J. Nig. Soc. Phys. Sci. 5 (2023) 1576 Journal of the Nigerian Society of Physical Sciences Isolation, Characterization, Antimicrobial and Theoretical Investigation of Some Bioactive Compounds Obtained from the Bulbs of Calotropisprocera M. E. Khana,∗, C. E. Elumb, A. O. Ijeomahb, P. J. Amejia, I. G. Osigbemhec, E. E. Etimf, J. V. Anyamb, A. Abeld, C. T. Agbere a Department of Chemistry, Federal University Lokoja, Kogi State, Nigeria b Department of Chemistry, Federal University of Agriculture Makurdi, Benue State, Nigeria c Department of Industrial Chemistry, Federal University Lokoja, Kogi State, Nigeria d Department of chemistry College of Education Hong, Adamawa State, Nigeria e Department of Chemistry, Benue State University, Makurdi, Benue State, Nigeria f Department of Chemical Sciences, Federal University Wukari, Taraba State, Nigeria Abstract This study characterizes the bioactive molecules from the bulb of Calotropisprocera and investigates the antimicrobial activities of the crude extracts. Theoretical studies on the two isolated compounds in the crude extract were also accomplished.The bulbs were air dried, pulverized, and subjected to extraction procedures by maceration using 500 mL each of normal-hexane, ethyl acetate and methanol. The crude extracts were further tested onmicroorganisms and phytochemical screening using standard procedures. In addition, the bioactive compounds in the extract were screened against DNA gyrase of two Gram negative bacterial species; Escherichia coli and Salmonella typhiusing Molecular Docking simulation techniques and further subjected to ADMET profiling,using the Swiss ADME online server. The Crude ethyl acetate extract has the highest effective activity against Escherichia coli (MIC 2.5mg / mL and MBC/MFC 5mg / mL), Staphylococcus aureus (MIC 2.5mg/mL), Candida albicans, Salmonella typhi and Candida stellafoidea (MIC 5mg/mL). β-Amyrin acetate and Taraxasterol are the two phytochemicals in the purified white crystalline fractions and were found to fasten to the active sites of DNA gyrase of the Gram negative bacterial species via hydrophobic and hydrogen bond interactions, with binding activity value of -9.6 kcal/mol and -9.5 kcal/mol, respectively. Also, ADMET investigations of the compounds revealed their sound oral bioavailability and excellent pharmacokinetic and toxicity profiles. The findings of this study could provide a platform for discovering safe and potent antibiotics against pathogenic microbes ravaging our society. DOI:10.46481/jnsps.2023.1576 Keywords: Phytochemical Screening, Anti-microbial screening, Calotropisprocera, Pharmacokinetics Article History : Received: 26 May 2023 Received in revised form: 23 June 2023 Accepted for publication: 24 June 2023 Published: 23 August 2023 © 2023 The Author(s). Published by the Nigerian Society of Physical Sciences under the terms of the Creative Commons Attribution 4.0 International license (https://creativecommons.org/licenses/by/4.0). Further distribution of this work must maintain attribution to the author(s) and the published article’s title, journal citation, and DOI. Communicated by: E. A. Emile ∗Corresponding author tel. no: +234 7031667488 Email address: khandora2000@gmail.com, muluh.khan@fulokoja.edu.ng (M. E. Khan ) 1. Introduction Natural products are the most successful precursors of future drug leads [1-5]. It has been, it is, and will continue to 1 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 2 provide distinctive structural variety that presents opportuni- ties for discovering, mainly new low molecular weight lead molecules [6]. Biosynthesis and a critical analysis of proteins, fatty acids, nucleic acids and carbon derivatives is known as primary metabolism and the molecules are called primary metabolites [7]. The mechanism by which an organism biosynthesizes bioactive molecules or secondary metabolites is a unique one [8]. These metabolites generally, are not actually needed for growth, development or reproduction of an organism but are produced either for adapting the organism to its surrounding environment or to act possibly for defence mechanism against predators in assisting survival of the organism [9]. Biosyn- thesis of these metabolites is gained from the fundamental processes of photosynthesis, glycolysis and the Krebs cycle to procure biosynthetic intermediates which, ultimately, results in the formation of these secondary metabolites [7]. Though the number of building blocks is limited, the formation of new secondary metabolites is infinite. Utmost important constructing blocks employed in the biosynthesis of these metabolites are those obtained from the intermediates: Acetyl coenzyme A (Acetyl-CoA), Shikimic acid, Mevalonic acid and 1-Deoxyxylulose-5-phosphate. These are involved in countless biosynthetic pathways, involving numerous different mechanisms, actions reactions, inactions and interactions. Calotropisprocera, asmall tree is widely spread in tropical and subtropical Africa [10]. It is called different names in English and locally, like “Sodom apple, Usher, Dead Sea apple, Swallow-wort, Giant milk weed ”Tumfafiya or Baabaa ambalee in Hausa, Ewe-Bomubomu in Yoruba, Otokwuru in Igbo and Konzar Atumbagh in Tiv [11]. The plant is hardy, pubescent, evergreen, erect, a compact shrub up to 4.5 m tall, and is covered with cottony tomentum. The stem is usually simple, rarely branched, woody at base and covered with fissures. Various parts of this plant have the ability to produce large quantities of latex