Skip to content
BY-NC-ND 4.0 license Open Access Published by De Gruyter Open Access July 18, 2018

Density functional theory calculations, vibration spectral analysis and molecular docking of the antimicrobial agent 6-(1,3-benzodioxol-5-ylmethyl)-5-ethyl-2-{[2-(morpholin-4-yl)ethyl] sulfanyl}pyrimidin-4(3H)-one

  • Maha S. Almutairi , S. Soumya , Reem I. Al-Wabli , I. Hubert Joe and Mohamed I. Attia EMAIL logo
From the journal Open Chemistry

Abstract

Vibrational spectral analysis and quantum chemical computations based on density functional theory have been performed on the antimicrobial agent 6-(1,3-benzodioxol-5-ylmethyl)-5-ethyl-2-{[2-(morpholin- 4-yl)ethyl]sulfanyl}pyrimidin-4-(3H)-one.The equilibrium structural geometry, various bonding features and harmonic vibrational wavenumbers of the title compound have been investigated using DFT-B3LYP function at 6-311++G(d, p) basis set. The detailed interpretations of the vibrational spectra have been carried out with the aid of VEDA 4 program. The various intramolecular interactions of the title compound have been exposed by natural bond orbital analysis. The FT-IR and FT-Raman spectra of the title molecule have been recorded and analyzed. Blue-shifting of the C-H wavenumber along with a decrease in the C-H bond length attribute for the formation of the C-H...O hydrogrn bonding provide an evidence for a charge transfer interaction. Also, the distribution of natural atomic charges reflects the presence of intramolecular hydrogen bonding. The analysis of the electron density of HOMO and LUMO gives an idea of the delocalization and the low value of energy gap indicates electron transfer within the molecule. Moreover, molecular docking studies revealed the possible binding of the title molecule to different antimicrobial target proteins.

Graphical Abstract

1 Introduction

Heterocyclic compounds bearing nitrogen atoms in their structural skeletons are found in many biologically active natural products and display appreciable therapeutic applications: among them, pyrimidines which constitute eminent parts in the chemistry of nucleic acids [1]. The pyrimidine system constitutes the key pharmacophore of several non-nucleoside chemotherapeutic agents due to their ability to inhibit vital enzymes responsible for DNA biosynthesis, such as dihydrofolate reductase, thymidylate synthetase, thymidine phosphorylase and reverse transcriptase [2]. In addition, the multi-functionalized pyrimidines exhibit diverse pharmacological efficiencies including anticancer [3,4,5], antiviral [6], antitubercular [7], antibacterial [8], anti-fungal [9] and anti-inflammatory [10] activities. Uracils are pyrimidine derivatives which are considered as privileged structures in drug innovation process due to their wide spectrum of biological activities, synthetic accessibility and ability to give drug like properties [11,12,13].

On the other hand, a number of biologically active molecules have the 1,3-benzodioxole fragment in their chemical skeletons and they display a wide array of biological activities such as anticancer [14], antioxidant [15], antiprotozoal [16] and immunomodulatory [17] activities.

Molecular hybridization is a well documented effective strategy in synthetic medicinal chemistry to get bioactive new chemical entities [18]. Covalent combination of two or more drug pharmacophores into a single molecule gave molecular hybrids which might devoid of unwanted side effects with concomitant synergic effect and better activity in many cases [19].

Molecular hybridization of thiouracil nucleus and 1,3-benzodioxole fragment furnished the title compound 6-(1,3-benzodioxol-5-ylmethyl)-5-ethyl-2-{[2-( morpholin- 4-yl)ethyl]sulfanyl}pyrimidin-4(3H)-one which displayed antimicrobial activity against Bacillus subtilis, Bacillus cereus and Aspergillus niger with minimum inhibitory concentration (MIC) value of 0.19 μmol/mL [20]. Therefore, we were encouraged to carry out the current spectroscopic characterization and density functional theory (DFT) computations on the title compound aiming to get more insights into its electronic properties as well as its natural charge distribution which might influence the interaction with its target protein. Moreover, molecular docking studies explored the possible binding mode of the title compound to its target protein.

2 Materials and methods

2.1 General

The FT-Raman spectrum of the title compound was carried out on a Bruker RFS-27 FT-Raman spectrometer (Bruker, Billerica, MA, USA) in the region 4000-50 cm-1. The 1064 nm line of Nd:YAG laser operating at 100 mW power was used for excitation and the spectral resolution was 2 cm-1. The FT-IR spectrum of the title compound was recorded in the 4000-400 cm-1 range using a Perkin Elmer RXL spectrophotometer (Perkin-Elmer, Ayer Rajah Crescent Level, Singapore), with sample in KBr using pellet press method. The spectral resolution was 1 cm-1.

2.2 Synthesis

The title compound molecule was prepared as previously reported in the literature [20].

2.3 Computational details

All DFT Computations of the title compound have been performed using Gaussian ‘09 [21] program package at the Becke3-Lee-Yang-Parr (B3LYP) level with 6-311++G(d, p) basis set [22,23,24,25]. In order to correct the over-estimations arising from some negative factors such as basis set incompleteness and anharmonicity characters of the vibrational modes, the calculated wavenumbers were scaled using a uniform scaling factor of 0.9673 [26]. The distributions of assignment of the calculated wavenumbers were aided by VEDA4 program [27]. Natural charge analyses along with HOMO-LUMO have been used to elucidate information regarding charge transfer within the molecule. The Raman activities (Si) calculated in the harmonic frequency calculations were later converted to relative Raman intensities (Ii) using the following relationship derived from the basic theory of Raman scattering [28],

Ii=f(νoνi)4siνi[1exp(hcνikT)](1)

where v0 is the exciting wavenumber (in cm-1), vi is the vibrational wavenumber of the ith normal mode, h, c and k are the universal constants and ‘f’ is a suitably chosen common scaling factor for all the peak intensities. The simulated IR and Raman spectra were plotted using pure Lorentzian band shapes with a full width half maximum (FWHM) of 10 cm-1.

Ethical approval: The conducted research is not related to either human or animals use.

3 Results and Discussion

3.1 Synthesis

The title compound was prepared as depicted in Schemes 1 and 2 by adopting the reported procedure [20]. The spectral data of the title molecule were in agreement with the reported ones [29].

3.2 Optimized geometry

The optimized molecular structure of 6-(1,3-benzodioxol- 5-ylmethyl)-5-ethyl-2-{[2-(morpholin-4-yl)ethyl] sulfanylpyrimidin-4(3H)-one obtained from DFT calculations at B3LYP level with 6-311++G(d, p) basis set is shown in Figure 1 and the geometrical parameters obtained from optimized geometry and XRD data are presented in Table 1. The asymmetry of the benzene ring is evident from the negative deviation in the bond angles of C36-C35-C34 and C37-C38-C39 as well as the positive deviation of the remaining angles from the normal value of 120°, which might be attributed to the presence of 1,3-dioxolane group. The interesting structural features of the title compound are the decrease of the cyclic angles C36-C35-C34 and C37-C38-C39 along with the increase of the neighbouring angles around the ring indicating a charge transfer (CT) interaction in the 1,3-benzodioxole moiety. Similarly, the decrease of the bond angle of C39- C34-C35 by ~3.4° and the increase of the bond angle of C34-C39-C38 by ~2.42° is associated with CT interaction between the 1,3-benzodioxole and pyrimidinone groups. The DFT calculations also gave a decrease in the bond angle of C31-C34-C35 by 0.35° and an increase in the bond angle of C31-C34-C39 by 0.75° from 120° at C34 position. This asymmetry of exocyclic angles reveals the repulsion between the CH2 group and the phenyl ring. Hyperconjugation of the carbonyl group with the adjacent C18-C19 single bond, which is evident from the bond distance of 1.22 and 1.46 Å for C18=O23 and C18-C19, respectively.

