Jeganathan Manivannan*ab,
Thangarasu Silambarasanb,
Rajendran Kadarkarairaja and
Boobalan Rajab
aVascular Biology Lab, AU-KBC Research Centre, MIT campus, Anna University, Chennai 600 044, Tamil Nadu, India. E-mail: drjmaniau@gmail.com; Tel: +91 9894907931
bCardiovascular Biology Lab, Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
First published on 3rd September 2015
Natural compounds can interact with multiple cellular target proteins and may be prioritized as drug leads. There is a need for prioritization of compounds that protect cardiovascular systems from pathological conditions. Here we prioritize morin, veratric acid, piperine, syringic acid, vanillic acid, diosgenin, diosmetin and sinapic acid that were already identified as cardioprotective molecules in our previous studies through multi-level data integration. In this study, initially we predict targets of the above-mentioned compounds by reverse pharmacophore (PharmMapper) and structural similarity based target-screening methods. We also explored the compound–target pathways (Biocarta and KEGG) and disease relationships. Further, we chose public microarray transcriptomic data from GEO to prioritize important pathogenic targets (heart failure, cardiac hypertrophy, vascular dysfunction and atherosclerosis), and we explored the interaction potential of the above compounds on the targets via blind docking (AutoDock Vina). Moreover, the multi target action of compounds was revealed by target information retrieved from large-scale text mining and organized databases (HIT and TCMID). The drug likeness profile and toxicity prediction was achieved based on Lipinski's rule and structural similarity search (ProTox). The observed results have demonstrated that the multi target potential and less toxic nature mean these molecules can be prioritized as lead compounds for cardiovascular diseases.
Systems pharmacology provides powerful new tools and approaches for natural product lead discovery. Recently, with this approach Li et al. revealed a multiple drug–target prediction and validation, and network pharmacology techniques, to shed new lights on the effectiveness and mechanism of Compound Danshen Formula.7 Formerly, Yu et al. explored the mifepristone target pathway, which is a good example to identify chemotherapeutic potential seamlessly from systems pharmacology.8 Zhang et al. indicates the potential applications of multi-level and multi-targeting therapies in cancer treatment.9
Natural products have been considered as an important source of lead compounds for drug discovery and more than 50 percent of FDA-approved drugs were natural products or natural product derivatives.10 Shukla et al. suggests that supplementation of cardiovascular responsive natural products needs to be considered in all populations who have high prevalence of CVD.11 With the development of large-scale network analysis, researchers have recently begun to explore the action mechanism of bioactive compounds in the context of biological networks, e.g. drug–target network.10 Zheng et al. put forth novel strategies of systems pharmacology for multi-target drug discovery from natural products for CVD based on network pharmacology methods.12
In this study, we aimed to prioritize natural molecules, morin, veratric acid, piperine, syringic acid, vanillic acid, diosgenin, diosmetin and sinapic acid, which was explored as cardiovascular protective and antihypertensive based on our previous laboratory studies. In our laboratory, we previously explored the following results; morin attenuates hypertension and oxidative stress in deoxycorticosterone acetate-salt (DOCA-salt) induced hypertensive rats. Further, it was shown to be a cardiovascular protective molecule with biochemical and histopathological evidences.13,14 Another study with similar hypertensive rat model illustrates that diosmin treatment dose-dependently prevents hypertension and cardiovascular system from oxidative stress.15 Consequently, our experiment also proves that, diosmin pretreatment improves cardiac function after ischemia/reperfusion.16 With L-NAME model of pharmacological hypertension we explored the preventive effect of veratric acid,17,18 piperine,19 syringic acid,20 vanillic acid,21 and sinapic acid22 along with their antioxidant potential. Further, we indicated that sinapic acid prevents ischemia/reperfusion injury and coronary dysfunction in rat heart and attenuates oxidative stress dependent H9c2 cardiomyoblast cell injury.23 Apart from this, we demonstrated that diosgenin, a steroidal saponin prevents cardiovascular remodeling with its antioxidant efficacy.24,25
Based on our previous results, we hypothesize that, along with antioxidant potential these molecules may exhibit its cardioprotective effect, due to their interaction with number of pathological pathways. Consequently, we propose this study to explore the compound targets-pathway related to cardiovascular dysfunction of above mentioned compounds via multi way systems pharmacological approaches such as (1) target screening via pharmacophore mapping approach and pathway analysis; (2) transcriptomic (microarray) reanalysis based target prioritization and docking; (3) curated natural compound database mining; (4) drug likeness and toxicity prediction to prioritize them as cardiovascular protective lead molecules.