when cut or broken [12]. Calotropisprocera (Asclepiadaceae) is recognised by most traditional systems of medicine as ‘Madar” in Unani medicinal system. Earliest Hindu writers mentioned the plant and its primeval name occurred in the Vedic literature was “Arka” alluding to the form of leaves, which was used in the sacrificial cremation described by the Sanskrit writers [13]. The Latex exuded by Calotropisprocera is valued for its high medicinal and pharmaceutical activity due to its high content of bioactive compounds such as;- cardiac glycosides, alkaloids, terpenes, resins, lipids, flavonoids, tannins and steroids [14]. This latex possesses different biological activities including: anti-inflammatory, analgesic, antitumor, antiviral, hepatoprotective, antiulcer, anthelmintic, insecticidal, herbici- dal, antioxidant and spasmolytic activities [15]. Theoretical Chemistry concepts such as In silicomolecular docking, Pharmacokinetic and toxicity profiling has found im- mense applications in the discovery of new bioactive molecules for the treatment of various diseases [16-18]. Molecular dock- ing entails the prediction of the binding interaction of bioactive compounds (ligands) with the active sites of a target macro- molecule (receptor). The ligands with the most stable confor- mations with the target protein are the most promising drug candidates [19]. Molecular docking technique has been widely used in pharmaceutical research in recent years because of its fast and cost effectiveness in screening data base of bioac- tive ligands [20-25, 18]. Likewise, pharmacokinetic investiga- tion which is concerned with the fate of therapeutic ligands in the biological system forms an essential component of mod- ern drug discovery. This deals with the absorption, distribution, metabolism, excretion, and toxicity (i.e., ADMET) potentials of bioactive ligands. In silico ADMET profiling of drug can- didates helps to minimize attrition rates during the preclinical and clinical stages of drug development [26-28].This study is aimed at isolation, characterization and application of in vitro & in silico techniques to explore the bioactive molecules present in Bulbs of Calotropisprocera. 2. Methods 2.1. Sample Collection The fresh bulbs of the plant identified as Calotropisprocera (Figure 1) was collected from Millionaires’ Quarters, Lafia L. G A. of Nasarawa State Nigeria, in August, 2019 and was authen- ticated at the College of Forestry and Fisheries, Federal Univer- sity of Agriculture Makurdi, Benue State Nigeria with voucher No FH/0086, deposited at the College herbarium. The bulbs were washed with clean water to removed dirt and air dried at room temperature. The dried fruits were then pulverized into coarse powder with mortar and pestle, then sieved with a sieve of 0.55 mm pores and stored in cellophane bags at room tem- perature until required for experimental use. Figure 1: Plant and bulbs of Calotropisprocera 2.2. Extraction 500g of the plant material was weighed and extracted by the process of Maceration, allowing the pulverized powdered mate- rial to soak in an appropriate solvent in a closed vessel at room temperature. Three chosen solvents were employed, in the following order, normal-Hexane, Ethyl Acetate and Methanol. The plant sample was macerated with two litres of each of the solvents for 72 hours with agitation. The above was filtered and the filtrate concentrated under reduced atmospheric pres- sure using a rotary evaporator at 37OC, to recover some of the 2 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 3 solvent. This procedure was repeated for the ethyl acetate and methanol extracts. They were then allowed to dry. The vari- ous solvents extracts were coded for quick identification during further analysis. 2.3. Screening for Phytochemicals Crude extracts from the used solvents were phytochemi- cally evaluated for the presence of anthraquinones, saponins, tannins, steroids, terpenes, reducing sugars, flavonoids and al- kaloids using standard scrutiny as reported by [29, 30]. Phy- tochemical screening results showed the presence of saponins, tannins, steroids/sterols, tannins, terpenoids, flavonoids, reduc- ing Sugars, and cardiac glycosides. 2.4. Bioassay Antibacterial and antifungal activities of the normal- hex- ane, ethylacetate and methanol crude extracts were inves- tigated using clinical isolates of some pathogenic microbes such as: Staphylococcus aureus, Staphylococcus faecalis, Es- cherichia coli, Vancomycin enterococci, Neisseria gonorrhoae, Profeusmirabili, Pseudomonas aeruginosa, Salmonella typhi, Candida albicans, Candidakrusei and Candida stellafoidea. Dispersion method was used for screening the extracts. Mueller Hinton Agar was used as the growth medium for microorgan- isms, sterilized at 121°C for 15 min, poured into Sterile Petri Dishes and were cooled and solidified. The said crude ex- tracts (0.4 g) were allowed to dissolve in 10.0 mL of Dimethyl- sulphoxide (DMSO) to acquire a concentration of 40.0 mg/mL. A disinfected medium was then seeded with the standard Inocu- lum (0.1 mL) of test microorganisms and were equitably spread over the surface of the medium with sterile swabs. Using a 6mm standard cork-borer, a well was cut at the centre of each inoc- ulated medium. A concentration of 5.0 mg/mL of the already weighed crude extract was then launched into each well on the inoculated medium. The inoculated medium was incubated at 37OC for 24 hours, after which the medium was observed for the zones of inhibition