Scheme 1 Preparation of the intermediate compound 4.Reagents and conditions: i) NaBH4, methanol, RT, 18 h; ii) SOCl2, RT, 18 h; iii) NaCN, KI, DMF, RT, 18 h
Scheme 1

Preparation of the intermediate compound 4.

Reagents and conditions: i) NaBH4, methanol, RT, 18 h; ii) SOCl2, RT, 18 h; iii) NaCN, KI, DMF, RT, 18 h

Scheme 2 Preparation of the title molecule 8.Reagents and conditions: i) (1) Zn/THF (2) K2CO3 (3) HCl; ii) (1) NH2CSNH2, NaOEt, ethanol, reflux, 24 h (2) HCl; iii) N-(2-chloroethyl) morpholine hydrochloride, K2CO3, DMF, RT, 18 h.
Scheme 2

Preparation of the title molecule 8.

Reagents and conditions: i) (1) Zn/THF (2) K2CO3 (3) HCl; ii) (1) NH2CSNH2, NaOEt, ethanol, reflux, 24 h (2) HCl; iii) N-(2-chloroethyl) morpholine hydrochloride, K2CO3, DMF, RT, 18 h.

Figure 1 The optimized molecular structure of the title molecule based on the B3LYP/6-311++G(d, p) level of basis set.
Figure 1

The optimized molecular structure of the title molecule based on the B3LYP/6-311++G(d, p) level of basis set.

Table 1

The calculated optimized geometrical parameters of compound at B3LYP/6-311++G(d, p) level of basis set.

Bond lengthCalc. (Å)Expt. (Å)Bond angleCalc. (°)Expt. (°)Torsion angleCalc. (°)Expt. (°)
C1-C21.541.51C2-C1-N6108.64111.34N6-C1-C2-O339.29-58.69
C1-N61.471.46C 2-C1-H7109.07109.31N6-C1-C2-H9161.58179.61
C1-H71.090.97C2-C1-H8111.59109.31N6-C1-C2-H10-79.0762.19
C1-H81.100.97N6-C1-H7108.62109.43H7-C1-C2-O3157.5262.33
C2-O31.431.43N6-C1-H8111.29109.39H7-C1-C2-H9-80.18-58.79
C2-H91.100.97H7-C1-H8107.58107.98H7-C1-C2-H1039.1659.40
C2-H101.090.97C1-C2-O3112.94111.11H8-C1-C2-O3-83.77-179.67
O3-C41.421.42C1-C2-H9109.24109.39H8-C1-C2-H938.5359.40
C4-C51.531.51C1-C2-H10110.41109.40H8-C1-C2-H10157.87-58.79
C4-H111.100.97O3-C2-H9109.64109.43C2-C1-N6-C5-58.2754.39
C4-H121.090.97O3-C2-H10105.90109.38H7-C1-N6-C5-176.78-66.56
C5-N61.481.47H9-C2-H10108.59108.07C1-C2-O3-C419.5159.80
C5-H131.090.97C2-O3-C4113.39108.80H9-C2-O3-C4-102.56-170.34
C5-H141.100.97O3-C4-C5111.13111.84H10-C2-O3-C4140.47-61.08
S15-C161.781.75O3-C4-H11110.80109.25C2-O3-C4-C5-64.03-59.41
S15-C161.811.75O3-C4-H12106.56109.26C2-O3-C4-H12176.48179.52
C16-N171.361.37C5-C4-H11110.70109.45O3-C4-C5-N643.9857.39
C16-N211.291.30C5-C4-H12109.72109.24O3-C4-C5-H13-74.60-63.52
N17-C181.411.38H11-C4-H12107.79107.96O3-C4-C5-H14165.94178.28
N17-H221.010.92C4-C5-N6108.89110.99H11-C4-C5-N6-79.60-63.66
C18-C191.461.45C 4-C5-H13110.13109.37H11-C4-C5-H13161.83175.43
C18-O231.221.24C4-C5-H14109.04109.45H11-C4-C5-H1442.3657.23
C19-C201.371.37N6-C5-H13108.27109.45H12-C4-C5-N6161.57178.48
C19-C241.511.50N6-C5-H14111.58109.47H12-C4-C5-H1342.9957.57
C20-N211.381.38H13-C5-H14108.92108.05H12-C4-C5-H14-76.48-60.64
C20-C311.521.51C1-N6-C5115.38109.27C4-C5-N6-C116.74-53.38
C24-H251.090.97N6-C48-H49110.77108.91H13-C5-N6-C1136.4967.53
C24-H261.090.97C48-C51-S15113.23112.18H14-C5-N6-C1-103.66-174.21
C24-C271.541.53C51-S15-C16101.48101.53S15-C16-N17-C18-179.60178.67
C27-H281.090.96N17-C16-N21123.06123.63S15-C16-N17-H220.234.48
C27-H291.090.96C16-N17-C18123.17121.88N21-C16-N17-C180.591.81
C27-H301.090.96C16-N17-H22121.45120.58N21-C16-N17-H22-179.58-175.04
C31-H321.090.97C18-N17-H22115.37117.47S15-C16-N21-C20-179.53179.24
C31-H331.100.97N17-C18-C19113.72116.05N17-C16-N21-C200.26-1.28
C31-C341.521.52N17-C18-O23119.58119.72C16-N17-C18-C19-0.28-0.46
C34-C351.411.39C19-C18-O23126.70124.23C16-N17-C18-O23179.72179.66
C34-C391.401.40C18-C19-C20118.54117.45H22-N17-C18-C19179.88176.48
C35-C361.381.37C18-C19-C24115.32115.97H22-N17-C18-O23-0.12-3.39
C35-H431.080.93C20-C19-C24126.14126.57N17-C18-C19-C24179.41179.68
C36-C371.391.38C19-C20-N21123.55123.72O23-C18-C19-C24-0.59-0.45
C36-O401.381.38C19-C20-C31123.69123.87C18-C19-C20-C31-179.14-177.96
C37-C381.381.37N21-C20-C31112.75112.41C24-C19-C20-C310.581.05
C37-O421.381.38C16-N21-C20117.94117.24C18-C19-C24-C2782.6074.61
C38-C391.401.40C19-C24-H25107.44109.00C20-C19-C24-C27-97.13-104.42
C38-H461.080.93C19-C24-H26110.50108.96C19-C20-C31-C34125.59-11.61
C39-H471.080.93C19-C24-C27113.62113.05N21-C20-C31-C34-55.23-69.92
O40-C411.431.44H25-C24-H26106.72107.73C20-C31-C34-C39-58.13109.14
C41-O421.431.42C31-C34-C35119.12119.65H32-C31-C34-C35-112.75-129.46
C41-H441.090.97C31-C34-H39121.20120.75C31-C34-C35-C36178.35175.81
N6-C481.461.46C39-C34-C35119.64119.54C31-C34-C35-H43-1.68-4.19
C48-H501.100.97C31-C34-C39120.63120.75H43-C35-C36-C37-179.60-179.26
C48-H491.090.97C34-C39-C38122.19122.42C35-C36-C37-O42177.61178.27
C51-H521.090.97C34-C35-C36117.70117.36O40-C36-C37-C38-177.75179.17
C5-H531.090.97C37-C38-C39116.94116.60H44-C41-O42-C37-138.76-116.63

