Available online at http://www.urpjournals.com International Journal of Agricultural and Food Science Universal Research Publications. All rights reserved ISSN 2249-8516 Original Article Ajocin (Allicin+Ajoene) can inhibit the enzymatic activity of aflatoxin biosynthesis in peanuts and prevent human carcinogenic exposure Archana Prabahar1‡, Srividhya Vellingiri1, Sathishkumar Natarajan1†, Kalpana Raja1‡, Renganathan Gandhimeyyan1, Bharathi Nathan*1 Email: [email protected], Telephone 91 - 9486775001 1 Department of Plant Molecular Biology and Biotechnology, Centre for Plant Molecular Biology, Tamil Nadu Agricultural University, Coimbatore -641003, Tamil Nadu, INDIA † Kyung-Hee University, 1 Seocheon-dong, Giheung-gu, Yongin-si, Gyeonggi-do 449-701, Republic of Korea. ‡ Data Mining and Text Mining Laboratory, Department of Bioinformatics, School of Life Sciences, Bharathiar University, Coimbatore -641046, Tamilnadu, INDIA. Archana Prabahar: [email protected], Srividhya Vellingiri: [email protected] Sathishkumar Natarajan: [email protected], Kalpana Raja: [email protected] Renganathan Gandhimeyyan: [email protected], Bharathi Nathan: [email protected] Received 13 July 2012; accepted 30 July 2012 Abstract Aflatoxin, a naturally occurring mycotoxin, produced by Aspergillus parasiticus infects peanuts and many other legumes. Among 23 enzymatic reactions involved in aflatoxin biosynthesis, only 15 are identified so far. Versicolorin B synthase (vbs) is the key enzyme involved in aflatoxin biosynthesis. It converts hydroxy versiconal acetate (VHA) to versicolorin B (VERB), which is finally converted into aflatoxin in a series of reactions. We selected two naturally available compounds, allicin and ajoene present in onion and garlic, to analyse their inhibitory effect on aflatoxin biosynthesis. Additionally, we virtually derived a new compound called ajocin (allicin + ajoene) to study their synergistic effect in preventing aflatoxin biosynthesis. We aim to compare the inhibitory action of these compounds against 15 known enzyme targets through docking simulations using Autodock. The best inhibitory action is reported by ajocin against the key enzyme, vbs. In silico docking studies thus confirm the enhanced inhibitory property of ajocin and reduce the risk of aflatoxin (carcinogen) exposure to human. © 2011 Universal Research Publications. All rights reserved Keywords: Aflatoxin, Allicin, Ajoene, Mycotoxins, Peanuts, Contamination. 1.1 INTRODUCTION The filamentous fungi A. flavus and A. parasiticus belonging to genus Aspergillus, produce various secondary metabolites such as aflatoxins and many other mycotoxins by infecting the agricultural crops and stored grains [1]. Aflatoxins are the most toxic, potent carcinogenic compounds causing aflatoxicosis and cancer in mammals. These naturally occurring teratogenic compounds contaminate many crops including peanuts, corn, cotton and treenuts [2]. Aflatoxins toxicity on farm animals fed with contaminated peanut meals had reported to numerous death due to acute liver necrosis. In human, hepatitis B infection and liver cancer are common in many parts of the world where aflatoxin exposure is high [3]. The four major aflatoxins are B1, B2, G1 and G2. Other significant members include the two oxidative forms of aflatoxin B1 namely M1 and M2. All these polyketide-derived 81 furanocoumarin compounds are highly toxic to agricultural crops, farm animals and human [4]. Aflatoxin contamination in food may cause neural tube defects and many primary cancers. Aflatoxin B1 in food and human dietary exposure may contribute to the aetiology of human chronic diseases. [5-6] One of the active compounds of freshly crushed garlic homogenates, Allicin is found to exhibit strong antifungal effect. Other antimicrobial activities of allicin include antibacterial, antiparasitic and antiviral effects. Allicin is assumed to be the main component responsible for the inhibition of aflatoxin formation [7]. Though the compound is found in a number of onion types, the extract from garlic shows comparatively greater antifungal activity [8]. Allicin can be isolated from garlic cloves through steam distillation. Pure