Based on the previous studies,30,31 in this study we hypothesized that, one compound may interacts with multiple components of various cellular pathways and disease related pathways as many natural compounds had such as curcumin.32 Here, we have submitted the Mol2 converted compound file to PharmMapper and chosen top 100 (considered as potential targets) among top 300. Their associated pathways were dissected by DAVID (Database for Annotation, Visualization and Integrated Discovery) Bioinformatics online tools (http://www.david.abcc.ncifcrf.gov/).33 Cellular pathway association were obtained according to the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (http://www.genome.jp/kegg/) and Biocarta (http://www.biocarta.com/genes/index.asp)34 and the annotations of target–disease relationship terms were obtained from annotations of Genetic Association Database (genetic_association_db_disease).35 We selected the significant terms (P < 0.05) for further explanations about the targeting pathways.
Moreover, in order to explore the possible targets of compounds via structure likeness, in this study we utilizes two potential web servers that uses structural similarity scores to find the possible targets. First, we used SwissTargetPrediction (http://www.swisstargetprediction.ch)36 web server to predict the targets of bioactive molecules based on a combination of 2D and 3D similarity measures with known ligands. Second, we utilized SuperPred (http://prediction.charite.de)37 web server, connects chemical similarity (2D structure) of drug-like compounds with molecular targets.
Microarray study and GEO accession | Focused modulated biological processes and pathways | Important known targets of the pathway |
---|---|---|
a JNK, c-Jun N-terminal kinases; ACE, angiotensin converting enzyme; REN, renin; ROCK, Rho-associated kinase; ECE1, endothelin converting enzyme 1; GSK-3β, glycogen synthase kinase-3β; TGF-β, transforming growth factor-β; NF-κB, nuclear factor-κB; ERK, extracellular signal-regulated kinase; JAK2, Janus kinase 2; COX2, cyclooxygenase-2. | ||
Vascular dysfunction in SHR (GSE8051) | Vasoconstriction (BP); JNK cascade (BP); vasodilation (BP) | ACE (PDB 1O8A), REN (PDB 2V0Z), ROCK (PDB 2ESM), ECE1 (PDB 3DWB), |
Atherogenesis in ApoE deficient mice (GSE19286) | Canonical Wnt receptor signaling pathway (BP); Wnt receptor signaling pathway (BP) | GSK-3β (PDB 1UV5) |
Heart failure in SHR (GSE19210) | TGF-β signaling pathway; positive regulation of I-κB kinase/NF-κB cascade (BP) | TGF-βR1 (PDB 1PY5), NF-κB (PDB 1SVC) |
Ang II treated heart (GSE59437) | Inflammatory process (BP); positive regulation of ERK1 and ERK2 cascade (BP) | JAK2 (PDB 3RVG), ERK2 (PDB 1TVO), COX2 (PDB 6COX) |
To explore the putative binding site of compounds on prioritized target structure, we utilized a blind docking approach via AutoDock Vina in PyRx0.8 which is an easy-to-use user interface that significantly improves the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading.44 Recently, a molecular docking study utilizing the above tool explores the potential target of Tanshinone IIA for acute promyelocytic leukemia.30 In this study, the grid box was set large enough to cover the whole protein (targets) for blind docking similar to a previous study that explore HIV protease inhibitors.45–47
Additionally, Ligplot plus version 1.4 (http://www.ebi.ac.uk/thornton-srv/software/LigPlus)48 was used for the 2D visualization of ligand–receptor interaction. The Catalytic Site Atlas (CSA) (http://www.ebi.ac.uk/thornton-srv/databases/CSA)49 was used to retrieve the information of enzyme catalytic site residues in order to explore the possible orthosteric (binding at the active site) and allosteric (binding elsewhere) binding mode of ligands with receptor.