The contraction of the C1-H8 bond length (1.091 Å) from the other methylene C-H bonds and the H8….O42 bond distance of 2.59 Å, which is shorter than the van der Waals radii of 2.72 [30], indicate C-H…O hydrogen bonding. Similarly C41-H45 bond length (1.083 Å) is much shorter than any other C-H bonds. H45.....O40 bond length is only 2.012 Å, which is shorter than van der Waals radii, indicating hydrogen bonding. The hydrogen bridge tends to push the two hetero atoms closer to each other.

The linear fitting graphs (Figure 2) manifested the correlation between the experimental and computed results. The correlation between the calculated and experimental bond lengths and bond angles showed R2 values of 0.979 and 0.922, respectively. The statistical results revealed that the computed results are in good agreement with the experimental XRD values. Therefore, this computation method was considered to carry out the required vibrational studies, natural bond orbital and Frontier orbital energy analyses.

Figure 2 Linear curve fitting plots of the calculated and experimental bond lengths and bond angles parameters for the title compound.
Figure 2

Linear curve fitting plots of the calculated and experimental bond lengths and bond angles parameters for the title compound.

3.3 Natural bond orbital analysis

The natural bond orbital (NBO) analysis is proved to be an effective tool for chemical interpretation of hyperconjugative interactions and electron density transfer (EDT) from filled lone electron pairs of the n(Y) of the “Lewis base” Y into the unfilled anti bond σ*(X-H) of the “Lewis acid” X-H in X-H---Y hydrogen bonding systems [30]. NBO analysis provides a description of the structure of a conformer by a set of localized bonds, antibonds and Rydberg extra valence orbitals. Stabilizing interactions between the filled and unoccupied orbitals and destabilizing interactions between the filled orbitals can also be obtained from this analysis [31]. The lowering of orbital energy due to the interaction between the doubly occupied orbital and the unoccupied ones is a very convenient guide to interpret the molecular structure. In energetic terms, hyperconjugation is an important effect [32] in which an occupied Lewis-type natural bond orbital is stabilized by overlapping with a non Lewis-type orbital (either one-center Rydberg or two-center antibonding NBO). This electron delocalization can be described as a charge transfer from a Lewis valence orbital (donor), with a decreasing of its occupancy, to a non-Lewis orbital (acceptor). Several other types of valuable data, such as directionality, hybridization and partial charges, were analyzed in the output of NBO analysis. The second-order perturbation theory analysis of Fock matrix in the NBO of the title compound shows strong intramolecular hyperconjugative interactions, which are presented in Table 2.

Table 2

The most important interactions between ‘filled’ (donors) Lewis-type NBOs and ‘empty’ (acceptors) non-Lewis NBOs of the title compound.

Donor (i)ED (i)eAcceptor (j)ED (j)eE(2)[a] (kcal/mol)E(j)-E(i)[b](a.u.)F(i, j)[c] (a.u.)
π (C19-C20)1.77576π*(C16-N21)0.3660810.200.260.048
π (C19-C20)1.77576π*(C18-O23)0.3434925.900.280.079
n1 (O3)1.96421σ* (C4-H12)0.014022.480.960.044
n2 (O3)1.92515σ*(C4-C5)0.025476.130.660.057
n2 (O3)1.92515σ*(C4-H11)0.030045.830.670.057
n1 (N17)1.60742σ*(C16-N21)0.0196658.600.280.114
n1 (N17)1.60742σ*(C18-O23)0.0096746.000.300.106
n1 (O23)1.97888σ*(N17-C18)0.091841.021.070.030
n1 (O23)1.97888σ*(C18-C19)0.061392.711.150.050
n2 (O23)1.85977σ*(N17-C18)0.0918429.100.640.123
n2 (O23)1.85977π*(C18-C19)0.0613916.600.720.100
n2 (O40)1.86172σ* (C35-C36)0.3793725.300.350.090
n2 (O40)1.86172σ*(C41-H45)0.038606.140.680.059
n2 (O42)1.86390π* (C37-C38)0.3783324.900.350.089
n2 (O42)1.86390σ* (C41-H45)0.038606.220.680.060

The intramolecular hyperconjugative interactions are formed by the orbital overlap between π*(C-C) and π*(C-C) bond orbitals which results in an intramolecular charge transfer (ICT) causing stabilization of the system [33]. These interactions are observed as an increase in the electron density (ED) in C19-C20 anti-bonding orbital that weakens the respective bonds. The strong intramolecular hyperconjugative interaction of π electrons from C16-N21 bond to the π*(C19-C20) bond of pyrimidine ring increases ED. Similar effect is shown by π(C19-C20) to π*(C18-O23). Energy E(2) associated with hyperconjugative interactions n2(O3)→σ*(C4-H11), n2(O3)→σ*(C4-H12), n2(O42)→σ*(C41-H45) and n2(O40)→σ*(C41-H45) are obtained as 5.83, 2.48, 6.22, 6.14 kcal.mol-1, respectively which quantify the extend of intramolecular hydrogen bonding.

The differences in E(2) energies are reasonably due to the fact that the accumulation of electron density in the C-H bond is not only drawn from the n(O) of hydrogen-acceptor but also from the entire molecule. The orbital interaction energy for n2(O23)→σ*(C18-C19) is 16.6 kcal. mol-1 and n1(O23)→σ*(N17-C18) is 29.1 kcal.mol-1 which are higher values than the other delocalizations. This interaction is responsible for a pronounced increase of the O23 (1.97888e) orbital occupancy than the other occupancies, and it is possibly due to hyperconjugation between O23 and the pyrimidine ring. These ICTs around the rings can induce large bioactivity in the molecule. The p-character of the oxygen lone pair orbitals n2(O23) and n1(O23) is 99.89% and 40.34%, respectively, which is a very close to pure π-type lone pair orbital participates in electron donation to the σ*(C-O) orbital for the n1(N17)→σ*(C18-O23) interaction in the title molecule. Also, an intermolecular hyperconjugative interaction occurs in n2(S15)→σ*(C16-N21) which increases ED (0.01966e) that weakens the respective bond leading to stabilization of 23 kcal.mol-1. The n1(N17)→σ*(C16-N21) interaction shows the highest E(2) energy of 58.6 kcal.mol-1.