allicin is a volatile molecule and poorly miscible in aqueous solutions [9]. Synthetically, allicin can International Journal of Agriculture and Food Science 2012, 2(3): 81-89 be obtained by mild oxidation of diallyl disulfide [10]. Another semi-synthetic method based on the supercritical carbon dioxide extraction (SC-CO2) of onions and their quantitative analysis by gas chromatography-mass spectrometry evidenced the presence of 28 sulfur compounds, including allicin [11]. A second garlic-derived sulfur-containing compound, ajoene is reported to exhibit antifungal activity towards A. niger. Ajoene is a derivative of allicin that can be isolated from the methanolic extract of garlic homogenate. Other antibiotic activities reported by ajoene include antiviral and antiprotozoal activities [12]. Allicin and ajoene are reported to possess antifungal activity. We virtually derived a third compound called ajocin by combining the structural features of allicin and ajoene to analyze the synergistic effect. Docking simulation is the effective way to predict the potency and efficacy of the three selected compounds in the inhibition of aflatoxin synthesis. Figure 1: 3D Structure of alicin 1.2. Materials and Methods 1.2.1. Enzymes involved in Aflatoxin Biosynthesis Biosynthesis of aflatoxin is a multistep process starting from the two substrates namely, acetyl CoA and malonyl CoA [13-15]. Figure 1 depicts the steps involved in this biosynthesis. Protein sequences of the fifteen enzymes involved in aflatoxin biosynthesis were retrieved from Swissprot (www.expasy.org) [16]. Templates for these protein sequences were identified by performing BLAST [17-19]. Based on the score, e-value and percentage identity, best templates were identified for every individual sequence. Structures of these enzymes were retrieved from ModBase (http://modbase.compbio.ucsf.edu/modbasecgi/index.cgi), a database of comparative protein structure models [20]. Figure 3: 3D Structure of ajocin by binding hydrophobic (CH3) probes to the protein, and finds clusters of probes with the most favorable binding energy. These clusters are placed in rank order of the likelihood of being a binding site according to the sum total binding energies for each cluster. Figure 4: Representation of the docked model of vbs with allicin 1.2.4. Selection of inhibitors for the aflatoxin biosynthetic pathway enzymes Inhibitors such as Allicin and Ajoene were selected for the docking study. 2D and 3D structures of these inhibitors were retrieved from Pubchem [22] compound database (http://pubchem.ncbi.nlm.nih.gov/). Synergistic activity of allicin and ajoene together may produce better activity and enhance the inhibitory action towards aflatoxin biosynthesis. In order to test this experimentally, we combined the two molecules using Chemsketch and derived an inhibitor, ajocin. Three dimensional structures of allicin, ajoene and ajocin were shown in figure 1, 2 and 3 respectively. This inhibitor was designed based on the literature survey of zimmu extract prepared by the combination of allicin and ajoene compounds from onion and garlic [15]. Since these compounds exhibited more antifungal activity against aflatoxin species, we made an Figure 2: 3D Structure of ajoene attempt to test the ajocin compound to inhibit aflatoxin 1.2.2. Model Assessment and Validation biosynthesis in peanut plants. Models thus generated were subjected to model assessment 1.2.5.Physiochemical Properties and Drug likeness using SAVS server (http://nihserver.mbi.ucla.edu/ SAVES /). prediction Ramachandran Plot [21] generated from SAVS software Drug likeness may be defined as a complex balance of was analyzed and the quality of the model was then various molecular properties and structure features which checked. determine whether particular molecule is similar to the 1.2.3. Active Site Prediction known drugs. These properties mainly focus on Q-SiteFinder (http://www.modelling.leeds.ac.uk/qsitefinder/) hydrophobicity, electronic distribution, hydrogen bonding is a new method of ligand binding site prediction. It works characteristics, molecule size, flexibility and presence of International Journal of Agriculture and Food Science 2012, 2(3): 81-89 82 various pharmacophoric features which in turn influence the behavior of molecule in a living organism, including bioavailability, transport properties, affinity to proteins, reactivity, toxicity, metabolic stability and many others. 