Identifying Biomolecular Targets of the Anticancer Vitamin-E-δ-Tocotrienol Using a Computational Approach: Virtual Target Screening

A B S T R A C T

In recent years, evidence has mounted that a particular form of vitamin E (its δ-tocotrienol variant) may have cellular functions beyond that of an antioxidant, a role commonly ascribed to the tocotrienol class of compounds. In particular, numerous studies of δ-tocotrienol’s effect on cancer cells have identified it as a potent anticancer and antitumor agent. However, this important revelation of potential therapeutic use poses a series of new challenges, with arguably the most important being the elucidation of the precise mechanism of action responsible for the anticancer activity of δ-tocotrienol. As an initial step to address this question, we have used a computational tool, Virtual Target Screening (a molecular docking-based tool that identifies potential binding partners for small molecules), to identify potential biomolecular targets of δ-tocotrienol. Then, to gain a consensus as to the type of biomolecular entity that could be a target for δ-tocotrienol, we utilized PharmMapper and PASS (a ligand-based chemoinformatic approach), and ProBiS (a tool that analyses binding site similarities across known proteins). The results of our multipronged computational consensus-seeking approach showed that such a strategy can identify potential cellular targets of small molecules. This is evidenced by our identification of estrogen receptor-beta, a protein that has been previously shown to bind δ-tocotrienol, which elicited a cellular response. This study supports the use of such a computational approach as an initial step in target identification to avoid time-consuming, costly large-scale experimental screening, greatly reducing the experimental work to just one or a few candidate proteins.

Keywords

Natural products, docking, virtual target screening, structure-activity relation, vitamin E

Introduction

Vitamins, a subclass of natural products, have long been thought to play an important role in the physiology of living organisms. Many essential processes in the human body rely on the availability of vitamins. While there is little dispute over the importance of vitamins, their precise mode of action that manifests into health benefits often remains elusive. However, this is not surprising, as understanding the underpinnings of a biological effect of a small molecule requires knowledge of its biomolecular targets. Zeroing in on the precise mode of action of many vitamins is made more complicated by the fact that they can come in various forms and induce a range of physiological responses. Vitamin E is a good example of such complexity. Vitamin E is an essential lipid-soluble vitamin and an important macronutrient that has long been thought to have strong antioxidant effects without causing major toxicity in humans [1-3]. Its primary activity has been attributed to its ability to reduce free radicals to prevent lipid peroxidation of polyunsaturated fatty acids [4].

Vitamin E consists of 8 naturally occurring isomers: d-alpha-, d-beta-, d-gamma-, and d-delta-tocopherols and d-alpha-, d-beta-, d-gamma-, and d-delta-tocotrienols [5]. Among the constituents of vitamin E, tocopherols initially received a great deal of attention from the scientific community due to their potent antioxidant properties and their abundance in common food sources. However, the focus has now shifted toward tocotrienols, which, although rare in nature, have exhibited a number of physiological effects that include neuroprotective, antioxidant, anticancer, cholesterol-lowering, and other therapeutic activities [6-10]. The diversity and, at the same time, specificity of physiological responses to tocotrienols may mean that their modes of action differ from the broad antioxidant activity of tocopherols. A quick look at the structures of tocopherols and tocotrienols reveals three double bonds in the farnesyl isoprenoid tail of the tocotrienols, making it the less flexible of the two classes of compounds (Figure 1). This additional restraint on the conformational freedom of tocotrienols, however, may be the reason for the more specific range of activities. The significantly reduced number of allowable conformations of tocotrienols over tocopherols may result in an increased affinity of tocotrienols toward a specific group of binding partners. If true, the activity of tocotrienols may go beyond that of a broad antioxidant and into the signaling realm.

Figure 1: Comparison of the structures of δ-tocopherol and δ-tocotrienol.

Studies of vitamin E delta-tocotrienol (VEDT) have already proposed the notion that at least VEDT's anticancer activity may be attributed, not just to its antioxidant properties, but rather to its involvement in the signaling pathways of cancer [11-13]. Furthermore, VEDT was proposed to act as a mediating substance in antiproliferative and apoptotic mechanisms in carcinogenic tumor cells [5, 6, 13-17]. In this study, we attempt to gain insight into the possible modes of action of VEDT by using molecular modeling, cheminformatics and bioinformatics, with the goal of proposing likely binding partners of VEDT.

As an initial step directed at pinpointing molecular target(s) of VEDT, we used in silico approaches to interrogate protein space for potential targets. With the growing prominence of in silico target discovery as a viable tool in research campaigns, a number of methodologies have been developed to take advantage of computational efficiency and storage capabilities of computer systems to search for potential protein targets, while greatly reducing the scale and scope of experimental work required for such exploration [18-24].

Here, we used a number of different approaches to computationally probe for potential target proteins of VEDT. These approaches vary significantly yet aim to solve the same problem. This was done in part to decrease the effect of errors resulting from the use of any one individual tool. The results drawn from the consensus reached via the utilization of different approaches have also previously produced reliable results [25-28]. One method, called Virtual Target Screening (VTS), is based on the explicit modeling of intermolecular interactions between VEDT and its potential targets via a docking protocol designed specifically to search for targets of small drug-like or natural product-like compounds [29, 30]. The other methods, PharmMapper and PASS (Prediction of Activity Spectra of Substances), take chemoinformatic approaches for proposing potential targets of interest [31-35]. Here, the structure of VEDT and its pharmacophore were compared to a database of structures and pharmacophores of small molecules with known biomolecular targets. Target proteins were then proposed based on the structural similarities between database compounds and VEDT. Finally, we used the binding site analysis tool Protein Binding Sites Detection (ProBiS) to mine the Protein Data Bank (PDB) for proteins containing binding pockets similar to those that are likely to accommodate VEDT [36-41].

