tatton-Brown K, Rahman N. The NSD1 and EZH2 overgrowth genes, similarities

and differences. Am J Med Genet C Semin Med Genet 2013;163C:86–91.[60] Jobe EM, McQuate AL, Zhao X. Crosstalk among epigenetic pathways regulates

neurogenesis. Front Neurosci 2012;6:59.[61] Fujiki R, Hashiba W, Sekine H, Yokoyama A, Chikanishi T, Ito S, et al. GlcNAcylation

of histone H2B facilitates its monoubiquitination. Nature 2011;480:557–60.[62] Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, et al. Combinato-

rial patterns of histone acetylations and methylations in the human genome. NatGenet 2008;40:897–903.

[63] Denis H, Deplus R, Putmans P, YamadaM, Métivier R, Fuks F. Functional connectionbetween deimination and deacetylation of histones. Mol Cell Biol 2009;29:4982–93.

[64] Lombardi PM, Cole KE, Dowling DP, Christianson DW. Structure, mechanism, andinhibition of histone deacetylases and related metalloenzymes. Curr Opin StructBiol 2011;21:735–43.

[65] Peserico A, Simone C. Physical and functional HAT/HDAC interplay regulates pro-tein acetylation balance. J Biomed Biotechnol 2011;2011:371832.

[66] Bruzzone S, Parenti MD, Grozio A, Ballestrero A, Bauer I, Del Rio A, et al. Rejuvenat-ing sirtuins: the rise of a new family of cancer drug targets. Curr Pharm Des 2013;19:614–23.

[67] Roth M, Chen WY. Sorting out functions of sirtuins in cancer. Oncogene 2013;26:1–12.

[68] Libert S, Pointer K, Bell EL, Das A, Cohen DE, Asara JM, et al. SIRT1 activates MAO-Ain the brain to mediate anxiety and exploratory drive. Cell 2011;147:1459–72.

[69] Finkel T, Deng C-X, Mostoslavsky R. Recent progress in the biology and physiologyof sirtuins. Nature 2009;460:587–91.

364 F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

[70] Parenti MD, Bruzzone S, Nencioni A, Del Rio A. Selectivity hot-spots of sirtuin cata-lytic cores, Mol Biosyst; 2015. http://dx.doi.org/10.1039/C5MB00205B.

[71] Morris BJ. Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med2013;56:133–71.

[72] Hodawadekar SC, Marmorstein R. Chemistry of acetyl transfer by histone modify-ing enzymes: structure, mechanism and implications for effector design. Oncogene2007;26:5528–40.

[73] Stein RA. Epigenetic therapies— a new direction in clinical medicine. Int J Clin Pract2014;68:802–11.

[74] Copeland RA. Protein methyltransferase inhibitors as personalized cancer thera-peutics. Drug Discov Today Ther Strateg 2012;9:e83–90.

[75] Martinet N, Michel BY, Bertrand P, Benhida R. Small molecules DNA methyltrans-ferases inhibitors. MedChemComm 2011;3:263.

[76] Nimura K, Ura K, Kaneda Y. Histone methyltransferases: regulation of transcriptionand contribution to human disease. J Mol Med (Berl) 2010;88:1213–20.

[77] Daniel FI, Cherubini K, Yurgel LS, de Figueiredo MAZ, Salum FG. The role of epige-netic transcription repression and DNA methyltransferases in cancer. Cancer2011;117:677–87.

[78] Ferrari KJ, Pasini D. Regulation and Function of DNA and HistoneMethylations. CurrPharm Des 2013;19:719–33.

[79] Piunti A, Pasini D. Epigenetic factors in cancer development: polycomb group pro-teins. Future Oncol 2011;7:57–75.

[80] Yoo J, Medina-Franco JL. Discovery and optimization of inhibitors of DNA methyl-transferase as novel drugs for cancer therapy; 2011.

[81] Burgers WA, Fuks F, Kouzarides T. DNA methyltransferases get connected to chro-matin. Trends Genet 2002;18:275–7.

[82] Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, et al. Histone demethyla-tion mediated by the nuclear amine oxidase homolog LSD1. Cell 2004;119:941–53.

[83] Kooistra SM, Helin K. Molecular mechanisms and potential functions of histonedemethylases. Nat Rev Mol Cell Biol 2012;13:297–311.