which were measured with a transparent ruler [31]. Minimum inhibition concentration (MIC) of these extracts were carried out using Broth Agar Dilution method. Mueller Hinton Broth was prepared by dispensing 10.0 mL into test tubes and pasteurised at 37°C for 6 hours, then cooled. Mc- Farland’s turbidity standard scale number 0.5 was prepared to give a turbid solution. Normal Saline (10.0 mL) which was dispensed into sterile test tubes and the test microbes were in- oculated and incubated at 37OC for 24 hours. Thereafter, the tubes were observed for turbidity. The lowest concentration of extracts in the sterile broth that indicated no turbidity was recorded as the minimum inhibition concentration (MIC) [32]. The minimum bactericidal and minimum fungicidal concentra- tion (MBC and MFC) were carried out to determine the concen- tration of extracts that could stop growth of tested microorgan- isms. Mueller Hinton Agar was prepared, pasteurised at 121OC within 15 minutes, poured into sterile Petri dishes and allowed to cool. The contents of the test tubes with the evaluated MIC were then sub-cultured onto prepared media, incubated at 37OC for 24 hrs, after which the plates were visualised for any colony growth. MBC/MFC plates with lowest concentration of extract, without a colony growth were considered as the MBC/MFC [32]. 2.5. Spectroscopic Characterization 1H and 13CNMR spectra of β -Amyrin acetate (Cp17) and Taraxasterol (Cp35) were run using CDCl3 as solvent on Agilent-NMR 500MHz spectrophotometer at Strathclyde In- stitute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, United Kingdom. 2.6. Molecular Optimization and Docking Procedures Antimicrobial studies on the crude extracts revealed that they possess profound bioactivities against Escherichia coli and Salmonella typhi. Also, phytochemical screening divulged the presence of β -Amyrin acetate and Taraxasterol. Guided by these findings, the two bioactive ligands were subjected to ge- ometry optimization using the Semi-empirical (pm3) method of Spartan’14 software to obtain their minimum energy ge- ometries. Molecular docking simulation was used to screen the compounds against DNA gyraseof Escherichia coli and Salmonella typhi. The 3D structure of the target protease (PDB code: 5ztj) was retrieved from protein data bank at www. rcsb. org/pdb. The water molecules, hetero-atoms, and co- crystallized ligands attached to the retrieved protease were re- moved using the Biovia Discovery Studio interface. The target protein was further processed via the addition of Polar hydro- gens and Kollman charges with the aid of Auto Dock Vina tool v1.5.7. Finally, the docking calculation between the ligands and the target DNA gyrase was performed using PyRx software of Auto Dock Vina tool by centring the Vina search space at X: -26.4401 Å, Y: 23.0598 Åand Z: 22.0813 Åwith dimensions of X: 51.6292 Å, Y: 53.0044 Åand Z: 51.8136 Å[26-27, 33-34]. 2.7. Drug-likeness Estimation This is the evaluation of oral bioavailability of therapeutic compounds. Here, in silico technique forms a pivotal part of modern drug discovery owing to the fact that most drugs are ad- ministered via the oral route. The assessment of oral bioavail- ability of β -Amyrin acetate and Taraxastero were performed using the Lipinski’s rule of five and the Veber’s rule. Accord- ing to the Lipinski’s rule, a drug must have its molecular weight (MW) ≤ 500, number of hydrogen bond donors (HBD) ≤ 5, oc- tanol/ water partition coefficient Log P ≤ 5 and number of hy- drogen bond acceptors (HBA) ≤ 10 for it to be orally bioavail- able and violation of more than one of these indices could trans- late to poor drug-likeness potentials of a ligand. Veber’s rule, on the other hand stipulates that, for a drug to be orally bioavail- able, the number of rotatable bonds (NRB) must be < 10 and topological polar surface area (TPSA) must be < 140 Å2. The NRB, TPSA, MW, HBD, HBA, and Log P value of the bioac- tive ligands were computed using the Swiss ADME (http: //www.swissadme.ch/www.swissadme.ch/) online tool [26- 27, 33-34]. 3 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 4 2.8. ADMET Profiling The high failure rates of drug candidates at the late stage of drug development has made prediction of pharmacokinetic and toxicity profiles of therapeutic ligands at the early stage of drug development a necessity. The phytochemicals were profiled for their gastrointestinal (GI) absorption, blood brain barrier (BBB) permeation, P-glycoprotein (P-gp) substrate po- tentials, and cytochrome-P450 enzymes inhibition using the Swiss ADME online server at http://www.swissadme.ch/ index.php. Furthermore, Osiris Data Warrior V5.5.0 chemo- informatics program was used to perform in silico toxicity as- say on the ligands using the following toxicity endpoints; mu- tagenicity, reproductive effect, and irritating effect [26-27, 33- 34]. 