Materials and Methods

I VEDT Preparation

Three-dimensional coordinates of VEDT and δ-tocopherol were downloaded from PubChem (Link 1) in a structure data file format and were prepared using the LigPrep (LigPrep, Schrödinger, New York, NY) module of the suite of molecular modeling software (Suite 2012: Maestro, Schrödinger, New York, NY) with default settings corresponding to physiological pH.

II Virtual Target Screening

VTS is a Web-based software application deployed on a dedicated computer cluster in the Chemistry Department at the University of South Florida [29, 30]. VTS works by comparing the docking performance of the molecule of interest (MOI) with a given protein against the performance of a calibration set of small molecules docked into the same protein. The National Cancer Institute Diversity Set (Set 1 provided by the Developmental Therapeutics Program, Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD; collections are available at Link 2), which consists of 1990 drug-like compound structures, served as a calibration set and was docked into each of the 1451 protein structures currently in the VTS library. Docking and scoring were performed using Glide (Glide, Schrödinger, New York, NY). The resulting docking score, called the GScore, is based on Schrödinger’s proprietary calculation of ligand/protein energetics. GScores are typically negative values since mutual accommodation of the ligand and protein reflects less disruptive energy in the complex versus the state when the entities are not combined. MOIs (VEDT and δ-tocopherol as two separate VTS runs) were then docked into each of the proteins in the VTS library.

In addition to human protein structures, which comprise the majority of the VTS library (~1000), structures from other organisms ranging from bacteria to mammals were also included in the VTS library to allow future utility in other project types. Once docked, the docking performance of the MOI, as measured by their GScores, was compared to the average GScores of the calibration set compounds for each protein. A protein was considered a hit if the MOI’s docking score was better than the average docking score of the top 200 calibration compounds docked into the protein and was of particular significance if the MOI’s docking score was better than the top 20 average.

III PharmMapper

PharmMapper uses pharmacophore mapping to identify potential targets for the MOI. Six pharmacophore features, including hydrophobic center, positively charged center, negatively charged center, hydrogen bond acceptor, and donor vectors and the aromatic plane of an ensemble of the MOI conformations, are mapped onto a library of pharmacophores extracted from publicly available crystal structures of protein-ligand complexes. A fit score between the MOI's pharmacophores and those in the library is then calculated, and the desired number of best-fitting targets, N, are suggested. The structure data file of VEDT downloaded from PubChem was converted to Tripos mol2 file using OpenBabel software [42]. The resulting mol2 file was submitted to PharmMapper server at (Link 3).

IV Prediction of Activity Spectra of Substances

Prediction of Activity Spectra of Substances (PASS) predicts the biological activity of the MOI based on the “multilevel neighborhoods of atoms” structural descriptors of compounds and a training set of structure-activity relationship (SAR) data for over 60,000 chemical substances (SAR Base) [43]. For each biological activity, based on the similarity of multilevel neighborhoods of atoms descriptors of the MOI and the substances in the SAR Base, PASS outputs two probabilities: the probability of the MOI to exhibit the activity and the probability of the MOI to not exhibit the activity. To execute PASS prediction, a SMILES chemical identifier representing VEDT [CC1=C2C(=CC(=C1)O)CCC(O2)(C)CCC=C(C)CCC=C(C)CCC=C(C)C] was extracted from the PubChem entry for VEDT and submitted to (Link 4).

V Protein Binding Sites Detection

Protein Binding Sites Detection (ProBiS) identifies proteins structurally similar to the user-supplied protein of interest in the PDB. The algorithm behind the ProBiS server represents the entire proteins as graphs where vertices correspond to functional groups of surface amino acids and edges represent distance between these functional groups. Pairwise alignment of structural features of the protein of interest and the proteins in the PDB is then performed, with displayed results sorted by the statistical Z-scores and proteins most similar to protein of interest displayed on top. The PDB code for estrogen receptor β (ERβ) 1NDE was used as the input for the ProBiS search [44]. The choice of the above PDB code was based on results of the VTS screen.

Results

The VTS screens for both VEDT and δ-tocopherol have resulted in a number of proteins that were hits for VEDT but not for δ-tocopherol (Table 1). According to VTS, ERβ (PDB code 1NDE) was a top hit for VEDT, although not for δ-tocopherol (Figure 2). Thirty-three other proteins were also predicted as potential hits for VEDT but not for δ-tocopherol. This is consistent with experimental findings that support binding of VEDT to ERβ [8, 12]. Moreover, some patterns or at least consistencies could be observed by looking at the VTS hit list. In particular, a number of hormone and nuclear receptors were shown in the proposed target list. This is not surprising, however, as a ProBis search for binding sites similar to the ERβ revealed that the binding sites of a number of proteins such as Rxr-like protein, retinoic acid receptor Rxr-α, progesterone receptor, and others are indeed quite similar (Table 2).

Table 1: VEDT and δ-tocopherol VTS results.