[84] Weake VM, Workman JL. Histone ubiquitination: triggering gene activity. Mol Cell2008;29:653–63.

[85] Kim J, Guermah M, McGinty RK, Lee J-S, Tang Z, Milne TA, et al. RAD6-mediatedtranscription-coupled H2B ubiquitylation directly stimulates H3K4 methylationin human cells. Cell 2009;137:459–71.

[86] Lee J-S, Shukla A, Schneider J, Swanson SK, Washburn MP, Florens L, et al. Histonecrosstalk between H2B monoubiquitination and H3 methylation mediated byCOMPASS. Cell 2007;131:1084–96.

[87] Stoleru D, Peng Y, Agosto J, Rosbash M. Coupled oscillators control morning andevening locomotor behaviour of drosophila. Nature 2004;431:862–8.

[88] Messner S, Hottiger MO. Histone ADP-ribosylation in DNA repair, replication andtranscription. Trends Cell Biol 2011;21:534–42.

[89] Cepeda V, Fuertes MA, Castilla J, Alonso C, Quevedo C, Soto M, et al. Poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors in cancer chemotherapy. Recent PatAnticancer Drug Discov 2006;1:39–53.

[90] Jagtap P, Szabó C. Poly(ADP-ribose) polymerase and the therapeutic effects of itsinhibitors. Nat Rev Drug Discov 2005;4:421–40.

[91] Quénet D, El Ramy R, Schreiber V, Dantzer F. The role of poly(ADP-ribosyl)ation inepigenetic events. Int J Biochem Cell Biol 2009;41:60–5.

[92] Hakmé A, Wong H-K, Dantzer F, Schreiber V. The expanding field of poly(ADP-ribosyl)ation reactions. “Protein modifications: beyond the usual suspects” reviewseries. EMBO Rep 2008;9:1094–100.

[93] Mendes F, Groessl M, Nazarov AA, Tsybin YO, Sava G, Santos I, et al. Metal-basedinhibition of poly(ADP-ribose) polymerase—the guardian angel of DNA. J MedChem 2011;54:2196–206.

[94] Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. PARP inhibition: PARP1and beyond. Nat Rev Cancer 2010;10:293–301.

[95] Laing S, Unger M, Koch-Nolte F, Haag F. ADP-ribosylation of arginine. Amino Acids2011;41:257–69.

[96] Hottiger MO, Hassa PO, Lüscher B, Schüler H, Koch-Nolte F. Toward a unified no-menclature for mammalian ADP-ribosyltransferases. Trends Biochem Sci 2010;35:208–19.

[97] Hanover JA. Epigenetics gets sweeter: O-GlcNAc joins the “histone code”. ChemBiol 2010;17:1272–4.

[98] Hanover JA, Krause MW, Love DC. Post-translational modifications: bittersweetmemories: linking metabolism to epigenetics through O-GlcNAcylation. Nat RevMol Cell Biol 2012;13:312–21.

[99] Sakabe K, Wang Z, Hart GW. Beta-N-acetylglucosamine (O-GlcNAc) is part of thehistone code. Proc Natl Acad Sci U S A 2010;107:19915–20.

[100] Zhang S, Roche K, Nasheuer H-P, Lowndes NF. Modification of histones by sugar Β-N-acetylglucosamine (GlcNAc) occurs on multiple residues, including histone H3serine 10, and is cell cycle-regulated. J Biol Chem 2011;286:37483–95.

[101] AhmadW, Shabbiri K, Nazar N, Nazar S, Qaiser S, Shabbir Mughal MA. Human link-er histones: interplay between phosphorylation and O-Β-GlcNAc to mediate chro-matin structural modifications. Cell Div 2011;6:15.

[102] Anzilotti C, Pratesi F, Tommasi C, Migliorini P. Peptidylarginine deiminase 4 andcitrullination in health and disease. Autoimmun Rev 2010;9:158–60.

[103] Kobza K, Camporeale G, Rueckert B, Kueh A, Griffin JB, Sarath G, et al. K4, K9 andK18 in human histone H3 are targets for biotinylation by biotinidase. FEBS J2005;272:4249–59.

[104] Camporeale G, Shubert EE, Sarath G, Cerny R, Zempleni J. K8 and K12 are biotinyl-ated in human histone H4. Eur J Biochem 2004;271:2257–63.