3. Results 3.1. Phytochemical and Antimicrobial Screening Phytochemical and antimicrobial screening of the crude ex- tract results are presented in Tables 1 and 2, respectively. Table 1: Phytochemical screening of the bulb extacts of Calotropisprocera Class of Compound normal- Hexane Ethyl acetate Methanol Saponins + + + Tannins + + + Flavonoids + + + Steroids/Sterols + + + Alkaloids + + + Reducing Sugars + + + Cardiac glycosides + + + Anthraquinone + Key = (+) indicate, present, (-) indicate, below detectable limits 3.2. Elucidated Structures of the Bioactive Compounds Figure 2 presents elucidated structures of the two bioactive ligands present in the crude extract of the Bulbs of Calotropis- procera. Experimental and literature data for 1H and 13CNMR analysis are presented in Tables S1-S4 in the supplementary file (below). Figure 2: Elucidated Chemical Structures of Bioactive compounds in the Ex- tract 3.3. Molecular Docking Outcome The molecular docking simulation derived binding affinities and diagrams of interaction of the ligands with the active sites of the DNA gyrasetarget are presented in Figure 3. Figure 3: Binding affinity and Diagrams of interaction of the investigated bioac- tive ligands with the active sites of DNA gyrase 4. Oral Bioavailability and ADMET Profiles of the Phyto- chemicals Oral bioavailability of a therapeutic ligand is a function of some of its physicochemical properties. Tables 3 and 4 present 4 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 5 Table 2: Sensitivity/zone of inhibition (mm) of the crude Extracts Against microorganisms Test organism EA MEOH HEXANE Ciprofloxacin Tetracycline Fluconazole Nectricillin resistant Staph. Aureus S (24) S (21) S (20) R (0) S (29) R (0) Vancomycin resistant ente- rococci R (0) R (0) R (0) S (28) S (30) R (0) Staphylococcus aureus S (27) S (24) S (21) R (0) S (32) R (0) Escherichia coli S (28) S (23) S (20) S (35) R (0) R (0) Neisseria gonorrhea S (26) S (22) S (18) R (0) S (25) R (0) Profeus mirabilis R (0) R (0) R (0) S (32) R (0) R(0) Pseudomonas aeruginosa R (0) R (0) R (0) S (30) S (27) R (0) Salmonella typhi S(24) S (21) S (18) S (41) R (0) R (0) Candida albicans S (25) S (22) S (20) R (0) R (0) S (32) Candida krusei R (0) R (0) R (0) R (0) R (0) S (30) Candida stellafoidea S (23) S (20) S (17) R (0) R (0) S (34) Legend =� S= Sensitive, R= Resistance, EA= Ethyl acetate extract, MEOH= Methanol extract, HEXANE= n-hexane extract, Numeric value in brackets = diameter of zone of inhibition in millimetres; Drug concentration: Ciprofloxacin = 20 µ g, Tetracychine = 20 µg, Fluconazole = 20 µg the descriptors of oral bioavailability and ADMET profiles of the ligands, respectively. Table 3: Oral Bioavailability Profiles of the Phytochemicals Ligand Rule β-Amyrin acetate Taraxasterol Lipinski’s Yes Yes HBA 2 1 HBD 0 1 MW (gmol-1) 440.7 426.7 cLogP(o/w) 7.0 7.1 Veber’s Yes Yes NRB 2 0 TPSA ( Å2) 26.3 20.23 HBA; hydrogen bond acceptor, HBD; hydrogen bond donor, Mw; molecular weight, cLogP; consensus octanol water partition coefficient, NRB; number of rotatable bond, TPSA; topological polar surface area 5. Discussion Phytochemical screening of the crude extracts of the plant divulged the presence of reducing sugars, saponins, steroids, tannis, alkaloids and flavanoids. Anthraquinones were below detection level (Table 1). This is corroborated with the work of (35), when they analysed the root bark of Terminaliaschimpe- riana. Screening for Antimicrobials was carried out against Nec- tricillin resistant Staphylococcus aureus, Vancomycin resistant Enterococci, Escherichia coli, Profeus mirabilis, Salmonella typhi, Pseudomonas aeruginosa, Candida albicans, Neisseria gonorrhea, Candida krusei and Candida stellafoidea. Param- eters determined were the zone of inhibition (ZI), minimum inhibitory concentration (MIC) and minimum bacterici- dal/fungicidal concentration (MBC/MFC). The antibacterial and antifugal results obtained showed that ethyl acetate extract had the highest diameter of zones of inhibition (28 mm) against (35 mm) for the tested microbes. Over all, ethyl acetate has the highest efficacy and efficiency of the applied solvents, followed by methanol and the N. Hexane. Out of the eleven (11) microorganisms employed, seven (7) had various degrees of activities, with the exception of, Vancomycin resistant enterococci, Profeus mirabilis, Pseudomonas aeruginosa , and 5 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 6 Table 4: In silico Pharmacokinetic and Toxicity profiles of the Phytochemicals Ligand CYP450 Substrate GIA P-gp+ BBB Mutagenic Irritant Reproductive effect Tumorigenic LogKp (cm/s) β -Ac Yes Yes No No None None None None -2.6 Ta Yes Yes No No None None None None -2.4 GIA; gastrointestinal absorption, BBB; blood brain barrier penetration, P-gp+; P-glycoprotein substrate β-Ac; β -Amyrin acetate, Ta; Taraxasterol Candida krusei, all of which had zero activities, as seen in Table 2. The proton analysis (1H NMR spectra) of β -Amyrin acetate (Table S2), showed the presence of the trademark amyrinolefinic proton (1H-12) resonating as triplet at δ 5.16 ppm due to deshielding caused by the π bond. The proton NMR signals at δ 0.85, 0.98, 0.94, 0.81, 1.11, 0.85, 0.77, and 0.92 ppm depict eight methyl groups all singlets with an additional methyl group at δ 2.02 being the acetate methyl protons. At δ 4.47ppm, the double doublet is due to the lone proton on C-3 bonded equatorial to an acetate ester. Broad singlet at δ 0.85 ppm of C-29 and C-30 methyl protons is a pointer to the characteristics of β – amyrin. Furthermore, 13C NMR of the lone proton δ 4.47 (Table S1), is assigned H-3 by HSQC correlation with δ 80.35 and HMBC- 3J correlation to methyls δ 15.71 (C-23), δ 28.30 (C-24) and δ 170.94 ppm (C-1’) from the acetyl moiety. HSQC correlation of δ 21.53 to δ 2.02 (3H) is evident of electron rich δ 170.94 ppm (C-1’) of the acetyl moiety. The acetate is attached to the first hexacyclic component at C-3. δ 5.16 and correlates with δ 121.68 ppm on HSQC and is ascribed H-12. It shows a HMBC- 2J correlation with the secondary carbon δ 23.75 (C-11) and 3J quaternary carbon δ 42.07ppm (C-14). H-12 