3.4 Natural population analysis

Natural population analysis provides an effective method to calculate atomic charges and electron distribution within the molecule [34]. The net atomic charges of the title molecule, obtained by means of natural population analysis [35], are plotted in Figure 3. Very similar negative charges are noticed for the two oxygen atoms of the 1,3-benzodioxole group. A little more complicated situation is observed when the charges on the carbon atoms are considered. All hydrogen atoms have net positive charges and H22 (0.2073 e) shows more positive charge than the other hydrogen atoms due to its attachment with a nitrogen atom and with the oxygen atom of the pyrimidine ring N-H⋅ ⋅ ⋅ O via intramolecular hydrogen bonding. All carbon atoms are negatively charged except C5, C16, C18, C20, C36, C37 and C41 due to their attachments with electronegative nitrogen, sulphur or oxygen atoms.

Figure 3 Natural charge distribution chart of the title molecule.
Figure 3

Natural charge distribution chart of the title molecule.

3.5 Vibrational analysis

The vibrational spectral analysis of the title compound has been performed based on the characteristic vibrations of its carbonyl, methylene, methyl and N-H groups. The computed vibrational wavenumbers, their IR and Raman activities as well as the atomic displacements corresponding to the different normal modes are used to identify the vibrational modes unambiguously. The selected vibrational assignments are presented in Table 3. The experimental and simulated FT-IR and FT-Raman spectra of the title compound are given in Figures 4 and 5, respectively. Correlation graphs between theoretical and experimental wavenumbers are given in Figure 6. There is a good agreement between the calculated and experimental values with a correlation coefficient (R2) values = 0.999 for both IR and Raman.

Table 3

The selected vibrational wavenumbers (cm-1), measured infrared and Raman band positions (cm-1), and their tentative assignment.

Scaled Wavenumbers (cm-1)Experimental (cm-1)WavenumberIntensityAssignments with PED%
IRRamanIRRaman
3446-3474 vw52.130.36νss N17-H22(100)
3097-3198 vw2.870.27ϑss C38-H46(92)
3085-3106 vw3.480.28ϑss C35-H43(99), ϑss C52-H51(66)
3071-3066 w4.600.47ϑss C39-H47(92)
3023-3016 w28.260.85ϑss C41-H44(94)
3009--31.380.35ϑas CH2-1 (16), ϑas CH2-2(12), ϑss C48-H49(89)
29782967 w2973 s7.640.38ϑas CH3, ϑas CH2-1 (41), ϑas CH2-2(70),
29332933 w-20.640.36ϑss CH2-8(82)
29282928 s-33.740.24ϑss CH3 (45)
29212916 s2922 vs40.570.27ϑss CH2-3 (64)
2900-2875 m41.930.60ϑss CH2-4 (20), CH2-7(35)
28982863 w2825w61.160.47ϑss CH2-5 (90),
16771650 s1645 vw775.721.31ϑss C=O (75)
1607-1607 vw0.010.58ϑss C35-C36(25), ϑss C36-C37(23),ϑss C34-C39(20)
1592--7.320.32ϑss C35-C36(17), ϑss C37-C38(36), ϑss C34-C35(12)
15631545 m1541 m164.150.62ϑss N 21-06(32), ϑss O9-C20(37)
1516-1511 w405.070.34ϑss C19-C20(23),δsci H22-N17-C16(14)
14871483 s-25.130.91δsci CH2-7(80)
1455--1.100.71δsci CH2-3(11), δsci CH2-4(38),δsci CH2-8(37)
14401439 m1441 m11.940.27δsci CH2-1(53), δtwi CH3 (20), δsci CH2(8) (81)
1418-1403 vw7.3330.76δsci H22-N17-C16(42)
13781379 s1378 w2.760.38τC41-H44-O40-H45(76),δw H22-N17-C16(42)
1361-1361 vw7.0 81.80δr CH3(37)
13531357 s-6.490.67δsci H8-C1-N6(21), τH13-C5-N6-S15(18)
13201318 s-0.250.48δsci H8-C1-N6(27), tC4-H11-C5-H12(21)
13091307 s1310 w5.720.95δw H25-C24-C19(29), T H25-C24-C19-C18(11
11941246 vs-34.097.34νss N21-C20(12), ϑss C19-C24(ll), ϑss 08-09(10)
1175--44.880.19δsci H32-C31-C34(13), τ H33-C31-C34-C35(15)
1157--25.580.10δsci H43-C35-C36(25), δrH44-C41-O42(21)
1155-1148 w14.860.29ϑss N6-0(32), ϑss N6-C5(19)
11291111 vs1135 w86.530.11ϑss N17-C16(14), ϑss N17-C18(2l)
1089-1087 vw70.410.11ϑss 03-C2(17), ϑss C4-C5(17), ϑss Cl-C2(15)
1071--14.491.59τC4-C5-O3-H(11)
10401035 s1036 vw26.260.06ϑss C24-C27(22), δr CH3(10)
10089911002 vw4.780.16ϑss C4-C5(13),-ϑssCl-C2(14)
972--0.900.17δsci C2-C1-N6(15), τ C4-C5-O3-H(10), τ H13-C5-N6-S15(14)
965-954 vw4.501.23ϑss C24-C27(18)
909926 s907 vw19.290.22νss C24-C27(10), δC34-C39-C38(11)
887859 s-6.120.23δsci N17-C16-N21(12), τ C31-C20-C34-H32(12)
773788 s-36.991.61τ H46-C38-C39-C34(22), τ O23-C19-N17-C18(17)
750-736 w5.910.07τ H25-C24-C19-C18(19), τ023-C19-N17-C8(11),τw CH3(56)
723722 m-11.150.04C37-C36-O40(12), τ C35-C36-C37-C38(10)
681680 m685 vw3.990.06νss N6-C1(13), νss S15-N6(20)
575566 m568 vw6.210.70C5-C4-O3(12), τ H22-N17-C18-O23(10), τ C35-C34-C39-C38(21)
552-558 vw10.980.09δr N17-C18-O23(18)
523--5.390.05δsci C16-N21-C20(38)
488466 m-3.590.09δsci C37-C38-C39(22), ϑss S15-C51(13)
420--4.860.06δC36-C37-C38(32), δC31-C34-C35(11)
223-218 w0.810.59δC31-C34-C35(11), τ H30-C27-C24-C19(23)
202-197 w3.590.26δC19-C24-C27(12), τ C20-N21-C16-S15(19)
170-168 w1.671.00τ N6-C1-C2-O3(10)
88-88 vs2.810.66τ C5-N6-S15-C16(25)
71-70 vs1.910.47τ C20-N21-C16-N17(15)
  1. ν-stretching, δ-bending, τ-Torsion, s-strong, vs-very strong, m-medium, w-weak, vw-very weak, ss-symmetric stretching, as-asymmetric stretching, δsci-scissoring, δw-wagging, δrock-rocking, δtwi-twisting, τ-torsion.

Figure 4 Experimental (a) and simulated (b) IR spectra of the title molecule.
Figure 4

Experimental (a) and simulated (b) IR spectra of the title molecule.

Figure 5 Experimental (a) and simulated (b) Raman spectra of the title molecule.
Figure 5

Experimental (a) and simulated (b) Raman spectra of the title molecule.

Figure 6 Correlation graphs between theoretical and experimental wavenumbers of (a) FT-IR (b) FT-Raman of the title compound.
Figure 6

Correlation graphs between theoretical and experimental wavenumbers of (a) FT-IR (b) FT-Raman of the title compound.