1.2.6.Lipinski Rule (Rule of Five) Lipinski rule is a rule of thumb to evaluate drug likeness, or determine if a chemical compound with a certain pharmacological or biological activity has properties that would make it a likely orally active drug in humans. Lipinski's rule says that, in general, an orally active drug has no more than one violation of the following criteria: Not more than 5 hydrogen bond donors (nitrogen or oxygen atoms with one or more hydrogen atoms) Not more than 10 hydrogen bond acceptors (nitrogen or oxygen atoms) A molecular weight under 500 daltons An octanol-water partition coefficient log P of less than 5 The rule describes molecular properties important for a drug's pharmacokinetics in the human body, including their absorption, distribution, metabolism, and excretion ("ADME"). However, the rule does not predict whether the compound is pharmacologically active [23-24]. Figure 5: Representation of the docked model of vbs with ajoene. Table 1: Enzyme involved in aflatoxin biosynthesis with its sequence alignment and structural quality assessment. Template ID Ramachandran Plot (Percentage of Enzyme Name Swissprot ID Percentage Identity allowed regions) adhA P87017 1vl8 24.00% 85.5% avfA Q5VDD9 1xq6 16.00% 79.6% avnA Q12732 1nr6 19.00 % 88.8% estA Q5VD97 1ea5 14.00 % 81.2% fas_2 Q00681 1e5m 25.00% 84.8% norA Q00258 1lqa 28.00% 89.7% norB Q5VDD0 1pz1 25.00% 89.3% omtA Q12120 1fp2 23.00 % 86.2% omtB Q5VDD8 1fp1 21.00 % 90.3% ordA Q5VDD6 1pq2 20.00 % 88.2% pksA Q5VDF2 1e5m 23.00% 84.1% vbs Q5VDD5 1cf3 30.00% 87.2% ver-1 Q8J2U6 1ja9 50.00 % 91.8% verA Q5VDE2 1tqn 20.00% 87.8% verB Q5VDE0 1tqn 19.00% 86.9% 1.2.7. Bioactivity Pharmacological or biological activity is an expression describing the beneficial or adverse effects of a drug on living matter. Activity depends critically on fulfillment of the ADME criteria. Once a model is build, the bioactivity of screened molecules may be then calculated as a sum of activity contributions of fragments in these molecules. This provides a molecule activity score (a number, typically between -3 and 3). Molecules with the highest activity score have the highest probability to be active. Lipinski rule and Bioactivity of the small molecules were prescreened 83 using Molinspiration Tool. (http://www.molinspiration.com/ cgi-bin/properties) 1.2.8. ADME Screening ADMET refers to the absorption, distribution, metabolism, excretion, and toxicity properties of a molecule within an organism. Optimizing these properties during early drug discovery is crucial for reducing ADMET problems later in the development process. One of the most daunting hurdles a drug candidate must pass is having favorable ADMET characteristics. Such early identification helps to make research process more efficient and cost-effective by International Journal of Agriculture and Food Science 2012, 2(3): 81-89 Figure 6: Representation of the docked model of vbs with ajocin. allowing us to eliminate compounds with unfavorable ADMET characteristics. ADMET Descriptors in Discovery Studio include models for intestinal absorption, aqueous solubility, blood brain barrier penetration, plasma protein binding, cytochrome P450 2D6 inhibition and hepatotoxicity. ADMET screening is performed using Discovery Studio TOMKAT. 