S. no. | Compound | Biocarta | KEGG pathway |
---|---|---|---|
a NFAT, nuclear factor of activated T cells; VEGF, vascular endothelial growth factor; IL-2, interleukin-2; FXR, farnesoid X receptor; LXR, liver X receptor; PPAR, peroxisome proliferator-activated receptor; NOD, nucleotide-binding oligomerization domain. | |||
1 | Morin | Cell cycle; NFAT and hypertrophy of the heart (transcription in the broken heart) | Cell cycle; VEGF signaling pathway |
2 | Piperine | NFAT and hypertrophy of the heart (transcription in the broken heart); insulin signaling pathway; IL-2 signaling pathway | Focal adhesion; VEGF signaling pathway; aldosterone-regulated sodium reabsorption |
3 | Veratric acid | NFAT and hypertrophy of the heart (transcription in the broken heart); apoptotic signaling in response to DNA damage | VEGF signaling pathway; renin–angiotensin system |
4 | Syringic acid | — | VEGF signaling pathway |
5 | Vanillic acid | Ras signaling pathway; Ang II mediated activation of JNK pathway via Pyk2 dependent signaling | VEGF signaling pathway |
6 | Diosgenin | FXR and LXR regulation of cholesterol metabolism; NFAT and hypertrophy of the heart (transcription in the broken heart) | PPAR signaling pathway; aldosterone-regulated sodium reabsorption |
7 | Diosmetin | Cell cycle; NFAT and hypertrophy of the heart (transcription in the broken heart) | NOD-like receptor signaling pathway; cell cycle |
8 | Sinapic acid | Ang II mediated activation of JNK pathway via Pyk2 dependent signaling; actions of nitric oxide in the heart; ras signaling pathway | Focal adhesion; type II diabetes mellitus |
Compound | Target | Similar compounds (2D/3D) | Target class |
---|---|---|---|
Morin | Estrogen receptor (by homology) | 65/32 | Transcription |
Estrogen receptor β | 65/32 | Transcription | |
Solute carrier family 22 member 12 | 1/1 | Unclassified | |
Aldose reductase (by homology) | 25/68 | Enzyme | |
Arachidonate 5-lipoxygenase | 13/52 | Enzyme | |
Piperine | Amine oxidase [flavin-containing] A (by homology) | 74/8 | Enzyme |
Amine oxidase [flavin-containing] B | 76/9 | Enzyme | |
Histone deacetylase 1 | 47/4 | Enzyme | |
Histone deacetylase 2 (by homology) | 47/4 | Enzyme | |
Histone deacetylase 3 | 45/4 | Enzyme | |
D(2) dopamine receptor | 41/21 | Membrane receptor | |
D(3) dopamine receptor | 26/19 | Membrane receptor | |
Veratric acid | Carbonic anhydrase-1,2,4,9,12,(3,5A,5B,13-by homology) | 17/20; 17/20; 3/4; 10/9; 10/10; 17/20; 17/20; 17/20; 17/20 | Enzyme |
Tyrosine-protein kinase Lck | 3/1 | Tyr kinase | |
Tyrosine-protein kinase Fyn | 2/2 | Tyr kinase | |
Proto-oncogene tyrosine-protein kinase Src (by homology) | 2/2 | Tyr kinase | |
Syringic acid | Carbonic anhydrase-1,2,3,4,5A,5B,6,7,9,12,(13-by homology) | 5/20; 5/20; 5/20; 1/4; 5/20; 5/20; 2/6; 5/20; 3/9; 3/10; 5/20 | Enzyme |
Thiopurine S-methyltransferase | 1/4 | Enzyme | |
Vanillic acid | Carbonic anhydrase-1,2,3,4,5A,5B,6,9,12,14,(3,7,13-by homology) | 17/23; 17/23; 6/4; 17/23; 17/23; 6/6; 9/9; 8/10; 8/10; 17/23; 17/23; 17/23 | Enzyme |