[Native ligand] Protein

PDB Code

VEDT Result

δ-tocopherol Result

[Mon]Estrogen receptor beta

1NDE

Hit*

Not a Hit

[ZEB]Cytidine deaminase

1CTU

Hit*

Hit

[MOF]Progesterone receptor

1SR7

Hit*

Hit

[Proflavin]Alpha-thrombin

1BCU

Hit

Not a Hit

[CL3]Cell division protein zipA

1Y2G

Hit

Not a Hit

[D91]Coagulation factor x- heavy chain

1WU1

Hit

Not a Hit

[SRL]Orphan nuclear receptor pxr

1ILH

Hit

Not a Hit

[_U]Cytidine deaminase

1AF2

Hit

Hit

[TTB]Retinoic acid receptor beta

1XAP

Hit

Not a Hit

[442]Thyroid hormone receptor beta-1

1R6G

Hit

Not a Hit

[L79]Retinoic acid receptor rxr-alpha

1RDT

Hit

Not a Hit

[STR]Igg1-kappa db3 fab (light chain)

1DBB

Hit

Not a Hit

[E1P]Peptidyl-prolyl cis-trans isomerase a

1W8M

Hit

Not a Hit

[AO5]Methionine aminopeptidase 2

1R58

Hit

Not a Hit

[SDK]Cathepsin K

1AU0

Hit

Not a Hit

[FSN]Thrombin light chain

1OYT

Hit

Not a Hit

[R11]Coagulation factor x

1G2M

Hit

Not a Hit

[HEM]albumin (no iron)

1N5U

Hit

Not a Hit

[ANO]Igg1-kappa db3 fab (light chain)

1DBK

Hit

Not a Hit

[L10]Mitogen-activated protein kinase 14

1W82

Hit

Not a Hit

[AB8]Integrin alpha-I

1XDG

Hit

Not a Hit

[FPP]Protein farnesyltransferase

1FPP

Hit

Not a Hit

[IGN]Prothrombin

1K21

Hit

Not a Hit

[DHZ]Cytidine deaminase

1CTT

Hit

Not a Hit

[MSC]HIV-1 protease

1D4J

Hit

Not a Hit

[L08]Integrin alpha-I

1RD4

Hit

Not a Hit

[CI1031(Z34)]Coagulation factor XA

1FJS

Hit

Not a Hit

[BLN]Cathepsin s

1MS6

Hit

Not a Hit

[AIH]Estrogen receptor

1XP1

Hit

Not a Hit

[CMB]Blood coagulation factor xa

1LPZ

Hit

Not a Hit

[RTR]Coagulation factor xa- heavy chain

1NFY

Hit

Not a Hit

[EST]Estradiol receptor

1QKT

Hit

Not a Hit

[STU]Tyrosine-protein kinase zap-70

1U59

Hit

Not a Hit

[Palmitate]Lipocalin beta-lactoglobulin

1B0O

Hit

Hit

[AIU]Estrogen receptor

1XP6

Hit

Not a Hit

[AAY]Integrin alpha-I

1XDD

Hit

Not a Hit

[HYC]Type117 beta-hydroxysteroid dehydrogenase

1I5R

Hit

Not a Hit

[CIU]Epoxide hydrolase

1EK1

Hit

Not a Hit

[165]Prothrombin

1SB1

Hit

Hit

[CIU]Epoxide hydrolase 2-cytoplasmic

1VJ5

Hit

Hit

[HYF]Orphan nuclear receptor pxr

1M13

Hit

Hit

[337]Map kinase 14

3CTQ

Hit

Hit

Hit: proteins into which an MOI docked with a score that outperformed the top 200 molecules of the calibration set; Hit*: top hits (proteins into which an MOI docked with a score that outperformed the top 20 molecules of the calibration set).


Table 2: Results of the ProBis query based on the PDB ID 1NDE input structure.

PDB ID

Protein Name

2j7y

ESTROGEN RECEPTOR BETA

3uud

ESTROGEN RECEPTOR

3ltx

ESTROGEN RECEPTOR

2e2r

ESTROGEN-RELATED RECEPTOR GAMMA

3mnp

GLUCOCORTICOID RECEPTOR

2q1h

ANCCR

1xiu

RXR-LIKE PROTEIN

4fne

STEROID RECEPTOR 2

3k6p

STEROID HORMONE RECEPTOR ERR1

1g2n

ULTRASPIRACLE PROTEIN

4e2j

ANCESTRAL GLUCOCORTICOID RECEPTOR 2

3vhv

MINERALOCORTICOID RECEPTOR

1t7r

ANDROGEN RECEPTOR

2p1t

RETINOIC ACID RECEPTOR RXR-ALPHA

3ry9

ANCESTRAL GLUCOCORTICOID RECEPTOR 1

1z5x

ULTRASPIRACLE PROTEIN (USP) A HOMOLOGUE OF RXR

3dzu

RETINOIC ACID RECEPTOR RXR-ALPHA

1sr7

PROGESTERONE RECEPTOR

3plz

FTZ-F1 RELATED PROTEIN

1hg4

ULTRASPIRACLE

1lbd

RETINOID X RECEPTOR

3eyb

NUCLEAR HORMONE RECEPTOR RXR

4iqr

HEPATOCYTE NUCLEAR FACTOR 4-ALPHA

2gl8

RETINOIC ACID RECEPTOR RXR-GAMMA

2q60

RETINOID X RECEPTOR

4j5x

RETINOIC ACID RECEPTOR RXR-ALPHA, NUCLEAR RECEPTOR COACTIVATOR 1

1lv2

HEPATOCYTE NUCLEAR FACTOR 4-GAMMA

3dzu

PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR GAMMA

3kmr

RETINOIC ACID RECEPTOR ALPHA

The list reveals structures in the Protein Data Bank that contain binding site similar to that of ERβ as represented by the structure under the PDB ID 1NDE.