1.2.9.Docking Studies Docking studies were performed using Autodock 4.0. The Graphical User Interface program "Auto-Dock Tools" was used to prepare, run, and analyze the docking simulations. Kollman united atom charges, solvation parameters and polar hydrogens were added into the receptor PDB file for the preparation of protein in docking simulation. AutoDock [25-27] requires pre-calculated grid maps, one for each atom type present in the flexible molecules being docked and it stores the potential energy arising from the interaction with rigid macromolecules. This grid must surround the region of interest in the rigid macromolecule. The grid box size was set at 60, 60 and 60 A° (x, y, and z) to include all the amino acid residues that present in rigid macromolecules. AutoGrid 4.0 Program, supplied with AutoDock 4.0 was used to produce grid maps. The spacing between grid points was 0.375 angstroms. The Lamarckian Genetic Algorithm (LGA) [28] 23 was chosen search for the best conformers. During the docking process, a maximum of 10 conformers was considered. The population size was set to 150 and the individuals were initialized randomly. Maximum number of energy evaluation was set to 500000, maximum number of generations 1000, maximum number of top individual that automatically survived set to 1, mutation rate of 0.02, crossover rate of 0.8, Step sizes were 0.2 Å for translations, 5.0° for quaternions and 5.0° for torsions. Cluster tolerance 84 0.5 Å, external grid energy 1000.0, max initial energy 0.0, max number of retries 10000 and 10 LGA runs were performed. All the AutoDock docking runs were performed in Intel(R) Xeon(R) CPU 5150 @ 2.66GHz, 2GB RAM in Apple system. Autodock results were analyzed to study the interactions and the binding energy of the docked structure. 1.3. Results and Discussion 1.3.1. Preparation of Receptor and Ligand molecules Fifteen enzyme sequences involved in aflatoxin biosynthetic pathway were retrieved from Swissprot. BLASTP search was performed against PDB [29] database in order to identify suitable templates with known structures. Templates identified were listed in Table 1 with their respective percentage identity. Models were retrieved for the target proteins from the template structures using Modbase [30-32]. These models were further refined by performing energy minimization. Model assessment was performed using SAVS. RAMPAGE analysis of the selected models showed the presence of more than 80% of amino acid residues in favored and allowed regions of Ramachandran plot. Modeled proteins thus obtained were used for the docking study [33-34]. Active sites of the modeled proteins were identified using Q-Site finder and listed in Table 2. Based on the literature study, small molecules such as allicin, ajoene and the combined compound ajocin were chosen as inhibitors against aflatoxin biosynthetic enzymes. 1.3.2. Prescreening of Ligands for Lipinski rule / ADMET / Bioavailability The molecular weight, LogP value, number of donors and acceptors for small molecule inhibitors Ac, Aj and Ajc were generated using Molinspiration tool. All these three molecules satisfied Lipinski rule. Bioactivities of these molecules were also predicted. Molecules with the highest activity score have the highest probability to be active. Hence Ajc is found to be more active. Molecular Properties and bioactivity were shown in Table 5. ADME screening is performed using Discovery Studio TOMKAT. ADMET descriptors like BBB (Blood Brain Barrier) penetration and HIA (Human Intestinal Absorption), CYP450 2D6, Lipinski rule of five, plasma protein binding, aqueous solubility and hepatotoxicity were found for these inhibitor molecules and tabulated in Table 6. All the three molecules satisfied ADMET characteristics and hence could be used for biological process. 1.3.3. Protein – Ligand Docking Molecular docking simulation studies were performed for aflatoxin biosynthetic enzymes individually with prescreened inhibitors namely allicin, ajoene and ajocin using AutoDock 4.0. Docked structures were analyzed and the interacting residues were identified. Residues interacting in the docking study were found to reside in the active site of the macromolecules which is well depicted in Table 4. The residues interacting at the active site regions were highlighted in the Table 2. Docked structures were ranked based on the binding energy. Binding energy of the best conformation for each of the enzyme with the inhibitor molecules were tabulated in Table 3. The hydrogen bonds formed during the interaction were listed in Table 4 along with the hydrogen bond distance. International Journal of Agriculture and Food Science 2012, 2(3): 81-89 Table 2: Enzymes and its active sites Active sites predicted by docking interactions are high-lighted