bonds to the electron rich alkenyl carbon (C-12) and splitting with δ 1.80 (H-11) thus, resolved as triplets. δ 33.42 (C-29) axial and δ 23.86 ppm (C-30) equatorial correlations to 1H-19a, 1H-19b and 3H-28 indicates their association to ring E of amyrin. Moreover, δ 0.85 (C-30 & C-29) 2J correlation to the quaternary δ 31.10 ppm (C-20) indicates the β – functionality of these methyls. The compound / molecule was established as β – Amyrin Acetate (Figure 2A), on the basis of spectral data (1D1H-NMR, 13C NMR, 2D HSQC, HMBC spectroscopy, melting point 242OC, and TLC. More so, comparison with literature data (36-37), confirmed the molecule as β –Amyrin Acetate. Studies show this compound to possess profound anti-inflammatory, anti-malarial and anti-rheumatism activities (38) and (39). Spectroscopic signals in the PMR (Table S4) and 13C NMR (Table S3) spectra of Taraxasterol were assigned com- pletely using one- and two-dimensional (2D) NMR methods (HSQC and HMBC). The weak-field region of the PMR of the compound contains signals for three protons. Based on cross-peaks in the HSQC, the first two protons at 5.10 ppm belong to C-30 with Chemical shift 107.25 ppm. The doublet of doublets at 4.70 ppm corresponds with the proton on C-3 (80.75 ppm) of ring A. Cross-peaks with C-2 and a quaternary C atom at 37.74 ppm (C-4) are found in the HMBC spectrum for H-3. Cross-peaks with protons at 0.78 ppm (1H, triplet; corresponds with the C atom with Chemical Shift (CS) 55.42 ppm) at HSQC. The protons with Chemical shift 0.91 ppm in the HSQC correspond with signals for C atoms with Chemical Shifts 28.06 and 16.36 ppm of the gem-dimethyls C-23 and C-24. Their protons also correlate with tertiary (55.42 ppm) and quaternary (37.74 ppm) C atoms C-5and C-4, respectively. Their characteristic atoms C-6 (18.37 ppm) and C-7 (34.13 ppm) are found using the HSQC spectra. Protons H-1 and H-5 and protons of the methyl with Chemical Shift 0.86 ppm, give correlations in the HMBC spectrum with a quaternary C atom with δ 38.01 ppm. These are C-10 and methyl C-25. In the HSQC spectrum, the last signal correlates with H-9 with δ1.33 ppm which, in turn, correlates with C-10 and yet another quaternary C atom with Chemical Shift 40.62 ppm (C-8), C-8 couples with methyl protons at 0.99 ppm (on C-26 with δ16.36 ppm). The doublet for methyl protons H-29 at 1.08 ppm in the HSQC spectrum corresponds with the C atom at 25.60 ppm(C-29). Corresponding cross-peaks in the HMBC spectrum between methyl protons in ring D at 1.08 ppm and C atoms at 38.30 (C-19) and 154.65 ppm (C-20) in addition to C-29 at 25.60 ppm and a proton at 2.15 ppm (H-19) confirm that the assignments were correct. Methyl protons with CS 0.94 ppm, which correspond in the HSQC spectrum with the C atom at 19.58 ppm (C-28) have correlations in the HMBC spectrum with C atoms at 34.41 and 48.70 ppm (Tables S3 and S4). These assignments in comparison with literature data (40-41), confirmed the compound to be taraxasterol (Figure 2B). The presence of carbon spectrum in 145ppm and 178ppm at the 13C NMR indicate the presence of fatty acid impurities. This compound was reported to have many biological properties, including anti-inflammatory and anti-tumour activities [42]. Oral bioavailability (drug-likeness) prediction for therapeu- tic ligands is a fundamental evaluation in novel drug discovery and development owing to the fact that oral delivery remains the most common path of drug delivery into the systemic circulation [34]. This essential parameter was assessed for the two phytochemicals taking cognisance of Lipinski’s rule of five and the Veber’s rule. The results, thus, presented in Table 3, implies that they obey both rules. Thus, the extract could be taken through the oral route. A major cause of high attrition rate in drug discovery and development is poor pharmacokinetic and toxicity profiles of drug candidates. A way of circumventing this challenge is early prediction of ADMET profiles of drug candidates. Table 4 presents the in silico ADMET profile of the two investigated 6 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 7 phytochemicals. All the ligands were found to possess good gastrointestinal absorption. The blood brain barrier (BBB) is a layer of endothelial cell that demarcate the brain from blood [44]. The assessment of BBB permeating potentials of the compounds shown in Table 4 portrayed that none of the compounds can penetrate the BBB and as such would not have any influence on the Central Nervous System. Also, P-glycoproteins (P-gp) are intracellular and extracel- lular membrane transporters of xenobiotic in the body [45]. It reduces cellular concentrations of its substrates leading to their poor pharmacokinetic profiles. All the investigated phytochemicals were found to be none substrates of P-gp (Table 4). Furthermore, Cytochrome 450 (CYP450) monooxygenase is a group of enzymes central to the metabolism and excretion of drugs. The bioactive ligands were screened against five isoforms of the enzyme. The result (Table 4) revealed that all the phytochemicals are substrate of CYP450 enzyme indicative of their high probabilities of been bio-transformed and eventually made bio available upon oral administration [46]. Another important pharmacokinetic parameter worthy of consideration especially for therapeutic compounds that re- quires transdermal administration is the skin permeability (LogKp). The LogKp data of β -Amyrin acetateand Taraxas- terol presented in Table 4 revealed that both the compounds have poor skin penetration potentials due to the negative values of their LogKp (45). Additionally, the toxicity profiles (Table 4) of the compounds revealed that none of them is mutagenic, tumorigenic, irritating, or pose any adverse effect on the repro- ductive system. 