Figure 2: Docked pose of VEDT as identified by VTS. VEDT, coloured in magenta, forms hydrogen bonds (yellow dashed lines) with Glu-305 and Arg-346.

PharmMapper prediction resulted in 300 proposed pharmacophores, with those fitting the flexible alignment with VEDT ranked on top. These results were examined for consistency with the best VEDT hits from the VTS screen. Based on the pharmacophore derived from the PDB structure 1NDE, ERβ, which was the best hit in the VTS screen, was ranked 51.

PASS prediction resulted in 500 proposed activities, which VEDT statistically is more likely to exhibit than not. The results were ranked by the probability that VEDT exhibits a given activity. Similarly, these results were examined for consistency with the VTS screen and PharmMapper prediction.

According to the PASS prediction, there are several activities related either to estrogen modulation or ERβ specifically (Table 3). Among the estrogen-related activities, VEDT is likely to serve as an estrogen antagonist with the highest probability, followed by ERβ antagonist, followed by estrogen agonist, followed by ERβ agonist. Therefore, VEDT is more likely to serve as an antagonist of estrogen and ERβ activity rather than their agonist.

Table 3: PASS prediction results.

Rank

Pa

Pi

Activity

213

0,177

0,022

Estrogen antagonist

234

0,148

0,024

Estrogen receptor beta antagonist

247

0,140

0,026

Estrogen agonist

371

0,048

0,021

Estrogen beta receptor agonist

Predicted activities with relation to the estrogen modulation or action of ERβ. Rank: overall PASS rank of this activity by VEDT; Pa: Probability that VEDT exhibits the given activity; Pi: Probability that VEDT does not exhibit the given activity.


Discussion

I Consensus Between Computational Studies

We identified potential molecular targets of VEDT via a combination of molecular modeling (docking), cheminformatics (SAR) techniques, and bioinformatics in the form of binding site analyses. Our results were consistent in identifying ERβ as a potential target of VEDT. All three approaches (VTS, PharmMapper, and PASS) identified ERβ modulation as one of the potential activities of VEDT. In particular, with VTS, ERβ was identified by PDB ID 1NDE as a top target from a list of 1451 protein structures. Although PharmMapper and PASS algorithms operate on a substantially larger data space, both identified ERβ as one of the top potential targets, although results did not rank it as high as with VTS. In addition, PharmMapper exhibited consistency in preference toward the conformation of the ERβ represented by 1NDE, suggesting a possible antagonistic mechanism of action [12, 44]. The hypothesis of the antagonistic mode of action can also be favored based on the results of PASS, which favored an antagonistic activity of VEDT toward ERβ than that of an agonistic.

II Consistency with Experimental Data

Although no experimental data were considered during the initial VTS screening of VEDT, a subsequent literature search revealed that Comitato and colleagues had performed both in vitro binding analyses and molecular docking studies to identify a high-affinity interaction between VEDT and ERβ, which was the first reported evidence of such an association [11, 12]. The consistency of experimental data with the computational approaches reported in this work is encouraging and gives validity to the use of tools such as VTS as an initial step to probe for potential molecular targets of compounds with an unknown mode of action. In addition, the techniques described in this work allow for additional inferences regarding the precise modulating effects of the MOI on its target. Although Comitato et al., based on the docking studies, favored an agonistic mode of action and seemed to contradict the preference toward an antagonistic activity favored by our results, the combination of different approaches reported here can be beneficial in facilitating the investigation of the true mode of action of the MOI.

Overall, the use of VTS as a first step, particularly in combination with other computational methods for consensus, is a viable strategy in helping to identify potential targets of natural products and other chemical substances before proceeding with extensive experimental work. Moreover, utilization of bioinformatics approaches like ProBis can help gain additional insight into potential targets and help identify structures for further inclusion into the VTS and cheminformatics screens. This multipronged consensus approach may prove especially valuable in cases where it is not feasible or is otherwise prohibitive to conduct experimental studies to address this question.

Computational approaches in drug discovery and development have faced skepticism in the past since, in some cases, needed virtual protein structures were not available or of poor resolution. In addition, there have been difficulties in developing accurate scoring algorithms for compound docking in virtual screening and difficulties accounting for protein flexibility (e.g. active site dynamics). Experimental screening has been considered more reliable since it is performed in a relevant in vitro or even in vivo setting. But virtual protein availability has greatly improved, protein conformational dynamics can be modeled with some accuracy, HPC (parallel processing on clusters) is more readily available and available virtual compound libraries provide greater breadth of chemical space to test (e.g. the ZINC databases).

When we consider virtual screening and/or VTS as a first pass at screening, compared to experimental screening, the accuracy is about the same or even better with the virtual approaches. Virtual is certainly the cheaper and faster approach to find “hit” compounds in virtual screening or to find “hit” proteins in VTS. VTS attained 72% accuracy in a test case of kinases and known kinase inhibitors with our prototype VTS system [Santiago, et al., 2012] and we believe VTS can be improved to 80-90% accuracy with planned improvements, such as incorporating machine learning to track protein promiscuity to down-weight proteins that tend to bind many compounds or incorporating low-mode vibrational analysis that allows the virtual protein/ligand complex more flexibility and in vivo-like dynamics. Experimental approaches can be costly and are hampered sometimes by poor availability of physical protein and viable assays. And experimental work can have issues with individual compounds such that we consider the accuracy of experimental approaches, as a first pass at screening, to be around 70%.