Thiopurine S-methyltransferase | 4/4 | Enzyme | |
Diosgenin | Oxysterols receptor LXR-β | 12/18 | Transcription factor |
Oxysterols receptor LXR-α | 12/18 | Transcription factor | |
3-Hydroxy-3-methylglutarylcoenzyme A reductase | 100/74 | Enzyme | |
Diosmetin | Aldose reductase (by homology) | 24/70 | Enzyme |
NADPH oxidase 4 | 8/8 | Enzyme | |
Xanthine dehydrogenase/oxidase | 2/14 | Enzyme | |
Sinapic acid | Carbonic anhydrase-1,2,5A,6,7,9,12,(3,5B,13-by homology) | 29/11; 29/11; 29/11; 2/7; 29/11; 7/10; 5/10; 29/11; 29/11; 29/11 | Enzyme |
Epidermal growth factor receptor | 5/16 | Tyr kinase | |
Receptor tyrosine-protein kinase erbB-2 | 5/16 | Tyr kinase | |
Receptor tyrosine-protein kinase erbB-3 (by homology) | 5/16 | Tyr kinase |
PDB ID | Morin | Piperine | Veratric acid | Syringic acid | Vanillic acid | Diosgenin | Diosmetin | Sinapic acid |
---|---|---|---|---|---|---|---|---|
a Docking score/interaction potential of compounds with targets (kcal mol−1). | ||||||||
1O8A | −8.6 | −9.0 | −6.3 | −6.0 | −6.3 | −11.8 | −8.4 | −6.3 |
1PY5 | −9.8 | −8.7 | −6.2 | −6.1 | −6.1 | −11.0 | −10.1 | −7.0 |
1SVC | −6.4 | −6.5 | −5.0 | −4.8 | −4.7 | −8.1 | −6.5 | −5.1 |
1TVO | −8.4 | −7.2 | −5.4 | −5.5 | −5.6 | −10.1 | −8.1 | −5.7 |
1UV5 | −8.9 | −8.6 | −6.2 | −5.8 | −5.9 | −9.3 | −9.1 | −6.7 |
2ESM | −7.8 | −7.7 | −5.8 | −5.4 | −5.7 | −8.9 | −7.7 | −5.9 |
2V0Z | −8.0 | −7.8 | −5.1 | −5.2 | −5.8 | −10.6 | −7.7 | −6.1 |
3DWB | −8.1 | −8.5 | −6.2 | −6.5 | −6.3 | −9.9 | −8.4 | −6.9 |
3RVG | −8.3 | −7.9 | −5.8 | −5.8 | −6.0 | −9.3 | −8.6 | −6.4 |
6COX | −8.8 | −8.6 | −6.2 | −6.0 | −6.3 | −10.2 | −9.1 | −6.7 |
The surrounding amino acid residues of conformations that shown highest interaction potential (ligand–receptor complex) were illustrated along with the catalytic residues of the enzymes in ESI Table 2.† While focusing on the inhibitory relationship at the enzyme level, we found that majority of the ligand–receptor interactions were allosteric and only few of them were shown to be directly contacted (possible hydrogen bonds) with receptor catalytic residues. From the catalytic site atlas we could directly derived the information of enzyme catalytic residues only for ACE and others were predicted by homology. From the results we observed that, diosmetin and morin have shown possible hydrogen bonding contact with His353 and His513 residues of ACE. Vanillic acid and veratric acid were shown to be interacted near to the cyclooxygenase active site residues (Tyr385, Gln203 and His207). Further morin, piperine and sinapic acid were shown hydrogen bond interaction with Arg738, His732, Glu608 and Asp671 residues of ECE1. Furthermore, piperine conformation was shown to be associated with the renin active site environment (Asp215). Diosmetin shown hydrogen bond contact with Asp202 residue of ROCK. Diosgenin and piperine shown to be interacted near the catalytic residue Gln185 of Gsk-3β. In the case of TGFβR1, diosmetin showing possible hydrogen bonding contact with Lys337. Diosgenin binds near to the environment of catalytic residues Lys151 and Ser153 of ERK2.