[105] Tong L, Denu JM. Function and metabolism of sirtuin metabolite O-acetyl-ADP-ribose. Biochim Biophys Acta 2010;1804:1617–25.

[106] Barski A, Cuddapah S, Cui K, Roh T-Y, Schones DE, Wang Z, et al. High-resolutionprofiling of histone methylations in the human genome. Cell 2007;129:823–37.

[107] Hurd PJ, Bannister AJ, Halls K, DawsonMA, Vermeulen M, Olsen JV, et al. Phosphor-ylation of histoneH3Thr-45 is linked to apoptosis. J Biol Chem2009;284:16575–83.

[108] Cieśla J, Frączyk T, Rode W. Phosphorylation of basic amino acid residues in pro-teins: important but easily missed. Acta Biochim Pol 2011;58:137–48.

[109] Wang Y, Wysocka J, Sayegh J, Lee Y-H, Perlin JR, Leonelli L, et al. Human PAD4 reg-ulates histone arginine methylation levels via demethylimination. Science 2004;306:279–83.

[110] Messner S, Altmeyer M, Zhao H, Pozivil A, Roschitzki B, Gehrig P, et al. PARP1 ADP-ribosylates lysine residues of the core histone tails. Nucleic Acids Res 2010;38:6350–62.

[111] Hassa PO, Haenni SS, Elser M, Hottiger MO. Nuclear ADP-ribosylation reactions inmammalian cells: where are we today and where are we going? Microbiol MolBiol Rev 2006;70:789–829.

[112] Kothapalli N, Camporeale G, Kueh A, Chew YC, Oommen AM, Griffin JB, et al. Bio-logical functions of biotinylated histones. J Nutr Biochem 2005;16:446–8.

[113] Dehennaut V, Leprince D, Lefebvre T. O-GlcNAcylation, an epigenetic mark. Focuson the histone code, TET family proteins, and Polycomb group proteins, FrontEndocrinol 2014;5:1–7.

[114] Talele TT, Khedkar SA, Rigby AC. Successful applications of computer aided drugdiscovery: moving drugs from concept to the clinic. Curr Top Med Chem 2010;10:127–41.

[115] Van Drie JH. Computer-aided drug design: the next 20 years. J Comput Aided MolDes 2007;21:591–601.

[116] Borhani DW, Shaw DE. The future of molecular dynamics simulations in drug dis-covery. J Comput Aided Mol Des 2012;26:15–26.

[117] Moura Barbosa AJ, Del Rio A. Freely accessible databases of commercial compoundsfor high-throughput virtual screenings. Curr Top Med Chem 2012;12:866–77.

[118] Del Rio A, Barbosa AJM, Caporuscio F, Mangiatordi GF. CoCoCo: a free suite ofmulticonformational chemical databases for high-throughput virtual screeningpurposes. Mol Biosyst 2010;6:2122–8.

[119] Del Rio A, Barbosa AJM, Caporuscio F. Use of large multiconformational databaseswith structure-based pharmacophore models for fast screening of commercialcompound collections. J Cheminform 2011;3:P27.

[120] JanzenWP,Wigle TJ, Jin J, Frye SV. Epigenetics: tools and technologies. Drug DiscovToday Technol 2010;7:e59–65.

[121] Jones P. Development of second generation epigenetic agents. MedChemComm2012;3:135.

[122] Mai A. Targeting epigenetics in drug discovery. ChemMedChem 2014;9:415–7.[123] Masciarelli S, Quaranta R, Iosue I, Colotti G, Padula F, Varchi G, et al. A small-

molecule targeting the MicroRNA binding domain of argonaute 2 improves theretinoic acid differentiation response of the acute promyelocytic leukemia cellline NB4. ACS Chem Biol 2014;9:1674–9.

[124] Schmidt MF. Drug targetmiRNAs: chances and challenges. Trends Biotechnol 2014;32:578–85.

[125] Malumbres M. miRNAs and cancer: an epigenetics view. Mol Aspects Med 2013;34:863–74.

[126] Sippl W, Jung M. Epigenetic drug discovery special issue. Bioorg Med Chem 2009;19:3603–4.

[127] Heinke R, Carlino L, Kannan S, Jung M, Sippl W. Computer- and structure-basedlead design for epigenetic targets. Bioorg Med Chem 2011;19:3605–15.

[128] Medina-Franco JL, Caulfield T. Advances in the computational development of DNAmethyltransferase inhibitors. Drug Discov Today 2011;16:418–25.