Experimental assaying can be affected by: 1) cell permeability; 2) compound solubility; 3) compound concentration; 4) proper solvents & buffers; 5) protein availability; 6) protein degradation; 7) potential modifications, degradation or sequestration of the MOI; 8) technical competency (e.g. pipetting errors) and availability of validated assays, reagents and equipment; 9) proper analysis and interpretation of results. Commercial services that test compounds against a panel of related protein targets (e.g. kinase panels) can be used but they are expensive and services that have comprehensive protein collections are an exception rather than the rule.

Typically, in experimental screening, 14% of compounds may be insoluble and 30-40% have poor solubility [45, 46]. So, by comparison between computational and experimental for a first pass, the accuracy of computational screening is most encouraging. With VTS, compound concentration, purity and solubility of the virtual MOI and protein structures are irrelevant as are degradation and aberrant modifications of the virtual structures. Also, the virtual MOI with 100% solubility, has zero toxicity, zero photosensitivity and maintains absolute stereochemistry (i.e. no racemic drift). Of course, VTS results need experimental confirmation but the experimental work is greatly reduced by the VTS filtering out insignificant proteins and by virtual screening filtering out irrelevant compounds. Later in drug development, solubility and permeability issues of a compound, which can be interrelated depending on the intended therapeutic context, can be addressed in compound optimization with experimental assays and computational modeling, including additional VTS runs.

Acknowledgments

We thank Rasa Hamilton (Moffitt Cancer Center) for editorial assistance.

Funding

The authors declare no competing financial interest. This work has been supported in part by the Chemical Biology Core at the H. Lee Moffitt Cancer Center & Research Institute, a comprehensive cancer center designated by the National Cancer Institute, supported under NIH Grant P30-CA76292.

Author Contributions

The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript.

Abbreviations

ERβ: Estrogen Receptor β
MOI: Molecule of Interest
PASS: Prediction of Activity Spectra of Substances
PDB: Protein Data Bank
ProBis: Protein Binding Sites Detection
SAR: Structure-Activity Relationship
VEDT: Vitamin E Delta-Tocotrienol
VTS: Virtual Target Screening

Article Info

Article Type
Research Article
Publication history
Received: Wed 15, Jul 2020
Accepted: Tue 28, Jul 2020
Published: Wed 30, Dec 2020
Copyright
© 2023 Wesley H. Brooks. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Hosting by Science Repository.
DOI: 10.31487/j.COR.2020.08.12

Author Info

Corresponding Author
Wesley H. Brooks
Department of Chemistry, University of South Florida, Tampa, Florida, USA

Figures & Tables

Table 1: VEDT and δ-tocopherol VTS results.

[Native ligand] Protein

PDB Code

VEDT Result

δ-tocopherol Result

[Mon]Estrogen receptor beta

1NDE

Hit*

Not a Hit

[ZEB]Cytidine deaminase

1CTU

Hit*

Hit

[MOF]Progesterone receptor

1SR7

Hit*

Hit

[Proflavin]Alpha-thrombin

1BCU

Hit

Not a Hit

[CL3]Cell division protein zipA

1Y2G

Hit

Not a Hit

[D91]Coagulation factor x- heavy chain

1WU1

Hit

Not a Hit

[SRL]Orphan nuclear receptor pxr

1ILH

Hit

Not a Hit

[_U]Cytidine deaminase

1AF2

Hit

Hit

[TTB]Retinoic acid receptor beta

1XAP

Hit

Not a Hit

[442]Thyroid hormone receptor beta-1

1R6G

Hit

Not a Hit

[L79]Retinoic acid receptor rxr-alpha

1RDT

Hit

Not a Hit

[STR]Igg1-kappa db3 fab (light chain)

1DBB

Hit

Not a Hit

[E1P]Peptidyl-prolyl cis-trans isomerase a

1W8M

Hit

Not a Hit

[AO5]Methionine aminopeptidase 2

1R58

Hit

Not a Hit

[SDK]Cathepsin K

1AU0

Hit

Not a Hit

[FSN]Thrombin light chain

1OYT

Hit

Not a Hit

[R11]Coagulation factor x

1G2M

Hit

Not a Hit

[HEM]albumin (no iron)

1N5U

Hit

Not a Hit

[ANO]Igg1-kappa db3 fab (light chain)