6. Conclusions Crude extracts of the bulbs of Calotropisprocerawere in- vestigated for the antimicrobial activities against eight bacteria and three fungi species. The extracts were found to exhibit sig- nificant inhibitory activities against the microbes with the crude ethyl acetate extract having the highest effective activity against Escherichia coli (MIC 2.5mg/mL and MBC/MFC 5mg/mL), Staphylococcus aureus (MIC 2.5mg/mL), Candida albicans, Salmonella typhiand Candida stellafoidea(MIC 5mg/mL). The phytochemical screening of the extracts confirmed the presence of β -Amyrin acetate and Taraxasterol. Theoretical studies on the binding interaction of the two identified phytochemicals with the active sites of DNA gyrase of Escherichia coli and Salmonella typhi revealed that the compounds bind to the tar- get macromolecule via hydrophobic and hydrogen bond inter- actions with binding affinity of -9.6 kcal/mol and -9.5 kcal/mol for β -Amyrin acetate and Taraxasterol, respectively. The phy- tochemicals were found to be better inhibitors of DNA gyrase when compared with the standard Ciprofloxacin ligand which binds to the target macromolecule with binding affinity of -7.6 kcal/mol. In addition, in silico drug-likeness and ADMET in- vestigations on the compounds showed that they obey both the Lipinski’s rule of five and the Veber’s rule in addition to dis- playing excellent pharmacokinetic and toxicity profiles. Acknowledgements The authors acknowledge the technical support of Strath- clyde Institute of Pharmacy and Biomedical Sciences, Uni- versity of Strathclyde Glasgow, United Kingdom and College of Forestry and Fisheries, Federal University of Agriculture Makurdi, Benue State Nigeria. References [1] B. B. Mishra, & V. K. Tiwari, “Natural products: An evolving role in future drug discovery”, Eur. J. Med. Chem. 46 (2011) 4769. [2] J. Rey-Ladino, A. G. Ross, A. W. Cripps, D. P. McManus, & R. Quinn, “Natural products and the search for novel vaccine adjuvant”, Vaccine 29 (2011) 6464. [3] G. M. Cragg, & D. J. Newman, “Biodiversity: A continuing source of novel drug leads”, Pure Appl. Chem. 77 (2005) 7. [4] B. Haefner, “Drugs from the deep: Marine natural products as drug can- didates”, Drug Discov. Today 8 (2003) 536. [5] M. S. Butler, “The role of natural product in chemistry in drug discovery”, Journal of Natural Product chemistry 67 (2004) 2141. [6] D. A. Dias, S. Urban, & U. Roessner, “A historical overview of natural products in drug discovery”, Metabolites 2 (2012) 303. [7] P. M. Dewick, “Medicinal Natural Products: A Biosynthentic Approach”, 2nd. John Wiley and Son; West Sussex, UK, (2002). [8] R. A. Maplestone, M. J. Stone, & D. H. Williams, “The evolutionary role of secondary metabolites- A review”, Gene 115 (1992) 151. [9] S. M. Colegate, R. J. Molyneux, “Bioactive Natural Products: Detec- tion, Isolation and Structure Determination”, CRC Press; Boca Raton, FL, USA, (2008). [10] R. M. I. Sabrin, A. M. Gamal, A. S. Lamiaa, M. Y. B. Laetitia, K. Robert, T. A. Y. Diaa, “Cytotoxic and Antibacterial Effects of Aqueous Extract of Calotropis procera Latex”, International Journal of Pharmacy And Phar- maceutical Research 16 (2019) 400. [11] N. Tour, & G. Talele, “Anti-inflammatory and gastromuscal protective effects of Calotropis procera (Asclepiadaceae) stem bark”, J. Nat. Med. 65 (2011) 598, doi:10-1007/s11418-011-0522-1. [12] H. B. Fernandes, D. L. Machado, J. M. Dias, T. V. Brito, J. A. Batista, R. O. Silva, A. Pereira, G. P. Ferreira, M. V. Ramos, J. V. R. Medeiros, K. S. Aragão, R. A. Ribeiro, A. L. R. Barbosa, & J. S. Oliveira, “Laticifer proteins from Plumeria pudica inhibit the inflammatory and nociceptive responses by decreasing the action of inflammatory mediators and pro- inflammatory cytokines”, Revista Brasileira de Farmacognosia 25 (2015) 269. [13] M. Yelne, P. Sharma, T. Dennis, “Database on medicinal plants used in ayurveda”, central council for research in ayurveda and siddha, New Delhi 2 (2000) 69. [14] S. R. Ibrahim, G. A. Mohamed, L. A. Shaala, L. M. Banuls, G. V. Goi- etsenoven, R. Kiss, “New ursane-type triterpenes from the root bark of Calotropisprocera”, Phytochem Lett 5 (2012) 490. [15] H. Shrikant, S. Ashwinikumar, D. Mayur, J. Shreeram, K. Kisan, H. Man- ish, “Colloids Surf., B: Biointerfaces”, Nat. Med. 95 (2011) 284. [16] T. T. Talele, A. K. Santosh, & C. R. Alan, “Successful Applications of Computer Aided Drug Discovery: moving Drugs from Concept to the Clinic”, Current Topics in Modern Chemistry 10 (2010) 127. [17] R. Devi, S. J. Singh, J. Powel, E. A. Fulton, E. Igbinedion, & K. Reels, “Internet-based interventions for secondary prevention of coronary heart disease (Review)”, Cochrane Database of Systematic Reviews 12 (2015) CD009386. [18] M. Z. Nunes, D. Bernardi, C. A. Baronio, J. Pastinato, M. Baldin, & M. Botton, “A laboratory bioassay method to assess the use of toxic bait on anastrpha fraterculus (Weidemann 1830)”, Neotrop Entomol. 1 (2019) 124. 