[129] Yoo J, Medina-Franco JL. Inhibitors of DNA methyltransferases: insights from com-putational studies. Curr Med Chem 2012:37–40.

[130] Zhang L, Wang X, Li X, XuW. Discovery of a series of small molecules as potent his-tone deacetylase inhibitors. J Enzyme Inhib Med Chem 2014;29:333–7.

[131] Kalyaanamoorthy S, Chen Y-PP. A steeredmolecular dynamicsmediated hit discov-ery for histone deacetylases. Phys Chem Chem Phys 2014;16:3777–91.

[132] Kalyaanamoorthy S, Chen Y-PP. Energy based pharmacophore mapping of HDACinhibitors against class I HDAC enzymes. Biochim Biophys Acta Proteins Proteomics2013;1834:317–28.

[133] Zhao L, Xiang Y, Song J, Zhang Z. A novel two-step QSAR modeling work flow topredict selectivity and activity of HDAC inhibitors. Bioorg Med Chem Lett 2013;23:929–33.

[134] Thangapandian S, Sundarapandian T, John S, Shalini J, Sakkiah S, Sugunadevi S,et al. Docking-enabled pharmacophore model for histone deacetylase 8 inhibitorsand its application in anti-cancer drug discovery. J Mol Graph Model 2010;29:382–95.

[135] Nair SB, Teli MK, Pradeep H, Rajanikant GK. Computational identification of novelhistone deacetylase inhibitors by docking based QSAR. Comput Biol Med 2012;42:697–705.

[136] Tang H, Wang XS, Huang X-P, Roth BL, Butler KV, Kozikowski AP, et al. Novel inhib-itors of human histone deacetylase (HDAC) identified by QSARmodeling of knowninhibitors, virtual screening, and experimental validation. J Chem Inf Model 2009;49:461–76.

[137] Chen Y, Li H, Tang W, Zhu C, Jiang Y, Zou J, et al. 3D-QSAR studies of HDACs inhib-itors using pharmacophore-based alignment. Eur J Med Chem 2009;44:2868–76.

[138] Vadivelan S, Sinha BN, Rambabu G, Boppana K, Jagarlapudi SARP. Pharmacophoremodeling and virtual screening studies to design some potential histonedeacetylase inhibitors as new leads. J Mol Graph Model 2008;26:935–46.

[139] Park H, Kim S, Kim YE, Lim S-J. A structure-based virtual screening approach to-ward the discovery of histone deacetylase inhibitors: identification of promisingzinc-chelating groups. ChemMedChem 2010;5:591–7.

[140] Kannan S, Melesina J, Hauser A, Chakrabarti A, Heimburg T, Schmidtkunz K, et al.Discovery of inhibitors of Schistosoma mansoni HDAC8 by combining homologymodeling. J Chem Inf Model 2014;54:3005–19.

365F. Andreoli, A. Del Rio / Computational and Structural Biotechnology Journal 13 (2015) 358–365

[141] Zeng F-Q, Peng S-M, Li L, Mu L-B, Zhang Z-H, Zhang Z-Y, et al. Structure-based iden-tification of drug-like inhibitors of p300 histone acetyltransferase. Acta Pharm Sin2013;48:700–8.

[142] Bowers EM, Yan G, Mukherjee C, Orry A, Wang L, Holbert MA, et al. Virtual ligandscreening of the p300/CBP histone acetyltransferase: identification of a selectivesmall molecule inhibitor. Chem Biol 2010;17:471–82.

[143] Tortorici M, Borrello MT, Tardugno M, Chiarelli LR, Pilotto S, Ciossani G, et al. Pro-tein recognition by short peptide reversible inhibitors of the chromatin-modifying LSD1/CoREST lysine demethylase. ACS Chem Biol 2013;8:1677–82.

[144] Robertson JC, Hurley NC, Tortorici M, Ciossani G, Borrello MT, Vellore NA, et al.Expanding the druggable space of the LSD1/CoREST epigenetic target: new poten-tial binding regions for drug-like molecules, peptides, protein partners, and chro-matin. PLoS Comput Biol 2013;9:e1003158.

[145] Ogasawara D, Itoh Y, Tsumoto H, Kakizawa T, Mino K, Fukuhara K, et al. Lysine-specific demethylase 1-selective inactivators: protein-targeted drug deliverymechanism. Angew Chem Int Ed 2013;52:8620–4.