1DBK

Hit

Not a Hit

[L10]Mitogen-activated protein kinase 14

1W82

Hit

Not a Hit

[AB8]Integrin alpha-I

1XDG

Hit

Not a Hit

[FPP]Protein farnesyltransferase

1FPP

Hit

Not a Hit

[IGN]Prothrombin

1K21

Hit

Not a Hit

[DHZ]Cytidine deaminase

1CTT

Hit

Not a Hit

[MSC]HIV-1 protease

1D4J

Hit

Not a Hit

[L08]Integrin alpha-I

1RD4

Hit

Not a Hit

[CI1031(Z34)]Coagulation factor XA

1FJS

Hit

Not a Hit

[BLN]Cathepsin s

1MS6

Hit

Not a Hit

[AIH]Estrogen receptor

1XP1

Hit

Not a Hit

[CMB]Blood coagulation factor xa

1LPZ

Hit

Not a Hit

[RTR]Coagulation factor xa- heavy chain

1NFY

Hit

Not a Hit

[EST]Estradiol receptor

1QKT

Hit

Not a Hit

[STU]Tyrosine-protein kinase zap-70

1U59

Hit

Not a Hit

[Palmitate]Lipocalin beta-lactoglobulin

1B0O

Hit

Hit

[AIU]Estrogen receptor

1XP6

Hit

Not a Hit

[AAY]Integrin alpha-I

1XDD

Hit

Not a Hit

[HYC]Type117 beta-hydroxysteroid dehydrogenase

1I5R

Hit

Not a Hit

[CIU]Epoxide hydrolase

1EK1

Hit

Not a Hit

[165]Prothrombin

1SB1

Hit

Hit

[CIU]Epoxide hydrolase 2-cytoplasmic

1VJ5

Hit

Hit

[HYF]Orphan nuclear receptor pxr

1M13

Hit

Hit

[337]Map kinase 14

3CTQ

Hit

Hit

Hit: proteins into which an MOI docked with a score that outperformed the top 200 molecules of the calibration set; Hit*: top hits (proteins into which an MOI docked with a score that outperformed the top 20 molecules of the calibration set).


Table 2: Results of the ProBis query based on the PDB ID 1NDE input structure.

PDB ID

Protein Name

2j7y

ESTROGEN RECEPTOR BETA

3uud

ESTROGEN RECEPTOR

3ltx

ESTROGEN RECEPTOR

2e2r

ESTROGEN-RELATED RECEPTOR GAMMA

3mnp

GLUCOCORTICOID RECEPTOR

2q1h

ANCCR

1xiu

RXR-LIKE PROTEIN

4fne

STEROID RECEPTOR 2

3k6p

STEROID HORMONE RECEPTOR ERR1

1g2n

ULTRASPIRACLE PROTEIN

4e2j

ANCESTRAL GLUCOCORTICOID RECEPTOR 2

3vhv

MINERALOCORTICOID RECEPTOR

1t7r

ANDROGEN RECEPTOR

2p1t

RETINOIC ACID RECEPTOR RXR-ALPHA

3ry9

ANCESTRAL GLUCOCORTICOID RECEPTOR 1

1z5x

ULTRASPIRACLE PROTEIN (USP) A HOMOLOGUE OF RXR

3dzu

RETINOIC ACID RECEPTOR RXR-ALPHA

1sr7

PROGESTERONE RECEPTOR

3plz

FTZ-F1 RELATED PROTEIN

1hg4

ULTRASPIRACLE

1lbd

RETINOID X RECEPTOR

3eyb

NUCLEAR HORMONE RECEPTOR RXR

4iqr

HEPATOCYTE NUCLEAR FACTOR 4-ALPHA

2gl8

RETINOIC ACID RECEPTOR RXR-GAMMA

2q60

RETINOID X RECEPTOR

4j5x

RETINOIC ACID RECEPTOR RXR-ALPHA, NUCLEAR RECEPTOR COACTIVATOR 1

1lv2

HEPATOCYTE NUCLEAR FACTOR 4-GAMMA

3dzu

PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR GAMMA

3kmr

RETINOIC ACID RECEPTOR ALPHA

The list reveals structures in the Protein Data Bank that contain binding site similar to that of ERβ as represented by the structure under the PDB ID 1NDE.


Table 3: PASS prediction results.

Rank

Pa

Pi

Activity

213

0,177

0,022

Estrogen antagonist

234

0,148

0,024

Estrogen receptor beta antagonist

247

0,140

0,026

Estrogen agonist

371

0,048

0,021

Estrogen beta receptor agonist

Predicted activities with relation to the estrogen modulation or action of ERβ. Rank: overall PASS rank of this activity by VEDT; Pa: Probability that VEDT exhibits the given activity; Pi: Probability that VEDT does not exhibit the given activity.


Science Repository

Figure 1: Comparison of the structures of δ-tocopherol and δ-tocotrienol.


Science Repository

Figure 2: Docked pose of VEDT as identified by VTS. VEDT, coloured in magenta, forms hydrogen bonds (yellow dashed lines) with Glu-305 and Arg-346.