Compound | Target | Identification method |
---|---|---|
Morin | Apoptosis regulator BAX | STITCH |
Endothelin-1 | Text mining | |
Xanthine dehydrogenase/oxidase | Text mining | |
Arachidonate 5-lipoxygenase | Text mining | |
Piperine | Tumor necrosis factor | STITCH, text mining |
Inhibitor of NF-κB kinase subunit α | Text mining | |
Inhibitor of NF-κB kinase subunit α | Text mining | |
Intercellular adhesion molecule 1 | Text mining | |
Interleukin-6 | Text mining | |
Veratric acid | Not-available | |
Syringic acid | Prostaglandin G/H synthase 2 | STITCH |
C–C motif chemokine 16 | STITCH | |
Vanillic acid | Nitric oxide synthase, endothelial | Text mining |
Prostaglandin G/H synthase 2 | STITCH | |
Diosgenin | Superoxide dismutase [Cu–Zn] | Text mining |
Catalase | Text mining | |
Vascular endothelial growth factor A | Text mining | |
Fatty acid synthase | Text mining | |
Lipoprotein lipase | STITCH | |
Diosmetin | Prostaglandin G/H synthase 2 | STITCH |
Tumor necrosis factor | STITCH | |
Sinapic acid | Choline O-acetyltransferase | Text mining |
Compound | Morin | Piperine | Veratric acid | Syringic acid | Vanillic acid | Diosgenin | Diosmetin | Sinapic acid |
---|---|---|---|---|---|---|---|---|
milog![]() |
1.881 | 3.332 | 1.495 | 1.204 | 1.187 | 5.932 | 2.282 | 1.265 |
TPSA | 131.351 | 38.777 | 55.767 | 75.995 | 66.761 | 38.696 | 100.129 | 75.995 |
n atoms | 22 | 21 | 13 | 14 | 12 | 30 | 22 | 16 |
MW | 302.238 | 285.343 | 182.175 | 198.174 | 168.148 | 414.63 | 300.266 | 224.212 |
nON | 7 | 4 | 4 | 5 | 4 | 3 | 6 | 5 |
nOHNH | 5 | 0 | 1 | 2 | 2 | 1 | 3 | 2 |
n violations | 0 | 0 | 0 | 0 | 0 | 1 | 0 | 0 |
n rotb | 1 | 3 | 3 | 3 | 2 | 0 | 2 | 4 |
MV | 240.084 | 267.741 | 162.136 | 170.154 | 144.608 | 419.383 | 249.594 | 197.571 |
S. no. | Compound | LD50 (mg kg−1) | Toxicity class |
---|---|---|---|
1 | Morin | 3919 | 5 – may be harmful if swallowed (2000 < LD50 ≤ 5000 mg kg−1) |
2 | Piperine | 330 | 4 – harmful if swallowed (300 < LD50 ≤ 2000 mg kg−1) |
3 | Veratric acid | 2000 | 4 – harmful if swallowed (300 < LD50 ≤ 2000 mg kg−1) |
4 | Syringic acid | 1700 | 4 – harmful if swallowed (300 < LD50 ≤ 2000 mg kg−1) |
5 | Vanillic acid | 2000 | 4 – harmful if swallowed (300 < LD50 ≤ 2000 mg kg−1) |
6 | Diosgenin | 8000 | 6 – non-toxic (LD50 > 5000 mg kg−1) |
7 | Diosmetin | 3919 | 5 – may be harmful if swallowed (2000 < LD50 ≤ 5000 mg kg−1) |
8 | Sinapic acid | 1772 | 4 – harmful if swallowed (300 < LD50 ≤ 2000 mg kg−1) |
By means of target prediction via structural similarity (SwissTargetPrediction), we found that carbonic anhydrase (CA) isozymes might be the main target of our study compounds. Here veratric acid, vanillic acid, sinapic acid and syringic acid have shown more similarity with 2D/3D structure of ligands that targets CA. CA and its inhibitors are relevant to many physiological processes and diseases. An earlier study suggest that the vasodilator effect of thiazide diuretics results primarily from inhibition of vascular smooth muscle cell CA and inhibition of CA isoenzymes (I, II, III, IV, V, IX and XII), expressed in the cells of cardiovascular system may involved in vasodilation and other cardiovascular regulations.65,66 Veratric acid may targets the Src family members Lck and Fyn, which are known to be involved in ox-LDL and H2O2-mediated activation of intracellular signaling events in vascular smooth muscle cells67 and also in CD36 scavenger receptors mediated foam cell formation in atherosclerosis.68 In the case of diosgenin, because of its structural relevancy it may be the inhibitor of 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase and LXRs. Statins (inhibitor of HMG-CoA reductase) treatment is regarded as one of the most effective