7 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 8 [19] Y. Liu, M. Kumar, G. G. Katul, & A. Porporato, “Reduced resilience as a potential early warning signal of forest mortality”, Nature Climate Change 9 (2019) 880. [20] H. L. Malou, C. R. Susana, & M. L. M. Luis, “Current Challenges to- ward In Vitro Cellular Validation of Inorganic Nanoparticles”, American Chemical Society 28 (2017) 212. [21] G. Mugumbate, V. Mendes, M. Blaszczyk, M. Sabbah, G. Papadatos, J. Lelievre, L. Ballell, D. Barros, C. Abell, T.L. Blundell, & J.P. Over- ington, “Target Identification of Mycobacterium tuberculosis Phenotypic Hits Using a Concerted Chemogenomic, Biophysical, and Structural Ap- proach”, Sec. Experimental Pharmacology and Drug Discovery 8 (2017) 681, https://doi.org/10.3389/fphar.2017.00681. [22] M. Wójcikowski, P. Ballester, & P. Siedlecki, “Performance of machine- learning scoring functions in structure-based virtual screening”, Scientific Report 7 (2017) 46710, https://doi.org/10.1038/srep46710. [23] J. K. Carpenter, L. A. Andrews, S. M. Witcraft, M. B. Powers, J. A. J. Smits, & S. G. Hofmann, “Cognitive behavioral therapy for anxiety and related disorders: A meta-analysis of randomized placebo-controlled tri- als”, Depress Anxiety 6 (2018) 502, doi: 10.1002/da.22728. [24] Z. Dutkiewicz, & R. Mikstacka, “Structure-Based Drug Design for Cy- tochrome P450 Family 1 Inhibitors”, Bioinorg Chem Appl. 2018 (2018) 3924608, doi:10.1155/2018/3924608. [25] S. Surabhi, & B. Singh, “Computer aided drug de- sign: an overview”, Jddt Internet 8 (2023) 504, https://jddtonline.info/index.php/jddt/article/view/1894 [26] J. P. Ameji, U. Uzairu, G. A. Shallangwa, S. Uba, “Design, pharma- cokinetic profiling, and assessment of kinetic and thermodynamic stabil- ity of novel anti-salmonella typhiimidazole analogues’, Bulletin of the National Research Centre 47 (2023) 6, https://doi.org/10.1186/ s42269-023-00983-5 [27] J. P. Ameji, U. Uzairu, G. A. Shallangwa, S. Uba, “Molecular dock- ing simulation, drug-likeness assessment, and pharmacokinetic study of some cephalosporin analogues against a penicillin-binding protein of Salmonella typhimurium”, The Journal of Antibiotics 6 (2023) 211, https://doi.org/10.1038/s41429-023-00598-y [28] E. Daoud, C. Caimino, M. A. Akeroyd, A. J. Norena, & D. M. Baguley, “The utility of economic measures to quantify the burden of tinnitus in affected individuals: A scoping review”, Pharmacoecon open 6 (2021) 21. [29] F. Z. Sabri, M. Belarbi, S. Sabri, M. S. A. Muneer, “Phytochemical screening and identification of some compounds from mallow”, J Nat Prod Resour 2 (2012) 512. [30] L. U. Akacha, M. E. Khan, J. Anyam, J. Igoli, T. A. Anyiin, J. Y. Dikko, “Phytochemical screening and Antimicrobial activity of Bryophyllum pin- natum Extracts”, British Biotechnology Journal 16 (2016) 1. [31] K. O. Akinyemi, O. Oladapo, C. E. Okwara, C. C. Ibe, K. A. Fasure, “Screening of crude extracts of six medicinal plants used in Southwest Nigerian unorthodox medicine for antimethicillin resistant Staphylococ- cus aureus activity”, BMC Complementary and Alternative Medicine 5 (2005) 6. [32] G. H. Shahidi, “Evaluation of antibacterial properties of Iranian medic- inal plants against Micrococcus aureus, Serratiamarcescens, Klebsiella pneunomiae and Bordella bronchoseptica”, Asian Journal of Sciences 3 (2004) 82. [33] J. P. Ameji, U. Uzairu, G. A. Shallangwa, S. Uba, “Obstructing Salmonella typhi’s virulence in eukaryotic cells through design of its SipB protein antagonists”, Journal of Taibah University Medical Sciences 18 (2023) 726e736. [34] J. P. Ameji, U. Uzairu, G. A. Shallangwa, S. Uba, “Virtual screening of novel pyridine derivatives as effective inhibitors of DNA gyrase (GyrA) of salmonella typhi”, Current Chemistry Letters 12 (2022) 1, DOI:~10. 5267/j.ccl.2022.10.002 [35] M. E. Khan, L. M. Bala, M. Maliki, “Phytochemical Analyses of Termi- naliaschimperiana (Combretaceae) Root Bark Extract to Isolate Stigmas- terol”, Adv. J. Chem. A 2 (2019) 327. [36] D. Ercil, M. K. Sakar, E. D. Olmo, A. S. Feliciano, “Chemical con- stituents of Rinariaaucheri”, Turkish Journal of Organic Chemistry 28 (2004) 133. [37] O. A. Ushie, H. M. Adamu, L. Y. Chindo, “Beta-Amyrin-3- acetate detected in Methanolic extract of Chrysophyllumalbidium”, Journal of Chemical Society of Nigeria 43 (2018) 89. [38] N. N. Okoye, D. L. Ajaghaka, N. Okeke, E. E. Ilodigwe, C. S. Nworu, & F. C. B. Okoye, “Beta -Amyrin and alpha -amyrin acetate isolated from the stem bark of Alstoniaboonei display profound anti-inflammatory ac- tivity”, Journal of pharmaceutical Biology 52 (2014) 1478. [39] L. Hernandez-Vaquez, S. Mangas, J. Palazon, & A. Navarrro-Ocana, “Valuable medicinal plants and resins: Commercial phytochemicals with bioactive properties”, Ind. Crop. prod. 31 (2010) 476. [40] W. F. Reynold, J. F. Sawyer, R. G. Enrique, L. I. Escobar, M. A. Chavez, J. N. Schoolery, “Total assignment of the spectrum of taraxasteryl acetate by 13C - 13C connectivity experiments and determination of the stereochem- istry of taraxasterol by X- ray diffraction”, Canadian journal of chemistry 63 (1985) 1048. [41] E. R. Shakurova, T. I Parfenova, R. S. Sufiyarva, A. Z. Khalilova, V. R. Khmetora, S. A. Bashkator, “Synthesis and anti-inflammatory activ- ity of acyl derivatives of taraxasterol”, Pharmaceutical chemistry Journal 42 (2008) 319. [42] K. Sharma, & R. Zafar, “Occurrence of taraxerol and taraxasterolin medicinal plants”, Pharmacognosy Review 9 (2015) 19. [43] Y. Wang, J. Xing, Y. Xu, N. Zhou, J. Peng, Z. Xiong, X. Liu, X. Luo, C. Luo, K. Chen, & M. Zheng, “In silico ADME/T modelling for rational drug design”, Q Rev. Biophys. 