[146] Itoh Y, Ogasawara D, Ota Y, Mizukami T, Suzuki T. Synthesis, LSD1 inhibitory activ-ity, and LSD1 binding model of optically pure lysine-PCPA conjugates. ComputStruct Biotechnol J 2014;9:e201402002.

[147] Schmitt ML, Ladwein KI, Carlino L, Schulz-Fincke J, Willmann D, Metzger E, et al.Heterogeneous antibody-based activity assay for lysine specific demethylase 1(LSD1) on a histone peptide substrate. J Biomol Screen 2014;19:973–8.

[148] Pulla VK, Alvala M, Sriram DS, Viswanadha S, Sriram D, Yogeeswari P. Structure-based drug design of small molecule SIRT1 modulators to treat cancer and meta-bolic disorders. J Mol Graph Model 2014;52:46–56.

[149] Uciechowska U, Schemies J, Neugebauer RC, Huda E-M, Schmitt ML, Meier R, et al.Thiobarbiturates as sirtuin inhibitors: virtual screening, free-energy calculations,and biological testing. ChemMedChem 2008;3:1965–76.

[150] Parenti MD, Grozio A, Bauer I, Galeno L, Damonte P, Millo E, et al. Discovery of noveland selective SIRT6 inhibitors. J Med Chem 2014;57:4796–804.

[151] Heinke R, Spannhoff A, Meier R, Trojer P, Bauer I, Jung M, Sippl W, et al. Virtualscreening and biological characterization of novel histone arginine methyltransfer-ase PRMT1 inhibitors. ChemMedChem 2009;4:69–77.

[152] Deschamps N, Simões-Pires CA, Carrupt P-A, Nurisso A. How the flexibility ofhuman histone deacetylases influences ligand binding: an overview. Drug DiscovToday 2015:1–7.

[153] Vellore NA, Baron R. Epigenetic molecular recognition: a biomolecular modelingperspective. ChemMedChem 2014;9:484–94.

[154] Ran T, Zhang Z, Liu K, Lu Y, Li H, Xu J, et al. Insight into the key interactions ofbromodomain inhibitors based on molecular docking, interaction fingerprinting,molecular dynamics and binding free energy calculation. Mol BioSyst 2015;11:1295–304.

[155] Chen K, Zhang X, Wu Y-D, Wiest O. Inhibition and mechanism of HDAC8 revisited.J Am Chem Soc 2014:1–8.

[156] Pottel J, Therrien E, Gleason JL, Moitessier N. Docking ligands into flexible and sol-vated macromolecules. 6. Development and application to the docking of HDACsand other zinc metalloenzymes inhibitors. J Chem Inf Model 2014;54:254–65.

[157] Chen K, Xu L, Wiest O. Computational exploration of zinc binding groups for HDACinhibition. J Org Chem 2013;78:5051–5.

[158] Smadbeck J, PetersonMB, Zee BM, Garapaty S, Mago A, Lee C, et al. De novo peptidedesign and experimental validation of histone methyltransferase inhibitors. PLoSOne 2014;9.

[159] Lötsch J, Schneider G, Reker D, ParnhamMJ, Schneider P, Geisslinger G, et al. Com-mon non-epigenetic drugs as epigenetic modulators. Trends Mol Med 2013;19:742–53.

[160] vel Szic KS, Ndlovu MN, Haegeman G, Vanden Berghe W. Nature or nurture: letfood Be your epigenetic medicine in chronic inflammatory disorders. BiochemPharmacol 2010;80:1816–32.

[161] Rajendran P, Williams DE, Ho E, Dashwood RH. Metabolism as a key to histonedeacetylase inhibition. Crit Rev Biochem Mol Biol 2011;46:181–99.

[162] Gerhäuser C. Cancer cell metabolism, epigenetics and the potential influence of di-etary components — a perspective. Biomed Res 2012;23:1–21.

[163] Khan SI, Aumsuwan P, Khan IA,Walker LA, Dasmahapatra AK. Epigenetic events as-sociated with breast cancer and their prevention by dietary components targetingthe epigenome. Chem Res Toxicol 2012;25:61–73.

[164] NyströmM. Diet and epigenetics in colon cancer. World J Gastroenterol 2009;15:257.