3.5.1 Methyl group vibrations

Methyl groups are generally referred to as electron donating substituents in the aromatic ring systems. The methyl hydrogen atoms in the molecule are simultaneously subjected to hyperconjugation and back donation, which cause the decrease of stretching wavenumbers and IR intensities [36]. Symmetric methyl stretching vibrations are normally observed between the wavenumbers 2952 and 2972 cm-1, while the asymmetric stretching usually occurs between 2862 and 2882 cm1, respectively [37]. The methyl asymmetric stretching vibration of the title compound appears at 2973 cm1 as strong band in its FT- Raman spectrum, whereas its symmetric methyl stretching mode is observed at 2928 cm-1 in its FT-IR spectrum. The increase in wavenumber by 91 cm-1 (blue shifting) suggests the presence of intermolecular hydrogen bonding.

The asymmetrical bending vibrations usually occur in the range of 1450-1470 cm-1, while the symmetrical bending vibrations appear in the range of 1365-1385 cm-1 [33, 38]. The asymmetrical methyl vibrations generally overlap with the scissoring vibrations of the methylene groups. The asymmetrical bending vibrations of the title compound were identified at 1439 cm-1 as medium FT-IR band and at 1441 cm-1 as medium FT-Raman band. The absorption band arising from the symmetrical bending of the C-H bonds is very stable in position when the methyl group is attached to another carbon atom.

The rocking mode of the methyl group usually appears in the region of 1070-1010 cm1 [35] and it is observed at 1361 cm-1 as very weak band in the FT-IR spectrum of the title compound. Its methyl wagging vibrations occurred at 736 cm-1 as weak bands in its FT-Raman spectrum.

3.5.2 Methylene group vibrations

The asymmetrical stretching and symmetrical stretching bands of the methylene groups occur near 2926 and 2855 cm-1, respectively [39]. The wavenumber of the methylene stretching is increased when the methylene group is a part of a strained ring. The qualitative interpretation of intensities must rely on the understanding of some basic aspects of intramolecular charge distribution and on their effects on the IR intensities. The title compound has nine methylene groups. The CH2(1) and CH2(2) asymmetric stretching modes manifest their characteristic bands at 2967 and 2973 cm-1 as a weak and strong bands in the FT-IR and FT-Raman spectra of the title compound, respectively. CH2(3) symmetric stretching band occurs at 2916 cm-1 in the FT-IR spectrum of the title compound as strong band and at 2922 cm-1 in its FT-Raman spectrum as a very strong band.

CH2(4) and CH2(7) show a medium symmetric stretching band at 2875 cm-1 in the FT-Raman spectrum with a blue shifting of 20 cm-1 along with increase in the wavenumber of bending vibration by 18 cm-1 indicating the possibility of C41-H45...O42 intramolecular hydrogen bond. This possibility can be confirmed from the optimized geometry and NBO analysis of the title compound. CH2(8) symmetric stretching mode occurs as a weak band at 2933 cm-1 Electronic effects including back-donation can shift the position and alter the intensity of methylene vibrations. The blue-shifting of the methylene stretching wavenumbers, point out to the influence of backdonation in the ring. The spectral analysis reveals that the methylene symmetric stretching modes are shifted to higher wavenumbers, probably because of electronic effects resulting from the influence of the methylene group on its adjacent groups.

The scissoring bands in the FT-IR spectra of hydrocarbons occur nearly at constant positions near 1465 cm-1. The scissoring modes of methylene groups of the title compound are observed as strong band at 1483 cm-1 in its FT-IR spectrum and as medium bands at 1441 and 1439 cm-1 in the FT-Raman and FT-IR spectra, respectively. Moreover, the vibrational bands corresponding to the twisting, wagging, and rocking vibrations of the methylene groups and the in-plane and out-of plane deformation modes have also been observed and supported by DFT computations.

3.5.3 Carbonyl group vibrations

The intensity of carbonyl group bands can increase due to the conjugation or formation of hydrogen bonds. The carbonyl stretching vibrations in aromatic compounds are expected in the region of 1700-1640 cm-1 [40].

The carbon-oxygen double bond is formed by π-π bond between carbon and oxygen and the lone pair of electrons on the oxygen atom affects the nature of carbonyl group. A very weak carbonyl stretching band is formed at 1645 cm-1 in the FT-Raman spectrum and at 1650 cm-1 in the FT-IR spectrum of the title compound. NCO rocking is identified as weak bands in the FT-Raman spectrum of the title molecule at 558 cm-1.

3.5.4 Aromatic CH vibration

Aromatic CH vibrations occur above 3000 cm-1 [40]. A very strong FT-IR band is noted at 3129 cm-1 and a very weak FT- Raman band at 3198 cm-1 corresponding to the aromatic CH stretching. FT-Raman weak bands are also found at 3106, 3066 and 3016 cm-1. The intensity enhancement of C-H stretching wavenumber and decrease in intensity of the C-H bending wavenumber could be attributed to the conjugation with ring π system.

3.5.5 NH vibrations

The N-H stretching vibrations generally occur in the region of 3500-3300 cm-1 [40]. The FT-IR band appeared at 3464 cm-1 has been assigned to N-H stretching vibration. The red shifting is further enhanced by the reduction in the NH bond order values due to donor-acceptor interaction. The N-H in-plane bending mode is observed as a medium band in the FT-IR spectrum at 1497 cm-1 and as a weak band at 1511 cm-1 in FT-Raman spectrum.

3.5.6 C-S vibrations

The FT-IR stretching mode of the C-S bond usually occurs at 696 and 671 cm-1 and Kaur et al. [41] reported that the C-S stretching mode appeared at 672 cm-1. In the title compound, the C-S stretching band is observed at 466 cm-1 in FT-IR spectrum as a medium band.

3.5.7 C-N and C-C vibrations

The C-N stretching vibrations generally occur in the region of 1350-1250 cm-1. Symmetric stretching of C16-N17 and C18-N17 shows a weak band in the FT-Raman at 1135 cm-1. Similarly, C1-N6 and C5-N6 shows a weak band in the FT-Raman at 1148 cm-1.

3.6 HOMO-LUMO energy gap

HOMO and LUMO are called the frontier orbitals since they determine the way the molecule interacts with other species. HOMO is the orbital that could act as an electron donor, since it is the outermost (highest energy) orbital containing electrons, while the LUMO is the orbital that could act as an electron acceptor, since it is the innermost (lowest energy) orbital that has room to accept electrons. A single orbital may be both the HOMO and the LUMO [42]. The lowest singlet transition is the transition from the HOMO orbital to the LUMO orbital [33] and the geometrical relaxation can be understood by analyzing the nodal patterns of the HOMO (-4.15 eV) and LUMO (-1.55 eV) orbitals with HOMO-LUMO energy gap of 0.2.6 eV for the title compound (Figures 7 and 8). HOMO-LUMO gap is small because the guest atom orbital(s) are only partially occupied and leading to a large stabilization of the LUMO due to the strong electron-accepting ability of the electron-acceptor group. HOMO localizes on the 1,3- benzodioxole moiety and the LUMOs are localized on the pyrimidine ring. Consequently, an ED transfer occurs from the aromatic part of the p-conjugated path in the electron-donor side to its electron-withdrawing part. Highly delocalized HOMO indicates that the electrons can more readily move around the molecule and hence an improved intramolecular charge transfer (ICT) [43].