methods for the stabilization of vulnerable atherosclerotic plaques, and beneficial in patients with coronary heart disease.69 In addition, LXRs are target of oxysterols and LXRs lie at the intersection of lipid metabolism, inflammation and pathways involved in progression of atherosclerotic lesions and CVD.70 The next compound piperine may be the potential inhibitor of histone deacetylases (HDACs) 1, 2 and 3. A previous study indicates that class I HDACs promote pathological cardiac hypertrophy and among the class I HDACs, HDAC2 is activated by hypertrophic stresses.71 Along with the above, it also targets the monoamine oxidase A (MAOA) that regulates the metabolism of key neurotransmitters that has been associated with cardiovascular risk factors.72 While specifically focusing on morin, one of its important predicted targets is estrogen receptor beta (ERbeta). It was previously indicated that, 17-beta-estradiol (E2) acts mainly through ERbeta and alleviates the important signaling in Ang II induced cardiac hypertrophy and fibrosis in female mice.73 Another important target of morin was arachidonate 5-lipoxygenase, which is related with the biosynthesis of lipid inflammatory mediators leukotrienes within the atherosclerotic lesion and inhibitors of 5-lipoxygenase pathway was now evaluated in clinical trials of patients with cardiovascular disease.74 While focusing on diosmetin, prediction results have shown that NADPH oxidases and xanthine dehydrogenase/oxidase may be its targets. Current knowledge on redox signaling pathways indicates that above two enzymes plays vital role in the pathogenesis of cardiac hypertrophy.75,76 In case of sinapic acid, the target may be the class of tyrosine kinases such as epidermal growth factor receptor (EGFR) and receptor tyrosine-protein kinase (erbB-2) and their signaling modulation contributes to the development of atherosclerosis, cardiac hypertrophy and hypertension.77 Along with the above, SuperPred based structural similarity search identify some new targets and many were overlapped with the targets predicted by SwissTargetPrediction method. It indicates vanillic acid may target arachidonate 15-lipoxygenase and veratric acid may target phosphodiesterase-4. Previous literatures have shown that, macrophage 12/15-lipoxygenase plays a dominant role in the development of atherosclerosis by promoting endothelial inflammation and foam cell formation78 and suggest that inhibition of phosphodiesterase-4 ameliorates hypertension-induced impairment of learning and memory functions.79
In drug–target discovery, virtual screening method is essential and have been applied as a complement to experimental techniques to rapid screening and predicting the location of functional binding pockets of ligands on targets and the blind docking calculations mediates the prediction of the above on the entire protein surface.80,81 In this study, docking receptors were prioritized components of important pathways deregulated (up regulated) during cardiovascular pathogenic progression obtained from reanalysis of microarray experiments. Similarly, an earlier study identified S100A8 as a prospective biomarker for kidney cancer from microarray-based transcriptomics experiments and docking analysis have showed that aspirin, celecoxib, dexamethasone and diclofenac binds to S100A8 and may inhibit downstream signaling in kidney cancer.82 Additionally, an earlier study proposed a possible mechanism of activation of acetylcholine binding protein via the “blind docking” of allosteric modulators.83 In the identical way, in this study, we performed blind docking of ligands via AutoDock Vina on prioritized pathway targets. All the compounds have shown good affinity