References

  1. Constantina Constantinou, Andreas Papas, Andreas I Constantinou (2008) Vitamin E and cancer: An insight into the anticancer activities of vitamin E isomers and analogs. Int J Cancer 123: 739-752. [Crossref]
  2. Ammu Radhakrishnan, Dulanthi Tudawe, Srikumar Chakravarthi, Gan Seng Chiew, Nagaraja Haleagrahara (2014) Effect of gamma-tocotrienol in counteracting oxidative stress and joint damage in collagen-induced arthritis in rats. Exp Ther Med 7: 1408-1414. [Crossref]
  3. Chandan K Sen, Savita Khanna, Sashwati Roy (2006) Tocotrienols: Vitamin E beyond tocopherols. Life Sci 78: 2088-2098. [Crossref]
  4. G van Poppel, H van den Berg (1997) Vitamins and cancer. Cancer Lett 114: 195-202. [Crossref]
  5. Mokenge P Malafa, Frida D Fokum, Arian Mowlavi, Mary Abusief, Michele King (2002) Vitamin E inhibits melanoma growth in mice. Surgery 131: 85-91. [Crossref]
  6. Suryatheja Ananthula, Parash Parajuli, Fathy A Behery, Alaadin Y Alayoubi, Khalid A El Sayed et al. (2014) Oxazine derivatives of gamma- and delta-tocotrienol display enhanced anticancer activity in vivo. Anticancer Res 34: 2715-2726. [Crossref]
  7. Ji Liu, Eunice Yuen Ting Lau, Jiezhong Chen, Joan Yong, Kai Dun Tang et al. (2014) Polysaccharopeptide enhanced the anti-cancer effect of gamma-tocotrienol through activation of AMPK. BMC Complement Altern Med 14: 303. [Crossref]
  8. Kazuhiro Nakaso, Naoko Tajima, Yosuke Horikoshi, Masato Nakasone, Takehiko Hanaki et al. (2014) The estrogen receptor beta-PI3K/Akt pathway mediates the cytoprotective effects of tocotrienol in a cellular Parkinson's disease model. Biochim Biophys Acta 1842: 1303-1312. [Crossref]
  9. Magda Nasr, Noha Nafee, Hoda Saad, Amani Kazem (2014) Improved antitumor activity and reduced cardiotoxicity of epirubicin using hepatocyte-targeted nanoparticles combined with tocotrienols against hepatocellular carcinoma in mice. Eur J Pharm Biopharm 88: 216-225. [Crossref]
  10. Paul W Sylvester, Sumit J Shah (2005) Mechanisms mediating the antiproliferative and apoptotic effects of vitamin E in mammary cancer cells. Front Biosci 10: 699-709. [Crossref]
  11. Raffaella Comitato, Guido Leoni, Raffaella Canali, Roberto Ambra, Kalanithi Nesaretnam et al. (2010) Tocotrienols activity in MCF-7 breast cancer cells: involvement of ERbeta signal transduction. Mol Nutr Food Res 54: 669-678. [Crossref]
  12. Raffaella Comitato, Kalanithi Nesaretnam, Guido Leoni, Roberto Ambra, Raffaella Canali et al. (2009) A novel mechanism of natural vitamin E tocotrienol activity: involvement of ERbeta signal transduction. Am J Physiol Endocrinol Metab 297: E427-E437. [Crossref]
  13. Lean Teik Ng, Liang Tzung Lin, Chiu Lan Chen, Hsiu Wen Chen, Shu Jing Wu et al. (2014) Anti-melanogenic effects of delta-tocotrienol are associated with tyrosinase-related proteins and MAPK signaling pathway in B16 melanoma cells. Phytomedicine 21: 978-983. [Crossref]
  14. Pamela J Hodul, Yanbin Dong, Kazim Husain, Jose M Pimiento, Jiandong Chen et al. (2013) Vitamin E delta-tocotrienol induces p27(Kip1)-dependent cell-cycle arrest in pancreatic cancer cells via an E2F-1-dependent mechanism. PLoS One 8: e52526. [Crossref]
  15. Kazim Husain, Barbara A Centeno, Dung Tsa Chen, William J Fulp, Marta Perez et al. (2013) Prolonged survival and delayed progression of pancreatic intraepithelial neoplasia in LSL-KrasG12D/+;Pdx-1-Cre mice by vitamin E delta-tocotrienol. Carcinogenesis 34: 858-863. [Crossref]
  16. Kazim Husain, Barbara A Centeno, Dung Tsa Chen, Sunil R Hingorani, Said M Sebti et al. (2013) Vitamin E delta-tocotrienol prolongs survival in the LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) transgenic mouse model of pancreatic cancer. Cancer Prev Res (Phila) 6: 1074-1083. [Crossref]
  17. Masao Yamasaki, Misato Nishimura, Yoichi Sakakibara, Masahito Suiko, Kazuhiro Morishita et al. (2014) Delta-tocotrienol induces apoptotic cell death via depletion of intracellular squalene in ED40515 cells. Food Funct 5: 2842-2849. [Crossref]
  18. Mario Köster, Søren Lykke Andersen, Yasser A Elnakady, Klaus Gerth, Peter Washausen et al. (2003) Ratjadones inhibit nuclear export by blocking CRM1/exportin 1. Exp Cell Res 286: 321-331. [Crossref]
  19. Jacqueline C Hargis, Sai Lakshmana Vankayala, Justin K White, H Lee Woodcock (2014) Identification and Characterization of Noncovalent Interactions That Drive Binding and Specificity in DD-Peptidases and beta-Lactamases. J Chem Theory Comput 10: 855-864. [Crossref]
  20. Gary T Pauly, Natalia A Loktionova, Qingming Fang, Sai Lakshmana Vankayala, Wayne C Guida et al. (2008) Substitution of aminomethyl at the meta-position enhances the inactivation of O6-alkylguanine-DNA alkyltransferase by O6-benzylguanine. J Med Chem 51: 7144-7153. [Crossref]
  21. Brian K Shoichet (2004) Virtual screening of chemical libraries. Nature 432: 862-865. [Crossref]
  22. G C Terstappen, A Reggiani (2001) In silico research in drug discovery. Trends Pharmacol Sci 22: 23-26. [Crossref]