Figure 7 HOMO plot of the title compound.
Figure 7

HOMO plot of the title compound.

Figure 8 LUMO plot of the title compound.
Figure 8

LUMO plot of the title compound.

3.7 Molecular docking

Molecular docking process involves the prediction of ligand orientation into its targeted binding site [44]. The current study was performed using AutoDock 4.2 program [45]. The title molecule was energy minimized based on the DFT method. Three antimicrobial target proteins with PDB ID: 3QGT (antimalarial) [46], 3ACX and 1VQQ (antibacterial) [47, 48] were selected for the present docking analysis. The three-dimensional structural coordinate files of the target proteins were downloaded from the research collaboratory for structural bioinformatics (RCSB) protein data bank. Grid box size was built 90 x 90 x 90 points as set to be a catalytic site for all target proteins. The binding free energy for interactions of the ligand with proteins 3QGT, 3ACX and 1VQQ are -4.69, -3.94 and -3.29 kcal mol-1, respectively. The best result was obtained with the 3QGT protein for antimalarial activity of the title compound.

Fifty conformations of the title molecule were obtained and the top-ranked complex was identified which proved the best fitting. Molecular docking results with different target proteins are shown in Table 4. Pictorial representation of the best possible binding pose of the title compound with 3QGT protein is shown in Figure 9. Docking analysis showed that the carbonyl group in the pyrimidine ring and the oxygen atom of the morpholine group in title compound interact with the ARG77 and ASN66 residues of the target protein, respectively. The title compound forms two hydrogen bonds with ARG77 and ASN66 with distances of 2.5 and 1.8 Å, respectively. The docking results revealed the ability of the title compound for binding to the antibacterial target proteins (3ACX and 1VQQ) and hence it exhibited antimicrobial potential which is consistent with its experimental antimicrobial activity [20]. In addition, prediction of the binding mode of the title compound to the antimalarial target protein (3QGT) was also explored and hence its possible antimalarial potential.

Table 4

Molecular docking results with different target proteins.

Target protein IDDocking energy (kcal.mol-1)Atoms of ligands involved in dockingAmino acid residues involved in docking interactionBond distance of hydrogen bonds (Å)No. of hydrogen bonds
3QGT-4.69C4-O3ASN661.82
C18-O23ARG772.5
3ACX-3.94N17-H22VAL1941.72
C18-O23LEU1892.7
1VQQ-3.29N17-H22SER2502.02
C4-O3GLU2462.8

Figure 9 Binding pose diagram of the title compound to its target protein.
Figure 9

Binding pose diagram of the title compound to its target protein.

4 Conclusions

A comprehensive FT-IR and FT-Raman spectroscopic investigations as well as DFT computations were carried out on the antimicrobial agent 6-(1,3-benzodioxol-5- ylmethyl)-5-ethyl-2-{[2-(morpholin-4-yl)ethyl]sulfanyl} pyrimidin-4-(3H)-one. The geometry optimization of the title molecule exposed its non-planarity. Blue shifting of the C-H wavenumber along with a decrease in bond length of C-H attribute the formation of C-H…O hydrogen bonding. The possibilities of hydrogen bonding were explained with the aid of natural charge analysis. NBO analysis showed a pronounced increase in the lone pair orbital occupancy and hyperconjugation interactions leading to molecular stabilization. HOMO-LUMO energy gap suggested the possibility of intramolecular charge transfer within the title molecule. Molecular docking results predicted the antimalarial potential of the title compound due to its ability to interact with an antimalarial target protein (3QGT). The results of the current study could support the development of new potent thiouracil-bearing candidates in the antimicrobial research area.

Acknowledgment

The authors would like to extend their sincere appreciation to the Deanship of Scientific Research at King Saud University for its funding of this research through the Research Group Project No. RGP-196.

  1. Conflict of interest: Authors state no conflict of interest.

References

[1] Pałasz A., Cież D., In search of uracil derivatives as bioactive agents. Uracils and fused uracils: Synthesis, biological activity and applications. Eur. J. Med. Chem., 2015, 97, 582-611.10.1016/j.ejmech.2014.10.008Search in Google Scholar PubMed

[2] Al-Deeb O.A., Al-Turkistani A.A., Al-Abdullah E.S., El-Brollosy N.R., Habib E.E., El-Emam A.A., Pyrimidine-5-carbonitriles–part III: synthesis and antimicrobial activity of novel 6-(2-substituted propyl)-2, 4-disubstituted pyrimidine-5-carbonitriles. Heterocycl. Commun., 2013, 19, 411-419.10.1515/hc-2013-0139Search in Google Scholar

[3] Conejo-García A., García-Rubiño M.E., Marchal J.A., Núñez M.C., Ramírez A., Cimino S., García M.Á., Aránega A., Gallo M.A., Campos J.M., Synthesis and anticancer activity of (RS)-9-(2,3- dihydro-1,4-benzoxaheteroin-2-ylmethyl)-9H-purines. Eur. J. Med. Chem., 2011, 46, 3795-3801.10.1016/j.ejmech.2011.05.046Search in Google Scholar PubMed

[4] López-Cara L.C., Conejo-García A., Marchal J.A., Macchione G., Cruz-López O., Boulaiz H., García M.A., Rodríguez-Serrano F., Ramírez A., Cativiela C., New (RS)-benzoxazepin-purines with antitumour activity: the chiral switch from (RS)-2, 6-dichloro-9-[1-(p-nitrobenzenesulfonyl)-1,2,3,5-tetrahydro-4,1- benzoxazepin-3-yl]-9H-purine. Eur. J. Med. Chem., 2011, 46, 249-258.10.1016/j.ejmech.2010.11.011Search in Google Scholar PubMed

[5] Morales F., Ramírez A., Conejo-Garcia A., Morata C., Marchal J.A., Campos J. M., Anti-proliferative activity of 2, 6-dichloro- 9-or 7-(ethoxycarbonylmethyl)-9H-or 7H-purines against several human solid tumour cell lines. Eur. J. Med. Chem., 2014, 76, 118-124.10.1016/j.ejmech.2014.02.012Search in Google Scholar PubMed

[6] Parveen H., Hayat F., Salahuddin A., Azam A., Synthesis, characterization and biological evaluation of novel 6-ferrocenyl-4-aryl-2-substituted pyrimidine derivatives. Eur. J. Med. Chem., 2010, 45, 3497-3503.10.1016/j.ejmech.2010.04.023Search in Google Scholar PubMed

[7] Pisal M.M., Nawale L.U., Patil M.D. Bhansali S.G., Gajbhiye J.M., Sarkar D., Chavan S.P., Borate H.B., Hybrids of thienopyrimidinones and thiouracils as anti-tubercular agents: SAR and docking studies. Eur. J. Med. Chem., 2017, 127, 459-469.10.1016/j.ejmech.2017.01.009Search in Google Scholar PubMed

[8] Taher A.T., Abou-Seri S.M., Synthesis and bioactivity evaluation of new 6-aryl-5-cyano thiouracils as potential antimicrobial and anticancer agents. Molecules 2012, 17, 9868-9886.10.3390/molecules17089868Search in Google Scholar PubMed PubMed Central