on microarray prioritized pathway targets, which indicates the plausible cardioprotective nature of the compounds. When we reanalyzed the microarray data of vascular dysfunction in SHR, among important/prioritized pathways, we chosen biological process terms (GO) vasoconstriction and vasodilation. It is well known that, vascular dysfunction is one of the major targets of hypertension and its associated complications. We plausibly chosen components of vascular contraction pathway such as ACE, renin, ROCK and ECE1 as prioritized targets based on previous literature supports.84–86 ACE plays an important physiological role in regulation of blood pressure by converting Ang I to Ang II, a potent vasoconstrictor.84,85 Consequently, the inhibition of ACE activity is a major target in the prevention of hypertension and marine-derived natural ACE inhibitors are proposed as novel therapeutic drug candidates to treat hypertension.87 RhoA/ROCK, known regulators of vascular tone which belong to serine/threonine (Ser/Thr) protein kinase family, is reported to involve in the organization of the actin cytoskeleton, consequently in vascular regulation.88 Supportively, a previous study indicates that, virtual screening approach can able to predict ROCK1 interacting compound that also prove its inhibitory activity in vivo.89 In the view of endothelin converting enzyme, hyperactivation of the endothelin system has been implicated in the pathogenesis of various cardiovascular disorders including myocardial infarction, restenosis, hypertension, heart failure and chagas cardiopathy; and there is growing interest in blockade of endothelin formation.90 From the microarray reanalysis of atherogenesis process, we prioritize an important central regulator (target) from Wnt signaling pathway (GSK-3β). Existence of a common mechanism of accelerated atherosclerosis involving endoplasmic reticulum stress signaling through activation of GSK-3β and valproate supplementation blocked GSK-3β activation and attenuated the development of atherosclerosis.91,92 From the enriched pathways of heart failure microarray data, we mainly focused on TGF-β signaling pathway, consequently its component TGF-β type 1 receptor (TGF-βR1). In cellular systems, TGF-β signals via a classical pathway, binding to TGF-β type 2 receptor (TGF-βR2) to activate TGF-βR1 and subsequent signaling. In myocyte, TGF-β stimulation induces hypertrophy, whereas TGF-β1 deficiency restrict Ang II-mediated cardiac hypertrophy.93–95 Next, we targeted the NF-κB signaling pathway that enriched for heart failure, because prolonged activation of NF-κB appears to be promotes heart failure by triggering chronic inflammation.96 Additionally, from pathways significantly deregulated during Ang II induced hypertrophy, we prioritize components of inflammatory and mitogen activated kinase signaling. Here we had chosen JAK2, COX2 and ERK2 as targets, since their functional importance in the above pathways.97,98 Previous literatures have shown that, JAK2 kinase plays an important role in left ventricular remodeling during pressure overload hypertrophy and suggests that HDAC2 might be a downstream effector of JAK2 to mediate cardiac hypertrophic response by pressure overload or Ang-II.99,100 COX2 is an important mediator of inflammation, cardiac hypertrophy and failure, further in cardiomyocytes, it involves in Ang II-induced oxidative stress and also contributes inflammatory cardiovascular changes.101–103 Since the above mentioned targets are important mediators of major cardiovascular pathogenic events, we postulate the study compounds as cardiovascular protective agents because of their predicted interaction with the targets (docking affinity).