for each of the target enzyme molecule. Enzyme adhA avanA avfA estA fas2 norA norB omtA omtB ordA pksA vbs ver1 85 Active Sites Site 1 GLY22, GLY23, ALA24, SER25,GLY26, ILE27, GLY28, LEU29, ALA45, ASP46, ILE47, GLN48, GLU51, CYS70, ASP71, VAL72, THR73, SER74, HIS113, ILE114, LEU115, ALA 116, VAL136, MET172, GLY173, SER174, TYR187, LYS191, PRO217, ILE220, THR222, PRO223, LEU224, ILE225 Site 5 TYR95 , ALA296, GLY297, THR300, LEU304, PRO360, VAL361, GLN364, SER365, ILE368, LEU389, GLN391, PRO428, PHE429, SER430, ILE431, ARG434, ASN435, CYS436, ILE437 Site 4 LEU7, GLY8, ALA9, THR10, LEU34, VAL35, ARG36, GLY61, THR62, SER63, THR64, LEU69, VAL83, ALA84, GLN85, ASN86, SER88, PRO89, LEU94, SER98 Site 2 THR40, LEU41, ALA42, THR43, HIS85, GLY86, GLY87, GLY88, TRP89, VAL90, MET91, GLY92, GLY93, VAL94, GLU116, TYR117, ARG118, LYS119, TYR125, ALA128, ALA 155, ASP229, ASP230, THR233, TYR248 Site 3 GLY87, GLY88, SER154, ALA155, ASN158, ALA183, PRO184, ILE185, HIS188, ASP190, ALA191, LEU244, LYS246, ILE249, TYR283, PRO284, SER287 Site 1 ALA975, PHE976, SER978, ALA979, LEU993, SER994, GLU995, ILE1072, LYS1073, ILE1079, VAL1137, GLN1138, LEU1139, LEU1140, CYS1141, ARG1142, GLY1143, ALA1146, LEU1151, PRO1152, ILE1153, TYR1154, GLY1155, ILE1156, ILE1157, LEU1263, ASN 1264, GLY1265, LEU1267, GLN1268, ASP1271, THR1272 Site 1 GLY33, THR34, MET35, SER36, PHE37, ASN39, GLY40, TRP41, ASP69, TYR74, GLN75, TRP232, GLY233, VAL234, LEU235, GLY236, ARG237, GLY238, ALA243, GLU244, GLU245, ARG248 , GLU249, LYS252, GLN283, VAL296, ILE297, GLY298, GLY299, ARG300, ASN308 Site 1 GLY28, ALA29, MET30, THR31, PHE32, PHE40, ASP64, THR65, TYR69, LYS96, HIS 143 , TRP144, SER173, ASP174, GLN199, PRO226, TYR227, GLY228, VAL229, LEU230, ASN231, GLN232, GLY233, ILE298, VAL299, GLY300, VAL301 Site 1 VAL155, THR156, GLY157, ALA160, LEU161, VAL162, GLY163, LEU206, PHE207, ASP208, PHE221, ASP222, LEU223, GLY224, MET225, GLY226, GLY227, THR228, ALA230, ASP252, VAL253, GLY254, GLY255, GLY256, ARG257, GLY258, HIS259, LEU260, ASP 277, LEU278, VAL281, ILE282, ILE298, ARG313, SER314, ILE315, HIS317, ASP318, TYR 319, VAL368, VAL369, THR371, LEU372 Site 1 GLY192, HIS193, LEU194, MET195, ALA196, GLU198, ARG199, PRO200, SER202, GLY228, GLY229, GLY230, PHE231, GLY232, GLN233, GLN234, ARG289, HIS290, HIS293, ASP320, GLU321, MET322, THR336, MET340, GLU348, GLN376 Site1: LEU116, ALA117, MET132, GLU135, ILE136, MET179, MET180, VAL182, ALA183, TYR184, TYR186, THR187, SER270, ILE271, ARG274, LEU291, THR294, ASN295, GLU297, PHE298, VAL299, GLY301, GLY302, SER303, THR306, CYS440, PRO441, GLY442, ARG 443, VAL445, THR446, LYS449 Site 6: MET91 , SER366, PRO367, LEU368, GLY369, ALA370, HIS372, PRO393, ASN394, ILE395, PHE398, PHE431, VAL432, PHE433, GLY434, PHE435, ARG438, ILE439, CYS440 THR496, ASP499, TRP500, THR503, ASN504, ALA542, CYS543, GLY581, LEU582, LYS584, GLY585, PHE587, TYR597, TYR604, SER640, MET642, SER643, GLU644, SER645, MET646, THR647, ARG648, GLN654, HIS678, GLY679, THR680, GLY681, THR682, VAL684, GLY685, ASP686, VAL688, GLU689, HIS721, GLU723, PHE790, SER791, ALA792, ALA793, GLY794, GLY795, ASN796 Site 1 VAL80, GLY81, GLY82, GLY83, THR84, ALA85, ILE104, GLU105, ALA106, GLY107, TRP141, TYR144, TYR158, MET159, GLN160, GLY161, LYS162, THR163, GLY165, GLY166, SER167, THR168, THR314, LEU315, VAL316, SER350, ALA351, GLY352, VAL353, MET354, ARG355, SER356, GLN358, ASN511, TYR578, ALA579, GLY580, ASP612, ALA 613 Site 10 GLN390, ASP391, THR392, ALA492, LEU493, MET494, TYR577, ALA579, GLY580, VAL581, PHE618, ALA619, ILE620, ASP621, GLY622, PRO624 Site 1 THR15, GLY16, ALA17, GLY18, ARG19, GLY20, ILE21, GLY22, ALA23, ASN39,TYR 40, ALA41, International Journal of Agriculture and Food Science 2012, 2(3): 81-89 verA verB HIS42, SER43, ALA46, ASN94, ALA95, GLY96, ILE97, VAL98, THR142, SER 143, SER144, ASN145, THR146, HIS155, TYR158, LYS162, VAL186, ALA187, PRO188, GLY189, ALA190, ILE191, THR193, ASP194, MET195, PHE196, VAL199, SER200, TYR 203 Site 1 ARG78, LEU81, VAL84, MET90, MET91, THR92, TRP98, ARG102, PHE109, THR152, SER155, ILE156, VAL159, VAL160, TRP184, THR185, VAL211, MET212, TYR243, LEU267, ARG268, VAL 269, PHE270, LEU271, PHE272, ALA273, GLY274, ARG275, THR 