[9] Mai A., Rotili D., Massa S., Brosch G., Simonetti G., Passariello C., Palamara A.T., Discovery of uracil-based histone deacetylase inhibitors able to reduce acquired antifungal resistance and trailing growth in Candida albicans. Bioorg. Med. Chem. Lett. 2007, 17, 1221-1225.10.1016/j.bmcl.2006.12.028Search in Google Scholar PubMed

[10] Keche A.P., Hatnapure G.D., Tale R.H., Rodge A.H., Birajdar S.S., Kamble V.M., A novel pyrimidine derivatives with aryl urea, thiourea and sulfonamide moieties: synthesis, antiinflammatory and antimicrobial evaluation. Bioorg. Med. Chem. Lett. 2012, 22, 3445-3448.10.1016/j.bmcl.2012.03.092Search in Google Scholar PubMed

[11] Parker W.B., Enzymology of purine and pyrimidine antimetabolites used in the treatment of cancer. Chem. Rev., 2009, 109, 2880-2893.10.1021/cr900028pSearch in Google Scholar PubMed PubMed Central

[12] Saladino R., Crestini C., Palamara A.T., Danti M.C., Manetti F., Corelli F., Garaci E., Botta M., Synthesis, biological evaluation, and pharmacophore generation of uracil, 4-(3H)-pyrimidinone, and uridine derivatives as potent and selective inhibitors of parainfluenza 1 (Sendai) virus. J. Med. Chem., 2001, 44, 4554-4562.10.1021/jm010938iSearch in Google Scholar PubMed

[13] Zhi C., Long Z.-Y., Manikowski A., Brown N.C., Tarantino P.M., Holm K., Dix E.J., Wright G.E., Foster K.A., Butler M.M., Synthesis and antibacterial activity of 3-substituted-6-(3-ethyl- 4-methylanilino) uracils. J. Med. Chem., 2005, 48, 7063-7074.10.1021/jm050517rSearch in Google Scholar PubMed

[14] Leite A.C.L., da Silva K.P., de Souza I.A., de Araújo J.M., Brondani D.J., Synthesis, antitumour and antimicrobial activities of new peptidyl derivatives containing the 1, 3-benzodioxole system. Eur. J. Med. Chem., 2004, 39, 1059-1065.10.1016/j.ejmech.2004.09.007Search in Google Scholar PubMed

[15] Joshi R., Kumar M.S., Satyamoorthy K., Unnikrisnan M., Mukherjee T., Free radical reactions and antioxidant activities of sesamol: pulse radiolytic and biochemical studies. J. Agric. Food Chem., 2005, 53, 2696-2703.10.1021/jf0489769Search in Google Scholar PubMed

[16] Cotinguiba F., Regasini L.O., da Silva Bolzani V., Debonsi H.M., Passerini G.D., Cicarelli R.M.B., Kato M.J., Furlan M., Piperamides and their derivatives as potential antitrypanosomal agents. Med. Chem. Res., 2009, 18, 703.10.1007/s00044-008-9161-9Search in Google Scholar

[17] Bezerra D.P., Castro F.O. D., Alves A.P.N., Pessoa C., Moraes M.O.D., Silveira E.R., Lima M.A.S., Elmiro F.J.M., Alencar N., Mesquita R.O., In vitro and in vivo antitumor effect of 5-FU combined with piplartine and piperine. J. Appl. Toxicol., 2008, 28, 156-163.10.1002/jat.1261Search in Google Scholar PubMed

[18] Viegas-Junior C., Danuello A., da Silva Bolzani V., Barreiro E.J., Fraga C.A.M., Molecular hybridization: a useful tool in the design of new drug prototypes. Curr. Med. Chem., 2007, 14, 1829-1852.10.2174/092986707781058805Search in Google Scholar PubMed

[19] Tietze L.F., Bell H.P., Chandrasekhar S., Natural product hybrids as new leads for drug discovery. Angew. Chem. Int. Ed. 2003, 42, 3996-4028.10.1002/anie.200200553Search in Google Scholar PubMed

[20] Attia M.I., El-Brollosy N.R., Kansoh A.L., Ghabbour H.A., Al-Wabli R.I. Fun H.-K., Synthesis, Single crystal x-ray structure, and antimicrobial activity of 6-(1, 3-benzodioxol-5-ylmethyl)-5- ethyl-2-{[2-(morpholin-4-yl)ethyl]sulfanyl}pyrimidin-4 (3H)-one. J. Chem., 2014, 2014, Article ID 457430.10.1155/2014/457430Search in Google Scholar

[21] Frisch MJ, Trucks GW, Schlegel HB, Scuseria GE, Robb MA, Cheeseman JR, Scalmani G, Barone V, Mennucci B, Petersson GA, Nakatsuji H, Caricato M, Li X, Hratchian HP, Izmaylov AF, Bloino J, Zheng G, Sonnenberg JL, Hada M, Ehara M, Toyota K, Fukuda R, Hasegawa J, Ishida M, Nakajima T, Honda Y, Kitao O, Nakai H, Vreven T, Montgomery Jr JA, Peralta JE, Ogliaro F, Bearpark M, Heyd JJ, Brothers E, Kudin KN, Staroverov VN, Kobayashi R, Normand J, Raghavachari K, Rendell A, Burant JC, Iyengar SS, Tomasi J, Cossi M, Rega N, Millam JM, Klene M, Knox JE, Cross JB, Bakken V, Adamo C, Jaramillo J, Gomperts R, Stratmann RE, Yazyev O, Austin AJ, Cammi R, Pomelli C, Ochterski JW, Martin RL, Morokuma K, Zakrzewski VG, Voth GA, Salvador P, Dannenberg JJ, Dapprich S, Daniels AD, Farkas O, Foresman JB, Ortiz JV, Cioslowski J, Fox DJ, Gaussian-09, Revision A.02, Gaussian, Inc., Wallingford CT, 2009.Search in Google Scholar

[22] Lee C., Yang W., Parr R.G., Development of the Colle-Salvetti correlation-energy formula into a functional of the electron density. Phys. Rev. B, 1988, 37, 785.10.1103/PhysRevB.37.785Search in Google Scholar

[23] Becke A.D., Density-functional thermochemistry. IV. A new dynamical correlation functional and implications for exact-exchange mixing. J. Chem. Phys., 1996, 104, 1040-1046.10.1063/1.470829Search in Google Scholar

[24] Almutairi M.S., Muthu S., Prasana J.C., Chandralekha B., Al-Ghamdi A.R., Attia M. I. Comprehensive spectroscopic (FT-IR, FT-Raman, 1H and 13C NMR) identification and computational studies on 1-acetyl-1H-indole-2,3-dione. Open Chem., 2017, 15, 225-237.10.1515/chem-2017-0026Search in Google Scholar

[25] Al-Wabli R.I., Govindarajan M., Almutairi M.S., Attia M.I. A combined experimental and theoretical study on vibrational and electronic properties of (5-methoxy-1H-indol-1-yl) (5-methoxy-1H-indol-2-yl)methanone. Open Chem., 2017, 15, 238-246.10.1515/chem-2017-0027Search in Google Scholar