Along with the above, we did computational prioritization of compounds via database mining, which is a recently developed attractive approach for in silico drug discovery. The advantage of use of already curated database over traditional literature mining is reduced time consuming and we can obtain molecular target information including direct or indirect activation and up/down regulated genes under the treatment of individual ingredients.50–52 Similar to the current study, a previous bioinformatics analysis explored the antirheumatic effects of Huang-Lian-Jie-Du-Tang via ingredients target mining in HIT database with a network perspective.104 In this study, we have focused on HIT and TCMID database search and it explored the possible cardiovascular effects of study compounds in a detailed way. In case of morin, notably, it was shown to modulate endothelin-1 and xanthine oxidase. The source study has shown that, morin has an anti-hypertensive effect in high fat-induced hypertensive rats and it suppressed mRNA expression of endothelin-1 in the thoracic aorta.105 Another study demonstrated it as an inhibitor of xanthine oxidase106 and lipoxygenase (LOX).107 The LOX metabolites from arachidonic acid and linoleic acid have been implicated in atherosclerosis, inflammation and its inhibition decreases neointimal formation following vascular injury.108 Targets of diosgenin shows that it is associated with superoxide dismutase (SOD), catalase (CAT) and the results of the source study suggests that diosgenin could be a very useful compound to control hypercholesterolemia by both improving the lipid profile and modulating oxidative stress.109 Consistently, a previous analysis also supports that it increases the activity of antioxidant enzymes network and prevent oxidative stress in diabetic animals.110 While focusing on vanillic acid, it was shown to be directly activating the vascular master regulator eNOS. From the database search, the source study postulates that vanillic acid enhances eNOS expression moderately.111 Furthermore, vanillic acid also have shown to be an effective inhibitor of the activity of catalase oxidase, which is involved in oxidative stress mediated carcinogenic initiation.112 When focusing on diosmetin, the STITCH compound–target interaction has shown that, it may targets COX2 and tumor necrosis factor-α (TNF-α). While focusing on syringic acid in the similar way it shows that, it may also targets COX2, thus both of it may acts as anti-inflammatory molecules.113 When focusing on piperine, the results have shown that it interacts with TNF-α and NF-κB, the source study have shown that, treatment with piperine reduced the level of nitrite in the lipopolysaccharides (LPS) stimulated BALB/c mice via inhibition of TNF-α production.114 Also it blocks the phosphorylation and degradation of IκBα by attenuating TNF-α induced IκB kinase activity and expression of intercellular adhesion molecule (ICAM),115,116 which in turn suggests piperine or its structural analogues could be used for the development of new anti-inflammatory molecules. Along with the above, the cardiovascular protective role of piperine extends since it target on interleukin 6 (IL-6), in which piperine treatment significantly reduced the IL-6 production in B16F-10 cells.117 Collectively, hence, the targets such as COX2, TNF-α, eNOS, SOD, CAT, LOX and endothelin-1 are major regulators of cardiovascular pathogenesis and these compounds may be utilized for cardioprotective measures.
Because of toxicity, drug candidates entering clinical trials have only 8% chance of becoming marketed drugs and about 20% of failures in the late drug development.118,119 Animal trials are currently the major method for determining the possible toxic effects of drug candidates. In silico toxicity prediction methods represent an alternative approach to experimental and nowadays, the interest has shifted to the above.57,120 The oral toxicity prediction results obtained in this study illustrates that, the study compounds may have very less or no toxicity. The LD50 value of them was very high when compared with the already indicated pharmacological dose in animal models. Along with this, drug-like properties assessed by preliminary screening of compounds with the Lipinski's rule of 5 demonstrated drug likeness of study compounds and a recent study also indicated via the above rule to use natural products as a drug-like molecule resource for drug development.4 As a result, we can propose that, the current study compounds can be used as drug leads for further preclinical manipulations.
The target disease relationship study unveiled that, the target predicted from genetic association of disease, revealed an over-representation of cardiovascular diseases and ontology terms related to coronary artery diseases and myocardial infarction. Furthermore, the target disease relationship from text mining database TCMSP also supported the above.
Footnote |
† Electronic supplementary information (ESI) available. See DOI: 10.1039/c5ra10761j |
This journal is © The Royal Society of Chemistry 2015 |