277, THR278, PRO344, SER345, SER347, MET348, ARG352, ALA414, PHE415, GLU416, PRO419, ARG420, SER421, CYS422, ILE423, GLY424, GLN425, LEU427, ALA428, GLU431, LEU432 Site 2 ARG78, LEU81, GLU145, THR148, THR149, TRP184, THR185, PHE187, ARG275, ASN276, THR277, SER279, SER280, TYR284, PRO344, SER347, MET348, ARG349, GLU350, GLY351, ARG352, TRP373, ARG413, GLU416, GLY475, GLY477, ALA479, HIS480 Site 1 PHE135, ILE178, SER181, ILE182, LEU185, LYS267, MET294, VAL295, ALA297, GLY 298, SER299, THR301, THR302, ALA305, LEU306, ILE351, LEU355, TYR358, PRO359, ALA360, VAL361, HIS391, GLN429, PRO430, TRP431, SER432, CYS438, ILE439, GLY 440, ARG441, LEU443, ALA444, TYR445, GLU447, VAL448 Site 3 VAL171, GLU300, THR301, ALA303, SER304, ALA305, SER307, GLY308, ALA309, THR354, LEU355, TYR358, PRO359, ALA360, PHE471, GLN474, ILE479, TRP480, ALA481, LYS482, ARG483, GLU484, LEU485 Table 3: Enzymes and its binding energy with small molecules. Binding Energy Enzyme Allicin Ajoene -3.9 -5.89 adhA -4.04 -4.87 avanA -4.24 -4.58 avfA -4.81 -6.09 estA -4.36 -3.76 fas2 -4.59 -4.99 norA -3.85 -5.35 norB -3.7 -4.78 omtA Ajocin -8.3 -7.23 -6.06 -4.58 -1.26 -6.13 -6.58 -6.42 omtB -3.35 -4.24 -6.41 ordA -4.12 -4.48 -6.71 pksA -3.25 -1.85 54.7 vbs -4.37 -4.7 -7.74 ver1 -3.84 -4.94 -6.77 verA verB -4.21 -4.24 -4.91 -5.06 -7.49 -7.54 Table 4: Interaction of the enzymes with small molecules Hydrogen bond between aflatoxin biosynthetic enzymes and ligands with its binding distance Enzyme Binding Energy Ajoene Allicin Hydrogen bond adhA avanA avfA Lys 191: HZ1:Mol1:03 ARG 434:HH21 LIG1:O ALA 84: HN LIG1:O estA GLY86:HN LIG1: O TYR283: HH LIG1:S fas2 GLN1268:HE22 LIG1:O 86 Combined Compound Binding Hydrogen bond Distance Binding Distance Hydrogen bond Binding Distance 2.055 AdhA:ILE220:HN:Mol1:O 10 1.786 HIS113:HE2:lig1:O 1.920 1.951 ALA792:HN:mol1:O3 2.183 ARG434:HH21:lig1:O 1.986 2.035 1.943 GLN85:HN:MOL1:O10 2.121 1.938 LYS119:HZ3:MOL1:O10 1.834 ALA84:HN:Lig1:O EstA:GLY86:HN:Lig1: O EstA:TYR283:HN:Lig1: S Fas2:GLN1268:HE22:LI G1:O 2.042 1.856 SER132: HG:MOL1:O10 1.710 International Journal of Agriculture and Food Science 2012, 2(3): 81-89 2.034 2.081 1.91 norA norB omtA omtB GLU245:OE1 LIG1:SGLY298:HN LIG1:O GLY228:HN:MOL1: O3 HIS259:HN:MOL1:O 3 SER202:HG:MOL1:O 3 1.827 GLY233:HN:MOL1:O10 1.852 GLU298:HN: LIG1:O 1.921 2.108 GLN232:HN:MOL1:O10 2.121 LYS96:HZ2:LIG1:O 2.225 2.184 ARG313:HH11:MOL1:O10 1.859 ARG313:HH11:LIG1:O 1.705 1.872 HIS290:HD1:MOL1:O10 1.835 THR336:HG1:LIG1:O 1.943 ALA370:HN:MOL1:O10 HIS371:HE2:MOL1:O10 SER366:HG:LIG1:O 2.046 THR729:OG1:LIG1:S 2.999 1.961 2.032 ordA LYS449:HZ3:MOL1: O3 1.827 pksA ALA792:HN:mol1:O3 2.166 HIS721:NE2:MOL1:S5 vbs ILE620:HN:MOL1:03 1.885 GLN160:HE22:MOL1:S5 SER167:HN:MOL1:O10 ver1 ILE191:HN:MOL1:03 ARG78:HH12:MOL1: 03 ALA481:HN::MOL1: 03 1.922 GLY22:HN:MOL1:O10 2.2 1.93 1.739 GLY424:HN:MOL1:O10 1.970 ILE439:HN:MOL1:O10 verA verB 2.044 2.87 2.172 GLN160: HE22:LIG1:O ALA172: HN :LIG1:O ILE191:HN:LIG1:O 2.022 1.931 2.071 ALA479:HN:LIG1:O 1.778 1.757 GLY440:HN:LIG1:O 1.933 Table 5: Molecular Properties and Bioactivity Descriptors Allicin Ajoene Ajocin MOLECULAR PROPERTIES miLogP 2.064 0.026 2.725 TPSA 17.071 20.228 40.456 natoms 9 13 22 MW 162.279 235.419 406.77 nON 1 1 2 nOHNH 0 1 2 nviolations 0 0 0 nrotb 5 8 17 volume 145.506 210.596 375.404 BIOACTIVITY PREDICTION GPCR ligand -1.93 -0.94 -0.29 Ion channel modulator -2.02 -0.94 -0.51 Kinase inhibitor -2.58 -1.50 -0.68 Nuclear receptor ligand -2.59 -1.41 -0.29 Table 6: ADME Properties Ligand Log P BBB Hepatotoxicity Ajoene Alicin Ajocin 2.32 1.77 4.87 0.23 0.66 0.69 0 0 0 Hepatotoxicity Probability 0.35 0.32 0.16 The activity of Ajc compund was found to be higher than Ac and Aj compounds in most of the enzyme inhibitory process. VERB synthase gene (vbs) plays the key role in the synthesis of aflatoxin. Based on the literature study, the 87 CYP2D 6 0 0 0 CYP2D6 Probability 0.14 0.02 0.44 PSA_2D Solubility 20.82 20.82 40.63 -1.59 -1.08 -2.99 function of vbs gene is found to convert VHA to VERB in A. parasiticus. This is a key step in aflatoxin formation since it closes the bisfuran ring of aflatoxin. This ring is required for binding to DNA and gives aflatoxin its mode International