[26] Scott A.P., Radom L., Harmonic vibrational frequencies: an evaluation of Hartree–Fock, Møller–Plesset, quadratic configuration interaction, density functional theory, and semiempirical scale factors. J. Phys. Chem. 1996, 100, 16502-16513.10.1021/jp960976rSearch in Google Scholar

[27] Jamróz M.H., Vibrational energy distribution analysis (VEDA): scopes and limitations. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2013, 114, 220-230.10.1016/j.saa.2013.05.096Search in Google Scholar PubMed

[28] Keresztury G., Raman Spectroscopy: Theory in: JM Chalmers, PR Griffiths (Ed.), Hand book of vibrational spectroscopy, vol. 1. Wiley: 2002.Search in Google Scholar

[29] Attia M.I., Kansoh A.L., El-Brollosy N.R., Antimicrobial pyrimidinones II: synthesis and antimicrobial evaluation of certain novel 5, 6-disubstituted 2-(substituted amino) alkylthiopyrimidin-4(3H)-ones. Monatsh. Chem.-Chem. Mon. 2014, 145, 1825-1837.10.1007/s00706-014-1253-2Search in Google Scholar

[30] Amalanathan M., Rastogi V., Joe I.H., Palafox M., Tomar R., Density functional theory calculations and vibrational spectral analysis of 3, 5-(dinitrobenzoic acid). Spectrochim. Acta A Mol. Biomol. Spectrosc., 2011, 78, 1437-1444.10.1016/j.saa.2011.01.023Search in Google Scholar PubMed

[31] Edwin B., Amalanathan M., Joe I.H., Vibrational spectra and natural bond orbital analysis of organic crystal L-prolinium picrate. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2012, 96, 10-17.10.1016/j.saa.2012.04.062Search in Google Scholar PubMed

[32] Reed A.E., Curtiss L.A., Weinhold F., Intermolecular interactions from a natural bond orbital, donor-acceptor viewpoint. Chem. Rev., 1988, 88, 899-926.10.1021/cr00088a005Search in Google Scholar

[33] Xavier T., Rashid N., Joe I.H., Vibrational spectra and DFT study of anticancer active molecule 2-(4-bromophenyl)-1H- benzimidazole by normal coordinate analysis. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2011, 78, 319-326.10.1016/j.saa.2010.10.013Search in Google Scholar

[34] Benzon K., Varghese H.T., Panicker C.Y., Pradhan K., Tiwary B.K., Nanda A.K. Van Alsenoy C., Spectroscopic and theoretical characterization of 2-(4-methoxyphenyl)-4, 5-dimethyl- 1H-imidazole 3-oxide. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2015, 151, 965-979.10.1016/j.saa.2015.07.020Search in Google Scholar

[35] Mulliken R.S., Electronic population analysis on LCAO–MO molecular wave functions. I. J. Chem. Phys., 1955, 23, 1833-1840.10.1063/1.1740588Search in Google Scholar

[36] Gussoni M., Castiglioni C., Ramos M.N., Rui M., Zerbi G., Infrared intensities: from intensity parameters to an overall understanding of the spectrum. J. Mol. Struct., 1990, 224, 445-470.10.1016/0022-2860(90)87033-TSearch in Google Scholar

[37] King W., Mast G., Blanchette P., Infrared intensity sum rule and effective charges. J. Chem. Phys., 1972, 56, 4440-4446.10.1063/1.1677887Search in Google Scholar

[38] Benzon K., Varghese H.T., Panicker C.Y., Pradhan K., Tiwary B.K., Nanda A.K., Van Alsenoy C., Spectroscopic investigation (FT-IR and FT-Raman), vibrational assignments, HOMO–LUMO, NBO, MEP analysis and molecular docking study of 2-(4-hydroxyphenyl)-4, 5-dimethyl-1H-imidazole 3-oxide. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2015, 146, 307-322.10.1016/j.saa.2015.03.063Search in Google Scholar PubMed

[39] Lin-Vien D., Colthup, N.B., Fateley W.G., Grasselli J.G., The handbook of infrared and Raman characteristic frequencies of organic molecules, Elsevier: 1991.Search in Google Scholar

[40] Smith B.C., Infrared spectral interpretation: a systematic approach, CRC press, 1998.Search in Google Scholar

[41] Kaur M., Mary Y.S., Panicker C.Y., Varghese H.T., Yathirajan H., Byrappa K., Van Alsenoy C., Vibrational spectroscopic (FT-IR, FT-Raman) and quantum chemical calculations of 1-(5, 5-dioxido-10H-phenothiazin-10-yl) ethanone. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2014, 120, 445-455.10.1016/j.saa.2013.10.032Search in Google Scholar PubMed

[42] Joe I.H., FT-IR, Raman and DFT study of 2-amino-5-fluorobenzoic acid and its biological activity with other halogen (Cl, Br) substitution. Spectrochim. Acta A Mol. Biomol. Spectrosc., 2011, 79, (2), 332-337.10.1016/j.saa.2011.02.037Search in Google Scholar PubMed

[43] Atalay Y., Avci D., Başoğlu A., Linear and non-linear optical properties of some donor–acceptor oxadiazoles by ab initio Hartree-Fock calculations. Struct. Chem. 2008, 19, 239-246.10.1007/s11224-007-9278-3Search in Google Scholar

[44] Kitchen D.B., Decornez H., Furr J.R., Bajorath J., Docking and scoring in virtual screening for drug discovery: methods and applications. Nat. Rev. Drug Discov., 2004, 3, 935-949.10.1038/nrd1549Search in Google Scholar PubMed

[45] Morris G.M., Huey R., Lindstrom W., Sanner M.F., Belew R.K., Goodsell D.S., Olson A.J., AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem., 2009, 30, 2785-2791.10.1002/jcc.21256Search in Google Scholar PubMed PubMed Central

[46] Kamchonwongpaisan S., Quarrell R., Charoensetakul N., Ponsinet R., Vilaivan T., Vanichtanankul J., Tarnchompoo B., Sirawaraporn W., Lowe G., Yuthavong Y., Inhibitors of multiple mutants of Plasmodium f alciparum dihydrofolate reductase and their antimalarial activities. J. Med. Chem. 2004, 47, 673-680.10.1021/jm030165tSearch in Google Scholar PubMed

[47] Kotaiah Y., Nagaraju K., Harikrishna N., Rao C.V., Yamini L., Vijjulatha M., Synthesis, docking and evaluation of antioxidant and antimicrobial activities of novel 1,2,4-triazolo[3,4-b][1,3,4] thiadiazol-6-yl)selenopheno[2,3-d]pyrimidines. Eur. J. Med. Chem., 2014, 75, 195-202.10.1016/j.ejmech.2014.01.006Search in Google Scholar PubMed

[48] Acebrón I., Chang M., Mobashery S.; A Hermoso J., The allosteric site for the nascent cell wall in penicillin-binding protein 2a: an Achilles’ heel of methicillin-resistant Staphylococcus aureus. Curr. Med. Chem. 2015, 22, 1678-1686.10.2174/0929867322666150311150215Search in Google Scholar PubMed PubMed Central

Received: 2018-01-14
Accepted: 2018-04-13
Published Online: 2018-07-18

© 2018 Maha S. Almutairi et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 License.

Downloaded on 11.6.2024 from https://www.degruyter.com/document/doi/10.1515/chem-2018-0067/html
Scroll to top button