48 (2015) 488. [44] G. M. Levin, “P-glycoprotein: why this drug transporter may be clinically important”, Curr Psychiatry 11 (2012) 38. [45] T. S. Maliehe, P. H. Tsilo, & J. S. Shandu, “Computational Evaluation of ADMET Properties and Bioactive Score of Compounds from Encephalar- tosferox”, Pharmacogn J. 12 (2020) 1357. [46] M. F. Khan, M. A. Bari, M. K. Islam, M. S. Islam, M. S. Kayser, N. Na- har, M. Al-Faruk, & M. A. Rashid, “The natural anti-tubercular agents: In silico study of physicochemical, pharmacokinetic and toxicological prop- erties”, J. App. Pharm. Sci. 7 (2017) 034. Appendix Supplementary file: 8 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 9 Table S1: 13C NMR Experimental and Literature Data of β-Amyrin acetate Position Experimental data 13C (δppm) Okoyeet al., 2014 13C (δ ppm) Ushieet al., 2018 13C (δ ppm) Chaturved- ula&rakash, 2013 13C (δ ppm) Abdullahiet al., 2017 13C (δ ppm) Wau, 2016 13C (δ ppm) Ercilet al., 2004 13C (δ ppm) 1 38.91 38.79 38.70 39.30 38.60 38.40 38.14 2 26.70 27.44 28.80 28.30 23.90 23.70 27.51 3 80.35 79.24 79.10 79.60 80.60 81.10 79.12 4 38.91 38.99 38.5 39.10 38.60 37.80 38.88 5 55.40 55.37 55.5 55.10 51.50 55.30 55.27 6 18.24 18.58 18.80 18.80 18.20 18.70 18.44 7 32.85 32.85 33.10 33.20 33.30 32.80 33.03 8 40.05 40.21 38.80 39.80 37.80 39.90 38.82 9 47.59 47.43 49.20 48.00 43.30 47.40 47.81 10 37.08 37.15 36.70 37.40 36.80 36.80 37.00 11 23.75 23.75 22.70 23.90 22.70 23.70 23.48 12 121.68 121.93 116.80 122.20 129.80 122.30 121.82 13 145.21 145.41 142.70 145.70 142.70 145.50 145.28 14 42.07 41.92 41.30 42.10 42.80 41.60 42.18 15 26.36 26.36 27.40 26.00 27.90 27.00 26.12 16 26.80 27.14 27.10 26.30 26.20 26.20 27.37 17 32.70 32.70 49.20 32.60 32.50 32.60 32.04 18 48.04 47.84 33.30 48.10 55.60 47.50 47.22 19 46.85 47.03 48.70 47.30 40.80 46.99 46.93 20 31.10 31.30 29.10 31.30 41.70 31.30 31.34 21 38.20 37.35 35.10 34.30 31.30 34.80 34.83 22 34.56 34.94 37.50 37.40 42.80 37.20 37.26 23 15.71 15.71 15.43 16.10 16.70 16.85 28.22 24 28.30 28.31 29.70 28.20 29.70 28.40 15.47 25 16.01 15.80 15.40 16.10 16.60 16.00 15.72 26 16.92 17.01 17.54 17.20 16.10 17.00 16.97 27 26.21 26.62 21.32 24.40 18.10 26.00 25.56 28 28.60 28.62 28.00 28.20 27.90 28.80 28.44 29 33.42 33.56 33.70 33.90 27.50 33.40 33.44 30 23.86 23.91 25.90 23.80 25.30 23.60 23.48 11 170.94 171.53 170.91 171.00 173.71 171.10 171.01 21 21.53 21.53 29.80 21.53 21.20 21.30 21.80 9 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 10 Table S2: 1H Experimental and Literature Data NMR of β− Amyrin acetate Position Experi- mental data 1H (δ ppm) Okoyeet al., 2014 1H (δ ppm) Ushieet al., 2018 1H (δ ppm) Abdullahiet al., 2017 1H (δ ppm) Ercilet al., 2004 1H (δ ppm) Chaturvedula and Prakash, 2013 1H (δ ppm) Wau, 2016 1H (δ ppm) 1 1.61 1.49 1.31 1.65 2 1.64 1.55 1.67 1.60 1.64 3 4.47 3.20 3.20 4.47 3.23 3.26 4.50 4 - - - - - - 5 0.81 0.71 0.86 0.81 0.87 0.69 0.87 6 1.60 1.53 1.58 1.57 1.54 7 1.52 1.28 1.36 8 - - - - - - - 9 1.63 1.95 1.65 1.58 1.58 10 - - - - - - 11 1.80 1.84 1.96 2.25 1.86 12 5.16 5.16 5.50 4.83 5.12 5.18 5.18 13 - - - - - 14 - - - - - 15 2.00 1.99 2.00 16 0.79 1.60 0.97 17 - - - - 18 2.28 1.89 2.04 2.23 1.97 19 1.63 1.59 1.93 2.24 1.67 20 - - - 2.27 21 1.72 1.66 1.31 1.13 1.38 22 0.88 1.63 - 1.39 23 0.77 0.77 0.82 0.90 0.83 0.77 0.86 24 0.98 0.98 0.84 0.84 0.84 0.91 0.87 25 0.92 0.92 0.93 0.91 0.94 0.94 0.97 26 0.94 0.94 0.95 0.98 0.79 0.76 0.96 27 1.11 1.11 0.97 1.04 0.97 1.21 1.13 28 0.81 0.81 1.00 0.83 0.99 1.09 0.83 29 0.85 0.85 1.10 0.86 0.85 0.87 30 0.85 0.85 1.20 0.87 0.78 0.87 11 - - - - - - 21 2.02 2.01 2.07 1.57 2.06 2.05 10 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 11 Table S3: The 13C NMR Experimental and Literature Data of Taraxasterol position Experimental data 13C (δ ppm) Khalilovet al., 2003 13C (δ ppm) Reynoldet al., 1985 13C (δ ppm) Alavi and Yekta, 2008 13C (δ ppm) Shakurova et al., 2008 13C (δ ppm) Mahato and Kundu, 1994 13C (δ ppm) 1 38.45 38.40 38.44 38.90 38.80 2 23.60 23.60 23.70 27.70 23.70 3 80.75 80.80 80.96 79.00 79.00 4 37.74 37.70 37.79 38.70 80.94 38.80 5 55.42 55.40 55.40 55.20 55.40 6 18.37 18.10 18.18 18.40 18.30 7 34.13 33.90 33.99 34.00 34.10 8 40.62 40.80 40.91 40.80 40.90 9 50.47 50.30 50.39 50.40 50.50 10 37.00 37.00 37.04 37.00 37.10 11 21.50 21.40 21.46 21.50 21.50 12 25.52 25.50 26.15 26.10 26.20 13 38.80 38.80 39.15 39.20 39.20 14 41.85 41.90 42.03 42.10 42.00 15 26.60 26.60 26.64 26.50 26.70 16 39.00 39.10 38.29 38.40 38.30 17 34.41 34.40 34.53 34.40 34.50 18 48.70 48.60 48.63 48.70 48.70 19 38.30 38.30 39.28 39.40 39.40 20 154.65 154.40 154.64 154.60 154.54 154.70 21 25.62 25.40 25.61 25.50 25.60 22 38.93 39.30 38.85 38.90 38.90 23 28.06 27.80 27.94 28.00 28.78 28.00 24 16.36 16.40 16.51 15.40 16.48 15.40 25 15.51 15.40 16.34 16.90 14.54 16.80 26 16.36 16.20 15.89 16.00 16.11 15.90 27 14.85 14.60 14.72 14.90 14.09 14.80 28 19.58 26.10 19.49 19.40 19.53 19.50 29 25.06 19.40 25.49 25.50 25.94 25.50 30 107.25 107.00 107.12 107.20 107.23 107.10 11 Khan et al. / J. Nig. Soc. Phys. Sci. 5 (2023) 1576 12 Table S4: 1H NMR Experimental and Literature Data of Taraxasterol Postion Experimental data 1H (δ ppm) Khalilovet al., 2003 1H (δ ppm) Alavi and Yekta, 2008 1H (δ ppm) Mahoto and Kundu, 1994 1H (δ ppm) 1 0.92 0.92 2 1.67 1.67 3 4.70 4.70 3.22 3.22 4 - - 5 0.78 0.78 0.77 6 1.52 1.46 7 1.35 1.35 8 - - 9 1.33 1.33 10 - - 11 1.51 1.48 12 1.65 1.65 1.03 13 1.56 1.56 14 - - 15 1.65 1.65 0.93 16 1.17 1.26 17 - - 18 0.99 0.99 19 2.15 2.15 20 - - 21 2.48 2.48 22 1.41 1.41 23 0.91 0.91 0.77 0.71 24 0.91 0.91 0.85 0.77 25 0.86 0.86 0.86 0.86 26 0.99 0.99 1.02 0.97 27 0.96 0.96 0.93 28 0.94 0.94 0.85 0.98 29 1.08 1.08 1.02 0.96 30 5.10 4.79 4.62 4.66 12