Journal of Agriculture and Food Science 2012, 2(3): 81-89 of action as a mutagen. Despite other enzymes involved in the aflatoxin synthesis, vbs was found to be the most important target for enzyme inhibition to control aflatoxin synthesis. Docking studies were performed with these genes against the inhibitors allicin, ajoene and ajocin. The action of the inhibitor ajocin was best proved in the inhibition of vbs gene with a binding energy of -7.74 by the formation of two hydrogen bond interactions at the active site region. This enzyme inhibitory process is required to stop aflatoxin synthesis and to prevent peanut infection. Based on our in silico study and literature studies of zimmu compound in sorghum plants, we suggest that ajocin compound would contribute the maximum effect to inhibit the fungal infection caused by aflatoxins in peanut plants by preventing aflatoxin synthesis. 1.4. Conclusion: In this work, a molecular docking simulation study was carried out for all the fifteen enzymes involved in the aflatoxin biosynthesis using the inhibitors allicin, ajoene and ajocin. Enzyme inhibitory process was required to stop the synthesis of aflatoxins. From this in silico study and previously reported experimental data in literature, we found that vbs gene present in the aflatoxin biosynthetic pathway is known to be responsible for the production of aflatoxins. Hence, we conclude that ajocin would be the best possible component to inhibit the vbs gene which controls the biosynthesis of aflatoxins and prevent human carcinogenic exposure. Acknowledgments Authors would like to thank the faculties of Department of Plant Pathology, Centre for Plant Protection Studies, Tamil Nadu Agricultural University for providing information about Zimmu extract derived from alicin and ajoene which remained as an initiative for this In silico docking study. References 1. Y. Jiujiang, E.C. Thomas, C.N. William, W.B. Joan, Aspergillus flavus genomics: gateway to human and animal health, food safety, and crop resistance to diseases, Revista Iberoamericana de Micología. 22 (2005) 194-202. 2. C.C. Bruce, J.M. Russell, F.S. Thomas, Current Research on Reducing Pre- and Post-harvest Aflatoxin Contamination of U.S. Almond, Pistachio, and Walnut, Journal of Toxicology:ToxinReviews.22(2003)225-266. 3. E.D.A. Anne, M.H. Thomas, Mycotoxins in Plant Pathogenesis, Molecular Plant Microbe Interactions. 10( 2) (2007) 147–152. 4. Y. Jiujiang, D. Bhatnagar, C.E. Kenneth, Aflatoxin biosynthesis, Rev Iberoam Micol. 19 (2002) 191-200. 5. G. Sun, S. Wang, X. Hu, J. Su, Y. Zhang, Y. Xie, H. Zhang, L. Tang, J.S. Wang, Co-contamination of aflatoxin B1 and fumonisin B1 in food and human dietary exposure in three areas of China, Food additives and contaminants. 28(4) (2011) 461-470. 6. D.J. Groopman, W.N. Gerald, Aflatoxin and Hepatocellular Carcinoma, Chemical Carcinogenesis, Current Cancer Research. (2011) 113-133. 7. S. Ankri, D. Mirelman, Antimicrobial properties of allicin from garlic,Microbes Infect.1(2)(1999)125-129. 8. G.H. Bronwyn, D.L. Larry, Antimicrobial effects of 88 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Allium sativum L. (garlic), Allium ampeloprasum L. (elephant garlic), and Allium cepa L. (onion), garlic compounds and commercial garlic supplement products, Phytotherapy Research. 5(4) (1991) 154-158. C. Cavallito, J.H. Bailey, Allicin, the antibacterial principle of Allium sativum. Isolation, physical properties and antibacterial action, J. Am. Chem. Soc. 66 (1944) 1944-1952. H.P. Koch, L.D. Lawson, Garlic, the science and therapeutic application of Allium sativum L. and related species, in: Retford D.C. (Ed.), Williams and Wilkins, Baltimore. (1996) 1-233. N.K. Sinha, D.E. Guyer, D.A. Gage, C.T. Lira, Supercritical carbon dioxide extraction of onion flavors and their analysis by gas chromatography-mass spectrometry,J.Agric.Food Chem.40(5)(1992)842-845. R. Naganawa, N. Iwata, K. Ishikawa, H. Fukuda, T. Fujino, A. Suzuki, Inhibition of Microbial Growth by Ajoene, a Sulfur-Containing Compound Derived from Garlic, Applied and Environmental Microbiology. 