  23. Sai Lakshmana Vankayala, Jacqueline C Hargis, H Lee Woodcock (2012) Unlocking the binding and reaction mechanism of hydroxyurea substrates as biological nitric oxide donors. J Chem Inf Model 52: 1288-1297. [Crossref]
  24. Sai Lakshmana Vankayala, Jacqueline C Hargis, H Lee Woodcock (2013) How does catalase release nitric oxide? A computational structure-activity relationship study. J Chem Inf Model 53: 2951-2961. [Crossref]
  25. P S Charifson, J J Corkery, M A Murcko, W P Walters (1999) Consensus scoring: A method for obtaining improved hit rates from docking databases of three-dimensional structures into proteins. J Med Chem 42: 5100-5109. [Crossref]
  26. Abdulilah Ece, Fatma Sevin (2013) The discovery of potential cyclin A/CDK2 inhibitors: a combination of 3D QSAR pharmacophore modeling, virtual screening, and molecular docking studies. Med Chem Res 22: 5832-5843.
  27. Douglas R Houston, Malcolm D Walkinshaw (2013) Consensus docking: improving the reliability of docking in a virtual screening context. J Chem Inf Model 53: 384-390. [Crossref]
  28. Xiaofeng Ji, Yuan Zheng, Wei Wang, Jun Sheng, Jianhua Hao et al. (2013) Virtual screening of novel reversible inhibitors for marine alkaline protease MP. J Mol Graph Model 46: 125-131. [Crossref]
  29. Yuri Pevzner, Daniel Santiago, Jacqueline L von Salm, Rainer Metcalf, Kenyon G Daniel et al. (2014) Virtual target screening to rapidly identify potential protein targets of natural products in drug discovery. AIMS Mol Sci 1: 49-66.
  30. Daniel N Santiago, Yuri Pevzner, Ashley A Durand, MinhPhuong Tran, Rachel R Scheerer et al. (2012) Virtual target screening: validation using kinase inhibitors. J Chem Inf Model 52: 2192-2203. [Crossref]
  31. Zhuo Chen, Xin Wang, Weiping Zhu, Xianwen Cao, Linjiang Tong et al. (2011) Acenaphtho[1,2-b]pyrrole-based selective fibroblast growth factor receptors 1 (FGFR1) inhibitors: design, synthesis, and biological activity. J Med Chem 54: 3732-3745. [Crossref]
  32. Xiaofeng Liu, Sisheng Ouyang, Biao Yu, Yabo Liu, Kai Huang et al. (2010) PharmMapper server: a web server for potential drug target identification using pharmacophore mapping approach. Nucleic Acids Res 38: W609-W614. [Crossref]
  33. Yu V Borodina, D A Filimonov, Vladimir Poroikov (1996) Computer-aided prediction of prodrug activity using the pass system. Pharm Chem J 30: 760-763.
  34. D A Filimonov, V V Poroĭkov, E I Karaicheva, R K Kazarian, A P Budunova et al. (1995) The computerized prediction of the spectrum of biological activity of chemical compounds by their structural formula: the PASS system. Prediction of Activity Spectra for Substance. Eksp Klin Farmakol 58: 56-62. [Crossref]
  35. Alexey Lagunin, Dmitry Filimonov, Vladimir Poroikov (2010) Multi-targeted natural products evaluation based on biological activity prediction with PASS. Curr Pharm Des 16: 1703-1717. [Crossref]
  36. Janez Konc, Tomo Cesnik, Joanna Trykowska Konc, Matej Penca, Dušanka Janežič (2012) ProBiS-database: precalculated binding site similarities and local pairwise alignments of PDB structures. J Chem Inf Model 52: 604-612. [Crossref]
  37. Janez Konc, Dusanka Janezic (2010) ProBiS: a web server for detection of structurally similar protein binding sites. Nucleic Acids Res 38: W436-W440. [Crossref]
  38. Janez Konc, Dusanka Janezic (2010) ProBiS algorithm for detection of structurally similar protein binding sites by local structural alignment. Bioinformatics 26: 1160-1168. [Crossref]
  39. Janez Konc, Dusanka Janezic (2012) ProBiS-2012: web server and web services for detection of structurally similar binding sites in proteins. Nucleic Acids Res 40: W214-W221. [Crossref]
  40. H M Berman, J Westbrook, Z Feng, G Gilliland, T N Bhat et al. (2000) The Protein Data Bank. Nucleic Acids Res 28: 235-242. [Crossref]
  41. F C Bernstein, T F Koetzle, G J Williams, E F Meyer Jr, M D Brice et al. (1977) The Protein Data Bank: a computer-based archival file for macromolecular structures. J Mol Biol 112: 535-542. [Crossref]
  42. Noel M O'Boyle, Michael Banck, Craig A James, Chris Morley, Tim Vandermeersch et al. (2011) Open Babel: An open chemical toolbox. J Cheminform 3: 33. [Crossref]
  43. Filimonov D, Poroikov V, Borodina Y, Gloriozova T (1999) Chemical Similarity Assessment through Multilevel Neighborhoods of Atoms: Definition and Comparison with the Other Descriptors. J Chem Inf Model 39: 666-670.
  44. Brad R Henke, Thomas G Consler, Ning Go, Ron L Hale, Dana R Hohman et al. (2002) A new series of estrogen receptor modulators that display selectivity for estrogen receptor beta. J Med Chem 45: 5492-5505. [Crossref]
  45. C A Lipinski (2000) Drug-like properties and the causes of poor solubility and poor permeability. J Pharm Toxicol Methods 44: 235-249. [Crossref]
  46. Christopher Andrew Lipinski (2002) Poor Aqueous Solubility – an Industry wide problem in drug discovery. Am Pharm Rev 1-3.