62(11) (1996) 4238–4242. Y. Jiujiang, C. Perng-Kuang, C.K. Ehrlich, W. J. Cary, B. Deepak, E.C. Thomas, A.P. Gary, E.L. John, W.P. Charles, B.W. Joan, Clustered Pathway Genes in Aflatoxin Biosynthesis, Applied and Environmental Microbiology. 70(3) (2004)1253-1262. J.A. Anderson, Enzymes in aflatoxin biosynthesis, World Journal of Microbiology and Biotechnology. 8(1) (1992) 96-98. M. Karthikeyan, R. Sandosskumar, R. Radhajeyalakshmi, S. Mathiyazhagan, S.E.Khabbaz, K. Ganesamurthy, B. Selvi, R. Velazhahan, Effect of formulated zimmu (Allium cepa L. × Allium sativum L.) extract in the management of grain mold of sorghum, J Sci Food Agric. 87 (2007) 2495–2501 N. Guex, M.C. Peitsch, Swiss-Model and the SwissPdbViewer: An environment for comparative protein modeling, Electrophoresis. 18 (1997) 2714-2723. S.F. Altschul, W. Gish, W. Miller, E.W. Myers, D.J. Lipman, Basic local alignment search tool, J. Mol. Biol, 215 (1990) 403-410. J. Ye, S. McGinnis, T.L. Madden, BLAST: improvements for better sequence analysis, Nucleic Acids Research. 34 (1990) W6-W9. M. Johnson, I. Zaretskaya, Y. Raytselis, Y. Merezhuk , S. McGinnis, T.L. Madden, NCBI BLAST:a better web interface,Nucleic Acids Research.36(2008)W5-W9. D. Eramian, N. Eswar, M.Y. Shen, A. Sali, How well can the accuracy of comparative protein structure models be predicted?, Protein Science. 17 (2008) 1881-1893. G.N. Ramachandran,C.Ramakrishnan,V. Sasisekharan, Conformation of polypeptides and proteins, Journal of Molecular Biology. 7 (1963) 95–99. E.E. Bolton, Y. Wang, P.A. Thiessen, S.H. Bryant, PubChem: integrated platform of small molecules and biological activities, Annu Rep Comput Chem. 4 (2008) 217–241. C.A. Lipinski, Drug-like properties and the causes of poor solubility and poor permeability,Journal of International Journal of Agriculture and Food Science 2012, 2(3): 81-89 24. 25. 26. 27. 28. 29. Pharmacological and Toxicological Methods, 44 (2000) 235-249. C.A. Lipinski, F. Lombardo, B.W. Dominy, P.J. Feeney, Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings, Advanced Drug Delivery Reviews. 23(1-3) (1997) 3-25. G.M. Morris, D.S. Goodsell, R.S. Halliday, R. Huey, W.E. Hart, R.K. Belew, A.J. Olson, Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function, Journal of Computational Chemistry, 19(14) (1998) 1639-1662. D.S. Goodsell, G.M. Morris, A.J. Olson, Automated docking of flexible ligands: applications of AutoDock, Journal of Molecular Recognition. 9(1) (1996) 1–5. M. Rarey, B. Kramer, T. Lengauer, G. Klebe, A fast flexible docking method using an incremental construction algorithm, Journal of Molecular Biology. 261 (1996) 470–89. G. Jones, P. Willett, R.C. Glen, A.R. Leach, R. Taylor, Development and validation of a genetic algorithm for flexible docking, Journal of Molecular Biology. 267 (1997) 727–48. H.M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T.N. Bhat, H. Weissig, I.N. Shindyalov, P.E. Bourne, The Protein Data Bank, Nucleic Acids Research. 28(1) (2000) 235-242. 30. A. Schlessinger, E. Geier, H. Fan, J.J. Irwin, B.K. Shoichet, K.M. Giacomini, A. Sali, Structure-based discovery of prescription drugs that interact with the norepinephrine transporter, NET, Proceedings of the National Academy of Sciences.108(38)(2011)1581015815. 31. I. Yoko, H. Takanori, In Silico 3D Structure Prediction of Activated GPCRs as a Drug Target, Proceedings of 2011 International Conference on Bioscience, Biochemistry and Bioinformatics, IPCBEE, Singapore. 5 (2011). 32. S. Grosdidier, J. Fernández-Recio, Identification of hot-spot residues in protein-protein interactions by computational docking, BMC Bioinformatics. 9 (2008) 447. 33. P. Archana, N. Sathishkumar, N. Bharathi, In Silico docking analysis of curcumin – inhibitor for obesity, International Journal of Pharma and Bio Sciences. 1(4) (2010) 224-235. 34. N. Sathishkumar, S. Sathiyamoorthy, M. Ramya, D.U. Yang, H.N. Lee, D.C. Yang, Molecular docking studies of anti-apoptotic BCL-2, BCL-XL, and MCL-1 proteins with ginsenosides from Panax ginseng, Journal of Enzyme Inhibition and Medicinal Chemistry (2011) (In press). Source of support: Nil; Conflict of interest: None declared 89 International Journal of Agriculture and Food Science 2012, 2(3): 81-89
© Copyright 2025 Paperzz